12.07.2015 Views

Reproduction in Domestic Animals

Reproduction in Domestic Animals

Reproduction in Domestic Animals

SHOW MORE
SHOW LESS
  • No tags were found...

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

<strong>Reproduction</strong> <strong>in</strong> <strong>Domestic</strong> <strong>Animals</strong>Official Organ of European Society for <strong>Domestic</strong> Animal <strong>Reproduction</strong>, European Veter<strong>in</strong>arySociety of Small Animal <strong>Reproduction</strong> and Spanish Society of Animal <strong>Reproduction</strong>The Organis<strong>in</strong>g Committee wishes to acknowledge and expressits gratitude to the ma<strong>in</strong> sponsors of ICAR 2008 Congress:Editor-<strong>in</strong>-ChiefProf. Dr. H. Rodriguez-Márt<strong>in</strong>ezDivision of <strong>Reproduction</strong>Department of Cl<strong>in</strong>ical SciencesFaculty of Veter<strong>in</strong>ary Medic<strong>in</strong>e and Animal ScienceSwedish University of Agricultural Sciences (SLU)Ullsvägen 14C, Cl<strong>in</strong>ical Centre. P.O. Box 7054,Ultuna SE-750 07 Uppsala,SwedenTelephone: +46-(0) 18672172Fax: +46-(0) 18673545E-mail: heriberto.rodriguez@kv.slu.seAssociate EditorsProf. Dr. W.A. K<strong>in</strong>gUniversity of GuelphDept. of Biomedical SciencesGuelph, Ont. N1G 2W1, CanadaProf. Dr. E. Mart<strong>in</strong>ez-GarciaVeter<strong>in</strong>ary Teach<strong>in</strong>g HospitalDept. of Vet. Pathology, University of MurciaCampus de Esp<strong>in</strong>ardo, 30500 Murcia, Spa<strong>in</strong>Prof. Dr. M. McGowanSchool of Veter<strong>in</strong>ary ScienceUniversity of QueenslandSt. Lucia QLD 4072l, AustraliaProf Dr. José Luiz Rigo RodriguesUniversidade Federal do Rio Grande do SulFaculdade de Veter<strong>in</strong>áriaCaixa Postal 1500491501-970 Porto Alegre RS, BrazilProf. Dr. E. SatoLaboratory of Animal ProductionGraduate School of Agricultural ScienceTohoku UniversityAoba-Ku, Sendai 981-8555, JapanPr<strong>in</strong>ciple sponsorEditorial Advisory BoardW. R. Allen, Newmarket – J. Aurich, Vienna – F. W. Bazer, Texas – H. Bertsch<strong>in</strong>ger, Pretoria – P. Bols, Antwerp – G. Brem, Vienna – B. Brück, Frederiksberg –F. Camillo, Pisa – D. Cavestany, Montevideo – M. A. Crowe, Dubl<strong>in</strong> – H. Dobson, Liverpool – G. Evans, Sydney – A. Fontbonne, Maisons–Alfort – G. Foxcroft, Alberta –R. Geisert, Oklahoma – T. Greve, Frederiksberg – B. Hoffman, Gießen – T. Katila, Hels<strong>in</strong>ki – B. Kemp, Wagen<strong>in</strong>gen – G. Kilian, Pennsylvania – A. Kunavongkrit,Bangkok – C. L<strong>in</strong>de–Forsberg, Uppsala – C. Maxwell, Sydney – K. Niwa, Okayama – J. Nöthl<strong>in</strong>g, Pretoria – K. Okuda, Okayama – A. R. Peters, Potters Bar –D. Rath, Neustadt – L. Renato de França, Belo Horizonte – E. Seidel, Ft. Coll<strong>in</strong>s – L. Solti, Budapest – T. A. E. Stout, Utrecht – S. S. Suarez, Ithaca – C. Taman<strong>in</strong>i,Bologna – J. Vasquez, Murcia – E. Watson, Ed<strong>in</strong>burgh – G. v.d. Weijden, Utrecht – A. Ziecik, OlsztynThe Journal is <strong>in</strong>dexed by: AgBiotech News and Information; Agricola C R I S; Animal Breed<strong>in</strong>g Abstracts; BIOBASE; Biological Abstracts; BIOSIS Previews;Chemical Abstracts; Current Contents; Dairy Science Abstracts; Derwent Biotechnology Abstracts; Food Science and Technology Abstracts; ForestProducts Abstracts; Forestry Abstracts; Index Veter<strong>in</strong>arius; MEDLINE; Nutrition Abstracts and Reviews; Personal Alert; Pig News & Information; PoultryAbstracts; Protozoological Abstracts; Referativnyi Zhurnal; Review of Medical and Veter<strong>in</strong>ary Mycology; Science Citation Index; Soybean Abstracts (Onl<strong>in</strong>eEdition); Veter<strong>in</strong>ary Bullet<strong>in</strong>; Wildlife Review Abstracts; World Agricultural Economics and Rural Sociology Abstracts.<strong>Reproduction</strong> <strong>in</strong> <strong>Domestic</strong> <strong>Animals</strong> is the official organ for the European Society for <strong>Domestic</strong> Animal <strong>Reproduction</strong>, the European Veter<strong>in</strong>ary Society ofSmall Animal <strong>Reproduction</strong> and the Spanish Society of Animal <strong>Reproduction</strong>.<strong>Reproduction</strong> <strong>in</strong> <strong>Domestic</strong> <strong>Animals</strong> publishes comprehensive <strong>in</strong>formation concern<strong>in</strong>g physiology, pathology and biotechnology of reproduction, pay<strong>in</strong>g specialattention to applied and cl<strong>in</strong>ical research. The evaluation of papers is made by a team of specialists who decide about acceptance, changes or rejection. For author’s<strong>in</strong>structions please visit the journals homepage www.blackwellpublish<strong>in</strong>g.com/rdaResponsible accord<strong>in</strong>g to Berl<strong>in</strong> press law: Prof. Dr. H. Rodriguez-Márt<strong>in</strong>ez.Publication Rights Contributions published <strong>in</strong> this journal are protected by copyright. All rights, whether of the whole or part of the published material are reserved,specifically those of translation, repr<strong>in</strong>t<strong>in</strong>g, re-use of illustrations, reproduction on microfilms or by other means. Duplication of this journal or parts thereof is forbidden.Copy<strong>in</strong>g one article or parts thereof is only permitted under the provisions of the German Copyright Law of September 9,1965, <strong>in</strong> its version of June 24, 1985, and acopyright fee must always be paid. Violations fall under the prosecution act of the German Copyright Law. Legally permitted copies are to be marked with the sourceof the orig<strong>in</strong>al as well as with the name of the copier.Disclaimer The Publisher, Societies and Editors cannot be held responsible for errors or any consequences aris<strong>in</strong>g from the use of <strong>in</strong>formation conta<strong>in</strong>ed <strong>in</strong> this journal;the views and op<strong>in</strong>ions expressed do not necessarily reflect those of the Publisher, Societies and Editors, neither does the publication of advertisements constituteany endorsement by the Publisher, Societies and Editors of the products advertised.Author material archive policy Please note that unless specifically requested, Blackwell Publish<strong>in</strong>g will dispose of all hardcopy or electronic material submitted twomonths after publication. If you require the return of any material submitted, please <strong>in</strong>form the editorial office or production editor as soon as possible if you have notyet done so.Copyright and Photocopy<strong>in</strong>g Journal compilation © 2008 Blackwell Verlag. All rights reserved. No part of this publication may be reproduced, stored or transmitted <strong>in</strong>any form or by any means without the prior permission <strong>in</strong> writ<strong>in</strong>g from the copyright holder. Authorization to photocopy items for <strong>in</strong>ternal and personal use is granted bythe copyright holder for libraries and other users registered with their local <strong>Reproduction</strong> Rights Organisation (RRO), e.g. Copyright Clearance Center (CCC), 222Rosewood Drive, Danvers, MA 01923, USA (www.copyright.com), provided the appropriate fee is paid directly to the RRO. This consent does not extend to other k<strong>in</strong>dsof copy<strong>in</strong>g such as copy<strong>in</strong>g for general distribution, for advertis<strong>in</strong>g or promotional purposes, for creat<strong>in</strong>g new collective works or for resale. Special requests should beaddressed to: journalsrights@oxon.blackwellpublish<strong>in</strong>g.comSubscription <strong>in</strong>formation <strong>Reproduction</strong> <strong>in</strong> <strong>Domestic</strong> <strong>Animals</strong> is published six times a year. Subscription prices for 2008 are: Premium <strong>in</strong>stitutional: £594 (Europe),£653 (Rest of World), $1098 (The Americas); Personal: £177 (Europe, non Euro Zone), €266 (Europe, Euro Zone), £195 (Rest of World), $328 (The Americas).Customers <strong>in</strong> the UK should add VAT at 7%; customers <strong>in</strong> the EU should also add VAT at 7%, or provide a VAT registration number or evidence of entitlement toexemption. Customers <strong>in</strong> Canada should add 6% GST or provide evidence of entitlement to exemption. The Premium <strong>in</strong>stitutional price <strong>in</strong>cludes onl<strong>in</strong>e access to thecurrent and all available previous year electronic issues. For other pric<strong>in</strong>g options <strong>in</strong>clud<strong>in</strong>g access <strong>in</strong>formation and terms and conditions, please visit www.blackwellpublish<strong>in</strong>g.com/rdaBack issues S<strong>in</strong>gle issues from current and recent volumes are available at the current s<strong>in</strong>gle issue price from Blackwell Publish<strong>in</strong>g Journals. Earlier issues may beobta<strong>in</strong>ed from Periodicals Service Company, 11 Ma<strong>in</strong> Street, Germantown, NY 12526, USA. Tel: +1 518 537 4700, Fax: +1 518 537 5899, Email: psc@periodicals.com.Publisher<strong>Reproduction</strong> <strong>in</strong> <strong>Domestic</strong> <strong>Animals</strong> is published by Blackwell Verlag GmbH, Kurfürstendamm 58, 10707 Berl<strong>in</strong>, Germany. Tel: (+49 30) 327 906-65, Fax:(+49 30) 327 906-77. Email: verlag@blackwell.de. Internet homepage: www.blackwell.deBlackwell Verlag is now part of Wiley-BlackwellDelivery Terms and Legal TitlePrices <strong>in</strong>clude delivery of pr<strong>in</strong>t journals to the recipient's address. Delivery terms are Delivered Duty Unpaid (DDU); the recipient is responsible for pay<strong>in</strong>gany import duty or taxes. Legal title passes to the customer on despatch by our distributors.Journal Customer Services:For order<strong>in</strong>g <strong>in</strong>formation, claims and any enquiry concern<strong>in</strong>g your journal subscription please contact your nearest office:UK: Email: customerservices@blackwellpublish<strong>in</strong>g.com; Tel: +44 (0) 1865 778315; Fax: +44 (0) 1865 471775USA: Email: customerservices@blackwellpublish<strong>in</strong>g.com; Tel: +1 781 388 8599 or 1 800 835 6770 (Toll free <strong>in</strong> the USA & Canada); Fax: +1 781 388 8232or Fax: +44 (0) 1865 471775Asia: Email: customerservices@blackwellpublish<strong>in</strong>g.com; Tel: +65 6511 8000; Fax: +44 (0) 1865 471775Production editor Jovencio Laoag Jr. Tel: (+65) 6511 8188, Fax: (+65) 65118288, Email: RDA@oxon.blackwellpublish<strong>in</strong>g.comJournal publication manager Suzanne Albrecht. Tel: (+49 30) 327 906-65, Fax: (+49 30) 327 906-77. Email:suzanne.albrecht@ber.blackwellpublish<strong>in</strong>g.comAccess to this journal is available free onl<strong>in</strong>e with<strong>in</strong> <strong>in</strong>stitutions <strong>in</strong> the develop<strong>in</strong>g world through the AGORA <strong>in</strong>itiative with the FAO and the HINARI <strong>in</strong>itiativewith the WHO. For <strong>in</strong>formation, visit www.ag<strong>in</strong>ternetwork.org or www.health<strong>in</strong>ternetwork.org.REPRODUCTION IN DOMESTIC ANIMALS (ISSN 0936-6768) is published six times a year. US mail<strong>in</strong>g agent: Mercury Airfreight International Inc., 365 Blair Road, Avenel,NJ 07001, USA. Periodical postage paid at Rahway, NJ. Postmaster: Send all address changes to REPRODUCTION IN DOMESTIC ANIMALS, Blackwell Publish<strong>in</strong>g Inc.,Journals Subscription Department, 350 Ma<strong>in</strong> St., Malden, MA 02148-5020.Information for Authors Please send your manuscript to the Editor-<strong>in</strong>-Chief Prof. Dr. H. Rodriguez-Márt<strong>in</strong>ez and f<strong>in</strong>d <strong>in</strong>structions for authors on the journal homepagewww.blackwellpublish<strong>in</strong>g.com/rdaOur policy is to use permanent paper from mills that operate a susta<strong>in</strong>able forestry policy, and which has been manufactured from pulp that is processed us<strong>in</strong>g acidfreeand elementary chlor<strong>in</strong>e-free practices. Furthermore, we ensure that the text paper and cover board used <strong>in</strong> all our journals has met acceptable environmentalaccreditation standards.Note to NIH Grantees Pursuant to NIH mandate, Wiley-Blackwell will post the accepted version of contributions authored by NIH grant-holders to PubMed Centralupon acceptance. This accepted version will be made publicly available 12 months after publication. For further <strong>in</strong>formation, see www.wiley.com/go/nihmandate.ISSN 0936–6768 (Pr<strong>in</strong>t) ISSN 1439-0531 (Onl<strong>in</strong>e)Pr<strong>in</strong>ted <strong>in</strong> S<strong>in</strong>gapore by Markono Pr<strong>in</strong>t Media Pte Ltd.For submission <strong>in</strong>structions, subscription and all other <strong>in</strong>formation visit: www.blackwellpublish<strong>in</strong>g.com/rdaThis journal is available onl<strong>in</strong>e at Blackwell Synergy. Visit www.blackwell-synergy.com to search the articles and register for table of contents e-mail alerts.M<strong>in</strong>itüb Abfüll- und Labortechnik GmbH & Co. KGGold sponsorsIMV TechnologiesIntervet/Scher<strong>in</strong>g-PloughSilver sponsorAlltech Hungary Ltd.


<strong>Reproduction</strong><strong>in</strong> <strong>Domestic</strong> <strong>Animals</strong>Vol. 43 • Supplement 2 • July 2008Proceed<strong>in</strong>gs of the16th International Congress on Animal<strong>Reproduction</strong>13–17 July 2008 – Budapest, HungaryGuest Editor:Prof. Gyula HuszeniczaGuest Editorial Board:Dr. Judit BarnaProf. Andras D<strong>in</strong>nyesDr. Ferenc Fl<strong>in</strong>kProf. Gyorgy GaborProf. Miklos MezesProf. Jozsef RatkyProf. Laszlo SoltiProf. Laszlo Wekerle


<strong>Reproduction</strong><strong>in</strong> <strong>Domestic</strong> <strong>Animals</strong>Table of Contents Volume 43 · Supplement 2 · July 2008 · 1-422ORIGINAL ARTICLESC. GALLI, G. LAZZARIThe Manipulation of Gametes and Embryos <strong>in</strong> Farm <strong>Animals</strong> 1-7D. BLACHE, G. B. MARTIN, S. K. MALONEYTowards Ethically Improved Animal Experimentation <strong>in</strong> the Study of Animal <strong>Reproduction</strong> 8-14S. M. RHINDEndocr<strong>in</strong>e Disruptors and Other Food-contam<strong>in</strong>at<strong>in</strong>g Environmental Pollutants as Risk Factors <strong>in</strong>Animal <strong>Reproduction</strong> 15-22J. E. P. SANTOS, T. R. BILBY, W. W. THATCHER, C. R. STAPLES, F. T. SILVESTRELong Cha<strong>in</strong> Fatty Acids of Diet as Factors Influenc<strong>in</strong>g <strong>Reproduction</strong> <strong>in</strong> Cattle 23-30M. C. LUCYFunctional Differences <strong>in</strong> the Growth Hormone and Insul<strong>in</strong>-like Growth Factor Axis <strong>in</strong> Cattle and Pigs:Implications for Post-partum Nutrition and <strong>Reproduction</strong> 31-39P. CHEMINEAU, D. GUILLAUME, M. MIGAUD, J. C. THIÉRY, M. T. PELLICER-RUBIO, B. MALPAUXSeasonality of <strong>Reproduction</strong> <strong>in</strong> Mammals: Intimate Regulatory Mechanisms and Practical Implications 40-47M. MIHM, A. C. O. EVANSMechanisms for Dom<strong>in</strong>ant Follicle Selection <strong>in</strong> Monovulatory Species: A Comparison of Morphological,Endocr<strong>in</strong>e and Intraovarian Events <strong>in</strong> Cows, Mares and Women 48-56D. J. SKARZYNSKI, G. FERREIRA-DIAS, K. OKUDARegulation of Luteal Function and Corpus Luteum Regression <strong>in</strong> Cows: Hormonal Control, ImmuneMechanisms and Intercellular Communication 57-65H. J. BERTSCHINGER, D. G. A. MELTZER, A. VAN DYKCaptive Breed<strong>in</strong>g of Cheetahs <strong>in</strong> South Africa – 30 Years of Data from the de WildtCheetah and Wildlife Centre 66-73M. DEHNHARD, S. NAIDENKO, A. FRANK, B. BRAUN, F. GÖRITZ, K. JEWGENOWNon-<strong>in</strong>vasive Monitor<strong>in</strong>g of Hormones: A Tool to Improve <strong>Reproduction</strong> <strong>in</strong> Captive Breed<strong>in</strong>g of theEurasian Lynx: Hormone Monitor<strong>in</strong>g <strong>in</strong> Breed<strong>in</strong>g Programmes of Mammals 74-82J. A. LONGReproductive Biotechnology and Gene Mapp<strong>in</strong>g: Tools for Conserv<strong>in</strong>g Rare Breeds of Livestock 83-88B. BERGLUNDGenetic Improvement of Dairy Cow Reproductive Performance 89-95J. L. M. R. LEROY, T. VANHOLDER, A. T. M. VAN KNEGSEL, I. GARCIA-ISPIERTO, P. E. J. BOLSNutrient Prioritization <strong>in</strong> Dairy Cows Early Postpartum: Mismatch Between Metabolism and Fertility? 96-103R. S. ROBINSON, A. J. HAMMOND, D. C. WATHES, M. G. HUNTER, G. E. MANNCorpus Luteum-Endometrium-Embryo Interactions <strong>in</strong> the Dairy Cow: Underly<strong>in</strong>g Mechanisms andCl<strong>in</strong>ical Relevance 104-112N. C. FRIGGENS, M. BJERRING, C. RIDDER, S. HØJSGAARD, T. LARSENImproved Detection of Reproductive Status <strong>in</strong> Dairy Cows Us<strong>in</strong>g Milk Progesterone Measurements 113-121D. R. NOTTERGenetic Aspects of <strong>Reproduction</strong> <strong>in</strong> Sheep 122-128R. J. SCARAMUZZI, G. B. MARTINThe Importance of Interactions Among Nutritions, Seasonality and Socio-sexual Factors <strong>in</strong> theDevelopment of Hormone-free Methods for Controll<strong>in</strong>g Fertility 129-136K. M. MORTONDevelopmental Capabilities of Embryos Produced In Vitro from Prepubertal Lamb Oocytes 137-143E. AXNÉRUpdates on Reproductive Physiology, Genital Diseases and Artificial Insem<strong>in</strong>ation <strong>in</strong> the <strong>Domestic</strong> Cat 144-149H. LINDEBERG<strong>Reproduction</strong> of the Female Ferret (Mustela putorius furo) 150-156J. DE GIER, N. J. BEIJERINK, H. S. KOOISTRA, A. C. OKKENSPhysiology of the Can<strong>in</strong>e Anoestrus and Methods for Manipulation of Its Length 157-164G. C. W. ENGLAND, K. M. MILLARThe Ethics and Role of AI with Fresh and Frozen Semen <strong>in</strong> Dogs 165-171A. CARATY, I. FRANCESCHINIBasic Aspects of the Control of GnRH and LH Secretions by Kisspept<strong>in</strong>: Potential Applications forBetter Control of Fertility <strong>in</strong> Females 172-178R. FAYRER-HOSKENControll<strong>in</strong>g Animal Populations Us<strong>in</strong>g Anti-Fertility Vacc<strong>in</strong>es 179-185T. E. ADAMS, I. BOIMEThe Expand<strong>in</strong>g Role of Recomb<strong>in</strong>ant Gonadotrop<strong>in</strong>s <strong>in</strong> Assisted <strong>Reproduction</strong> 186-192


T. A. L. BREVINI, S. ANTONINI, G. PENNAROSSA, F. GANDOLFIRecent Progress <strong>in</strong> Embryonic Stem Cell Research and Its Application <strong>in</strong> <strong>Domestic</strong> Species 193-199B. M. A. O. PERERA<strong>Reproduction</strong> <strong>in</strong> <strong>Domestic</strong> Buffalo 200-206P. S. BRAR, A. S. NANDAPostpartum Ovarian Activity <strong>in</strong> South Asian Zebu Cattle 207-212M. J. R. PARANHOS DA COSTA, A. SCHMIDEK, L. M. TOLEDOMother-Offspr<strong>in</strong>g Interactions <strong>in</strong> Zebu Cattle 213-216B. S. PRAKASH, M. SARKAR, M. MONDALAn Update on <strong>Reproduction</strong> <strong>in</strong> Yak and Mithun 217-223F. X. DONADEU, H. G. PEDERSENFollicle Development <strong>in</strong> Mares 224-231M. A. HAYES, B. A. QUINN, N. D. KEIRSTEAD, P. KATAVOLOS, R. O. WAELCHLI, K. J. BETTERIDGEProte<strong>in</strong>s Associated With the Early Intrauter<strong>in</strong>e Equ<strong>in</strong>e Conceptus 232-237Z. ROTHHeat Stress, the Follicle, and Its Enclosed Oocyte: Mechanisms and Potential Strategies to ImproveFertility <strong>in</strong> Dairy Cows 238-244K. - P. BRÜSSOW, J. RÁTKY, H. RODRIGUEZ-MARTINEZFertilization and Early Embryonic Development <strong>in</strong> the Porc<strong>in</strong>e Fallopian Tube 245-251S. PYÖRÄLÄMastitis <strong>in</strong> Post-Partum Dairy Cows 252-259M. G. DISKIN, D. G. MORRISEmbryonic and Early Foetal Losses <strong>in</strong> Cattle and Other Rum<strong>in</strong>ants 260-267N. MANABE, F. MATSUDA-MINEHATA, Y. GOTO, A. MAEDA, Y. CHENG, S. NAKAGAWA, N. INOUE, K. WONGPANIT, H. JIN, H. GONDA, J. LIRole of Cell Death Ligand and Receptor System on Regulation of Follicular Atresia <strong>in</strong> Pig Ovaries 268-272F. F. BARTOL, A. A. WILEY, C. A. BAGNELLEpigenetic Programm<strong>in</strong>g of Porc<strong>in</strong>e Endometrial Function and the Lactocr<strong>in</strong>e Hypothesis 273-279K. C. CAIRES, J. A. SCHMIDT, A. P. OLIVER, J. DE AVILA, D. J. MCLEANEndocr<strong>in</strong>e Regulation of the Establishment of Spermatogenesis <strong>in</strong> Pigs 280-287I. DOBRINSKIMale Germ Cell Transplantation 288-294N. RAWLINGS, A. C. O. EVANS, R. K. CHANDOLIA, E. T. BAGUSexual Maturation <strong>in</strong> the Bull 295-301A. DINNYES, X. C. TIAN, X. YANGEpigenetic Regulation of Foetal Development <strong>in</strong> Nuclear Transfer Animal Models 302-309R. C. BOTT, D. T. CLOPTON, A. S. CUPPA Proposed Role for VEGF Isoforms <strong>in</strong> Sex-Specific Vasculature Development <strong>in</strong> the Gonad 310-316B. K. WHITLOCK, J. A. DANIEL, R. R. WILBORN, T. H. ELSASSER, J. A. CARROLL, J. L. SARTINComparative Aspects of the Endotox<strong>in</strong>- and Cytok<strong>in</strong>e-Induced Endocr<strong>in</strong>e Cascade Influenc<strong>in</strong>gNeuroendocr<strong>in</strong>e Control of Growth and <strong>Reproduction</strong> <strong>in</strong> Farm <strong>Animals</strong> 317-323C. R. BARB, G. J. HAUSMAN, C. A. LENTSEnergy Metabolism and Lept<strong>in</strong>: Effects on Neuroendocr<strong>in</strong>e Regulation of <strong>Reproduction</strong> <strong>in</strong> the Gilt and Sao 324-330C. GALLI, I. LAGUTINA, R. DUCHI, S. COLLEONI, G. LAZZARISomatic Cell Nuclear Transfer <strong>in</strong> Horses 331-337D. RATH, L. A. JOHNSONApplication and Commercialization of Flow Cytometrically Sex-Sorted Semen 338-346J. M. VAZQUEZ, J. ROCA, M. A. GIL, C. CUELLO, I. PARRILLA, I. CABALLERO, J. L. VAZQUEZ, E. A. MARTÍNEZLow-Dose Insem<strong>in</strong>ation <strong>in</strong> Pigs: Problems and Possibilities 347-354C. B. A. WHITELAW, S. G. LILLICO, T. KINGProduction of Transgenic Farm <strong>Animals</strong> by Viral Vector-Mediated Gene Transfer 355-358A. C. O. EVANS, N. FORDE, G. M. O'GORMAN, A. E. ZIELAK, P. LONERGAN, T. FAIRUse of Microarray Technology to Profile Gene Expression Patterns Important for <strong>Reproduction</strong> <strong>in</strong> Cattle 359-367J. P. KASTELIC, J. C. THUNDATHILBreed<strong>in</strong>g Soundness Evaluation and Semen Analysis for Predict<strong>in</strong>g Bull Fertility 368-373G. C. ALTHOUSESanitary Procedures for the Production of Extended Semen 374-378B. LEBOEUF, J. A. DELGADILLO, E. MANFREDI, A. PIACÈRE, V. CLÉMENT, P. MARTIN, M. PELLICER,P. BOUÉ, R. DE CREMOUXManagement of Goat <strong>Reproduction</strong> and Insem<strong>in</strong>ation for Genetic Improvement <strong>in</strong> France 379-385N. KOSTEREVA, M. -C. HOFMANNRegulation of the Spermatogonial Stem Cell Niche 386-392P. MERMILLOD, R. DALBIÈS-TRAN, S. UZBEKOVA, A. THÉLIE, J. - M. TRAVERSO, C. PERREAU,P. PAPILLIER, P. MONGETFactors Affect<strong>in</strong>g Oocyte Quality: Who is Driv<strong>in</strong>g the Follicle? 393-400K. KIKUCHI, N. KASHIWAZAKI, T. NAGAI, M. NAKAI, T. SOMFAI, J. NOGUCHI, H. KANEKOSelected Aspects of Advanced Porc<strong>in</strong>e Reproductive Technology 401-406B. OBACKClimb<strong>in</strong>g Mount Efficiency – Small Steps, Not Giant Leaps Towards Higher Clon<strong>in</strong>g Success <strong>in</strong> Farm <strong>Animals</strong> 407-416P. LOI, K. MATZUKAWA, G. PTAK, Y. NATAN, J. FULKA JR, A. ARAVNuclear Transfer of Freeze-Dried Somatic Cells <strong>in</strong>to Enucleated Sheep Oocytes 417-422


M<strong>in</strong>itüb:ProductsforArtificial Insem<strong>in</strong>ation andEmbryo Transfer


WhitePartners <strong>in</strong> <strong>Reproduction</strong>Research and Practice


Reprod Dom Anim 43 (Suppl. 2), 1–7 (2008); doi: 10.1111/j.1439-0531.2008.01136.xISSN 0936-6768The Manipulation of Gametes and Embryos <strong>in</strong> Farm <strong>Animals</strong>C Galli 1,2 and G Lazzari 11 Laboratorio di Tecnologie della Riproduzione, Istituto Sperimentale Italiano Lazzaro Spallanzani, CIZ srl, Cremona, Italy; 2 Dip. Cl<strong>in</strong>icoVeter<strong>in</strong>ario, Universita` di Bologna, Bologna, ItalyContentsThis paper summarizes the major advances <strong>in</strong> farm animalassisted reproduction <strong>in</strong> the last 20 years with particularattention to the contributions of the authors. A ma<strong>in</strong> emphasisis on the biology of the oocyte, <strong>in</strong>clud<strong>in</strong>g a description ofmethods for isolation of develop<strong>in</strong>g follicles and culture of thecorrespond<strong>in</strong>g oocytes. Milestones that have led to optimizationof procedures for maturation of fully grown oocytes,fertilization, <strong>in</strong>tracytoplasmic sperm <strong>in</strong>jection and embryoculture <strong>in</strong> sheep, cattle, pigs and horses are described. Thecurrent status of nuclear transfer, clon<strong>in</strong>g and embryonic stemcell generation and culture is also summarized for all majorfarm animal species. It is concluded that the manipulation ofearly development <strong>in</strong> farm animals is of crucial importance foragricultural purposes and that reproductive biotechnologies <strong>in</strong>farm animals are expected to play an <strong>in</strong>creas<strong>in</strong>g role <strong>in</strong> the nextdecades due to the grow<strong>in</strong>g demand for agricultural productsfrom the emerg<strong>in</strong>g economies worldwide. In the biomedicalfield large animals represent <strong>in</strong>creas<strong>in</strong>gly important researchmodels especially <strong>in</strong> the stem cell field for creat<strong>in</strong>g geneticallymodified animals for specific purposes. F<strong>in</strong>ally, the successfultranslation of large animal research <strong>in</strong> the applied contextrequires solid science, long-term resource commitment from<strong>in</strong>volved <strong>in</strong>stitutions, and vision, dedication and entrepreneurialskills from the scientists <strong>in</strong>volved.IntroductionDespite the many scientific advances that have characterizedreproduction research, the science of reproductionreta<strong>in</strong>s an aura of wonder. This feel<strong>in</strong>g is captured <strong>in</strong>Harvey’s phrase: ‘ex ovo omnia’ and this fundamentaltruth expla<strong>in</strong>s the central role that reproduction has <strong>in</strong>biology of all life <strong>in</strong>clud<strong>in</strong>g our own. In livestockbreed<strong>in</strong>g, as well as for companion animals and wildlifeconservation, reproduction and selection allow ma<strong>in</strong>tenanceand propagation of species and breeds. Mostimportantly, farm animal reproduction is a centralphenomenon required for genetic selection to improveproductive ⁄ reproductive traits <strong>in</strong> the next generation.Genomics, proteomics and marker-assisted selectionwould all be limited technologies if not exploited throughreproduction. The development of assisted reproductiontechniques, as advanced tools for animal breed<strong>in</strong>g, hasbe<strong>in</strong>g <strong>in</strong>strumental for the acquisition of <strong>in</strong>-depth knowledgeof the molecular mechanisms of the reproductionprocesses, for study<strong>in</strong>g gametes and embryos and forallow<strong>in</strong>g the manipulation of early development <strong>in</strong> farmanimals. The strong driv<strong>in</strong>g force beh<strong>in</strong>d the advancementof reproductive technologies has been the need foragricultural and, more recently, biomedical applications.In many <strong>in</strong>stances, the technical achievements havesurpassed the full understand<strong>in</strong>g of the underly<strong>in</strong>gbiological mechanisms for those achievements.In this paper, we will discuss the <strong>in</strong> vitro reproductivetechnologies that have had the greatest impact onanimal breed<strong>in</strong>g and highlight our contributions to theirdevelopment. For this reason, it was not the aim of thepaper to make an extensive review of the publishedliterature but rather to mention the work and publicationsthat have directly <strong>in</strong>spired our work. We apologize<strong>in</strong> advance for those citations that are not <strong>in</strong>cluded.At the beg<strong>in</strong>n<strong>in</strong>g of our career <strong>in</strong> science, <strong>in</strong> the mid-1980s, the only embryo technology that was applied <strong>in</strong>practice was superovulation and embryo transfer [multipleovulation embryo transfer (MOET)]. At that time,soon became apparent that the MOET technique hadmany limits and other ways could be developed forproduc<strong>in</strong>g embryos required by the cattle <strong>in</strong>dustry. Asem<strong>in</strong>al paper published by Staigmiller and Moor (1984)drew our attention, as it demonstrated that immatureoocytes, after <strong>in</strong> vitro maturation (IVM), could beconverted <strong>in</strong>to viable embryos and offspr<strong>in</strong>g at a level ofefficiency that was remarkably high. Our <strong>in</strong>terest wasparticularly attracted to the female germ l<strong>in</strong>e. On theone hand, we dreamed about the possibility of exploit<strong>in</strong>gthe large pool of grow<strong>in</strong>g oocytes <strong>in</strong> the ovaries and,on the other hand, we aimed at rescu<strong>in</strong>g those fullygrown oocytes present <strong>in</strong> medium to large size folliclesthat exist throughout the reproductive lifetime of femalecattle. The aim was to overcome the physiologicallimitation of conventional breed<strong>in</strong>g, especially <strong>in</strong> themonotocycous Bos taurus, and make a better use offemale gametes that were otherwise dest<strong>in</strong>ed to wastageby atresia. In this way, the genetic impact of females ofsuperior genotype and performance on a given populationcould be greatly amplified. Indeed the oocyte is themost <strong>in</strong>terest<strong>in</strong>g cell <strong>in</strong> biology and may play the mostcrucial role <strong>in</strong> successful breed<strong>in</strong>g. A significant proportionof successful embryo development can be relatedback to the oocyte (Staigmiller and Moor 1984) and theproduction of large number of good quality oocytesus<strong>in</strong>g <strong>in</strong> vitro techniques opened a new era <strong>in</strong> embryotechnologies for farm animals.In this paper, we will review our work of the last20 years. Efforts started with a specific <strong>in</strong>terest <strong>in</strong> theoocyte and evolved <strong>in</strong>to build<strong>in</strong>g novel approaches toembryo-related biotechnologies rang<strong>in</strong>g from control ofmeiosis, <strong>in</strong> vitro fertilization (IVF), embryo culture,nuclear transfer, stem cells and genetic modification oflarge domestic animals. We always worked <strong>in</strong> anultrashort feedback loop between fundamental researchand technology application, <strong>in</strong> close contact with theend users of the technologies that we were work<strong>in</strong>g on.Such contacts have served as a strong rem<strong>in</strong>der thatany new technology has to have application. In ourÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


2 C Galli and G Lazzaricircumstances, the f<strong>in</strong>al result was the birth of liveoffspr<strong>in</strong>g at an acceptable rate, with several differenttechnologies and across a number of different speciesthat <strong>in</strong>cluded cattle, sheep, horses, pigs and buffalo.From the Very Beg<strong>in</strong>n<strong>in</strong>g: The Grow<strong>in</strong>g OocyteIn mammals, only a t<strong>in</strong>y fraction of the large number ofprimordial follicles generates progeny; the vast majorityare lost dur<strong>in</strong>g through atresia. For many years, several<strong>in</strong>vestigators isolated and <strong>in</strong>vestigated the biology of<strong>in</strong>tact primordial follicles <strong>in</strong> rodents (Roy and Greenwald1985) and eventually grew them successfully <strong>in</strong>culture (Eppig and O’Brien 1996). This task provedmuch more difficult <strong>in</strong> large animals because of the largesize and extensive connective tissue of the ovary.Nonetheless, we developed (Lazzari et al. 1992) aprotocol for isolation of large numbers of primordialoocytes from the ovary of newborn piglets based onenzymatic digestion and separation by centrifugal elutriation.Later, oocytes were obta<strong>in</strong>ed from secondaryfollicles of cattle and successful development with thebirth of live calves was achieved, albeit at a low rate(Miyano and Manabe 2007). Culture requirements forgrow<strong>in</strong>g a complex tri-dimensional structure such as anovarian follicle are very complex. The only progress thathas been made <strong>in</strong>volves culture <strong>in</strong> vivo of th<strong>in</strong> slices ofthe ovary follow<strong>in</strong>g transplantation <strong>in</strong> humans (Meirowet al. 2007). This technology has potential applicationsfor patients undergo<strong>in</strong>g certa<strong>in</strong> therapies that causeoocyte damage.The hypothesis that the ovarian reservoir of germ cellsis replenished by haematopoietic stem cells mov<strong>in</strong>g <strong>in</strong>tothe ovaries (Johnson et al. 2005) is an <strong>in</strong>trigu<strong>in</strong>g onethat suggests new approaches for develop<strong>in</strong>g largenumbers of oocytes. However, no progeny has beenobta<strong>in</strong>ed from such migrat<strong>in</strong>g cells, and thus the longestablishedview that the number of oocytes is fixed atbirth is true (Eggan et al. 2006). A more realisticapproach for the production of new female gametes isthrough differentiation of embryonic stem (ES) cellsand, <strong>in</strong>deed, development of oocyte-like cells from EScells has been reported (Hubner et al. 2003; Ko andScholer 2006). In spite of these astonish<strong>in</strong>g results, thecomplexity of epigenetic regulation of germ cell development<strong>in</strong> mammals (Sasaki and Matsui 2008) sets thisambitious target very far off.Oocyte Maturation In Vitro across Species andTimeFrom the late 1970s, it became clear that it was possibleto exploit the oocyte reservoir present <strong>in</strong> secondaryfollicles of farm animals. Early on, successful developmentwas obta<strong>in</strong>ed by matur<strong>in</strong>g the oocyte <strong>in</strong>side thefollicle (Moor and Trounson 1977). The subsequentunderstand<strong>in</strong>g of the crucial role of the surround<strong>in</strong>gfollicular ⁄ cumulus cells dur<strong>in</strong>g the <strong>in</strong>itial phases ofmaturation (Moor et al. 1981) leads to the developmentof a co-culture system for IVM of extrafollicular oocytesthat resulted <strong>in</strong> a high proportion of such oocytesdevelop<strong>in</strong>g <strong>in</strong>to live lambs after <strong>in</strong> vivo transfer (Staigmillerand Moor 1984). After the successful achievementof IVM <strong>in</strong> sheep, the work was expanded by<strong>in</strong>vestigat<strong>in</strong>g the effects of gonadotrop<strong>in</strong>s on oocytematuration (Galli and Moor 1991b). In addition, themodel was transferred to the cow at Animal BiotechnologyCambridge Ltd <strong>in</strong> UK, at Ovamass <strong>in</strong> Dubl<strong>in</strong>(Lu and Polge 1992; Carolan et al. 1994) and elsewhere(Xu et al. 1987; Goto et al. 1988). Similar developmentsoccurred <strong>in</strong> the pig (Moor et al. 1990), horse (Zhanget al. 1989) and other species as well. When IVF wasperfected, allow<strong>in</strong>g a deeper understand<strong>in</strong>g of oocytedevelopmental competence, other factors, such as folliclesize, were identified as important <strong>in</strong>dicators of oocytecompetence <strong>in</strong> the cow (Galli and Moor 1991a) andhorse (Galli et al. 2007). Studies on the prote<strong>in</strong> synthesisactivity with<strong>in</strong> the oocyte and on the <strong>in</strong>teractionsbetween follicular cells and oocytes (Staigmiller andMoor 1984; Downs et al. 1988; Buccione et al. 1990)contributed to the identification of active compoundssecreted by somatic cells (epidermal growth factor,<strong>in</strong>sul<strong>in</strong> growth factors, fibroblast growth factors, etc.)that promote competent maturation.The development of procedures for oocyte retrievalfrom live bov<strong>in</strong>e donors (Pieterse et al. 1991; Hasleret al. 1995; Galli et al. 2001), referred to as ovum pickup, opened an important area for the application ofoocyte maturation and embryo production <strong>in</strong> vitro forbreed<strong>in</strong>g purposes <strong>in</strong> cattle as well as other large speciesas buffalo (Galli et al. 1998) and horses (Galli et al.2002).Despite the high rate of nuclear maturation obta<strong>in</strong>ed<strong>in</strong> vitro, the developmental competence of maturedoocytes is variable. One likely source for much of thisvariation is the <strong>in</strong>tr<strong>in</strong>sic quality of the oocytes recoveredfrom the ovaries. To overcome the problem caused byharvest<strong>in</strong>g of suboptimal oocytes, it was tested whethera period of prematuration <strong>in</strong> vitro to ma<strong>in</strong>ta<strong>in</strong> oocytesarrested <strong>in</strong> prophase I could <strong>in</strong>crease their developmentalcompetence follow<strong>in</strong>g maturation. Butyrolactoneand roscovit<strong>in</strong>e, two specific prote<strong>in</strong> k<strong>in</strong>ases <strong>in</strong>hibitors,were used for keep<strong>in</strong>g oocytes arrested <strong>in</strong> Germ<strong>in</strong>alVesicle (GV) for a 24-h culture period (Ponderato et al.2001). This meiosis block was fully reversible as demonstratedby resumption of meiosis and normal embryonicdevelopment follow<strong>in</strong>g transfer of embryos derivedfrom prematured oocytes (Ponderato et al. 2002). Yet,this strategy did not improve oocyte competence andfound very limited application.Manipulat<strong>in</strong>g the Mature Egg for EmbryoProductionFollow<strong>in</strong>g the sem<strong>in</strong>al work <strong>in</strong> laboratory animals(Yanagimachi and Chang 1964), IVF was later developed<strong>in</strong> farm animal species. The birth of the first IVFcalf derived from <strong>in</strong> vivo matured oocytes (Brackettet al. 1982) was followed by significant advances <strong>in</strong> IVFwhen hepar<strong>in</strong> was used as capacitat<strong>in</strong>g agent for bullsperm (Parrish et al. 1986). At about the same time, IVFbecame a reality <strong>in</strong> other large species except the horsewhere success has been only exceptional. Indeed, onlytwo foals have been reported as the result of IVF(Palmer et al. 1991). Assisted reproduction of thehorse eventually benefited from the development ofÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Embryo Biotechnologies <strong>in</strong> Farm <strong>Animals</strong> 3<strong>in</strong>tracytoplasmic sperm <strong>in</strong>jection (ICSI) <strong>in</strong> humans(Palermo et al. 1992). The use of <strong>in</strong>tracytoplasmicsperm <strong>in</strong>jection (ICSI) has been very efficient as a wayto circumvent failure of IVF (Galli et al. 2007). In otherfarm animals like cattle (Goto and Yanagita 1995; Galliet al. 2003c), sheep (Catt et al. 1996) and pigs (Kolbeand Holtz 2000), the efficiency of ICSI for embryoproduction rema<strong>in</strong>s lower than for IVF and thereforethe technique is not used rout<strong>in</strong>ely. In pigs, IVF ischaracterized by a high <strong>in</strong>cidence of polyspermy thatcompromises embryonic development. This problem hasbeen overcome <strong>in</strong> part by improv<strong>in</strong>g the quality of<strong>in</strong> vitro matured oocytes rather than by chang<strong>in</strong>g IVFconditions.In recent years, separation of X and Y-bear<strong>in</strong>g spermby flow cytometry is f<strong>in</strong>d<strong>in</strong>g wide application <strong>in</strong> reproductivetechnologies. When comb<strong>in</strong>ed with IVF or ICSI,a synergy can occur to <strong>in</strong>crease the number of embryosof the desired sex comb<strong>in</strong>ation (Seidel 2003; Cran 2007)produced by assisted reproductive techniques moreeffectively than older technologies based on sex<strong>in</strong>g ofcells collected by embryo biopsy.Manipulaton of Embryo DevelopmentThe goal of <strong>in</strong>creas<strong>in</strong>g efficiency of <strong>in</strong> vitro embryoproduction (IVP), especially <strong>in</strong> cattle, has been thedriv<strong>in</strong>g force for much of the applied research <strong>in</strong> embryobiology and culture; scores of papers have beenpublished on methods for improv<strong>in</strong>g the yield of IVPembryos <strong>in</strong> cattle. Yet, it became soon obvious thatmerely count<strong>in</strong>g the number of blastocysts was not anaccurate measure of the quality of the overall procedureand of the viability of the embryos (Gandolfi and Moor1987). For many years, these constra<strong>in</strong>ts on the cultureof viable cattle embryos <strong>in</strong> vitro were overcome by us<strong>in</strong>g<strong>in</strong> vivo culture <strong>in</strong> the oviduct of surrogate sheep (Galliand Lazzari 1996). In particular, a major concern forembryos produced by IVP was the so-called largeoffspr<strong>in</strong>g syndrome (LOS), first <strong>in</strong> sheep and then <strong>in</strong>cattle (Young et al. 1998). The use of serum supplementationand coculture were recognized as the primarycause of LOS. Later, an extensive field study (vanWagtendonk-de Leeuw et al. 2000) demonstrated thatthe <strong>in</strong>cidence of LOS was greatly reduced us<strong>in</strong>g a culturemedium based on Synthetic oviduct Fluid (Tervit et al.1972) and the <strong>in</strong>dustry took up this method <strong>in</strong> most<strong>in</strong>stances. These observations were accompanied by alarge set of studies look<strong>in</strong>g at the effects of <strong>in</strong> vitroculture on molecular and cellular markers of embryodevelopment, metabolism, gene expression and viability.Together with others, we <strong>in</strong>vestigated the cause of LOSby compar<strong>in</strong>g different <strong>in</strong> vitro culture systems with<strong>in</strong> vivo culture of bov<strong>in</strong>e embryos <strong>in</strong> the sheep oviduct.We set up an experiment compar<strong>in</strong>g <strong>in</strong> vivo culture <strong>in</strong> thesheep oviduct with <strong>in</strong> vitro culture <strong>in</strong> the presence ofserum or high levels of bov<strong>in</strong>e serum album<strong>in</strong> (BSA)(Lazzari et al. 2002). Results <strong>in</strong>dicated that embryosgrown <strong>in</strong> serum but also embryos grown <strong>in</strong> high levels ofBSA have alterations <strong>in</strong> the gene expression levels ofseveral developmentally important transcripts. In addition,we demonstrated that these embryos have morecells at the blastocyst stage on day 7 than IVM–IVFembryos developed <strong>in</strong> the sheep oviduct. Follow<strong>in</strong>gtransfer of day 7 blastocysts to recipients and recoveryafter 5 days, we showed that <strong>in</strong> vitro cultured embryosare larger on day 12 than those derived from sheepoviduct cultured up to day 7. Moreover, when the day12 embryos derived from <strong>in</strong> vitro culture, werere-transferred to new recipients, they gave rise tooffspr<strong>in</strong>g with an average birth weight higher than thecorrespond<strong>in</strong>g offspr<strong>in</strong>g derived from embryos cultured<strong>in</strong> the sheep oviduct. These f<strong>in</strong>d<strong>in</strong>gs clearly <strong>in</strong>dicatedthat <strong>in</strong> vitro culture can alter development at very earlystages and that the LOS is correlated to abnormallyadvanced embryonic growth and gene expressionpatterns at very early stages.Besides LOS, another important issue that has limitedthe use of <strong>in</strong> vitro produced embryos is their <strong>in</strong>consistentsurvival to freez<strong>in</strong>g and thaw<strong>in</strong>g. This limitation wasanother reason why we used for many years the sheepoviduct culture system because it allowed production ofIVM–IVF embryos that were undist<strong>in</strong>guishable from<strong>in</strong> vivo produced embryos <strong>in</strong> terms of viability andfreezability (Galli and Lazzari 1996; Lonergan and Fair2008). At present, recent improvements <strong>in</strong> mediaformulation (Gardner et al. 1994) have removed almostentirely the barrier of embryo freezability; pregnancyrate follow<strong>in</strong>g transfer of frozen-thawed <strong>in</strong> vitro producedcattle embryos is commercially viable. Importantvariations still exist across laboratories but those thathave really mastered the technology are now wellestablishedcommercial producers of IVP embryos.In vitro produced embryos account for approximatelyone-third of the cattle embryos produced worldwide(http://www.iets.org).A slightly different story has unfolded <strong>in</strong> the horse.For many years, equ<strong>in</strong>e-assisted reproduction laggedbeh<strong>in</strong>d other species. This was due <strong>in</strong> part to the lack of<strong>in</strong>terest of the horse <strong>in</strong>dustry <strong>in</strong> develop<strong>in</strong>g this technologybut also to a sort of reluctance to really translatequestions that had been asked and answered before <strong>in</strong>other species. Our work on equ<strong>in</strong>e reproduction datesback to the Cambridge period, <strong>in</strong> the late 1980s, whenwe obta<strong>in</strong>ed equ<strong>in</strong>e embryos from <strong>in</strong> vitro maturedoocytes transferred to the oviduct of <strong>in</strong>sem<strong>in</strong>ated mares(Zhang et al. 1989). The most recent work is summarized<strong>in</strong> a paper (Galli et al. 2007) where all the relevantsteps have been described <strong>in</strong>clud<strong>in</strong>g oocyte maturation,fertilization by ICSI, embryo culture and freez<strong>in</strong>g, nonsurgicaltransfer, and somatic cell nuclear transfer.Prometea, the first cloned horse, was derived totallyfrom <strong>in</strong> vitro procedures that had been optimized step bystep with dedication and perseverance, tak<strong>in</strong>g advantageand translat<strong>in</strong>g the experience derived from the establishedbov<strong>in</strong>e technology. Today, viable and freezableequ<strong>in</strong>e embryos can be produced efficiently by <strong>in</strong> vitrotechnologies (Colleoni et al. 2007). This success story isexpected to f<strong>in</strong>ally open up the horse <strong>in</strong>dustry to thesame opportunities that the cattle <strong>in</strong>dustry has exploitedfor several years.Nuclear Transfer and Clon<strong>in</strong>gThe birth of Dolly the sheep through clon<strong>in</strong>g by nucleartransfer (Wilmut et al. 1997) is an astonish<strong>in</strong>g accom-Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


4 C Galli and G Lazzariplishment that has attracted the attention of the generalpublic towards farm animal embryo-technologies. Thisis one of the best examples where the search of newreproductive techniques <strong>in</strong> farm animals has providedbasic knowledge not previously obta<strong>in</strong>ed <strong>in</strong> laboratoryanimals. As with many other areas of research, newconcepts and new techniques develop over time throughthe contribution of many <strong>in</strong>vestigators. The fundamentalwork on clon<strong>in</strong>g was done <strong>in</strong> amphybia <strong>in</strong> the 1960s(Gurdon 2006) followed by significant advancementsachieved with embryo clon<strong>in</strong>g of farm animals (Willadsen1986) when matured oocytes, <strong>in</strong>stead of zygotes,became the recipients of the donor nuclei. Ten yearslater, it was aga<strong>in</strong> farm animals that, with somatic cellnuclear transfer, gave a major contribution to the basicunderstand<strong>in</strong>g of cell reprogramm<strong>in</strong>g and epigeneticcontrol of mammalian development, and thereby open<strong>in</strong>ga new era <strong>in</strong> cell biology. Several farm animals werecloned before clon<strong>in</strong>g was demonstrated <strong>in</strong> laboratoryspecies. Most major domestic mammals have now beencloned albeit at a low efficiency. The success with whichdifferent mammals have been cloned was directlycorrelated to the availability of good quality matureoocytes and good quality embryos either after <strong>in</strong> vivo or<strong>in</strong> vitro culture. It was as early as 1990 at Cambridgethat we were generat<strong>in</strong>g embryos by nuclear transferwith somatic cells although the two pregnancies thatwere obta<strong>in</strong>ed did not develop to term (Galli et al.1991). In those early days, these k<strong>in</strong>ds of experimentswere looked at with high degree of scepticism until itwas, <strong>in</strong> fact, accomplished (Campbell et al. 1996; Wilmutet al. 1997). Thereafter, it was merely a comb<strong>in</strong>ationof already available technologies of gamete andembryo manipulation that lead to success <strong>in</strong> a variety ofspecies. In the horse, for example, it was not until suchtechnologies were developed that clon<strong>in</strong>g was achieved(Galli et al. 2003b; Lagut<strong>in</strong>a et al. 2005) <strong>in</strong> a consistentand reproducible way.In the clon<strong>in</strong>g of farm animals, there are significantspecies differences that have not been fully expla<strong>in</strong>ed.For example, we never observed <strong>in</strong> cloned horses(Lagut<strong>in</strong>a et al. 2005) and pigs (Lagut<strong>in</strong>a et al. 2006)the abnormalities, ma<strong>in</strong>ly associated with placentadysfunction, described <strong>in</strong> cattle and sheep. From apractical perspective, over the last 10 years, the efficiencyof somatic cell nuclear transfer as a whole has notimproved much and its practical application is limited tothe production of animals that have high added valuelike breed<strong>in</strong>g stock (Galli et al. 1999, 2003a; Lagut<strong>in</strong>aet al. 2005) or for generat<strong>in</strong>g transgenic founder animalswhen nuclear transfer is comb<strong>in</strong>ed with genetic eng<strong>in</strong>eer<strong>in</strong>gof somatic cells (Brophy et al. 2003; Kuroiwaet al. 2004; Wall et al. 2005; Lombardo et al. 2007).An understand<strong>in</strong>g of the constra<strong>in</strong>ts of nucleartransfer and development of the concept of reprogramm<strong>in</strong>gof fully differentiated somatic cells and thederivation of stem cells from cloned embryos (Wakayamaet al. 2001; Lazzari et al. 2006) has led to the ideaof reprogramm<strong>in</strong>g differentiated cells <strong>in</strong> vitro directlywithout the need of the oocyte. A set of four genes hasbeen demonstrated to be at the core of pluripotencywhen transfected somatic cells gave rise to <strong>in</strong>ducedpluripotent stem cells (iPS cells) (Takahashi and Yamanaka2006). These cells, of somatic orig<strong>in</strong>, are extraord<strong>in</strong>arybecause they carry all the properties of ES cellsderived from the early embryo. This research representsa major breakthrough for overcom<strong>in</strong>g the limited oocyteavailability <strong>in</strong> humans.Understand<strong>in</strong>g the molecular basis of pluripotencywill be a major step forward that is expected to lead toadvances <strong>in</strong> nuclear transfer technology and clon<strong>in</strong>g offarm animals for agricultural and biomedical applicationson a larger basis.Stem CellsAt this writ<strong>in</strong>g, we still await the first def<strong>in</strong>itive report ofthe derivation of farm animal ES cells. Over the years,from 1981, when mouse ES cells were first reported, tothis time, several laboratories and scientists all over theworld, <strong>in</strong>clud<strong>in</strong>g us, have attempted ES cell derivationfrom cattle, pig and sheep embryos (Galli et al. 1994;Keefer et al. 2007). The orig<strong>in</strong>al mouse approach hasbeen used extensively and f<strong>in</strong>ally proven unsuccessfulalthough a few reports have been published on thederivation of so-called ES-like cells (Notarianni et al.1991). Yet, the stemness (stam<strong>in</strong>ality) of these cellsappeared to be very limited and most likely theyrepresent trophoblastic cells given their epithelial nature,loss of OCT4 expression and limited differentiationpotential (Notarianni et al. 1991; Iwasaki et al. 2000;Keefer et al. 2007). A major and often reported problem<strong>in</strong> ES cell derivation from farm animal embryos is thetendency of the trophoblast and ⁄ or the endoderm toovergrow the <strong>in</strong>ner cell mass cells <strong>in</strong> culture even aftercareful dissection or isolation by immunosurgery (Keeferet al. 2007). This is a peculiarity of rum<strong>in</strong>ant and pigembryos and reflects the exuberant growth of thetrophoblast and the underly<strong>in</strong>g endoderm that occurs<strong>in</strong> vivo before implantation. The other well-knownproblem is the tendency for neural differentiation,generally followed by apoptosis, that is often observedespecially after plat<strong>in</strong>g of <strong>in</strong>ner cell mass cells <strong>in</strong> serumfreemedia. This tendency, which is a problem, alsorepresents an <strong>in</strong>terest<strong>in</strong>g biological system to exploreearly neurulation events <strong>in</strong> mammals (Lazzari et al.2006). We showed that bov<strong>in</strong>e <strong>in</strong>ner cell mass cells, bothfrom fertilized and nuclear transfer embryos, can bedifferentiated <strong>in</strong> an orderly manner towards the neuroectodermgiv<strong>in</strong>g rise to neural rosettes resembl<strong>in</strong>g thedevelop<strong>in</strong>g neural tube. From these rosettes highlyproliferat<strong>in</strong>g neural precursors can be obta<strong>in</strong>ed andfrom those a large variety of nervous system derivatives.In light of these reports and observations, it appearsthat the search for ES cells <strong>in</strong> farm animals shouldabandon the orig<strong>in</strong>al mouse procedure and pay moreattention to the recent f<strong>in</strong>d<strong>in</strong>gs for ES derivation <strong>in</strong>mouse and human. Advances <strong>in</strong> mouse and, morerecently, <strong>in</strong> human embryonic stem cell culture havedemonstrated that a number of different culture conditionscan support pluripotency of embryo-derived stemcells. Mouse ES cells can be grown not only <strong>in</strong> serumsupplementedculture media conta<strong>in</strong><strong>in</strong>g leukaemia<strong>in</strong>hibitory factor (LIF) and feeders but also <strong>in</strong> serumfreeand feeder-free culture by the addition of bonemorphogenetic prote<strong>in</strong> (BMP) molecules (Y<strong>in</strong>g et al.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Embryo Biotechnologies <strong>in</strong> Farm <strong>Animals</strong> 52003a). This second method has been developed follow<strong>in</strong>gthe f<strong>in</strong>d<strong>in</strong>g that mouse ES cells grown <strong>in</strong> a serumsupplementedmedium and LIF can be differentiated<strong>in</strong>to neuroectodermal cells by serum and LIF withdrawal(Y<strong>in</strong>g et al. 2003b). The role of BMP moleculesis to counteract and block the <strong>in</strong>duction of neuraldifferentiation and fix the undifferentiated state <strong>in</strong> aclever balance between conflict<strong>in</strong>g <strong>in</strong>ductive signall<strong>in</strong>gpathways. Other recent protocols, based on the stimulationof the nodal-activ<strong>in</strong> signall<strong>in</strong>g pathways, havebeen shown to ma<strong>in</strong>ta<strong>in</strong> the undifferentiated proliferationof human and mouse ES cells (James et al. 2005;Vallier et al. 2005; Brons et al. 2007; Ogawa et al. 2007).A particular attention should be paid to very recentreports of a novel stem cell type derived from mouseembryos and called epi stem cells (EpiSCs) (Brons et al.2007; Tesar et al. 2007). Interest<strong>in</strong>gly, mouse EpiSCshave been shown to be very similar to human ES cells <strong>in</strong>morphology, growth factor requirement and geneexpression (Brons et al. 2007; Tesar et al. 2007) whilemouse ES cells differ considerably from human ES <strong>in</strong>culture requirements for the ma<strong>in</strong>tenance of the undifferentiatedstate, growth rate and response to <strong>in</strong>ductivesignals. Another important difference between mouseES cells and EpiSCs is the fact that only the formerare capable of giv<strong>in</strong>g rise to chimeric offspr<strong>in</strong>g follow<strong>in</strong>gblastocyst <strong>in</strong>jection. Under the present status ofresearch, epiSC derivation <strong>in</strong> farm animals may proveeasier than true ES cell derivation and could representa significant step forward towards the understand<strong>in</strong>gof the requirements for ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g pluripotency<strong>in</strong> cultured <strong>in</strong>ner cells mass cells of farm animalembryos.From an applied perspective, embryonic stem cells <strong>in</strong>farm animals are important for several reasons but themost relevant is to provide a method to <strong>in</strong>troduceprecise genetic modification <strong>in</strong>to animals by homologousrecomb<strong>in</strong>ation of ES cells (Lombardo et al. 2007)followed by blastocyst <strong>in</strong>jection for chimera derivationand breed<strong>in</strong>g, or by somatic cell nuclear transfer. Asecond important objective is to provide large animalmodels <strong>in</strong> which the ES cell technology can be tested fortissue-specific differentiation (Brown et al. 2007) and celltherapy of various tissues and organs. Conventional EScells, carry<strong>in</strong>g all the properties of mouse ES cells, canserve both purposes but epiSCs can serve at least thesecond. Therefore, ES and epiSC research still deservesmajor research efforts and represents the frontier ofreproductive technologies <strong>in</strong> farm animals both foragriculture and biotechnological applications.ConclusionsThe manipulation of early embryonic development <strong>in</strong>farm animals is of foremost importance both foragriculture and biotechnology applications but also asa model to study some basic aspects of reproduction anddevelopmental biology. In the agricultural context,reproductive biotechnologies <strong>in</strong> farm animals areexpected to play an <strong>in</strong>creas<strong>in</strong>g role <strong>in</strong> the next decadesdue to the grow<strong>in</strong>g demand for agricultural productsfrom the emerg<strong>in</strong>g economies worldwide (see ‘TheEconomist’, The end of cheap food, 7–14 December2007). Modern reproductive biotechnologies, togetherwith the most recent molecular techniques of genomics,proteomics and transgenesis are the powerful tools thatwill provide the answers to the rapidly chang<strong>in</strong>g worldscenario of food demand. In the biomedical field, largeanimals represent <strong>in</strong>creas<strong>in</strong>gly important research modelsespecially <strong>in</strong> the stem cell field, for creat<strong>in</strong>g diseasemodels and genetically modified animals as potentialdonors of tissues and organs for xenotransplantation.These are the reasons why large animal research, thatshort-sighted worldwide research policies have neglected<strong>in</strong> recent years, will rega<strong>in</strong> a lead<strong>in</strong>g role <strong>in</strong> the com<strong>in</strong>gfuture.F<strong>in</strong>ally, successful translation of large animal research<strong>in</strong>to commercial enterprise requires solid science, longtermresource commitment, and extensive steps ofvalidation to reach the thresholds of reproducibilityand profitability. Therefore, strong scientific drive,vision and entrepreneurial skills are all needed forcontribut<strong>in</strong>g to progress <strong>in</strong> large animal reproductivesciences.AcknowledgementsA successful career <strong>in</strong> animal reproduction is the result of a full-timetotal dedication to the work but also of good tra<strong>in</strong><strong>in</strong>g, a stimulat<strong>in</strong>genvironment and <strong>in</strong>teraction with peers around the world. For thisreason, we want to acknowledge the many colleagues and collaboratorswhose support over the years has been <strong>in</strong>strumental to success <strong>in</strong>our work. In particular, we want to acknowledge Professor A Lauria,who dur<strong>in</strong>g our years as undergraduate students transmitted to us hisenthusiasm for animal reproduction, and especially Dr. Robert Moor,our mentor for many years, who gave us vision and <strong>in</strong>spired most ofthe work that we have been do<strong>in</strong>g <strong>in</strong> the last 20 years and we willprobably be do<strong>in</strong>g for the next 20 years.ReferencesBrackett BG, Bousquet D, Boice ML, Donawick WJ, EvansJF, Dressel MA, 1982: Normal development follow<strong>in</strong>g<strong>in</strong> vitro fertilization <strong>in</strong> the cow. Biol Reprod 27, 147–158.Brons IG, Smithers LE, Trotter MW, Rugg-Gunn P, Sun B,Chuva de Sousa Lopes SM, Howlett SK, Clarkson A,Ahrlund-Richter L, Pedersen RA, Vallier L, 2007: Derivationof pluripotent epiblast stem cells from mammalianembryos. Nature 448, 191–195.Brophy B, Smolenski G, Wheeler T, Wells D, L’Huillier P,Laible G, 2003: Cloned transgenic cattle produce milk withhigher levels of beta-case<strong>in</strong> and kappa-case<strong>in</strong>. Nat Biotechnol21, 157–162.Brown BD, Gentner B, Cantore A, Colleoni S, Amendola M,Z<strong>in</strong>gale A, Baccar<strong>in</strong>i A, Lazzari G, Galli C, Nald<strong>in</strong>i L, 2007:Endogenous microRNA can be broadly exploited to regulatetransgene expression accord<strong>in</strong>g to tissue, l<strong>in</strong>eage anddifferentiation state. Nat Biotechnol 25, 1457–1467.Buccione R, Schroeder AC, Eppig JJ, 1990: Interactionsbetween somatic cells and germ cells throughout mammalianoogenesis. Biol Reprod 43, 543–547.Campbell KH, McWhir J, Ritchie WA, Wilmut I, 1996: Sheepcloned by nuclear transfer from a cultured cell l<strong>in</strong>e. Nature380, 64–66.Carolan C, Monaghan P, Gallagher M, Gordon I, 1994: Effectof recovery method on yield of bov<strong>in</strong>e oocytes per ovary andtheir developmental competence after maturation, fertilizationand culture <strong>in</strong> vitro. Theriogenology 41, 1061–1068.Catt SL, Catt JW, Gomez MC, Maxwell WM, Evans G, 1996:Birth of a male lamb derived from an <strong>in</strong> vitro maturedÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


6 C Galli and G Lazzarioocyte fertilised by <strong>in</strong>tracytoplasmic <strong>in</strong>jection of a s<strong>in</strong>glepresumptive male sperm. Vet Rec 139, 494–495.Colleoni S, Barbac<strong>in</strong>i S, Necchi D, Duchi R, Lazzari G, GalliC, 2007: Application of ovum pick-up, <strong>in</strong>tracytoplasmicsperm <strong>in</strong>jection and embryo culture <strong>in</strong> equ<strong>in</strong>e practice. ProcAm Assoc Equ<strong>in</strong>e Pract (AAEP) 53, 554–559.Cran DG, 2007: XY sperm separation and use <strong>in</strong> artificial<strong>in</strong>sem<strong>in</strong>ation and other ARTs. Soc Reprod Fertil Suppl 65,475–491.Downs SM, Daniel SA, Eppig JJ, 1988: Induction of maturation<strong>in</strong> cumulus cell-enclosed mouse oocytes by folliclestimulat<strong>in</strong>ghormone and epidermal growth factor: evidencefor a positive stimulus of somatic cell orig<strong>in</strong>. J Exp Zool 245,86–96.Eggan K, Jurga S, Gosden R, M<strong>in</strong> IM, Wagers AJ, 2006:Ovulated oocytes <strong>in</strong> adult mice derive from non-circulat<strong>in</strong>ggerm cells. Nature 441, 1109–1114.Eppig JJ, O’Brien MJ, 1996: Development <strong>in</strong> vitro of mouseoocytes from primordial follicles. Biol Reprod 54, 197–207.Galli C, Lazzari G, 1996: Practical aspects of IVM ⁄ IVF <strong>in</strong>cattle. Anim Reprod Sci 42, 371–379.Galli C, Moor RM, 1991a: Development of immature bov<strong>in</strong>eoocytes <strong>in</strong>to viable embryos <strong>in</strong> vitro. Bull l’AssociationAnatom 228, 67–71.Galli C, Moor RM, 1991b: Gonadotroph<strong>in</strong> requirements forthe <strong>in</strong> vitro maturation of sheep oocytes and their subsequentembryonic development. Theriogenology 35, 1083–1093.Galli C, Laurie S, Lazzari G, Moor RM, 1991: Nucleartransplantation (by electrofusion) of cultured embryoniccells with Met II cytoplasts <strong>in</strong> the sheep. Annu Conf Ital SocVet Sci XLV, 299–303.Galli C, Lazzari G, Flechon JE, Moor RM, 1994: Embryonicstem cells <strong>in</strong> farm animals. Zygote 2, 385–389.Galli C, Duchi R, Crotti G, Lazzari G, 1998: Embryoproduction by Ovum Pick Up <strong>in</strong> Water Buffalo. Theriogenol49, 400.Galli C, Duchi R, Moor RM, Lazzari G, 1999: Mammalianleukocytes conta<strong>in</strong> all the genetic <strong>in</strong>formation necessary forthe development of a new <strong>in</strong>dividual. Clon<strong>in</strong>g 1, 161–170.Galli C, Crotti G, Notari C, Tur<strong>in</strong>i P, Duchi R, Lazzari G,2001: Embryo production by ovum pick up from livedonors. Theriogenology 55, 1341–1357.Galli C, Crotti G, Tur<strong>in</strong>i P, Duchi R, Mari G, Zavaglia G,Duchamp G, Daels P, Lazzari G, 2002: Frozen- thawedembryos produced by Ovum Pick Up of immature oocytesand ICSI are capable to establish pregnancies <strong>in</strong> the horse.Theriogenology 58, 705–708.Galli C, Vassiliev I, Lagut<strong>in</strong>a I, Galli A, Lazzari G, 2003a:Bov<strong>in</strong>e embryo development follow<strong>in</strong>g ICSI: effect ofactivation, sperm capacitation and pre-treatment with dithiothreitol.Theriogenology 60, 1467–1480.Galli C, Duchi R, Crotti G, Tur<strong>in</strong>i P, Ponderato N, Colleoni S,Lagut<strong>in</strong>a I, Lazzari G, 2003b: Bov<strong>in</strong>e embryo technologies.Theriogenology 59, 599–616.Galli C, Lagut<strong>in</strong>a I, Crotti G, Colleoni S, Tur<strong>in</strong>i P, PonderatoN, Duchi R, Lazzari G, 2003c: Pregnancy: a cloned horseborn to its dam tw<strong>in</strong>. Nature 424, 635.Galli C, Colleoni S, Duchi R, Lagut<strong>in</strong>a I, Lazzari G, 2007:Developmental competence of equ<strong>in</strong>e oocytes and embryosobta<strong>in</strong>ed by <strong>in</strong> vitro procedures rang<strong>in</strong>g from <strong>in</strong> vitromaturation and ICSI to embryo culture, cryopreservationand somatic cell nuclear transfer. Anim Reprod Sci 98, 39–55.Gandolfi F, Moor RM, 1987: Stimulation of early embryonicdevelopment <strong>in</strong> the sheep by co-culture with oviductepithelial cells. J Reprod Fertil 81, 23–28.Gardner DK, Lane M, Spitzer A, Batt PA, 1994: Enhancedrates of cleavage and development for sheep zygotescultured to the blastocyst stage <strong>in</strong> vitro <strong>in</strong> the absence ofserum and somatic cells: am<strong>in</strong>o acids, vitam<strong>in</strong>s, and cultur<strong>in</strong>gembryos <strong>in</strong> groups stimulate development. Biol Reprod50, 390–400.Goto K, Yanagita K, 1995: Normality of calves obta<strong>in</strong>ed by<strong>in</strong>tracytoplasmic sperm <strong>in</strong>jection. Hum Reprod 10, 1554.Goto K, Kajihara Y, Kosaka S, Koba M, Nakanishi Y,Ogawa K, 1988: Pregnancies after co-culture of cumuluscells with bov<strong>in</strong>e embryos derived from <strong>in</strong>-vitro fertilizationof <strong>in</strong>-vitro matured follicular oocytes. J Reprod Fertil 83,753–758.Gurdon JB, 2006: From nuclear transfer to nuclear reprogramm<strong>in</strong>g:the reversal of cell differentiation. Annu Rev CellDev Biol 22, 1–22.Hasler JF, Henderson WB, Hurtgen PJ, J<strong>in</strong> ZQ, McCauleyAD, Mower SA, Neely B, Shuey LS, Stokes JE, TrimmerSA, 1995: Production, freez<strong>in</strong>g and transfer of bov<strong>in</strong>e IVFembryos and subsequent calv<strong>in</strong>g results. Theriogenology 43,141–152.Hubner K, Fuhrmann G, Christenson LK, Kehler J, Re<strong>in</strong>boldR, De La Fuente R, Wood J, Strauss JF III, Boiani M,Scholer HR, 2003: Derivation of oocytes from mouseembryonic stem cells. Science 300, 1251–1256.Iwasaki S, Campbell KH, Galli C, Akiyama K, 2000:Production of live calves derived from embryonic stem-likecells aggregated with tetraploid embryos. Biol Reprod 62,470–475.James D, Lev<strong>in</strong>e AJ, Besser D, Hemmati-Brivanlou A, 2005:TGFbeta ⁄ activ<strong>in</strong> ⁄ nodal signal<strong>in</strong>g is necessary for the ma<strong>in</strong>tenanceof pluripotency <strong>in</strong> human embryonic stem cells.Development 132, 1273–1282.Johnson J, Bagley J, Skaznik-Wikiel M, Lee HJ, Adams GB,Niikura Y, Tschudy KS, Tilly JC, Cortes ML, Forkert R,Spitzer T, Iacom<strong>in</strong>i J, Scadden DT, Tilly JL, 2005: Oocytegeneration <strong>in</strong> adult mammalian ovaries by putative germcells <strong>in</strong> bone marrow and peripheral blood. Cell 122, 303–315.Keefer CL, Pant D, Blomberg L, Talbot NC, 2007: Challengesand prospects for the establishment of embryonic stem celll<strong>in</strong>es of domesticated ungulates. Anim Reprod Sci 98, 147–168.Ko K, Scholer HR, 2006: Embryonic stem cells as a potentialsource of gametes. Sem<strong>in</strong> Reprod Med 24, 322–329.Kolbe T, Holtz W, 2000: Birth of a piglet derived from anoocyte fertilized by <strong>in</strong>tracytoplasmic sperm <strong>in</strong>jection (ICSI).Anim Reprod Sci 64, 97–101.Kuroiwa Y, Kas<strong>in</strong>athan P, Matsushita H, Sathiyaselan J,Sullivan EJ, Kakitani M, Tomizuka K, Ishida I, Robl JM,2004: Sequential target<strong>in</strong>g of the genes encod<strong>in</strong>g immunoglobul<strong>in</strong>-muand prion prote<strong>in</strong> <strong>in</strong> cattle. Nat Genet 36, 775–780.Lagut<strong>in</strong>a I, Lazzari G, Duchi R, Colleoni S, Ponderato N,Tur<strong>in</strong>i P, Crotti G, Galli C., 2005: Somatic cell nucleartransfer <strong>in</strong> horses: effect of oocyte morphology, embryoreconstruction method and donor cell type. <strong>Reproduction</strong>130, 559–567.Lagut<strong>in</strong>a I, Lazzari G, Galli C, 2006: Birth of cloned pigs fromzona-free nuclear transfer blastocysts developed <strong>in</strong> vitrobefore transfer. Clon<strong>in</strong>g Stem Cells 8, 283–293.Lazzari G, Galli C, Moor RM, 1992: Centrifugal elutriation ofporc<strong>in</strong>e oocytes isolated from the ovaries of newborn piglets.Anal Biochem 200, 31–35.Lazzari G, Wrenzycki C, Herrmann D, Duchi R, Kruip T,Niemann H, Galli C, 2002: Cellular and molecular deviations<strong>in</strong> bov<strong>in</strong>e <strong>in</strong> vitro-produced embryos are related to thelarge offspr<strong>in</strong>g syndrome. Biol Reprod 67, 767–775.Lazzari G, Colleoni S, Giannelli SG, Brunetti D, Colombo E,Lagut<strong>in</strong>a I, Galli C, Broccoli V, 2006: Direct derivation ofneural rosettes from cloned bov<strong>in</strong>e blastocysts: a model ofearly neurulation events and neural crest specification <strong>in</strong>vitro. Stem Cells 24, 2514–2521.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Embryo Biotechnologies <strong>in</strong> Farm <strong>Animals</strong> 7Lombardo A, Genovese P, Beausejour CM, Colleoni S, LeeYL, Kim KA, Ando D, Urnov FD, Galli C, Gregory PD,Holmes MC, Nald<strong>in</strong>i L, 2007: Gene edit<strong>in</strong>g <strong>in</strong> human stemcells us<strong>in</strong>g z<strong>in</strong>c f<strong>in</strong>ger nucleases and <strong>in</strong>tegrase-defectivelentiviral vector delivery. Nat Biotechnol 25, 1298–1306.Lonergan P, Fair T, 2008: In vitro-produced bov<strong>in</strong>e embryos:deal<strong>in</strong>g with the warts. Theriogenology 69, 17–22.Lu K, Polge C, 1992: A summary of two-year’s results <strong>in</strong> largescale <strong>in</strong> vitro bov<strong>in</strong>e embryo production. Proceed<strong>in</strong>gs of the12th International Congress on Animal <strong>Reproduction</strong>, TheHague, pp. 1315–1317.Meirow D, Levron J, Eldar-Geva T, Hardan I, Fridman E,Yem<strong>in</strong>i Z, Dor J, 2007: Monitor<strong>in</strong>g the ovaries afterautotransplantation of cryopreserved ovarian tissue: endocr<strong>in</strong>estudies, <strong>in</strong> vitro fertilization cycles, and live birth. FertilSteril 87, 418 e417–418 e415.Miyano T, Manabe N, 2007: Oocyte growth and acquisition ofmeiotic competence. Soc Reprod Fertil Suppl 63, 531–538.Moor RM, Trounson AO, 1977: Hormonal and follicularfactors affect<strong>in</strong>g maturation of sheep oocytes <strong>in</strong> vitro andtheir subsequent developmental capacity. J Reprod Fertil49, 101–109.Moor RM, Osborn JC, Cran DG, Walters DE, 1981: Selectiveeffect of gonadotroph<strong>in</strong>s on cell coupl<strong>in</strong>g, nuclear maturationand prote<strong>in</strong> synthesis <strong>in</strong> mammalian oocytes. J EmbryolExp Morphol 61, 347–365.Moor RM, Mattioli M, D<strong>in</strong>g J, Nagai T, 1990: Maturation ofpig oocytes <strong>in</strong> vivo and <strong>in</strong> vitro. J Reprod Fertil Suppl 40,197–210.Notarianni E, Galli C, Laurie S, Moor RM, Evans MJ, 1991:Derivation of pluripotent, embryonic cell l<strong>in</strong>es from the pigand sheep. J Reprod Fertil Suppl 43, 255–260.Ogawa K, Saito A, Matsui H, Suzuki H, Ohtsuka S,Shimosato D, Morishita Y, Watabe T, Niwa H, MiyazonoK, 2007: Activ<strong>in</strong>-Nodal signal<strong>in</strong>g is <strong>in</strong>volved <strong>in</strong> propagationof mouse embryonic stem cells. J Cell Sci 120, 55–65.Palermo G, Joris H, Devroey P, Van Steirteghem AC, 1992:Pregnancies after <strong>in</strong>tracytoplasmic <strong>in</strong>jection of s<strong>in</strong>gle spermatozoon<strong>in</strong>to an oocyte. Lancet 340, 17–18.Palmer E, Bezard J, Magistr<strong>in</strong>i M, Duchamp G, 1991: In vitrofertilization <strong>in</strong> the horse. A retrospective study. J ReprodFertil Suppl 44, 375–384.Parrish JJ, Susko-Parrish JL, Leibfried-Rutledge ML, CritserES, Eyestone WH, First NL, 1986: Bov<strong>in</strong>e <strong>in</strong> vitro fertilizationwith frozen-thawed semen. Theriogenology 25, 591–600.Pieterse MC, Vos PL, Kruip TA, Wurth YA, van Beneden TH,Willemse AH, Taverne MA, 1991: Transvag<strong>in</strong>al ultrasoundguided follicular aspiration of bov<strong>in</strong>e oocytes. Theriogenology35, 857–862.Ponderato N, Lagut<strong>in</strong>a I, Crotti G, Tur<strong>in</strong>i P, Galli C, LazzariG, 2001: Bov<strong>in</strong>e oocytes treated prior to <strong>in</strong> vitro maturationwith a comb<strong>in</strong>ation of butyrolactone I and roscovit<strong>in</strong>e atlow doses ma<strong>in</strong>ta<strong>in</strong> a normal developmental capacity. MolReprod Dev 60, 579–585.Ponderato N, Crotti G, Tur<strong>in</strong>i P, Duchi R, Galli C, Lazzari G,2002: Embryonic and foetal development of bov<strong>in</strong>e oocytestreated with a comb<strong>in</strong>ation of butyrolactone I and roscovit<strong>in</strong>e<strong>in</strong> an enriched medium prior to IVM and IVF. MolReprod Dev 62, 513–518.Roy SK, Greenwald GS, 1985: An enzymatic method fordissociation of <strong>in</strong>tact follicles from the hamster ovary:histological and quantitative aspects. Biol Reprod 32, 203–215.Sasaki H, Matsui Y, 2008: Epigenetic events <strong>in</strong> mammaliangerm-cell development: reprogramm<strong>in</strong>g and beyond. NatRev Genet 9, 129–140.Seidel GE Jr, 2003: Sex<strong>in</strong>g mammalian sperm – <strong>in</strong>tertw<strong>in</strong><strong>in</strong>g ofcommerce, technology, and biology. Anim Reprod Sci 79,145–156.Staigmiller R, Moor R, 1984: Effect of follicle cells on thematuration and developmental competence of ov<strong>in</strong>e oocytesmatured outside the follicle. Gamete Res 9, 221–229.Takahashi K, Yamanaka S, 2006: Induction of pluripotentstem cells from mouse embryonic and adult fibroblastcultures by def<strong>in</strong>ed factors. Cell 126, 663–676.Tervit HR, Whitt<strong>in</strong>gham DG, Rowson LE, 1972: Successfulculture <strong>in</strong> vitro of sheep and cattle ova. J Reprod Fertil 30,493–497.Tesar PJ, Chenoweth JG, Brook FA, Davies TJ, Evans EP,Mack DL, Gardner RL, McKay RD, 2007: New cell l<strong>in</strong>esfrom mouse epiblast share def<strong>in</strong><strong>in</strong>g features with humanembryonic stem cells. Nature 448, 196–199.Vallier L, Alexander M, Pedersen RA, 2005: Activ<strong>in</strong> ⁄ Nodaland FGF pathways cooperate to ma<strong>in</strong>ta<strong>in</strong> pluripotency ofhuman embryonic stem cells. J Cell Sci 118, 4495–4509.van Wagtendonk-de Leeuw AM, Mullaart E, de Roos AP,Merton JS, den Daas JH, Kemp B, de Ruigh L, 2000: Effectsof different reproduction techniques: AI MOET or IVP, onhealth and welfare of bov<strong>in</strong>e offspr<strong>in</strong>g. Theriogenology 53,575–597.Wakayama T, Tabar V, Rodriguez I, Perry AC, Studer L,Mombaerts P, 2001: Differentiation of embryonic stem celll<strong>in</strong>es generated from adult somatic cells by nuclear transfer.Science 292, 740–743.Wall RJ, Powell AM, Paape MJ, Kerr DE, Bannerman DD,Pursel VG, Wells KD, Talbot N, Hawk HW, 2005:Genetically enhanced cows resist <strong>in</strong>tramammary Staphylococcusaureus <strong>in</strong>fection. Nat Biotechnol 23, 445–451.Willadsen SM, 1986: Nuclear transplantation <strong>in</strong> sheepembryos. Nature 320, 63–65.Wilmut I, Schnieke AE, McWhir J, K<strong>in</strong>d AJ, Campbell KH,1997: Viable offspr<strong>in</strong>g derived from fetal and adult mammaliancells. Nature 385, 810–813.Xu KP, Greve T, Callesen H, Hyttel P, 1987: Pregnancyresult<strong>in</strong>g from cattle oocytes matured and fertilized <strong>in</strong> vitro.J Reprod Fertil 81, 501–504.Yanagimachi R, Chang MC, 1964: In Vitro fertilization ofGolden Hamster Ova. J Exp Zool 156, 361–375.Y<strong>in</strong>g QL, Nichols J, Chambers I, Smith A, 2003a: BMP<strong>in</strong>duction of Id prote<strong>in</strong>s suppresses differentiation andsusta<strong>in</strong>s embryonic stem cell self-renewal <strong>in</strong> collaborationwith STAT3. Cell 115, 281–292.Y<strong>in</strong>g QL, Stavridis M, Griffiths D, Li M, Smith A, 2003b:Conversion of embryonic stem cells <strong>in</strong>to neuroectodermalprecursors <strong>in</strong> adherent monoculture. Nat Biotechnol 21,183–186.Young LE, S<strong>in</strong>clair KD, Wilmut I, 1998: Large offspr<strong>in</strong>gsyndrome <strong>in</strong> cattle and sheep. Rev Reprod 3, 155–163.Zhang JJ, Boyle MS, Allen WR, Galli C, 1989: Recent studieson <strong>in</strong> vivo fertilization of <strong>in</strong> vitro matured horse oocytes.Equ<strong>in</strong>e Veter<strong>in</strong> J 8, 101–104.Author’s address (for correspondence): C Galli, Laboratorio diTecnologie della, Riproduzione, Via Porcellasco 7 ⁄ f, 26100 Cremona,Italy. E-mail: cesaregalli@ltrciz.itConflict of <strong>in</strong>terest: All authors declare no conflict of <strong>in</strong>terests.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 8–14 (2008); doi: 10.1111/j.1439-0531.2008.01137.xISSN 0936-6768Towards Ethically Improved Animal Experimentation <strong>in</strong> the Study of Animal<strong>Reproduction</strong>D Blache 1 , GB Mart<strong>in</strong> 1 and SK Maloney 21 UWA Institute of Agriculture M082, The University of Western Australia; 2 Physiology-Biomedical, Biomolecular and Chemical Sciences,The University of Western Australia, Crawley, WA, AustraliaContentsThe ethics of animal-based research is a cont<strong>in</strong>u<strong>in</strong>g area ofdebate, but ethical research protocols do not prevent scientificprogress. In this paper, we argue that our current knowledge ofthe factors that affect reproductive processes providesresearchers with a solid foundation upon which they canconduct more ethical research and simultaneously producedata of higher quality. We support this argument by show<strong>in</strong>ghow a deep understand<strong>in</strong>g of the genetics, nutrition andtemperament of our experimental animals can improve compliancewith two of the ‘3 Rs’, reduction and ref<strong>in</strong>ement,simply by offer<strong>in</strong>g better control over the variance <strong>in</strong> ourexperimental model. The outcome is a better experimentaldesign, on both ethical and scientific grounds.Introduction<strong>Reproduction</strong> has a long history as a field of researchand, if anyth<strong>in</strong>g, there is actually an acceleration of<strong>in</strong>terest <strong>in</strong> the ma<strong>in</strong> goals of better reproductivetechnology and better fundamental understand<strong>in</strong>g ofthe biological processes. There is little to dist<strong>in</strong>guishbetween these goals because reproductive technologiesare frequently essential as tools for basic research, andbasic research underp<strong>in</strong>s reproductive technology. Indeal<strong>in</strong>g with the topic of this paper, we will illustrate ourarguments primarily with examples for farmed rum<strong>in</strong>ants,particularly sheep. We will not dwell on dist<strong>in</strong>ctionsbetween applied and fundamental research despitethe focus on production animals.Over the last 200 years, we have seen an <strong>in</strong>crease <strong>in</strong>the concerns of society about the ethics of animal-basedresearch, particularly the potential suffer<strong>in</strong>g of experimentalanimals. There has been <strong>in</strong>tense discussion of theethical dilemmas aris<strong>in</strong>g generally from the use ofanimals and this has led to a utilitarian framework,the cost ⁄ benefit analysis, for ethical assessment anddecision-mak<strong>in</strong>g. Scientists have no trouble argu<strong>in</strong>g thebenefits of animal experimentation and augment<strong>in</strong>g thedenom<strong>in</strong>ator of a cost ⁄ benefit analysis. However froman animal welfare viewpo<strong>in</strong>t, the best way to decreasethe cost ⁄ benefit ratio is to decrease the numerator, i.e. todecrease the cost of experimentation to the subjectanimals.In this paper, we are not aim<strong>in</strong>g to provide a recipefor improv<strong>in</strong>g the welfare of animals used <strong>in</strong> research,but to show that ‘ethical science is good science’(Somerville 2007). We will argue that, <strong>in</strong> the field ofreproduction, our current knowledge of the factors thataffect reproductive processes provides researchers with asolid foundation upon which they can comply withethical guidel<strong>in</strong>es and simultaneously produce data ofhigher quality. This paper is therefore not a manifestofor animal rights or animal liberation. We will onlydiscuss ethics and how to be ethical whilst do<strong>in</strong>g betterscience. We will beg<strong>in</strong> by def<strong>in</strong><strong>in</strong>g ethics and discuss<strong>in</strong>gwhy it has become more important and relevant <strong>in</strong>animal research. We will briefly review the ‘Pr<strong>in</strong>ciple of3 Rs’ and how it should be applied to research <strong>in</strong>reproduction, for which we will need to summarize ourknowledge of the complex of processes that control thereproductive system. F<strong>in</strong>ally, we will describe thepotential for improv<strong>in</strong>g the ethics of research <strong>in</strong> reproductionby consider<strong>in</strong>g the models used, particularly thegenetics, nutrition and temperament of the experimentalanimals. We will illustrate most of our po<strong>in</strong>ts us<strong>in</strong>g<strong>in</strong>formation about sheep because it is the focus of ourstudies, but the pr<strong>in</strong>ciples are broad so can be applied tomost species.Ethics and Animal-based Research?Everyday we make decisions about what we are will<strong>in</strong>gor not will<strong>in</strong>g to do. Most of such decisions are purelypersonal <strong>in</strong> that they do not impact on others. When ourdecisions impact on others, they are reasoned andassessed, at least <strong>in</strong> part, on moral grounds (LaFolette2007). When we plan experiments us<strong>in</strong>g animals, ourdecisions cannot be based on personal choice because, asa society, we have decided that animals are not objects,so we need to engage <strong>in</strong> moral debate to decide whetheran experiment is worth do<strong>in</strong>g. Ethics is a philosophicalprocess <strong>in</strong>volv<strong>in</strong>g critical evaluation of our actions andleads to moral truth (morally right or wrong; Bryantet al. 2005; Mepham 2005), so it deals with the moraldimension of decision-mak<strong>in</strong>g.A major po<strong>in</strong>t to consider with respect to ethics is thatmoral decisions are taken <strong>in</strong> reference to ethical theories.Why? Because ethical theories, such as consequentialismor deontology, provide ground rules for th<strong>in</strong>k<strong>in</strong>g aboutmoral issues. Ethical theory, like any theory, provides aframework for analys<strong>in</strong>g a question or fact critically.Animal research is ma<strong>in</strong>ly discussed <strong>in</strong> the context of theutilitarian theory, deontology be<strong>in</strong>g only used sparsely.The utilitarian theory, <strong>in</strong> short, leads to an assessment ofthe ethical value of a given experiment based oncost ⁄ benefit analysis (Vallentyne 2007). It is not alwayseasy to estimate accurately both the cost and the benefit,especially when consider<strong>in</strong>g experiments outside thelaboratory or with long-term consequences (Schmidtz2007). However, the benefits of animal experimentationcan be, for example, societal (medical breakthrough),scientific (new knowledge) or educational (tra<strong>in</strong><strong>in</strong>g ofÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Ethical Models for Study<strong>in</strong>g <strong>Reproduction</strong> 9research students), and they may even benefit theanimals themselves (new cures, better managementpractices). The cost side of the equation <strong>in</strong>cludesimpositions made on animals for the purpose of theexperimentation and can <strong>in</strong>clude the pa<strong>in</strong> of procedures,lack of environmental stimulation (e.g. lack of contactwith conspecifics), or food restriction. F<strong>in</strong>al assessmentof the cost–benefit ratio is fairly subjective becausesuffer<strong>in</strong>g is not easily quantified and the benefits areusually a ‘best-guess’. However, any attempt to decreasethe cost to the animals for a given benefit will <strong>in</strong>creasethe ethical value of an experiment. A good way todecrease the cost is to adopt more ‘humane’ approachesto animal experimentation. In this context, the ‘Pr<strong>in</strong>cipleof 3 Rs’ (3Rs) orig<strong>in</strong>ally proposed by Charles Hume, thefounder of the University Federation for AnimalWelfare (UFAW) <strong>in</strong> 1954, and later def<strong>in</strong>ed <strong>in</strong> greatdetail <strong>in</strong> the sem<strong>in</strong>al book by Russell and Burch (1959),a valuable resource. In brief, the 3 Rs’ areReplacement – substitution of <strong>in</strong>sentient material forconscious animals;Reduction – get the same amount of <strong>in</strong>formation fromfewer animals;Ref<strong>in</strong>ement – decrease the imposition on the animalsused <strong>in</strong> the research.The 3Rs is widely used by animal ethics committeesaround the world to assess animal-based research,<strong>in</strong>clud<strong>in</strong>g experimentation <strong>in</strong> the field of reproductivebiology. Replacement is seen as the ultimate ethicalsolution, but that does not mean that cell culture andmathematical models should replace live animal experimentation.Many theories can ultimately only be tested<strong>in</strong> whole organisms, as is very well demonstrated <strong>in</strong>research for drug discovery, even if some drugs tested onanimals turn out to be dangerous to humans (Fest<strong>in</strong>gand Altman 2002). In fact, cell l<strong>in</strong>es, such as immortalizedGnRH neurons, have been <strong>in</strong>tensively used to studythe regulation of the reproductive function (Wetsel1995).Knowledge <strong>in</strong> Reproductive Physiology: A Curseor a Bless<strong>in</strong>g?Regardless of the level of the <strong>in</strong>vestigation of thereproductive system, from cell physiology to behaviour,it is important for the researcher to be cognizant of themultiplicity of factors that <strong>in</strong>fluence the reproductiveprocess, as illustrated <strong>in</strong> Fig. 1. The complexity of the<strong>in</strong>puts is both a curse because perfect experimentaldesign requires the control of all of these factors, and abless<strong>in</strong>g because a deep knowledge of how these factors<strong>in</strong>fluence experimental outcomes gives us a strong graspof the factors that we need to control <strong>in</strong> our experimentaldesign. In account<strong>in</strong>g for all factors that affectreproduction, we account for more variability and thepower and precision of our experiments is <strong>in</strong>creased,improv<strong>in</strong>g the ethical outcomes of ref<strong>in</strong>ement andreduction (Howard 2002). Some factors are easier tocontrol than others, especially when large groups ofanimals are required. Importantly, many of these factorsare not <strong>in</strong>dependent so <strong>in</strong>teractions among them (e.g.nutrition, photoperiod and genotype; Blache et al. 2006,2007) can lead to complex outcomes but, because theyPhotoperiodicsignalsP<strong>in</strong>ealmelaton<strong>in</strong>GenotypeAnnualrhythmgeneratorOestrogenAromataseNegativefeedback‘Emotional reactivity’AppetitePhotoperiod-driven filterNutritionalsignalsMetabolicSensor“Pulse generator”GnRH pulsesLH, FSHSex steroidsSexual behaviourSocio-sexualsignalsOestrogenAromataseNegativefeedbackFig. 1. A schema describ<strong>in</strong>g the proposed relationships betweenemotional reactivity and external <strong>in</strong>puts (photoperiod, nutrition andsocial signals) and the ways that they <strong>in</strong>teract with genotype andsteroid feedback <strong>in</strong> the control of hypothalamo–pituitary–testicularaxis <strong>in</strong> the male sheep. The <strong>in</strong>teractions <strong>in</strong>dicated with broken l<strong>in</strong>es areeither hypothetical or demonstrated (see text) but the nature of thesepathways is not fully understood <strong>in</strong> rum<strong>in</strong>ants. Redrawn after Blacheet al. (2007)have been extensively studied, we have excellent opportunitiesto apply the 3Rs. Below, we will illustrate thispo<strong>in</strong>t by show<strong>in</strong>g how controll<strong>in</strong>g for genotype, nutritionand temperament can lead to more ethical animalexperimentation.Genetics and ethical experimentationDur<strong>in</strong>g the last decade, there has been a dramatic<strong>in</strong>crease <strong>in</strong> the use of genetically modified animals, suchas conventional <strong>in</strong>bred l<strong>in</strong>es, or l<strong>in</strong>es carry<strong>in</strong>g deletedgenes (knock-out), additional genes (knock-<strong>in</strong>), or overexpressedgenes (transgenic). In the UK, the number ofgenetically modified animals used <strong>in</strong> research more thanquadrupled between 1995 and 2006 and are they nowused <strong>in</strong> 34% of all procedures (Home Office 2007). Theyare considered better experimental models than theirwild-type counterparts because they exhibit less variabilityand ⁄ or they have a modification that allowsexperiments to target a specific function, gene ormolecule (Fest<strong>in</strong>g 1990; Fest<strong>in</strong>g and Altman 2002). Inreproduction, the knock-out rodent models have confirmedand <strong>in</strong>formed our understand<strong>in</strong>g of, for example,the role of sex steroid hormones <strong>in</strong> many of the stages ofthe reproductive process. This has been clearly demonstratedfor the oestrogen receptor <strong>in</strong> females and theandrogen receptor <strong>in</strong> males (De Gendt et al. 2004;Hewitt et al. 2005). Similarly, animals with an eng<strong>in</strong>eereddeficiency <strong>in</strong> FSH and LH production, or theirreceptors, or <strong>in</strong> metabolic hormone receptors, have alsocontributed to advances <strong>in</strong> our knowledge of reproductiveprocesses and how those processes <strong>in</strong>teract withother functions and systems (Kumar 2005). However,these models cannot completely substitute for ‘genetically<strong>in</strong>tact’ animals because of the plasticity,Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


10 D Blache, GB Mart<strong>in</strong> and SK Maloneyadaptability and redundancy of physiological systems,and because most systems are polygenic. In addition, <strong>in</strong>ethical terms, the creation of knock-outs is costly for theanimals themselves because a proportion of geneticallymodified animals will not be viable and will have to bedestroyed before be<strong>in</strong>g part of an experiment (NuffieldCouncil on Bioethics 2005; NHMRC Animal WelfareCommittee 2007). Estimat<strong>in</strong>g the relative ratio betweencost and benefit for the use of genetically modifiedanimals is not an easy task and, to our knowledge, hasnot been addressed, as recognized <strong>in</strong> a recent reviewabout the use of transgenic farm animals to improveproduction (Bacci 2007).A different strategy for reduc<strong>in</strong>g <strong>in</strong>ter-<strong>in</strong>dividualvariability, and thus address<strong>in</strong>g ref<strong>in</strong>ement and reduction,is the use of monozygotic tw<strong>in</strong>s (Biggers 1986).Tw<strong>in</strong> studies have been used successfully <strong>in</strong> fields ofresearch such as health and immunology, but it does notseem to be as advantageous <strong>in</strong> reproductive biology. Insheep, for example, we have studied gonadotroph<strong>in</strong>responses follow<strong>in</strong>g an <strong>in</strong>crease <strong>in</strong> nutrition, ovarianresponses to exogenous <strong>in</strong>hib<strong>in</strong> and the responses ofhypothalamo–pituitary axis to an opioid. For mostreproductive traits (ovulation rate, secretion of gonadotroph<strong>in</strong>sand sex steroids), randomly selected animalswere just as efficient for experimentation as geneticallyidentical tw<strong>in</strong>s (Celi et al. 2007). Monozygotic tw<strong>in</strong>sshowed an advantage only for live weight and scrotalcircumference. In addition, the reproductive biotechnologiesthemselves need to address ethical questionsconcern<strong>in</strong>g the overall imposition on animals neededto ga<strong>in</strong> the result (Church 1988). Aga<strong>in</strong>, there is nodirect estimate of the reduction <strong>in</strong> the use of animalsfollow<strong>in</strong>g clon<strong>in</strong>g. On the contrary, the variancebetween clones <strong>in</strong> both behavioural and physiologicalparameters can be <strong>in</strong>creased because of epigenetic andearly-life effects (Seidel 2001; Archer et al. 2003a,b).Nutritional <strong>in</strong>put <strong>in</strong>to reproduction and ethicalexperimentationThe relationship between the level of nutrition or, moregenerally, metabolic status and reproductive capacity isevident for all stages <strong>in</strong> the reproductive process:puberty, gamete production, conception and the survivalof embryos, fetuses and newborn (review: Rob<strong>in</strong>sonet al. 2006). The responses can be rapid, with anacute <strong>in</strong>crease <strong>in</strong> nutrient supply <strong>in</strong>duc<strong>in</strong>g multipleovulation <strong>in</strong> females (V<strong>in</strong>oles et al. 2005) and an<strong>in</strong>crease <strong>in</strong> GnRH output <strong>in</strong> adult males (Zhang et al.2004). The responses can also be delayed <strong>in</strong> time, such asthe effect of peri-conception under-nutrition on thelength of gestation (Bloomfield et al. 2003). In addition,nutrition of the mother can affect reproductive processes<strong>in</strong> her offspr<strong>in</strong>g (‘fetal programm<strong>in</strong>g’), such as thenumber of Sertoli cells (Bielli et al. 2002) and length ofgestation and ovulation rate (Rae et al. 2001). Foetalprogramm<strong>in</strong>g is a relatively new field of <strong>in</strong>vestigationand many of the impacts of nutritional imbalance <strong>in</strong>utero on the reproductive physiology of the offspr<strong>in</strong>g arenot yet known (Rh<strong>in</strong>d 2004). Thus, their potentialimpact on experimental design <strong>in</strong>, for example, a studydone with randomly selected animals, is currentlyimpossible to assess, although it is most likely that they<strong>in</strong>crease between-animal variation.Therefore, if we are to ensure that the <strong>in</strong>dividuals used<strong>in</strong> an experiment are homogenous, we need to considerthe metabolic status of not only the experimentalanimals but also their ancestors. Obviously, this phenomenonmight expla<strong>in</strong> discrepancies among the outcomesof comparable experiments. We have beenconfronted with this problem <strong>in</strong> our own research <strong>in</strong>tothe bra<strong>in</strong> sites where steroids exert negative feedback onGnRH pulse frequency <strong>in</strong> mature Mer<strong>in</strong>o rams (Blacheet al. 1997). Dur<strong>in</strong>g a prelim<strong>in</strong>ary trial, done <strong>in</strong> WesternAustralia, implantation of oestradiol <strong>in</strong>to the ventromedialnucleus of the hypothalamus decreased thefrequency of LH pulses. We then decided to repeat theexperiment with a larger number animals for statisticalvalidity, but <strong>in</strong> our collaborator’s laboratory <strong>in</strong> easternAustralia. The same treatment <strong>in</strong> the same bra<strong>in</strong>location had no effect. A few months after this apparentfailure, we repeated the experiment a third time <strong>in</strong> thewest, and obta<strong>in</strong>ed the same results as <strong>in</strong> the first study.The contrary results could not be attributed to photoperiodor genotype. The answer came from anotherstudy, done at the same time, <strong>in</strong> which we demonstratedthat <strong>in</strong>sul<strong>in</strong> was a very powerful stimulator of GnRHneuronal activity (Miller et al. 1995). When we measured<strong>in</strong>sul<strong>in</strong> <strong>in</strong> samples from the steroid implantationstudies, we discovered that the concentration was aboutthree times higher <strong>in</strong> the eastern rams (31.4 ±1.4 ng ⁄ ml) than <strong>in</strong> the western rams (9.1 ± 0.6 ng ⁄ ml).This was not due to differences <strong>in</strong> their nutrition dur<strong>in</strong>gthe experiments, but because the eastern rams had abetter body condition (fatter) than the western ramsbefore the start of the experiment. We have thusconcluded, a posteriori, that the pre-experimental,nutritional history of the animals, and thus their tonic<strong>in</strong>sul<strong>in</strong> concentrations, prevented the oestradiol implantfrom <strong>in</strong>hibit<strong>in</strong>g GnRH secretion <strong>in</strong> the eastern experiment.Thus, extensive knowledge of the history of theanimals can decrease the probability of conflict<strong>in</strong>gexperimental outcomes. As we learn more about, forexample, the epigenetic effects of nutrition (Rh<strong>in</strong>d 2004),this situation will become even more complex. Clearly,this is a particularly difficult issue for research done withfarm animals or wildlife.Animal emotion and ethical experimentationA major potential imposition on experimental animals is‘stress’ associated with the experimental protocol. Stressis an <strong>in</strong>tegral part of everyday life for all animals, withthe challenges of psychological or physical stressorsevok<strong>in</strong>g the normal adaptive responses that ma<strong>in</strong>ta<strong>in</strong>homeostasis (Matteri et al. 1984). However, these sameresponses can not only <strong>in</strong>terfere with an experimentaloutcome but also weigh heavily on the ‘cost’ side of thecost-benefit ratio. Thus, welfare can be improved byreduc<strong>in</strong>g any stress caused by experimental conditions,such as hous<strong>in</strong>g or the <strong>in</strong>teractions between experimentalanimals and human experimenters. Our aim shouldbe to <strong>in</strong>crease the freedom from fear and distress. This isvery relevant to studies <strong>in</strong> reproduction becausethe reproductive endocr<strong>in</strong>e axis can be profoundlyÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Ethical Models for Study<strong>in</strong>g <strong>Reproduction</strong> 11<strong>in</strong>fluenced by stress, as has been clearly demonstratedfor farm animals (e.g. Mart<strong>in</strong> et al. 1981; Caraty et al.1997; von Borell et al. 2007).If reduced stress <strong>in</strong> experimentalanimals can improve the cost ⁄ benefit ratio, thenit might be useful to select experimental animals on thebasis of their temperament or fear reaction (Mills et al.1997).What is temperament and how is it assessed?Temperament has been def<strong>in</strong>ed for humans as ‘naturecontroll<strong>in</strong>g the way he behaves, feels and th<strong>in</strong>ks’(Cannon 1927). For animals, many attempts have beenmade to apply similar criteria on the basis of ourperceptions of their ‘temperament’. We cannot knowwhat is tak<strong>in</strong>g place <strong>in</strong> the m<strong>in</strong>d of an animal, so weoften make assumptions about their ‘temperament’based on behavioural and neuroendocr<strong>in</strong>e characteristics.Boissy (1995) concluded that emotional reactivityhas a significant impact on the relationship of an animalto its environment and that non-human animals havepersonality, temperament or <strong>in</strong>dividual behaviours,amongst which fearfulness plays an important role <strong>in</strong>an <strong>in</strong>dividual’s behavioural response to threaten<strong>in</strong>gsituations. Temperament has also been def<strong>in</strong>ed byMurphy (1999) as the emotivity of ‘fearfulness andreactivity of an animal <strong>in</strong> response to humans andstrange, novel or threaten<strong>in</strong>g environments’. Measuresof temperament, or fearfulness, <strong>in</strong> domesticated speciesare thus far restricted mostly to sheep, cattle, chickensand horses, and have been designed on the basis of theabove two def<strong>in</strong>itions (Forkman et al. 2007). For cattle,such assessments have <strong>in</strong>cluded subjective measuressuch as observations of crush behaviour or objectivemeasures such as flight time. (Vois<strong>in</strong>et et al. 1997; Fellet al. 1999). For sheep, we have used a comb<strong>in</strong>ation oftwo objective behavioural tests, the ‘arena test’ and the‘box test’ (Murphy et al. 1994). Briefly, the ‘arena test’ isa motivational choice test that measures approach ⁄avoidance behaviour to humans (Murphy 1999). The‘box test’ is similar to isolation tests used <strong>in</strong> sheep andcattle (Cockram et al. 1994; Grignard et al. 2000) andprovides an objective measure of the degree of anxietyassociated with isolation. At the University of WesternAustralia, we have designed a selection <strong>in</strong>dex us<strong>in</strong>g bothtests and then genetically selected two l<strong>in</strong>es of sheepus<strong>in</strong>g males from the extremes of the variation. With<strong>in</strong>this framework, temperament has proven to be moderatelyheritable (h 2 0.3) and thus responds favourablyto selection (Murphy 1999) so, after more than 15generations, we now have ‘calm’ and ‘nervous’ l<strong>in</strong>es.How can selection for temperament improve ourexperiments on reproduction?In mammals, temperament, or emotional reactivity ortameness, affects reproductive capacity and reproductivephysiology, either directly or <strong>in</strong>directly (Price 2002). Insheep, direct effects of temperament have been demonstrated<strong>in</strong> the early stages of gestation and on thesurvival of newborn lambs. Early studies <strong>in</strong> our laboratoryshowed that calm ewes were better mothers thannervous ewes (Murphy et al. 1994) because they spendmore time with their lamb(s) and, when disturbed, havea shorter flight distance and return to their lamb(s)sooner. Consequently, the <strong>in</strong>cidence of lamb mortality,birth to wean<strong>in</strong>g, was about half for calm ewescompared to nervous ewes, and <strong>in</strong>dependent of thephysical status of the lambs (Murphy et al. 1994;Murphy 1999). Fecundity, and perhaps embryo survival,are higher <strong>in</strong> calm than <strong>in</strong> nervous ewes (Hartet al. 2008). Ewes of calm temperament also carriedmore tw<strong>in</strong> embryos than nervous ewes (1.39 embryos,n = 472 : 1.29 embryos, n = 302; p < 0.001). Selectionfor calm temperament also modifies the expressionof reproductive behaviour, lead<strong>in</strong>g to an <strong>in</strong>crease <strong>in</strong>proceptivity <strong>in</strong> maiden ewes (Gelez et al. 2003).Temperament also has <strong>in</strong>direct effects on the outcomes<strong>in</strong> other types of experimentation, <strong>in</strong>clud<strong>in</strong>gstudies with metabolic factors such as lept<strong>in</strong>, a hormone<strong>in</strong>volved <strong>in</strong> the regulation of metabolism and thesignall<strong>in</strong>g of the metabolic status to the reproductivesystem (Blache et al. 2007). Because lept<strong>in</strong> is also acytok<strong>in</strong>e and thus <strong>in</strong>volved <strong>in</strong> immune signall<strong>in</strong>g, wedesigned an experiment to study the lept<strong>in</strong> response toan immune challenge (Fig. 2). A response was observedonly <strong>in</strong> sheep that had been selected for nervousness.When the data from both calm and nervous sheep werecomb<strong>in</strong>ed, the response to the immune challenge wasnot significant, presumably because the temperamentvariance was now <strong>in</strong>corporated <strong>in</strong>to residual (background)variance, reduc<strong>in</strong>g the power of the analysis.This example illustrates perfectly how selection fortemperament could improve the level of ref<strong>in</strong>ement andreduction <strong>in</strong> an animal model. M<strong>in</strong>imiz<strong>in</strong>g temperamentdifferences thus can reduce the number of animals usedChange <strong>in</strong> plasmalept<strong>in</strong> (%)(a) LPS <strong>in</strong>jected at 0 h (b) Vehicle <strong>in</strong>jected at 0 h (c) Calm plus Nervous140130120110100900 1 2 3 4 5 6 0 1 2 3 4 5 6 0 1 2 3 4 5 6Time (h)Time (h)Time (h)Fig. 2. Change <strong>in</strong> plasma concentrations of lept<strong>in</strong> <strong>in</strong> sheep selected for calm (closed circles) or nervous temperament (closed squares) (a) dur<strong>in</strong>gfever <strong>in</strong>duced by <strong>in</strong>jection with lipopolysaccharide (0.4 lg ⁄ kg) or (b) after <strong>in</strong>jection with vehicle. The black bar <strong>in</strong>dicates the period dur<strong>in</strong>g whichthe selected l<strong>in</strong>es differed (p < 0.05). In the composite graph (c), calm and nervous animals have been pooled and there was no difference betweenvehicle (open circles) and <strong>in</strong>duced fever (closed circles) (Blache and Maloney, unpublished data)Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


12 D Blache, GB Mart<strong>in</strong> and SK MaloneyVoluntary feed<strong>in</strong>takeMetabolic fuelEnergy reserveEmotional reactivityHPA axisMetabolicHormones:lept<strong>in</strong>, orex<strong>in</strong>s<strong>in</strong>sul<strong>in</strong>,IGF-1, GH,T3, T4ThermogenesisImmune functionCellular ma<strong>in</strong>tenanceGrowth, locomotion<strong>Reproduction</strong>Fig. 3. Central role of metabolic hormones <strong>in</strong> the <strong>in</strong>teractions amongemotion, reproductive function, immune function, metabolic status,thermogenesis and other bodily functions. The sympathetic pathwayshave been omitted for clarityand limit the risk of conflict<strong>in</strong>g results s<strong>in</strong>ce, bydef<strong>in</strong>ition, a group of animals show<strong>in</strong>g a homogenousresponse to any stimulus will have less variability <strong>in</strong> anyparameter associated with that response.As described previously, the amount of availablemetabolic fuel needs to be ‘assessed’ by a control systemand then expenditure ‘directed’ towards reproductivefunction if and when required. For all systems, theassessment of energy levels and their partition<strong>in</strong>g isunder the control of metabolic hormones (Fig. 3).Thyroid concentrations were higher <strong>in</strong> a sub-populationof ‘calm’ than <strong>in</strong> a sub-population of ‘nervous’ animals<strong>in</strong> our selection l<strong>in</strong>es (Blache et al. 2002). We have alsoshown that the cortisol response to a novel object ishigher <strong>in</strong> nervous ewes than <strong>in</strong> calm ewes (Bickell et al.2008). These considerations apply equally to stressassociated with experimental protocols (Mart<strong>in</strong> et al.1981; Adams et al. 1993). The effects of stress andcortisol on the reproductive axis are numerous anddiverse – <strong>in</strong> the female, they can either <strong>in</strong>hibit orstimulate ovulation (Dobson and Smith 2000). Theeffect of temperament genetics on reproductive processeslikely <strong>in</strong>volves hormonal pathways. These l<strong>in</strong>ksare still to be demonstrated experimentally, but temperamentaffects the hormonal systems <strong>in</strong>volved <strong>in</strong> thecontrol of energy partition<strong>in</strong>g and this may be how it<strong>in</strong>fluences the outcomes of experimentation target<strong>in</strong>g therelationship between nutrition and reproduction.Aside from improv<strong>in</strong>g the experimental design andoutcomes, the welfare of experimental animals could beimproved by selection for temperament. Less reactiveanimals will have an improved freedom from fear anddistress s<strong>in</strong>ce animals that are less reactive to novelty willpresent reduced physiological and behavioural reactionsto novel experiences (Korte 2001), <strong>in</strong>clud<strong>in</strong>g exposure toa research environment.ConclusionIn <strong>in</strong>ference science, hypothesis test<strong>in</strong>g <strong>in</strong>volves manipulat<strong>in</strong>gone or a few parameters, while other factors thatcould <strong>in</strong>fluence the dependent parameters are controlled.In this paper, we have shown that multiplefactors that affect and control reproduction could affectthe outcomes of animal experimentation because theyaffect reproductive physiology, over both short and longtime scales, and even across generations. These factorsneed to be controlled. In addition, emotional reactivityhas the potential to alter experimental outcomes, just asgenotype can affect the outcomes of experimentation <strong>in</strong>which photoperiod or nutrition is manipulated. Byconsider<strong>in</strong>g the nutritional history and emotional characteristicsof the <strong>in</strong>dividual subjects, the variabilitywith<strong>in</strong> experimental groups can be reduced and two ofthe 3Rs (reduction, ref<strong>in</strong>ement) would be applied,lead<strong>in</strong>g to more ethical experimentation. This creates aw<strong>in</strong>–w<strong>in</strong> situation for researchers and experimentalanimals because more ethical science results <strong>in</strong> betterscience.AcknowledgementsWe would like to thank the generous assistance of the students andstaff of Animal Science (University of WA). Fund<strong>in</strong>g was supplied bythe National Health and Medical Research Council, the AustralianResearch Council, the Ada Bartholomew Medical Research Trust, andthe University of Western Australia.ReferencesAdams NR, Atk<strong>in</strong>son S, Mart<strong>in</strong> GB, Briegel JR, Boukhliq R,Sanders MR, 1993: Frequent blood sampl<strong>in</strong>g changes theplasma concentration of LH and FSH and the ovulationrate <strong>in</strong> Mer<strong>in</strong>o ewes. J Reprod Fertil 99, 689–694.Archer GS, D<strong>in</strong>dot S, Friend TH, Walker S, Zaunbrecher G,Lawhorn B, Piedrahita J, 2003a: Hierarchical phenotypicand epigenetic variation <strong>in</strong> cloned sw<strong>in</strong>e. Biol Reprod 69,430–436.Archer GS, Friend TH, Piedrahita J, Nevill CH, Walker S,2003b: Behavioural variation among cloned pigs. ApplAnimal Behav Sci 82, 151–161.Bacci ML, 2007: A brief overview of transgenic farm animals.Veter<strong>in</strong> Res Commun 31(Suppl. 1), 9–14.Bickell SL, Ferguson DM, Boissy A, Blache D, 2008: Sheepselected on their behavioural response to a social and humanchallenge differ also <strong>in</strong> their response to novelty. PhysiolBehav (<strong>in</strong> press).Bielli A, Pérez R, Pedrana G, Milton JTB, Lopez A,Blackberry M, Duncombe G, Rodriguez-Mart<strong>in</strong>ez H, Mart<strong>in</strong>GB, 2002: Low maternal nutrition dur<strong>in</strong>g pregnancyreduces the numbers of Sertoli cells <strong>in</strong> the newborn lamb.Reprod Fertil Dev 14, 333–337.Biggers JD, 1986: The potential use of artificially producedmonozygotic tw<strong>in</strong>s for comparative experiments. Theriogenology26, 1–25.Blache D, Tjondronegoro S, Blackberry MA, Anderson ST,Curlewis JD, Mart<strong>in</strong> GB, 1997: Gonadotroph<strong>in</strong> and prolact<strong>in</strong>secretion <strong>in</strong> castrated male sheep follow<strong>in</strong>g subcutaneousor <strong>in</strong>tracranial treatment with testicular hormones.Endocr 7, 235–243.Blache D, Blackberry MA, Van Cleeff JK, 2002: Does thethyroid axis play a role <strong>in</strong> domestication? 56th AnnualMeet<strong>in</strong>g of the American Institute of Biological Sciences,Wash<strong>in</strong>gton, DC, 22–24 March 2002, Abstract 27.Blache D, Zhang S, Mart<strong>in</strong> GB, 2006: Dynamic and <strong>in</strong>tegrativeaspects of the regulation of reproduction by metabolicstatus <strong>in</strong> male sheep. Reprod Nutr Dev 46, 379–390.Blache D, Chagas LM, Mart<strong>in</strong> GB, 2007: Nutritional <strong>in</strong>puts<strong>in</strong>to the reproductive neuroendocr<strong>in</strong>e control system – amultidimensional perspective. <strong>in</strong>: Juengel JI, Murray JF,Smith MF (eds), <strong>Reproduction</strong> <strong>in</strong> <strong>Domestic</strong> Rum<strong>in</strong>ants VI,Vol. Suppl 64. Nott<strong>in</strong>gham University Press, Nott<strong>in</strong>gham,UK, pp. 123–139.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Ethical Models for Study<strong>in</strong>g <strong>Reproduction</strong> 13Bloomfield FH, Oliver MH, Hawk<strong>in</strong>s P, Campbell M, PhillipsDJ, Gluckman PD, Challis Jr G, Hard<strong>in</strong>g JE, 2003: Apericonceptional nutritional orig<strong>in</strong> for non<strong>in</strong>fectious pretermbirth. Science 300, 606.Boissy A, 1995: Fear and fearfulness <strong>in</strong> animals. Q Rev Biol70, 165–191.von Borell E, Dobson H, Prunier A, 2007: Stress, behaviourand reproductive performance <strong>in</strong> female cattle and pigs.Hormones Behav 52, 130–138.Bryant J, Baggott la Velle L, Searle J, 2005: Introduction toBioethics. Wiley, Chichester.Cannon WB, 1927: The James–Lange theory of emotions: acritical exam<strong>in</strong>ation and an alteration. Am J Psychol 39,106–124.Caraty A, Miller DW, Delaleu B, Mart<strong>in</strong> GB, 1997: Stimulationof LH secretion <strong>in</strong> sheep by central adm<strong>in</strong>istration ofcorticotroph<strong>in</strong>-releas<strong>in</strong>g hormone. J Reprod Fertil 111, 249–257.Celi P, Walkden-Brown SW, Blache D, Szell AZ, Wilk<strong>in</strong>sonHM, Mart<strong>in</strong> GB, 2007: Tw<strong>in</strong> efficiency for reproductivevariables <strong>in</strong> monozygotic tw<strong>in</strong> sheep. Theriogenology 68,663–672.Church RB, 1988: Are problems posed by genetically eng<strong>in</strong>eeredanimals? Appl Anim Behav Sci 20, 73–82.Cockram MS, Ranson M, Imlah P, Goddard PJ, Burrells C,Harkiss GD, 1994: The behavioural, endocr<strong>in</strong>e and immuneresponses of sheep to isolation. Anim Production 58, 389–399.De Gendt K, Sw<strong>in</strong>nen JV, Saunders PT K, Schoonjans L,Dewerch<strong>in</strong> M, Devos A, Tan K, Atanassova N, Claessens F,Le´cureuil C, Heyns W, Carmeliet P, Guillou F, Sharpe RM,Verhoeven G, 2004: A Sertoli cell-selective knockout of theandrogen receptor causes spermatogenic arrest <strong>in</strong> meiosis.Proc Natl Acad Sci U S A 101, 1327–1332.Dobson H, Smith RF, 2000: What is stress, and how does itaffect reproduction? Anim Reprod Sci 60, 743–752.Fell LR, Colditz IG, Walker KH, Watson DL, 1999: Associationsbetween temperament, performance and immunefunction <strong>in</strong> cattle enter<strong>in</strong>g a commercial feedlot. Aust J ExpAgric 39, 795–802.Fest<strong>in</strong>g MFW, 1990: Use of genetically heterogeneous rats andmice <strong>in</strong> toxicological research: a personal perspective.Toxicol Appl Pharmacol 102, 197–204.Fest<strong>in</strong>g M, Altman DG, 2002: Guidel<strong>in</strong>es for the design andstatistical analysis of experiments us<strong>in</strong>g laboratory animals.ILAR 43, 244–258.Forkman B, Boissy A, Meunier-Salaun M-C, Canali E, JonesRB, 2007: A critical review of fear tests used on cattle, pigs,sheep, poultry and horses. Physiol Behav 91, 531–565.Gelez H, L<strong>in</strong>dsay DR, Blache D, Mart<strong>in</strong> GB, Fabre-Nys C,2003: Temperament and sexual experience affect femalesexual behaviour <strong>in</strong> sheep. Appl Anim Behav Sci 84, 81–87.Grignard L, Boissy A, Boiv<strong>in</strong> X, Garel JP, Le Ne<strong>in</strong>dre P, 2000:The social environment <strong>in</strong>fluences the behavioural responsesof beef cattle to handl<strong>in</strong>g. Appl Anim Behav Sci 68, 1–11.Hart K, Van Lier E, V<strong>in</strong>˜ oles C, Paganoni B, Blache D, 2008:Calm Mer<strong>in</strong>o ewes have more multiple pregnancies thannervous Mer<strong>in</strong>o ewes due to higher ovulation rate. Proc 16thInternational Congress on Animal <strong>Reproduction</strong> (<strong>in</strong> press).Hewitt SC, Harrell JC, Korach KS, 2005: Lessons <strong>in</strong> estrogenbiology from knockout and transgenic animals. Annu RevPhysiol 67, 285–308.Home Office, 2007: Statistics of Scientific Procedures on Liv<strong>in</strong>g<strong>Animals</strong>, Great Brita<strong>in</strong>, 2006. The Stationery Office, London,p. 47.Howard BR, 2002: Control of variability. ILAR 43, 191–193.Korte SM, 2001: Corticosteroids <strong>in</strong> relation to fear, anxietyand psychopathology. Neurosci Biobehav Rev 25, 117–142.Kumar RT, 2005: What have we learned about gonadotrop<strong>in</strong>function from gonadotrop<strong>in</strong> subunit and receptor knockoutmice? <strong>Reproduction</strong> 130, 293–302.LaFolette H, 2007: The Practice of Ethics. Blackwell Publish<strong>in</strong>g,Oxford.Mart<strong>in</strong> GB, Oldham CM, L<strong>in</strong>dsay DR, 1981: Effect of stressdue to laparoscopy on plasma cortisol levels, the preovulatorysurge of LH, and ovulation <strong>in</strong> the ewe. Theriogenology16, 39–44.Matteri RL, Watson JG, Moberg GP, 1984: Stress or acuteadrenocorticotroph<strong>in</strong> treatment suppresses LHRH-<strong>in</strong>ducedLH release <strong>in</strong> the ram. J Reprod Fertil 72, 385–393.Mepham B, 2005: Bioethics. Oxford University Press, Oxford.Miller DW, Blache D, Mart<strong>in</strong> GB, 1995: The role of<strong>in</strong>tracerebral <strong>in</strong>sul<strong>in</strong> <strong>in</strong> the effect of nutrition on gonadotroph<strong>in</strong>secretion <strong>in</strong> mature male sheep. J Endocr<strong>in</strong>ol 147,321–329.Mills AD, Beilharz RG, Hock<strong>in</strong>g PM, 1997: Genetic selection.In: Appleby MC, Hughes BO (eds), Animal Welfare. CABInternational, Wall<strong>in</strong>gford, pp. 219–231.Murphy PM, 1999: Maternal Behaviour and Rear<strong>in</strong>g Ability ofMer<strong>in</strong>o Ewes can be Improved by Strategic Feed SupplementationDur<strong>in</strong>g Late Pregnancy and Selection for CalmTemperament. The University of Western Australia, Perth.Murphy PM, Purvis IW, L<strong>in</strong>dsay DR, Le Ne<strong>in</strong>dre P, OrgeurP, Po<strong>in</strong>dron P, 1994: Measures of temperament are highlyrepeatable <strong>in</strong> Mer<strong>in</strong>o sheep and some are related to maternalbehaviour. Proc Austral Soc Anim Prod 20, 247–250.NHMRC Animal Welfare Committee, 2007: Guidel<strong>in</strong>es forthe Generation, Breed<strong>in</strong>g, Care and Use of GeneticallyModified and Cloned <strong>Animals</strong> for Scientific Purposes.Commonwealth of Australia, Canberra, p. 41.Nuffield Council on Bioethics, 2005: The Ethics of ResearchInvolv<strong>in</strong>g <strong>Animals</strong>. Nuffield Council on Bioethics, London,p. 376.Price EO, 2002: Animal <strong>Domestic</strong>ation and Behavior. CABInternational, Wall<strong>in</strong>gford, p. 320.Rae MT, Palassio S, Kyle CE, Brooks AN, Lea RG, MillerDW, Rh<strong>in</strong>d SM, 2001: Effect of maternal undernutritiondur<strong>in</strong>g pregnancy on early ovarian development andsubsequent follicular development <strong>in</strong> sheep fetuses. <strong>Reproduction</strong>,122, 915–922.Rh<strong>in</strong>d SM, 2004: Effects of maternal nutrition on fetal andneonatal reproductive development and function. AnimReprod Sci 82–83, 169–181.Rob<strong>in</strong>son JJ, Ashworth CJ, Rooke JA, Mitchell LM, McEvoyTG, 2006: Nutrition and fertility <strong>in</strong> rum<strong>in</strong>ant livestock.Animal Feed Sci Technol 126, 259–276.Russell WMS, Burch RL, 1959: The Pr<strong>in</strong>ciple of HumaneExperimental Technique. Methuen, London.Schmidtz D, 2007: A place for cost–benefit analysis. In:LaFolette H (ed), Ethics <strong>in</strong> Practice: An Anthology.Blackwell Publish<strong>in</strong>g, Oxford, pp. 669–679.Seidel GE, 2001: Clon<strong>in</strong>g, transgenesis, and genetic variance <strong>in</strong>animals. Clon<strong>in</strong>g Stem Cells 3, 251–256.Somerville M, 2007: Th<strong>in</strong>k<strong>in</strong>g Ethics,Do<strong>in</strong>g Ethics. EthicallyChallenged. The Miegunyah Press, Melbourne, pp. 68–98.Vallentyne P, 2007: Consequentialism. In: LaFolette H (ed),The Practice of Ethics. Blackwell Publish<strong>in</strong>g, Oxford, pp.22–30.V<strong>in</strong>oles C, Forsberg M, Mart<strong>in</strong> GB, Cajarville C, Repetto J,Meikle A, 2005: Short-term nutritional supplementation ofewes <strong>in</strong> low body condition affects follicle development dueto an <strong>in</strong>crease <strong>in</strong> glucose and metabolic hormones. <strong>Reproduction</strong>129, 299–309.Vois<strong>in</strong>et BD, Grand<strong>in</strong> T, Tatum JD, O’Connor SF, StruthersJJ, 1997: Feedlot cattle with calm temperaments have ahigher average daily weight ga<strong>in</strong>s the cattle with excitabletemperaments. J Anim Sci 75, 892–896.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


14 D Blache, GB Mart<strong>in</strong> and SK MaloneyWetsel WC, 1995: Immortalized hypothalamic lute<strong>in</strong>iz<strong>in</strong>ghormone-releas<strong>in</strong>g hormone (LHRH) neurons: a new toolfor dissect<strong>in</strong>g the molecular and cellular basis of LHRHphysiology. Cell Mol Neurobiol 15, 43–78.Zhang S, Blache D, Blackberry MA., Mart<strong>in</strong> GB, 2004:Dynamics of the responses <strong>in</strong> secretion of LH, lept<strong>in</strong> and<strong>in</strong>sul<strong>in</strong> follow<strong>in</strong>g an acute <strong>in</strong>crease <strong>in</strong> nutrition <strong>in</strong> maturemale sheep. Reprod Fertil Dev 16, 823–829.Conflict of <strong>in</strong>terest: D Blanche has received grants from AustralianResearch Council, Medical Research Council and Meat & LivestockAustralia; Much of G.B. Mart<strong>in</strong>’s research is funded by Meat &Livestock Australia; S. Maloney declares no conflict of <strong>in</strong>terest.Author’s address (for correspondence): D Blache, School of AnimalBiology M085, Faculty of Natural and Agricultural Sciences, TheUniversity of Western Australia, 35 Stirl<strong>in</strong>g Highway, Crawley, WA6009, Australia. E-mail: dbla@animals.uwa.edu.auÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 15–22 (2008); doi: 10.1111/j.1439-0531.2008.01138.xISSN 0936-6768Endocr<strong>in</strong>e Disruptors and Other Food-contam<strong>in</strong>at<strong>in</strong>g Environmental Pollutants asRisk Factors <strong>in</strong> Animal <strong>Reproduction</strong>SM Rh<strong>in</strong>dMacaulay Institute, Aberdeen, UKContentsPollutants of many chemical classes, derived primarily fromanthropogenic activities, are ubiquitous <strong>in</strong> the environment,persistent, biologically available and can exert adverse effectson the reproductive and other, <strong>in</strong>directly related, physiologicalsystems. Food is generally considered to be the major route ofanimal exposure <strong>in</strong> vertebrate species but the relative contributionsof other routes of exposure such as through lungs, gillsor sk<strong>in</strong> are not well studied and may be of importance forcerta<strong>in</strong> animal groups, depend<strong>in</strong>g on their immediate environment.<strong>Animals</strong> are particularly sensitive to exposure dur<strong>in</strong>gdevelopmental stages but the pattern of exposure to chemicalsis likely to be different to that of adults. Quantification of therisk posed by the <strong>in</strong>gestion of pollutants <strong>in</strong> food is complexand depends on many factors <strong>in</strong>clud<strong>in</strong>g species, diet composition,duration of exposure to the food, efficiency of pollutantabsorption, subsequent metabolism, sensitivity of targetorgans and stage of development. While the effects of highdoses of s<strong>in</strong>gle chemicals are proven, dietary exposure topollutants generally <strong>in</strong>volves prolonged, low-level exposure toa large number of compounds, each of which has differentchemical characteristics, exerts different biological effects andis present at vary<strong>in</strong>g concentrations. Thus, while exposure topollutants through feed is undoubtedly a significant risk factorfor many species and may be the most important one for manyterrestrial vertebrates, other routes of exposure may be moreimportant <strong>in</strong> other groups.IntroductionExposure to pollutants, and <strong>in</strong> particular to endocr<strong>in</strong>edisrupt<strong>in</strong>g compounds (EDCs) and potentially toxicmetals (PTMs) can <strong>in</strong>duce adverse physiological changes<strong>in</strong> animals of every group so far studied, from bacteria(Fox 2004) to humans (Toppari et al. 1996) and animalsof many phyla <strong>in</strong> between (Institute for Environmentand Health 1999). Many of the effects perta<strong>in</strong> to thereproductive system (Toppari et al. 1996; Paul et al.2005; Uzumcu and Zachow 2007, Fowler et al. 2008) butother physiological systems can be perturbed by exposure.These <strong>in</strong>clude the thyroid gland (Hansen 1998),neuroendocr<strong>in</strong>e system (Le<strong>in</strong> et al. 2007) and thereforebehaviour (Erhard and Rh<strong>in</strong>d 2004), the immune system(V<strong>in</strong>e et al. 2000; Daniel et al. 2001) and systems thatcontrol adipogenesis and nutrient partition<strong>in</strong>g (Newboldet al. 2007); disruption of these physiologicalsystems may also imp<strong>in</strong>ge on reproductive fitness.Evidence of relationships between exposure to pollutantsand reproductive effects is derived from observationalstudies of wildlife exposed, un<strong>in</strong>tentionally, topollutants (Smith 1981; Guillette et al. 1994; Institutefor Environment and Health 1999) and from empiricalstudies of laboratory and domestic animals or cellcultures (Gray et al. 2000; Meerts et al. 2001; Evanset al. 2004). The reproductive systems of animals ofmany phyla have been found to be perturbed throughmany mechnisms (Table 1). The first type of studyserved to demonstrate associations between EDC exposureand adverse effects on reproductive function whilethe latter has begun to elucidate mechanisms of action.However, even if they have <strong>in</strong>volved adm<strong>in</strong>istration ofchemicals <strong>in</strong> feed, some controlled studies are poorrepresentations of normal real-world exposure throughfood, be<strong>in</strong>g based on s<strong>in</strong>gle chemicals, often at very highconcentrations, and adm<strong>in</strong>istered for short periods.The aim of this paper is to review what is known ofthe risks of exposure to pollutants through the diet andto identify some pr<strong>in</strong>ciples that perta<strong>in</strong> to their effects onreproductive function <strong>in</strong> general, irrespective of speciesand habitat.What Classes of Chemical are Involved andHow Do They Operate?The acute toxicity of high levels of PTMs has probablybeen recognized s<strong>in</strong>ce metals were first smelted. However,recent reports <strong>in</strong>dicate that they may also exertmore subtle effects and that very low, environmentallyrelevant, concentrations of cadmium, copper, cobalt,nickel, lead, t<strong>in</strong> and mercury can each activate theoestrogen receptor a (Mart<strong>in</strong> et al. 2003; Henson andChedrese 2004) while methyl mercury can disrupt nonsteroidalreceptors (Klaper et al. 2006) and copper cancompromise olfactory or other senses <strong>in</strong> fish (Sandahlet al. 2007). Thus, each has the potential to disruptreproductive function.Organic EDCs, unlike PTMs which can also actthrough endocr<strong>in</strong>e disruption, are primarily anthropogenic<strong>in</strong> nature and have been produced <strong>in</strong> largeamounts only for approximately 60 years, only. Classesand sources of EDCs have been described previously(Rh<strong>in</strong>d 2005). They seldom <strong>in</strong>duce an acute toxic effectbut can affect reproductive function at concentrationsmany orders of magnitude below the toxic dose. Theseeffects are expressed through many mechanisms <strong>in</strong>clud<strong>in</strong>gb<strong>in</strong>d<strong>in</strong>g to the androgen receptor (DDT metabolites;Kelce et al. 1998), oestrogen receptor [alkyl phenols andpolychlor<strong>in</strong>ated biphenyls (PCBs); Dodge 1998] and arylhydrocarbon receptor [polycyclic aromatic hydrocarbons(PAHs); Haque et al. 2005]. In addition, PCBs canact directly on cellular systems via non-receptor-mediatedsystems (Li and Hansen 1996). It is noteworthy thatthe DNA of the oestrogen receptor appears to be highlyconserved over a very wide range of phyla and species,suggest<strong>in</strong>g that it is evolutionarily ancient (ThorntonÓ 2008 Macaulay Land Use Research Institute


16 SM Rh<strong>in</strong>dTable 1. Selected examples of reproductive perturbation, by environmental exposure to endocr<strong>in</strong>e disrupt<strong>in</strong>g pollutants, <strong>in</strong> species represent<strong>in</strong>gdifferent classes and phylaSpecies Chemical Effect ReferenceWhite footed mice (Peromyscus leucopus) PCB and cadmium Reduced testis size Batty et al. (1990)Seagull (Larus californicus) DDT Fem<strong>in</strong>ization of embryos Fry and Toone (1981)American alligator (Alligator mississippiensis) Dicofol, DDT Abnormal testes, phalli and testosterone Guillette et al. (1994)Dogwhelk (Nucella lapillus) Tributylt<strong>in</strong> Imposex Gibbs and Bryan (1987)Mayfly (Cloeon dipterum) Esfenvalerate (<strong>in</strong>secticide) Reduced larval survival Beketov and Liess (2005)Annelids Various Multiple Krajniak (2005)Nematode (Caenorhabditis elegans) Various Multiple Ho¨ ss and Weltje (2007)et al. 2003). S<strong>in</strong>ce many EDC actions are mediatedthrough this receptor, it may expla<strong>in</strong> why the <strong>in</strong>fluencesof these pollutants are present <strong>in</strong> so many animalgroups.Some EDCs act through multiple mechanisms ands<strong>in</strong>ce animals are usually exposed, simultaneously, tomany EDCs, those of different classes can act <strong>in</strong> concertwith each other (Rajapakse et al. 2002) and with PTMs(Bemis and Seegal 1999). Consequently, prediction ofrisk from environmental exposure is made more difficult.As a result of receptor-mediated signals, endocr<strong>in</strong>eperturbations or direct effects of pollutants on DNA,changes <strong>in</strong> expression of a wide range of genes can be<strong>in</strong>duced. The changes <strong>in</strong> gene function are probably<strong>in</strong>duced epigenetically (i.e. mitotically heritable changes<strong>in</strong> gene function may be caused without changes <strong>in</strong>DNA sequence) through processes such as DNA methylation(Crews and McLachlan 2007).What Factors Determ<strong>in</strong>e the Risk Associatedwith Pollutant Exposure?Risks associated with dietary pollutants <strong>in</strong> both vertebrateand <strong>in</strong>vertebrate species depend on the propertiesof the chemicals <strong>in</strong>volved s<strong>in</strong>ce these may determ<strong>in</strong>e theamount present <strong>in</strong> food, the likelihood of <strong>in</strong>gestion,uptake rate and bioavailability follow<strong>in</strong>g <strong>in</strong>gestion, andtarget organ sensitivity. Ow<strong>in</strong>g to the logistical difficultiesof measurements <strong>in</strong> small, <strong>in</strong>vertebrate animals,much of the evidence perta<strong>in</strong><strong>in</strong>g to tissue levels ofpollutants is derived from studies of vertebrates andparticularly mammals. Many basic pr<strong>in</strong>ciples concern<strong>in</strong>gexposure risk can be extrapolated, to an extent, tosmaller, <strong>in</strong>vertebrate species but it should be noted thatthe contribution of diet to pollutant burden may berelatively less important than exposure through thebody surface, particularly for species that live <strong>in</strong> water,sediment or soil. S<strong>in</strong>ce assessment of the relativeimportance of the respective routes of exposure islogistically extremely difficult, this is poorly understood.However, s<strong>in</strong>ce the health and reproductive efficiency ofsuch species is critical for ecosystem health and susta<strong>in</strong>ability,potential effects of pollutants on these animalgroups are of great importance.Persistence and biological availabilityWhile environmental concentrations of PTMs andEDCs are highly variable, they are generally low.Many rema<strong>in</strong> <strong>in</strong> the environment <strong>in</strong>def<strong>in</strong>itely but theirbiological availability determ<strong>in</strong>es whether or not theycan exert any adverse effects. Uptake of PTMs from thedigestive tract may be limited by sequestration with<strong>in</strong><strong>in</strong>gested soil or plants (Wilk<strong>in</strong>son et al. 2003) and differswith <strong>in</strong>dividual element and chemical speciation, animalspecies, diet composition (e.g. fibre content), vitam<strong>in</strong>status, dose, animal age, dietary chelators, the presenceof antagonistic elements, pregnancy and lactation (Institutefor Environment and Health 1998; Wilk<strong>in</strong>son et al.2003) (Fig. 1). Once absorbed, metallothione<strong>in</strong>, andother prote<strong>in</strong>s produced <strong>in</strong> the liver and gut wall, buffer<strong>in</strong>tracellular levels of metals and act as a storagemechanism, particularly <strong>in</strong> vertebrates (Wilk<strong>in</strong>son et al.2003).Gastro<strong>in</strong>test<strong>in</strong>al absorption of many EDCs is efficient<strong>in</strong> vertebrates (Wild and Jones 1992; Norstrom 2002)and once absorbed they rema<strong>in</strong>, generally, biologicallyavailable be<strong>in</strong>g sequestered little (


Dietary Pollutants as Risk Factors <strong>in</strong> <strong>Reproduction</strong> 17Level ofexposureClass of compound – volatility, hydrophobicity,degradability, solubility, b<strong>in</strong>d<strong>in</strong>g properties, etcTrophic level/biomagnificationEnvironmental concentrationUptakeDietary exposure(food/water)• Food type• Degradation <strong>in</strong> <strong>in</strong>test<strong>in</strong>al tract• Efficiency of absorption from tract• Dietary chelatorsNon-dietary exposure(sk<strong>in</strong>/<strong>in</strong>tegument/lungs/gills, etc)• Structure/permability of barrier• Active transport processes (?)• Octanol/water or octanol/air coefficient• SequestrationPost-uptakeWith<strong>in</strong> animal• Metabolism• Excretion• Lipid contentFig. 1. Determ<strong>in</strong>ants of level ofexposure to environmentalpollutants, of uptake by animals,rates of exposure of their organsand effects on themIndividual organ• Permeability of organ membrane(e.g. bra<strong>in</strong>; testis)• Sensitivity of target tissues/targetgenesOffspr<strong>in</strong>g• Susceptibility of gametes• Efficiency of trans-placentaltransfer• Fetal metabolism/excretionassociated gene expression. Furthermore, disruption ofthe germ cell l<strong>in</strong>e <strong>in</strong> one generation can be expressed <strong>in</strong>subsequent generations (Bøgh et al. 2001; Anway andSk<strong>in</strong>ner 2006; Edwards and Myers 2007), probablythrough altered gene methylation, and therefore geneexpression, dur<strong>in</strong>g the period of sex differentiation of thedevelop<strong>in</strong>g gonad (Anway and Sk<strong>in</strong>ner 2006). Althoughdemonstrated <strong>in</strong> vertebrates, these fundamental mechanismsare likely to perta<strong>in</strong> to most species.How Important is Exposure Through Ingestion?The complex relationship between rates of pollutant<strong>in</strong>gestion and effects exerted may be further complicatedby related factors such as additional uptake throughdr<strong>in</strong>k<strong>in</strong>g water, <strong>in</strong>halation or, particularly <strong>in</strong> smaller,<strong>in</strong>vertebrate species, through dermal absorption ofpollutants.Exposure through foodFood is generally considered to be the most importantroute of exposure <strong>in</strong> vertebrates, except, perhaps, at thelowest trophic levels (Fries 1995; Kavlock et al. 2002;Norstrom 2002). S<strong>in</strong>ce pollutants accumulate <strong>in</strong> thetissue of animals at each trophic level, the position of theanimal <strong>in</strong> the food cha<strong>in</strong> determ<strong>in</strong>es the exposure rate;ow<strong>in</strong>g to the effects of biomagnification (<strong>in</strong>creasedconcentrations as a result of food cha<strong>in</strong> energetics),the highest tissue concentrations are frequently recorded<strong>in</strong> carnivorous species at or near to the top of the foodcha<strong>in</strong> (Johnson et al. 1996; Darnerud et al. 2002; Davidand Gans 2003; Law et al. 2003; Naert et al. 2007). Forexample, <strong>in</strong> such species, concentrations of polybrom<strong>in</strong>ateddiphenyl ethers (PBDEs) often can be>100 lg ⁄ kg lipid (Darnerud et al. 2001) and PCBs>100 lg ⁄ kg dry matter based on the whole body(Johnson et al. 1996).Herbivores, be<strong>in</strong>g much closer to the primary sourceof food production, are not subject to multiple levels ofbiomagnification. Accord<strong>in</strong>gly, tissue accumulation (the<strong>in</strong>crease <strong>in</strong> concentration tissues attributable to uptake)is frequently much lower with PBDEs be<strong>in</strong>g


18 SM Rh<strong>in</strong>d<strong>in</strong>gestion’ for these may be very different from that ofadults, e.g. mammalian foetuses are nourished viamaternal blood and develop<strong>in</strong>g birds by egg yolk andwhile young mammals consume milk, adults eat animalor plant material. Similarly, larval stages of <strong>in</strong>sects andother <strong>in</strong>vertebrates may have very different diets toadults. In addition to differences <strong>in</strong> food source,differences <strong>in</strong> metabolism with age mean that it isunwise to extrapolate from effects of exposure <strong>in</strong> adultsto those <strong>in</strong> immature animals, because the relationshipbetween the amounts of pollutants ‘<strong>in</strong>gested’ at thedifferent stages and the respective tissue concentrationsmay differ with differences <strong>in</strong> metabolism. Furthermore,<strong>in</strong>direct measures of exposure of immature animals topollutants, for example, through measurements <strong>in</strong>mammalian milk or umbilical cord blood may be ofless value than tissue concentrations <strong>in</strong> the foetus ⁄ immatureanimal for the prediction of physiological effectss<strong>in</strong>ce the former does not take account of effects ofmetabolism and excretion <strong>in</strong> the offspr<strong>in</strong>g.While concentrations of pollutants <strong>in</strong> human (Dekon<strong>in</strong>gand Karmaus 2000; Darnerud et al. 2001) andsheep milk (Rh<strong>in</strong>d et al. 2007b), and human umbilicalcord blood, amniotic fluid and meconium (Dallaireet al. 2003; Barr et al. 2007) have been measured, thereare relatively few reports of foetal tissue concentrations(Bosse et al. 1996; Dekon<strong>in</strong>g and Karmaus 2000) <strong>in</strong> anyspecies and so direct assessment of risk to the foetus isnot possible. Concentrations of PTMs (Rh<strong>in</strong>d 2005) andseveral PCBs and PBDEs (Rh<strong>in</strong>d et al. 2007a) have beenshown to be lower <strong>in</strong> fetal sheep than <strong>in</strong> the correspond<strong>in</strong>gmaternal tissue but <strong>in</strong>dividual concentrationsdiffer greatly with congener (PCB < 20 ng ⁄ kg to3.8 lg ⁄ kg; PBDE < 20 ng ⁄ kg to 30 lg ⁄ kg liver drymatter), as does the relationship between adult andfoetal levels. Schecter et al. (2007) provided the firstreport of PBDE levels <strong>in</strong> human foetuses; mean totalPBDE concentrations were 23 ng ⁄ kg lipid.Interest<strong>in</strong>gly, sheep maternal and foetal concentrationsof neither PTMs (Rh<strong>in</strong>d et al. 2005a) nor PCBsand PBDEs (SM Rh<strong>in</strong>d, unpublished observations) weresignificantly correlated. However, related studies havealso shown that very small <strong>in</strong>creases <strong>in</strong> maternalexposure to pastures fertilized with sewage sludge whichconta<strong>in</strong>s multiple pollutants, result <strong>in</strong> changes <strong>in</strong> structureand function of the foetal testis (Paul et al. 2005)and ovary (Fowler et al. 2008). Collectively, theseobservations show that the relationships between pollutantexposure and effect are highly complex, that foetalexposure cannot be extrapolated from adult profiles,and that small <strong>in</strong>creases <strong>in</strong> exposure to multiple pollutantscan adversely affect foetal reproductive development.Immediately after birth, the ma<strong>in</strong> source of nutrient <strong>in</strong>mammals is milk. The transfer of PTMs to the offspr<strong>in</strong>gthrough milk is probably relatively unimportant (Wilk<strong>in</strong>sonet al. 2003) but exposure to lipophilic organicpollutants through milk may be significant, particularly<strong>in</strong> species such as mar<strong>in</strong>e mammals which have a highmilk fat content (Du<strong>in</strong>ker and Hillebrand 1979). On theother hand, studies compar<strong>in</strong>g milk concentrations atearly and late stages of lactation <strong>in</strong> sheep (periods of fatmobilization and deposition respectively) <strong>in</strong>dicate thatmilk concentrations of phthalate and alkyl phenol arenot elevated by <strong>in</strong>creased maternal fat mobilizationdur<strong>in</strong>g early lactation (Rh<strong>in</strong>d et al. 2007b) suggest<strong>in</strong>gthat maternal fat may not be a significant contributor toneonatal pollutant exposure <strong>in</strong> this species.Food-related exposureWhile food is generally regarded as the major source ofexposure <strong>in</strong> vertebrates, <strong>in</strong> some circumstances foodrelatedroutes may also be important. There is a risk forall species (vertebrate and <strong>in</strong>vertebrate) of exposure tocontam<strong>in</strong>ated water or air as a result of the processes offood acquisition and <strong>in</strong>gestion. For example, dairy cowsdr<strong>in</strong>k<strong>in</strong>g from canals <strong>in</strong> The Netherlands which arecontam<strong>in</strong>ated with sewage, and therefore a cocktail ofpollutants, have been shown to have reduced reproductiveefficiency and lower milk production (Meijer et al.1999). Most of the pollutants that are likely to bepresent <strong>in</strong> food are relatively <strong>in</strong>soluble <strong>in</strong> water and soare not normally <strong>in</strong>gested with dr<strong>in</strong>k<strong>in</strong>g water. However,if bound to suspended solids <strong>in</strong> the water, suchpollutants could be <strong>in</strong>gested. In addition, these pollutantsmay <strong>in</strong>teract with steroids from the contraceptivepill and alkyl phenols which are typically present <strong>in</strong> theaqueous component of sewage (Institute for Environmentand Health 1999; Bowman et al. 2003).Similarly, surface application of sewage sludge topastures has been shown to be associated with compromisedfoetal testis and ovary development (Paul et al.2005; Fowler et al. 2008). S<strong>in</strong>ce systemic uptake ofpollutants by plants is very limited (Wild and Jones1992), it is likely that the associated exposure topollutants results from <strong>in</strong>gestion of soil along with theherbage, particularly when the herbage mass is small (upto 30% of dry matter <strong>in</strong>take of sheep may be soil;Thornton and Abrahams 1983), as a result of surfacecontam<strong>in</strong>ation of the herbage with soil particles.In specific circumstances, air-borne pollutants l<strong>in</strong>kedto the food source could, at least theoretically, contributeto the body burden of pollutants, e.g. domesticanimals may <strong>in</strong>hale additional pollutants when graz<strong>in</strong>gpastures fertilized with sewage sludge, ow<strong>in</strong>g to theretention of volatilized chemicals with<strong>in</strong> the sward.Concentrations of many pollutant classes <strong>in</strong> normal airare measurable (David and Sandra 2001; U.S. CentralPollution Control Board Newsletter 2001; Lee et al.2004) and there is evidence that urban air-bornepollution can cause DNA damage <strong>in</strong> humans (Whyattet al. 1998). Thus, the potential for additional exposures,result<strong>in</strong>g from the process of acquir<strong>in</strong>g food,should be taken <strong>in</strong>to account.Age-related effectsThe degree of pollutant accumulation through fooddepends on animal age not only because of the differenceswith age <strong>in</strong> diet type but also because olderanimals may be exposed to pollutants for longer and, asshown <strong>in</strong> some species of fish, may accumulate more(Darnerud et al. 2001). Furthermore, it has been shownthat when exposure is prolonged, low, and apparentlyharmless, pollutant concentrations can exert biologicalÓ 2008 Macaulay Land Use Research Institute


Dietary Pollutants as Risk Factors <strong>in</strong> <strong>Reproduction</strong> 19effects at least equivalent to, and sometimes higher than,those <strong>in</strong>duced by much higher doses adm<strong>in</strong>istered overshort periods of time (Borman et al. 2000).Individual variation <strong>in</strong> exposureIn species as diverse as sheep (Rh<strong>in</strong>d et al. 2005b) and<strong>in</strong>sects from lake sediments (Kovats and Ciborowski1989) differences <strong>in</strong> tissue concentrations of variouspollutants of at least 10-fold between <strong>in</strong>dividuals fromthe same source are commonly observed. Similar environmentalvariation was reported <strong>in</strong> soil samples (foodcontam<strong>in</strong>ant and <strong>in</strong>vertebrate habitat) collected fromwith<strong>in</strong> experimental plots (Rh<strong>in</strong>d et al. 2002). Thisfurther complicates the prediction of responses of<strong>in</strong>dividual animals to pollutant exposure.In summary, there seems to be little doubt that thediet is a major source of exposure to pollutants <strong>in</strong> manyvertebrate species and while the effects on tissueconcentrations are greatest <strong>in</strong> carnivores near to thetop of the food cha<strong>in</strong>, significant tissue accumulationcan occur <strong>in</strong> herbivores. However, the relative contributionsof other factors such as uptake from thesurround<strong>in</strong>g media (air, water, soil or sediment) are lessclear and, <strong>in</strong> certa<strong>in</strong> circumstances and species, thesesources may be more important than dietary ones,particularly <strong>in</strong> small, soil-, sediment-, or water-dwell<strong>in</strong>gspecies.What Risk is Associated with Pollutants <strong>in</strong>Food?S<strong>in</strong>ce many studies of effects of pollutants on reproductivefunction frequently <strong>in</strong>volve transient, unusuallyhigh levels of exposure of wild or laboratory species ofanimals to a s<strong>in</strong>gle chemical, it is sometimes concludedthat normal, low, levels of exposure to pollutants areof no biological consequence. This argument is re<strong>in</strong>forcedby the fact that environmental concentrationsof some, but not all, pollutants are decl<strong>in</strong><strong>in</strong>g (Darnerudet al. 2001; Norstrom 2002), as are milk (Solomonand Weiss 2002) and tissue concentrations (Noakeset al. 2006). In fact, even tissue concentrations thatare at or near to background environmental concentrationsmay exert biological effects, particularly whenthey act <strong>in</strong> conjunction with a mixture of otherchemicals; ‘real-world’ exposure normally <strong>in</strong>volvesprolonged exposure to a mixture of compounds at lowconcentrations. Effects of such exposure patterns havebeen demonstrated by long-term studies of sheepma<strong>in</strong>ta<strong>in</strong>ed throughout their breed<strong>in</strong>g lives on pasturesfertilized with either conventional <strong>in</strong>organic fertilizeror sewage sludge. The latter conta<strong>in</strong>s large amounts ofboth <strong>in</strong>organic and organic pollutants, relative toenvironmental levels (Smith 1996; Stevens et al. 2003)and, when applied to pasture as a fertilizer, results <strong>in</strong>very small <strong>in</strong>creases <strong>in</strong> environmental levels of thesechemicals (Rh<strong>in</strong>d et al. 2002) and <strong>in</strong> the tissue levelsof sheep graz<strong>in</strong>g the pasture (Rh<strong>in</strong>d et al. 2005a,b,2007b). However, these small <strong>in</strong>creases are associatedwith disruption of reproductive and other functions(Erhard and Rh<strong>in</strong>d 2004; Paul et al. 2005; Fowleret al. 2008).Such studies clearly demonstrate that pollutants <strong>in</strong>feed can represent a risk to reproductive function but donot allow the quantification of that risk with respect toeither <strong>in</strong>dividual chemicals or <strong>in</strong>dividual animals. Thatwould require determ<strong>in</strong>ation of dose response relationshipsbetween the concentrations of each chemical, <strong>in</strong>each relevant tissue or organ and for each observedchange <strong>in</strong> structure or function. It is possible to achievethis, empirically, for a small number of <strong>in</strong>dividualchemicals but, at present, virtually impossible to do sofor the thousands of chemicals to which animals areexposed. Understand<strong>in</strong>g of mixture effects, will require<strong>in</strong>tegration of the observations from the wide range ofempirical approaches that are available us<strong>in</strong>g powerfulpredictive computer models (Suk et al. 2002).ConclusionsThere is no doubt that exposure to pollutants canadversely affect reproductive function <strong>in</strong> animals andthat food is likely to be the most important route ofexposure for many or most species, particularly forthose near the top of the food cha<strong>in</strong>. However,assessment of the risk is complicated by the fact that<strong>in</strong> the ‘real world’ food conta<strong>in</strong>s many different classesof pollutants, each at low and variable concentrations,and exposure to them occurs throughout life, <strong>in</strong>clud<strong>in</strong>gdur<strong>in</strong>g developmental stages which are particularlysensitive to their effects. In short, dietary pollutantsrepresent a significant risk factor for reproduction,under certa<strong>in</strong> circumstances, but precise quantificationof these risks for <strong>in</strong>dividuals is extremely difficult withcurrent knowledge.ReferencesAlbro PW, Lavenhar SR, 1989: Metabolism of di-(2-ethylhexyl)phthalate. Drug Metab Rev 21, 13–34.Anway MD, Sk<strong>in</strong>ner MK, 2006: Epigenetic transgenerationalactions of endocr<strong>in</strong>e disruptors. Endocr<strong>in</strong>ology 147, S43–S49.Bachour G, Fail<strong>in</strong>g K, Elmadfa SG, Brunn H, 1998: Speciesand organ dependence of PCB contam<strong>in</strong>ation <strong>in</strong> fish, foxes,roe deer, and humans. Arch Environ Contam Toxicol 35,666–673.Barr DB, Bishop A, Needham LL, 2007: Concentrtions ofxenobiotic chemicals <strong>in</strong> the maternal-fetal unit. ReprodToxicol 23, 260–266.Batty J, Leavitt RA, Biondot N, Pol<strong>in</strong> D, 1990: Anecotoxicological study of a population of the white footedmouse (Peromyscus leucopus) <strong>in</strong>habit<strong>in</strong>g a polychlor<strong>in</strong>atedbiphenyls-contam<strong>in</strong>ated area. Arch Environ Contam Toxicol19, 283–290.Beketov MA, Liess M, 2005: Acute contam<strong>in</strong>ation withesfenvalerate and food limitation: chronic effects on themayfly, Cloeon dipterum. Environ Toxicol Chem 24, 1281–1286.Bemis JC, Seegal RF, 1999: Polychlor<strong>in</strong>ated biphenyls andmethylmercury act synergistically to reduce rat bra<strong>in</strong> dopam<strong>in</strong>econtent <strong>in</strong> vitro. Environ Health Perspect 107, 879–885.Bøgh IB, Christensen P, Dantzer V, Groot M, Thøfner ICN,Rasmussen RK, Schmidt M, Greve T, 2001: Endocr<strong>in</strong>edisrupt<strong>in</strong>g compounds: effect of octylphenol on reproductionover three generations. Theriogenol 55, 131–150.Ó 2008 Macaulay Land Use Research Institute


20 SM Rh<strong>in</strong>dBorman SM, Christian PJ, Sipes IG, Hoyer PB, 2000:Ovotoxicity <strong>in</strong> female Fischer rats and B6 mice <strong>in</strong>duced bylow-dose exposure to three polycyclic aromatic hydrocarbons:comparison through calculation of an ovotoxic <strong>in</strong>dex.Toxicol Appl Pharmacol 167, 191–198.Bosse U, Bannert N, Niessen KH, Teufel M, Rose I1996: Fetalorgan and tissue content of organohalogens and polychlor<strong>in</strong>atedbiphenyl. Zbl Hyg 198, 331–339.Bowman JC, Readman JW, Zhou JL, 2003: Sorption of thenatural endocr<strong>in</strong>e disruptors, oestrone and estradiol-17B <strong>in</strong>the aquatic environment. Environ Geochem Health 25, 63–67.Crews D, McLachlan JA, 2007: Epigenetics, evolution, endocr<strong>in</strong>edisruption, health and disease. Endocr<strong>in</strong>ology147(Suppl.), S4–S10.Crofton KM, Craft ES, Hedge JM, Genn<strong>in</strong>gs C, Simmons JE,Carchman RA, Carter WH, deVito MJ, 2005: Thyroidhormone-disrupt<strong>in</strong>gchemicals: evidence for dose-dependentadditivity or synergism. Environ Health Perspect 113, 1549–1554.Dallaire F, Dewailly E, Muckle G, Ayotte P, 2003: Timetrends of persistent organic pollutants and heavy metals <strong>in</strong>umbilical cord blood of Inuit <strong>in</strong>fants born <strong>in</strong> Nunavik(Quebec, Canada) between 1994 and 2001. Environ HealthPerspect 111, 1660–1664.Daniel V, Huber W, Bauer K, Suesal C, Conradt C, Opelz G,2001: Associations of blood levels of PCB, HCHs, and HCBwith numbers of lymphocyte subpopulations, <strong>in</strong> vitrolymphocyte response, plasma cytok<strong>in</strong>e levels, and immunoglobul<strong>in</strong>autoantibodies. Environ Health Perspect 109, 173–178.Darnerud PO, Eriksen GS, Johannesson T, Larsen PB,Viluksela M, 2001: Polybrom<strong>in</strong>ated diphenyl ethers: occurrence,dietary exposure, and toxicology. Environ HealthPerspect 109(Suppl. 1), 49–68.David RM, Gans G2003: Summary of mammalian toxicologyand health effects of phthalate esters. In: Staples CA(ed.),An Assessment of the Potential Environmental Risks Posedby Phthalates. Handbook of Environmental Chemistry 3.Spr<strong>in</strong>ger, London, pp. 299–316.David F, Sandra P2001: Phthalate esters <strong>in</strong> the environment.Monitor<strong>in</strong>g program for the determ<strong>in</strong>ation of phthalates <strong>in</strong>air, vegetation, cattle feed, milk and fish <strong>in</strong> The Netherlands(1999–2001). ECPI -2001-10, Research Institute of Chromatography,Belgium.Dekon<strong>in</strong>g EP, Karmaus W, 2000: PCB exposure <strong>in</strong> utero andvia breast milk. A review. J Exp Anal Environ Epidemiol 10,285–293.Dodge JA, 1998: Structure ⁄ activity relationships. Pure ApplChem 70, 1725–1734.Du<strong>in</strong>ker JC, Hillebrand MThJ, 1979: Mobilisation of organochlor<strong>in</strong>esfrom female lipid tissue and transplacentaltransfer to foetus <strong>in</strong> a harbour porpoise (Phocoena phocoena)<strong>in</strong> a contam<strong>in</strong>ated area. Bull Environ Contam Toxicol23, 728–732.Edwards TM, Myers JP, 2007: Environmental exposures andgene regulation <strong>in</strong> disease etiology. Environ Health Perspect115, 1264–1270.Erhard H, Rh<strong>in</strong>d SM, 2004: Prenatal and postnatal exposureto environmental pollutants <strong>in</strong> sewage sludge alters emotionalreactivity and exploratory behaviour <strong>in</strong> sheep. SciTotal Environ 332, 101–108.Evans NP, North T, Dye S, Sweeney T, 2004: Differentialeffects of the endocr<strong>in</strong>e-disrupt<strong>in</strong>g compounds Bisphenol-Aand Octylphenol on gonadotroph<strong>in</strong> secretion, <strong>in</strong> prepubertalewe lambs. Domest Anim Endocr 26, 61–73.Fry DM, Toone CK, 1981: DDT-<strong>in</strong>duced fern<strong>in</strong>ization of gullembryos. Science 213, 922–924.Fowler PA, Dora N, McFerran H, Amezaga MR, Miller DW,Lea RG, Cash P, McNeilly AS, Evans NP, Cot<strong>in</strong>ot C,Sharpe RM, Rh<strong>in</strong>d SM, 2008: In-utero exposure to lowdoses of environmental pollutants disrupts fetal ovariandevelopment <strong>in</strong> sheep. Mol Human Reprod doi:0-1093 ⁄molehr ⁄ gan020.Fox JE, 2004: Chemical communication threatened by endocr<strong>in</strong>e-disrupt<strong>in</strong>gcompounds. Environ Health Perspect 112,648–653.Fries GF, 1995: A review of the significance of animal foodproducts as potential pathways of human exposures todiox<strong>in</strong>s. J Anim Sci 73, 1639–1650.Gandolfi F, Pocar P, Brev<strong>in</strong>i TAL, Fischer B, 2002: Impact ofendocr<strong>in</strong>e disrupters on ovarian function and embryonicdevelopment. Domest Anim Endocr 23, 189–201.Gibbs P, Bryan G, 1987: TBT pa<strong>in</strong>ts and the demise of thedog-whelk, Nucela lapillus (Gastropoda). Oceans 19, 1482–1487.Gray LE, Ostby J, Furr J, Price M, Veeramachaneni DNR,Parks L, 2000: Per<strong>in</strong>atal exposure to the phthalates DEHP,BBP, and DINP, but not DEP, DMP, or DOTP, alterssexual differentiation of the male rat. Toxicol Sci 58, 350–365.Guillette LJ, Gross TS, Masson JR, Matter JM, Percival HF,Woodward AR, 1994: Developmental abnormalities of thegonad and abnormal sex hormone concentrations <strong>in</strong> juvenilealligators from contam<strong>in</strong>ated and control lakes <strong>in</strong> Florida.Environ Health Perspect 102, 680–688.Hansen L, 1998: Stepp<strong>in</strong>g backward to improve assessment ofPCB congener toxicities. Environ Health Perspect 106, 171–189.Haque M, Francis J, Sehgal I, 2005: Aryl hydrocarbonexposure <strong>in</strong>duces expression of MMP-9 <strong>in</strong> human prostatecancer cell l<strong>in</strong>es. Cancer Lett 225, 159–166.Henson MC, Chedrese PJ, 2004: Endocr<strong>in</strong>e disruption bycadmium, a common environmental toxicant with paradoxicaleffects on reproduction. Exp Biol Med 229, 383–392.Ho¨ ss S, Weltje L, 2007: Endocr<strong>in</strong>e disruption <strong>in</strong> nematodes:effects and mechanisms. Ecotoxicology 16, 15–28.Institute for Environment and Health, 1998: IEH Report onFactors Affect<strong>in</strong>g the Absorption of Toxic Metals from theDiet (Report R8). MRC Institute for Environment andHealth, Leicester, UK.Institute for Environment and Health, 1999: Institute forEnvironment and Health Assessment of the EcologicalSignificance of Endocr<strong>in</strong>e Disruption: Effects on ReproductiveFunction and Consequences for Natural Populations(Assessment A4). MRC Institute for Environment andHealth, Leicester, UK.Johnson MS, Leah RT, Conor L, Rae C, Saunders S, 1996:Polchlor<strong>in</strong>ated biphenyls <strong>in</strong> small mammals from contam<strong>in</strong>atedlandfill sites. Environ Pollut 92, 185–191.Kavlock K, Boekelheide R, Chap<strong>in</strong> M, Cunn<strong>in</strong>gham E,Faustman E, Foster P, Golub M, Henderson R, H<strong>in</strong>bergI, Little R, Seed J, Shea K, Tabacova S, Tyl R, Williams P,Zacharewski T, 2002: NTP center for the evaluation of risksto human reproduction: phthalates expert panel report onthe reproductive and developmental toxicity of di(2-ethylhexyl)phthalate. Reprod Toxicol 16, 529–653.Kelce WR, Gray LE, Wilson EM, 1998: Anti-androgens asenvironmental endocr<strong>in</strong>e disruptors. Reprod Fert Dev 10,105–111.Kelly BC, Ikonomou MG, Blair JD, Mor<strong>in</strong> AE, Gobas FAPC,2007: Food web-specific biomagnification of persistentorganic pollutants. Science 317, 236–238.Klaper R, Rees CB, Drevnick P, Weber ED, Sandhe<strong>in</strong>rich M,Carvan MJ, 2006: Gene expression changes related toendocr<strong>in</strong>e function and decl<strong>in</strong>e <strong>in</strong> reproduction <strong>in</strong> fatheadÓ 2008 Macaulay Land Use Research Institute


Dietary Pollutants as Risk Factors <strong>in</strong> <strong>Reproduction</strong> 21m<strong>in</strong>now (Pimephales promelas) after dietary methylmercuryexposure. Environ Health Perspect 114, 1337–1343.Kovats ZE, Ciborowski JJH, 1989: Aquatic <strong>in</strong>sect adults as<strong>in</strong>dicators of organochlor<strong>in</strong>e contam<strong>in</strong>ation. J Great LakesRes 15, 623–634.Krajniak K, 2005: Annelid endocr<strong>in</strong>e disruptors and a surveyof <strong>in</strong>vertebrate FMRFamide-related peptides1. Integr CompBiol 45, 88–96.Law RJ, Alaee M, Allch<strong>in</strong> CR, Boon JP, Lebeuf M, Lepom P,Stern GA, 2003: Levels and trends of polybrom<strong>in</strong>ateddiphenylethers and other brom<strong>in</strong>ated flame retardants <strong>in</strong>wildlife. Environ Int 29, 757–770.Lee RGM, Thomas GO, Jones KC, 2004: PBDEs <strong>in</strong> theatmosphere of three locations <strong>in</strong> Western Europe. EnvironSci Technol 38, 699–706.Le<strong>in</strong> PJ, Yang D, Bachstetter AD, Tilson HA, Harry GJ,Mervis RF, Kodavanti PRS, 2007: Ontogenetic alterations<strong>in</strong> molecular and structural correlates of dendritic growthafter developmental exposure to polychlor<strong>in</strong>ated biphenyls.Environ Health Perspect 115, 556–563.Li M-H, Hansen LG, 1996: Enzyme <strong>in</strong>duction and acuteendocr<strong>in</strong>e effects <strong>in</strong> prepubertal female rats receiv<strong>in</strong>g environmentalPCB ⁄ PCDF ⁄ PCDD mixtures. Environ HealthPerspect 104, 712–722.Mart<strong>in</strong> MB, Reiter R, Pham T, Avellanet YR, Camara J,Lahm M, Pentecost E, Pratap K, Gilmore BA, Divekar S,Dagata RS, Bull JL, Stoica A, 2003: Estrogen-like activity ofmetals <strong>in</strong> Mcf-7 breast cancer cells. Endocr<strong>in</strong>ology 144,2425–2436.Meerts IATM, Letcher RJ, Hov<strong>in</strong>g S, Marsh G, Bergman A,Lemmen JG, van der Berg B, Brouwer A, 2001: In vitroestrogenicity of polybrom<strong>in</strong>ated diphenyl ethers, hydroxylatedPBDEs, and polybrom<strong>in</strong>ated bisphenol A compounds.Environ Health Perspect 109, 399–407.Meijer GAL, de Bree JA, Wagenaar JA, Spoelstra SF, 1999:Sewerage overflows put production and fertility of dairycows at risk. J Environ Qual 28, 1381–1383.Naert C, Van Peteghem C, Kupper J, Jenni L, Naegeli H,2007: Distribution of polychlor<strong>in</strong>ated biphenyls and polybrom<strong>in</strong>ateddiphenyl ethers <strong>in</strong> birds of prey from Switzerland.Chemosphere 68, 977–987.Newbold RR, Padilla-Banks E, Snyder RJ, Phillips TM,Jefferson WN, 2007: Developmental exposure to endocr<strong>in</strong>edisruptors and the obesity epidemic. Reprod Toxicol 23,290–296.Noakes PS, Taylor P, Wilk<strong>in</strong>son S, Prescott SL, 2006: Therelationship between persistent organic pollutants <strong>in</strong>maternal and neonatal tissues and immune responses toallergens: a novel exploratory study. Chemosphere 63,1304–1311.Norstrom RJ, 2002: Understand<strong>in</strong>g bioaccumulation of POPs<strong>in</strong> food webs. Environ Sci Pollut Res 9, 300–303.Paul C, Rh<strong>in</strong>d SM, Kyle CE, Scott H, McK<strong>in</strong>nell C, SharpeRM, 2005: Cellular and hormonal disruption of fetal testisdevelopment <strong>in</strong> sheep reared on pasture treated with sewagesludge. Environ Health Perspect 113, 1530–1587.Payne J, Scholze M, Kortenkamp A, 2001: Mixtures of fourorganochlor<strong>in</strong>es enhance human breast cancer cell proliferation.Environ Health Perspect 109, 391–397.Pocar P, Perazzoli F, Luciano A, Gandolfi F, 2001: In vitroreproductive toxicity of polychlor<strong>in</strong>ated biphenyls: effectson oocyte maturation and developmental competence <strong>in</strong>cattle. Mol Reprod Dev 58, 411–416.Rajapakse N, Silva E, Kortenkamp A, 2002: Comb<strong>in</strong><strong>in</strong>gxenoestrogens at levels below <strong>in</strong>dividual no-observed-effectconcentrations dramatically enhances steroid hormoneaction. Environ Health Perspect 110, 917–921.Reid BJ, Jones KC, Semple KT, 2000: Bioavailability ofpersistent organic pollutants <strong>in</strong> soils and sediments – aperspective on mechanism, consequences and assessment.Environ Pollut 108, 103–112.Rh<strong>in</strong>d SM, 2002: Endocr<strong>in</strong>e disrupt<strong>in</strong>g compounds and farmanimals: their properties, actions and routes of exposure.Domest Anim Endocr<strong>in</strong>ol 23, 179–187.Rh<strong>in</strong>d SM, 2005: Are endocr<strong>in</strong>e disrupt<strong>in</strong>g compounds athreat to farm animal health, welfare and productivity?Reprod Dom Anim 40, 282–290.Rh<strong>in</strong>d SM, Smith A, Kyle CE, Telfer G, Mart<strong>in</strong> G, Duff E,Mayes RW, 2002: Phthalate and alkyl phenol concentrations<strong>in</strong> soil follow<strong>in</strong>g applications of <strong>in</strong>organic fertiliser orsewage sludge to pasture and potential rates of <strong>in</strong>gestion bygraz<strong>in</strong>g rum<strong>in</strong>ants. J Environ Monit 4, 142–148.Rh<strong>in</strong>d SM, Kyle CE, Owen J, 2005a: Accumulation ofpotentially toxic metals <strong>in</strong> the liver tissue of sheep grazedon sewage sludge-treated pastures. Anim Sci 81, 107–113.Rh<strong>in</strong>d SM, Kyle CE, Telfer G, Duff EI, Smith A, 2005b: Alkylphenols and diethylhexyl phthalate <strong>in</strong> tissues of sheepgraz<strong>in</strong>g pastures fertilised with sewage sludge or <strong>in</strong>organicfertilizer. Environ Health Perspect 113, 447–453.Rh<strong>in</strong>d SM, Kyle CE, Mackie C, Telfer G, 2007a: PCB andPBDE accumulation <strong>in</strong> adult and fetal sheep graz<strong>in</strong>g sludgetreated pastures. Proc Soc Environ Toxicol Chem, Norwich.5–6 September, 2007.Rh<strong>in</strong>d SM, Kyle CE, Mackie C, Telfer G, 2007b: Effects ofexposure of ewes to sewage sludge-treated pasture onphthalate and alkyl phenol concentrations <strong>in</strong> their milk.Sci Total Environ 383, 70–80.Sandahl JF, Baldw<strong>in</strong> DH, Jenk<strong>in</strong>s JJ, Scholz NL, 2007: Asensory system at the <strong>in</strong>terface between urban stormwaterrunoff and salmon survival. Environ Sci Technol 41, 2998–3004.Schecter A, Johnson-Welch S, Tung KC, Harris TR, Papke O,Rosen R, 2007: Polybrom<strong>in</strong>ated diphenyl ether (PBDE)levels <strong>in</strong> livers of U.S. human fetuses and newborns. JToxicol Environ Health 70, 1–6.Smith BS, 1981: Male characteristics on female mud snailscaused by antifoul<strong>in</strong>g pa<strong>in</strong>ts. J Appl Toxicol 1, 22–25.Smith SR, 1996: Agricultural recycl<strong>in</strong>g of sewage sludge andthe environment CAB International. Wall<strong>in</strong>gford, UK.Stevens JL, Northcott GL, Stern GA, Tomy GT, Jones KC,2003: PAHs, PCBs, PCNs, organochlor<strong>in</strong>e pesticides, syntheticmusks, and polychlor<strong>in</strong>ated n-alkanes <strong>in</strong> UK sewagesludge: survey results and implications. Environ Sci Technol37, 462–467.Suk WA, Olden K, Yang RSH, 2002: Chemical mixturesresearch: significance and future perspectives. EnvironHealth Perspect 110(Suppl. 6), 891–892.Thornton I, Abrahams P, 1983: Soil <strong>in</strong>gestion – a majorpathway of heavy metals <strong>in</strong>to livestock graz<strong>in</strong>g contam<strong>in</strong>atedland. Sci Total Environ 28, 287–294.Thornton JW, Need E, Crews D, 2003: Ancient orig<strong>in</strong> ofsteroid receptors and reconstitution of the ancestral receptor.Science 301, 1714–1717.T<strong>in</strong>well H, Ashby J, 2004: Sensitivity of the immature ratuterotrophic assay to mixtures of estrogens. Environ HealthPerspect 112, 575–582.Toppari J, Larsen JC, Christiansen P, Giwercman A, GrandjeanP, Guillette LJ, Jegou B, Jensen TK, Jouannet P,Keid<strong>in</strong>g N, Leffers H, McLachlan JA, Meyer O, Muller J,Meyts ER, Scheike T, Sharpe R, Sumpter J, Skakkebaek NE,1996: Male reproductive health and environmental xenoestrogens.Environ Health Perspect 104(Suppl. 4), 741–803.U.S. Central Pollution Control Board Newsletter, 2001: http://www.cpcb.nic.<strong>in</strong>/aug2001vi.htmU.S. Department of Health and Services, 2005: Report onCarc<strong>in</strong>ogens, 11th edn. Public Health Service, NationalToxicology Program (http://ntp.niehs.nih.gov/ntp/roc/toc11.html).Ó 2008 Macaulay Land Use Research Institute


22 SM Rh<strong>in</strong>dUzumcu M, Zachow R, 2007: Developmental exposure toenvironmental endocr<strong>in</strong>e disruptors: consequences with<strong>in</strong>the ovary and on female reproductive function. ReprodToxicol 23, 337–352.V<strong>in</strong>e MF, Ste<strong>in</strong> L, Weigle K, Schroeder J, Degnan D, Chiu-kitJT, Hanchette C, Backer L, 2000: Effects on the immunesystem associated with liv<strong>in</strong>g near a pesticide dump site.Environ Health Perspect 108, 1113–1124.Watk<strong>in</strong>s JB, Klaassen CD, 1986: Xenobiotic biotransformation<strong>in</strong> livestock: comparison to other species commonlyused <strong>in</strong> toxicity test<strong>in</strong>g. J Anim Sci 63, 933–942.Whyatt RM, Santella RM, Jedrychowski W, Garte SJ, BellDA, Ottman R, Gladek-Yarborough A, Cosma G, YoungT-L, Cooper TB, Randall MC, Manchester DK, Perera FP,1998: Relationshp between ambient air pollution and DNAdamage <strong>in</strong> Polish mothers and newborns. Environ HealthPerspect 106(Suppl. 3), 821–826.Wild SR, Jones KC, 1992: Organic chemicals enter<strong>in</strong>gagricultural soils <strong>in</strong> sewage sludges: screen<strong>in</strong>g for theirpotential to transfer to crop plants and livestock. Sci TotalEnviron 119, 85–119.Wilk<strong>in</strong>son JM, Hill J, Phillips JC, 2003: The accumulation ofpotentially-toxic metals by graz<strong>in</strong>g rum<strong>in</strong>ants. Proc NutrSoc 62, 267–277.Author’s address (for correspondence): SM Rh<strong>in</strong>d, MacaulayInstitute, Craigiebucker, Aberdeen, AB15 8QH, UK. E-mail:s.rh<strong>in</strong>d@macaulay.ac.ukConflict of <strong>in</strong>terest: The author declares no conflict of <strong>in</strong>terest.Ó 2008 Macaulay Land Use Research Institute


Reprod Dom Anim 43 (Supp. 2), 23–30 (2008); doi: 10.1111/j.1439-0531.2008.01139.xISSN 0936-6768Long Cha<strong>in</strong> Fatty Acids of Diet as Factors Influenc<strong>in</strong>g <strong>Reproduction</strong> <strong>in</strong> CattleJEP Santos 1 , TR Bilby 2 , WW Thatcher 1 , CR Staples 1 and FT Silvestre 11 Department of Animal Sciences, University of Florida, Ga<strong>in</strong>esville, FL; 2 Department of Animal Sciences, University of Arizona, Tucson, AZ, USAContentsCattle are fed moderate amounts of long cha<strong>in</strong> fatty acids (FA)with the objective to enhance lactation and growth; however,recent <strong>in</strong>terest on lipid feed<strong>in</strong>g to cows has focused onreproduction, immunity and health. Increas<strong>in</strong>g the caloricdensity of the ration by fat feed<strong>in</strong>g has generally improvedmeasures of cow reproduction, but when milk yield and bodyweight losses were <strong>in</strong>creased by fat supplementation, positiveeffects on reproduction were not always observed. Feed<strong>in</strong>g fathas <strong>in</strong>fluenced reproduction by alter<strong>in</strong>g the size of thedom<strong>in</strong>ant follicle, hasten<strong>in</strong>g the <strong>in</strong>terval to first postpartumovulation <strong>in</strong> beef cows, <strong>in</strong>creas<strong>in</strong>g progesterone concentrationsdur<strong>in</strong>g the luteal phase of the oestrous cycle, modulat<strong>in</strong>guter<strong>in</strong>e prostagland<strong>in</strong> (PG) synthesis, and improv<strong>in</strong>g oocyteand embryo quality and developmental competence. Some ofthese effects were altered by the type of FA fed. Thepolyunsaturated FA of the n-6 and n-3 families seem to havethe most remarkable effects on reproductive responses ofcattle, but it is not completely clear whether these effects aremediated only by them or by other potential <strong>in</strong>termediatesproduced dur<strong>in</strong>g rumen biohydrogenation. Generally, feed<strong>in</strong>gfat sources rich <strong>in</strong> n-6 FA dur<strong>in</strong>g late gestation and earlylactation enhanced follicle growth, uter<strong>in</strong>e PG secretion,embryo quality and pregnancy <strong>in</strong> cows. Similarly, feed<strong>in</strong>gn-3 FA dur<strong>in</strong>g lactation suppressed uter<strong>in</strong>e PG release, andimproved embryo quality and ma<strong>in</strong>tenance of pregnancy.Future research ought to focus on methods to improve thedelivery of specific FA and adequately powered studies shouldbe designed to critically evaluate their effects on establishmentand ma<strong>in</strong>tenance of pregnancy <strong>in</strong> cattle.IntroductionRum<strong>in</strong>ant diets are supplemented with fat primarily to<strong>in</strong>crease energy concentration and to enhance animalperformance. Dairy and beef cattle diets, without anysupplemental fat, conta<strong>in</strong> approximately 2% long-cha<strong>in</strong>fatty acids (LCFA) of vegetable orig<strong>in</strong> that are predom<strong>in</strong>antlypolyunsaturated. Because of the high energydensity, fats are usually <strong>in</strong>corporated <strong>in</strong>to cattle rationsto improve production, growth and reproduction.Dur<strong>in</strong>g early lactation, when lactat<strong>in</strong>g cows undergo aperiod of nutrient deficit, it was <strong>in</strong>itially thought that<strong>in</strong>corporat<strong>in</strong>g supplemental fat to the diet wouldenhance energy <strong>in</strong>take and energy balance, which wasexpected to improve reproduction. Because early lactationcows mobilize large quantities of stored triacylglycerols<strong>in</strong> adipose tissue, concentrations of fatty acids(FA) <strong>in</strong> blood are usually high dur<strong>in</strong>g the first weeks oflactation (Drackley 1999). This has been suggested tocause an unbalance <strong>in</strong> substrate supply to the cow,which compromises appetite and overall energy <strong>in</strong>take(Drackley 1999). When fat is fed <strong>in</strong> early lactation, oftencows either consume less diet or production <strong>in</strong>creases,therefore fat feed<strong>in</strong>g early postpartum seldom altersenergy status even though a more energy dense ration isconsumed. Staples et al. (1998) <strong>in</strong>dicated that feed<strong>in</strong>g fatdid not alter the energy status of dairy cows andsuggested that reproductive responses were the result ofsupply<strong>in</strong>g LCFA and alter<strong>in</strong>g substrate availability tothe cow rather than simply an energy effect.As with other nutrients, certa<strong>in</strong> FA are essential formammals. In 1929, George O. Burr and his wife werethe first to describe the essentiality of FA <strong>in</strong> rats(Burr and Burr 1929, 1930). They observed that grow<strong>in</strong>grats fed diets low <strong>in</strong> fat ceased grow<strong>in</strong>g and experiencedhealth problems and irregular ovulation, which werethen reversed after feed<strong>in</strong>g fat sources rich <strong>in</strong> thepolyunsaturated FA C18:2 n-6 (l<strong>in</strong>oleic acid) andC18:3 n-3 (a-l<strong>in</strong>olenic acid) (Burr and Burr 1930).Therefore, the concept of essential FA was establishedand later understood that C18:2 n-6 and C18:3 n-3 couldnot be synthesized by mammalian cells because of lackof desaturase enzymes beyond the 9th C <strong>in</strong> the acylcha<strong>in</strong>. Because of the essentiality of FA and the role ofspecific FA on reproductive processes, it is possible thatreproduction <strong>in</strong> cattle may be more <strong>in</strong>fluenced by thetype of fat fed than fat feed<strong>in</strong>g per se. This is particularlyimportant and challeng<strong>in</strong>g as rum<strong>in</strong>ants extensivelyhydrogenate polyunsaturated FA, thereby limit<strong>in</strong>g thesupply of dietary unsaturated FA for absorption <strong>in</strong> thesmall <strong>in</strong>test<strong>in</strong>e.Feed<strong>in</strong>g Fat and Fatty Acids to CattleLipids are important molecules that serve as a source ofenergy and are critical components of the physical andfunctional structure of cells. Lipids present <strong>in</strong> cellmembranes such as FA <strong>in</strong> phospholipids play animportant role <strong>in</strong> regulat<strong>in</strong>g the properties and activitiesof cell membranes. Changes <strong>in</strong> cha<strong>in</strong> length, degree ofunsaturation and position of the double bonds <strong>in</strong> theacyl cha<strong>in</strong> of FA can have remarkable impacts on theirfunction and may play a role <strong>in</strong> reproduction <strong>in</strong> cattle(Staples et al. 1998; Mattos et al. 2000), although theexact mechanisms are still unclear. Potential mechanismsmay <strong>in</strong>clude improved dietary energy density(Ferguson et al. 1990), altered follicle development(Staples and Thatcher 2005), <strong>in</strong>creased concentrationsof progesterone (Staples et al. 1998), suppressed luteolyticsignals around maternal recognition of pregnancy(Mattos et al. 2000), and improved embryo quality(Cerri et al. 2004).The use of fat <strong>in</strong> diets of dairy cattle usually<strong>in</strong>creases the energy density of the ration and improveslactation and reproduction, although improvements <strong>in</strong>reproduction occur <strong>in</strong> spite of provision of calories(Staples et al. 1998). These effects might be mediatedby the FA composition of the fat source; however, aÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


24 JEP Santos, TR Bilby, WW Thatcher, CR Staples and FT Silvestremajor impediment to the study of FA and reproduction<strong>in</strong> cattle is the <strong>in</strong>ability to predict the delivery ofspecific lipids, particularly polyunsaturated FA to thesmall <strong>in</strong>test<strong>in</strong>e for absorption, and the specific needs ofdifferent tissues for FA to modulate reproduction.Microbial activity <strong>in</strong> the rumen results <strong>in</strong> lipolysis oftriacylglycerols and biohydrogenation of unsaturatedFA which dramatically reduces the amount of polyunsaturatedFA reach<strong>in</strong>g the small <strong>in</strong>test<strong>in</strong>e forabsorption. In fact, Juchem (2007) demonstrated thatmore than 70% of the C18:2 n-6 and more than 85%of C18:3 n-3 fed to lactat<strong>in</strong>g cows were biohydrogenated<strong>in</strong> the rumen when fed as unprotected oils oras Ca salts of long cha<strong>in</strong> FA (Ca-LCFA), respectively.Therefore, if specific unsaturated FA are important forreproduction <strong>in</strong> cattle, it is critical that future researchwith lipids and reproduction aim to improve the extentof delivery of unsaturated FA for absorption. In spiteof the difficulties to deliver polyunsaturated FA torum<strong>in</strong>ants, studies have generally <strong>in</strong>dicated that thepolyunsaturated FA of the n-6 (l<strong>in</strong>oleic acid) and n-3[a-l<strong>in</strong>olenic acid; eicosapentaenoic (EPA), C20:5 n-3;docosahexaenoic (DHA), C22:6 n-3] families are themost beneficial to improv<strong>in</strong>g reproduction <strong>in</strong> cows.Fatty Acids and Postpartum Uter<strong>in</strong>e HealthUter<strong>in</strong>e health is an important risk factor for subsequentfertility <strong>in</strong> lactat<strong>in</strong>g dairy cows. Dur<strong>in</strong>g the process ofparturition, eicosanoids are produced <strong>in</strong> substantialamounts and play an important role <strong>in</strong> regulation andcontrol of parturition, and expulsion of the placenta anduter<strong>in</strong>e contents through open<strong>in</strong>g of the cervixand contractions of the uterus. Prostagland<strong>in</strong> F 2a is animportant eicosanoid that regulates CL lifespan andmight <strong>in</strong>fluence retention of foetal membranes andsubsequent uter<strong>in</strong>e health. Uter<strong>in</strong>e synthesis of PGF 2ais regulated <strong>in</strong> part by substrate availability, andarachidonic acid (AA; C20:4 n-6) is the precursor forPGF 2a synthesis, so it is plausible to suggest that<strong>in</strong>crements <strong>in</strong> AA content of endometrial tissue shouldenhance uter<strong>in</strong>e PGF 2a secretion, which <strong>in</strong> turn may<strong>in</strong>fluence uter<strong>in</strong>e health.Burns et al. (2003) fed non-lactat<strong>in</strong>g beef cows n-3FA from fish meal and reduced the endometrialconcentration of AA and <strong>in</strong>creased those of EPA andtotal n-3 FA. Similar effects have been observed withlactat<strong>in</strong>g dairy cows fed <strong>in</strong>creas<strong>in</strong>g amounts of fishmeal or Ca-LCFA enriched <strong>in</strong> fish oil (Bilby et al.2006b; Moussavi et al. 2007). Because of <strong>in</strong>corporationof n-6 and n-3 FA primarily <strong>in</strong> the phospholipidcomponent of endometrial tissue, it is possible thatchanges <strong>in</strong> FA content of the endometrial tissue mightmodulate endometrial secretion of PGF 2a <strong>in</strong> cows.Feed<strong>in</strong>g approximately 2% of the ration as fish oil rich<strong>in</strong> n-3 FA reduced the peripheral blood concentrationsof PGF 2a metabolite (PGFM) <strong>in</strong>dicat<strong>in</strong>g reduceduter<strong>in</strong>e secretion of PGF 2a (Mattos et al. 2004). Incontrast feed<strong>in</strong>g supplemental fat pre-partum conta<strong>in</strong><strong>in</strong>gapproximately 30% of FA as C18:2 n-6 <strong>in</strong>creaseduter<strong>in</strong>e secretion of PGF 2a based on PGFM <strong>in</strong> blood(Cullens et al. 2004). Increased synthesis of PGF 2awhen cows were supplemented with n-6 FA pre-partummight enhance the potential for uter<strong>in</strong>e and immunecells to secrete eicosanoids which may <strong>in</strong>fluencepostpartum uter<strong>in</strong>e health and immuno-competenceof the cow. Collectively, these data <strong>in</strong>dicate thatfeed<strong>in</strong>g fat sources differ<strong>in</strong>g <strong>in</strong> FA profile dur<strong>in</strong>g thetransition period can <strong>in</strong>fluence the natural release ofPGF 2a by the uterus of the cow.Three studies exam<strong>in</strong>ed the effect of feed<strong>in</strong>g fat prepartumon postpartum health of dairy cows (Cullenset al. 2004; Juchem 2007; Silvestre, unpublished data).When cows were supplemented with Ca-LCFA rich <strong>in</strong>n-6 FA pre-partum, <strong>in</strong>cidence of postpartum diseases<strong>in</strong>clud<strong>in</strong>g reta<strong>in</strong>ed placenta, metritis and mastitis wasreduced (8.3% vs 42.9%) compared with cows not fedfat pre-partum (Cullens et al. 2004). Juchem (2007)supplemented the diet of 501 pre-partum dairy cowswith 2% Ca-LCFA of either palm oil or a blend ofC18:2 n-6 and trans-octadecenoic FA. Incidence ofreta<strong>in</strong>ed placenta did not differ between treatments(6.6%). Risk of uter<strong>in</strong>e disease was similar betweensources of FA, but cows fed the blend of C18:2 n-6and trans-octadecenoic FA had reduced the odds ofpuerperal metritis (8.8% vs 15.1%; adjusted oddsratio ¼ 0.53). Rate of uter<strong>in</strong>e <strong>in</strong>volution did not differ,and 91% of the cows had completed uter<strong>in</strong>e <strong>in</strong>volutionat the last ultrasonography on week 6 postpartum(Juchem 2007). In a similar attempt, Silvestre (unpublisheddata) fed 1167 pre-partum dairy cows 1.5% ofthe ration as Ca-LCFA of either palm oil or saffloweroil. Feed<strong>in</strong>g a fat source rich <strong>in</strong> C18:2 n6 enhancedmeasures of <strong>in</strong>nate immunity; however, <strong>in</strong>cidences ofreta<strong>in</strong>ed placenta (10.1%), metritis (17.4%), andpurulent cervical discharge (29%) did not differbetween treatments. These data suggest that, althoughfeed<strong>in</strong>g fat sources rich <strong>in</strong> n-6 FA may enhanceimmune responses and have pro-<strong>in</strong>flammatory effects,its impacts on uter<strong>in</strong>e health are subtle.Fatty Acids, Follicle Development andResumption of Postpartum CyclicityOne of the mechanisms by which fat feed<strong>in</strong>g mightimprove fertility <strong>in</strong> cattle is by <strong>in</strong>fluenc<strong>in</strong>g folliclegrowth and ovulation (Lucy et al. 1993). Lucy et al.(1991) replaced corn with Ca-LCFA <strong>in</strong> the diet fed todairy cows beg<strong>in</strong>n<strong>in</strong>g at parturition, and feed<strong>in</strong>gCa-LCFA <strong>in</strong>creased the number of medium (6–9 mm)sized follicles with<strong>in</strong> 25 days postpartum, and that offollicles >15 mm <strong>in</strong> a synchronized oestrous cycle. Inaddition, diameter of the largest (18.2 vs 12.4 mm)follicle was greater <strong>in</strong> cows fed Ca-LCFA. When thisstudy was repeated with isocaloric diets, similar effectswere observed (Lucy et al. 1993). Staples and Thatcher(2005) summarized the effects of supplemental fats onthe size of the dom<strong>in</strong>ant follicle (Table 1). On average,dom<strong>in</strong>ant follicle diameter was 3.2 mm larger, whichrepresents a 23% <strong>in</strong>crease <strong>in</strong> fat supplemented cows.Several studies have shown that dom<strong>in</strong>ant folliclediameter <strong>in</strong>creased <strong>in</strong> cows fed diets enriched <strong>in</strong>polyunsaturated FA compared with monounsaturatedFA, suggest<strong>in</strong>g differential effects of FA on folliclegrowth (Staples et al. 2000; Bilby et al. 2006a). Folliclesfrom cows abomasally <strong>in</strong>fused with yellow greaseÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Factors Influenc<strong>in</strong>g <strong>Reproduction</strong> <strong>in</strong> Cattle 25Table 1. Effect of supplemental fat on the diameter of the dom<strong>in</strong>antovarian follicle of lactat<strong>in</strong>g dairy cows (from Staples and Thatcher2005)ReferenceFatsourceExperimental dietsControl (mm)Fat (mm)Ambrose et al. (2006) Rolled flaxseeds 14.1 16.9Beam and Butler (1997) Tallow, yellow grease 11.0 13.5Bilby et al. (2006a) Ca-LCFA or flaxseed oil 15.0 16.5Lucy et al. (1991) Ca-LCFA 12.4 18.2Lucy et al. (1993) Ca-LCFA 16.0 18.6Oldick et al. (1997) Yellow grease 16.9 20.9Rob<strong>in</strong>son et al. (2002) Protected soybeans 13.3 16.9Staples et al. (2000) Soybean oil, fish oil 14.3 17.1Average 14.1 17.3Ca-LCFA = Ca salts of long cha<strong>in</strong> fatty acids from palm oil or a blend of palmand soyabean oils.Control vs fat was P < 0.10 for each study.grew faster to a larger diameter than follicles fromcows <strong>in</strong>fused with tallow (Oldick et al. 1997). Itappears that fat feed<strong>in</strong>g, but more importantly typeof fat, stimulates follicle growth <strong>in</strong> cows. The impact oflarger ovarian follicles on fertility because of fatsupplementation has not been def<strong>in</strong>ed, but cowsexperienc<strong>in</strong>g earlier postpartum ovulation have beenreported to have larger follicles (Beam and Butler1997). Therefore, it is possible that <strong>in</strong>creas<strong>in</strong>g thenumber and size of larger follicles by feed<strong>in</strong>g fat canreduce the <strong>in</strong>terval from calv<strong>in</strong>g to first postpartumovulation, which has been observed for postpartumbeef cows (Lammoglia et al. 1996, 1997; De Fries et al.1998).Although some studies have <strong>in</strong>dicated that feed<strong>in</strong>g fathastens follicle growth, which might <strong>in</strong>fluence resumptionof postpartum ovulation (Lammoglia et al. 1996,1997; De Fries et al. 1998), it is unclear whethersupplemental fats differ<strong>in</strong>g <strong>in</strong> FA profile have anydifferential effect on resumption of cyclicity. Juchem(2007) fed 699 multiparous cows either 400 g of FAfrom tallow or from Ca-LCFA conta<strong>in</strong><strong>in</strong>g palm and fishoils and observed no difference <strong>in</strong> proportion of cycl<strong>in</strong>gcows at 65 days postpartum (83.2% vs 82.2%, respectively).Subsequently, dairy cows supplemented withCa-LCFA of palm oil or a blend of C18:2 n-6 and transoctadecenoicFA from 25 days pre-partum to 80 dayspostpartum experienced a similar mean <strong>in</strong>terval to firstpostpartum ovulation (30.5 and 32.2 days, respectively;Juchem 2007). Recently, Silvestre (unpublished data) fedcows (n = 1055) either Ca-LCFA of palm oil or ofsafflower oil from 2 weeks pre-partum to 4 weekspostpartum, and then half of the cows <strong>in</strong> each transitiontreatment group were switched to either Ca-LCFA ofpalm oil or fish oil (264 ⁄ treatment). The proportions ofcyclic cows at 63 days postpartum were 84.2%, 79.5%,79.2% and 77.1% for cows fed palm oil ⁄ palm oil, palmoil ⁄ fish oil, safflower ⁄ palm oil, and safflower ⁄ fish oil,respectively, and they did not differ. Taken together,these data demonstrate that type of supplemental FA,whether more saturated or unsaturated does not <strong>in</strong>fluenceresumption of postpartum cyclicity <strong>in</strong> lactat<strong>in</strong>gdairy cows.Fatty Acids and OestradiolOestradiol has stimulatory effects on uter<strong>in</strong>e secretion ofPGF 2a (Knickerbocker et al. 1986), and can <strong>in</strong>crease thesensitivity of the CL to PGF 2a (Howard et al. 1990)which may enhance regression of the CL. Thus loweredplasma oestradiol may help prevent pre-mature CLregression and early embryonic mortality. Oldick et al.(1997) reported that abomasal <strong>in</strong>fusion of tallow oryellow grease reduced concentrations of plasma oestradiolon days 15 to 20 of a synchronized oestrous cyclecompared with cows <strong>in</strong>fused with glucose, a responsethat also has been observed <strong>in</strong> beef cows supplementedwith lipids (Hightshoe et al. 1991). Also, oestradiolconcentration was reduced <strong>in</strong> the follicular fluid frombeef cows fed soybean oil (Ryan et al. 1992). Although areduction <strong>in</strong> follicular oestradiol caused by fat feed<strong>in</strong>gmight potentially benefit CL lifespan, it may be detrimentalto expression of oestrus and uter<strong>in</strong>e prim<strong>in</strong>gdur<strong>in</strong>g prooestrus.Fat and Luteal FunctionImproved fertility <strong>in</strong> cattle has been associated with<strong>in</strong>creased circulat<strong>in</strong>g concentrations of progesteronedur<strong>in</strong>g the luteal phase before and after AI. Additionof fat to cattle diets has consistently shown to <strong>in</strong>creaseplasma cholesterol and cholesterol content <strong>in</strong> follicularfluid and <strong>in</strong> the CL (Staples et al. 1998; Williams 1989;Ryan et al. 1992; Hawk<strong>in</strong>s et al. 1995; Lammoglia et al.1996). Cholesterol serves as a precursor for the synthesisof progesterone by ovarian cells and both high and lowdensity lipoprote<strong>in</strong>s deliver cholesterol to ovarian tissuesfor steroidogenesis (Grummer and Carroll 1991).Hypercholesterolemia may <strong>in</strong>crease CL steroidogenesis;however, the <strong>in</strong>creased plasma progesterone concentrations<strong>in</strong> dairy and beef cows fed fat (Table 2) may beexpla<strong>in</strong>ed possibly by reduced progesterone clearance,not by <strong>in</strong>creased synthesis (Hawk<strong>in</strong>s et al. 1995).Fatty Acids, Oocyte Quality and MembraneCompositionCompetence of the oocyte and embryo is related to FAcomposition; specifically, phospholipid content of thecellular membrane plays a vital role <strong>in</strong> developmentdur<strong>in</strong>g and after fertilization. The amount of lipid <strong>in</strong> therum<strong>in</strong>ant oocyte is approximately 20-fold greater thanthat of the mouse (76 vs 4 ng) and consists (w ⁄ w) ofapproximately 50% triacylglycerol, 20% phospholipid,20% cholesterol and 10% free FA (McEvoy et al. 2000).Previous studies showed that C16:0 and C18:1 were themost abundant FA <strong>in</strong> the phospholipid fraction ofoocytes from cattle and may function as an energyreserve (Kim et al. 2001; Zeron et al. 2001). PolyunsaturatedFA comprised


26 JEP Santos, TR Bilby, WW Thatcher, CR Staples and FT SilvestreTable 2. Effect of supplemental fat on plasma progesterone concentrations<strong>in</strong> dairy and beef cattleMeasurementProgesterone,ng ⁄ mlControlp


Factors Influenc<strong>in</strong>g <strong>Reproduction</strong> <strong>in</strong> Cattle 27Feed<strong>in</strong>g n-3 FA can attenuate endometrial PGF 2aproduction. Feed<strong>in</strong>g fish meal to dairy cows attenuatedthe decrease <strong>in</strong> plasma progesterone concentrations2 days after PGF 2a <strong>in</strong>jection suggest<strong>in</strong>g changes <strong>in</strong> CLregression because of fish oil FA (Burke et al. 1996).Mattos et al. (2002) demonstrated that FA from fishmeal reduced plasma PGFM concentrations comparedwith unsupplemented cows after an oestradiol ⁄ oxytoc<strong>in</strong>challenge. Dairy cows fed fish oil dur<strong>in</strong>g the transitionperiod had greater EPA and DHA concentrations <strong>in</strong>caruncular tissues and reduced postpartum concentrationsof PGFM compared with cows fed olive oil(Mattos et al. 2004). Conversely, feed<strong>in</strong>g fat sources rich<strong>in</strong> n-6 FA <strong>in</strong>creased plasma PGFM after an oxytoc<strong>in</strong>challenge (Rob<strong>in</strong>son et al. 2002; Petit et al. 2004). Thussupplemental lipids can either <strong>in</strong>hibit or stimulate PGsecretion depend<strong>in</strong>g upon the specific FA.Incubation of bov<strong>in</strong>e endometrial cells with AA stimulatedPGF 2a production compared with cells not supplementedwith FA. On the other hand, cellssupplemented with n-3 FA had reduced secretion ofPGF 2a (Mattos et al. 2003). The mechanism by which n-3FA <strong>in</strong>hibit PGF 2a secretion may <strong>in</strong>volve decreas<strong>in</strong>g theavailability of AA precursor, <strong>in</strong>creas<strong>in</strong>g the concentrationof FA that compete with AA for process<strong>in</strong>g by PGHS-2,or <strong>in</strong>hibition of PGHS-2 (Mattos et al. 2000). Bilby et al.(2006b) concluded that n-3 FA supplementation tolactat<strong>in</strong>g dairy cows had little effect on the endometrialcomponents that regulate the PG cascade. Instead EPAand DHA exerted their regulatory effects as alternativesubstrates that reduced the lipid pools of AA. In supportof these conclusions, Burns et al. (2003) demonstratedthat feed<strong>in</strong>g fish meal reduced the endometrial concentrationof AA and <strong>in</strong>creased those of EPA and total n-3FA. Therefore, different FA can alter PGF 2a secretion by<strong>in</strong>fluenc<strong>in</strong>g FA availability <strong>in</strong> the endometrial tissue, andsupply<strong>in</strong>g FA that <strong>in</strong>hibit PGF 2a release by the uterusmight improve the mechanism of embryo preservation,which may benefit embryonic survival <strong>in</strong> cattle.Fatty Acids and Fertility of CowsStudies evaluat<strong>in</strong>g the effects of supplemental fat onreproductive performance of beef cattle are limited. Toour knowledge, no controlled trials have been conductedwith adequate number of animals to evaluate thepotential for fat supplementation to impact establishmentand ma<strong>in</strong>tenance of pregnancy of beef cows. DeFries et al. (1998) observed a tendency (p = 0.09) for<strong>in</strong>creased pregnancy <strong>in</strong> Brahman cows fed 5.2% fatcompared with cows fed 3.7% fat <strong>in</strong> the diet; however,the number of cows used <strong>in</strong> this study was limited toonly 20 per treatment.Feed<strong>in</strong>g fat to dairy cattle might improve pregnancyper AI (Table 3), although responses have not beenconsistent. When fat feed<strong>in</strong>g <strong>in</strong>creased postpartum bodyweight loss, primiparous cows fed fat had reducedpregnancy at first AI (Sklan et al. 1994). However,Ferguson et al. (1990) observed a 2.2-fold <strong>in</strong>creased odds(odds ratio = 2.2) of becom<strong>in</strong>g pregnant at first and allAI <strong>in</strong> lactat<strong>in</strong>g cows fed 0.5 kg ⁄ day of fat, which tended(p = 0.08) to enhance the proportion of pregnant cowsat the end of the study (93% vs 86.2%). In graz<strong>in</strong>g cows,Table 3. Effect of fat supplementation on pregnancy at first postpartumAI <strong>in</strong> lactat<strong>in</strong>g dairy cowsReferenceCowsFatsource andamountPregnancy perAI, %ControlFerguson et al. (1990) 253 0.5 kg of saturated 42.6 59.1 *free FAMcNamara et al. (2003) 201 0.32 to 0.36 kg 35.5 51.1 *of FA fromCa-LCFASch<strong>in</strong>goethe153 Oilseeds 46.5 42.0and Casper (1991)Schneider et al. (1988) 181 0.5 kg of Ca-LCFA 43.1 60.5Scott et al. (1995) 443 0.45 kg of Ca-LCFA 49.3 45.7Sklan et al. (1991) 99 2.6% of ration 41.5 39.2as Ca-LCFASklan et al. (1994) 102 2.5 of ration asCa-LCFAPrimiparous 73.7 * 33.3Multiparous 42.1 33.3FA = fatty acid; Ca-LCFA = Ca salts of long cha<strong>in</strong> fatty acids from palm oil.* With<strong>in</strong> a row, effect of supplemental fat (p < 0.05).supplementation with 0.35 kg of FA improved pregnancyafter the first postpartum AI, although theproportion of cows pregnant at the end of the study didnot differ (McNamara et al. 2003). Feed<strong>in</strong>g Ca-LCFA ofpalm oil improved pregnancy of dairy cows (Schneideret al. 1988; Sklan et al. 1991), but the authors did notreport statistical significance. On the other hand, othersdid not observe improvements on fertility of dairy cowssupplemented with Ca-LCFA (Sklan et al. 1994; Scottet al. 1995) or oilseeds (Sch<strong>in</strong>goethe and Casper 1991),which might be attributed to <strong>in</strong>creased milk yield andbody weight losses (Sklan et al. 1991, 1994).Because the benefits of feed<strong>in</strong>g fat may orig<strong>in</strong>ate fromspecific FA (Staples et al. 1998; Staples and Thatcher2005), and absorption of unsaturated FA is limited <strong>in</strong>rum<strong>in</strong>ants because of microbial biohydrogenation <strong>in</strong> therumen (Juchem 2007), studies have evaluated whetherfeed<strong>in</strong>g FA differ<strong>in</strong>g <strong>in</strong> the degree of saturation might<strong>in</strong>fluence fertility of dairy cows (Table 4). When cowswere fed 0.75 kg of fat from flaxseed, a source rich <strong>in</strong>C18:3 n-3, or sunflower seed, a source rich <strong>in</strong> C18:2 n-6,pregnancy tended (p = 0.07) to be greater for cows fedn-3 FA (Ambrose et al. 2006). However, a similarresponse to flaxseed was not observed by others (Petitand Twagiramungu 2006; Fuentes et al. 2008). Similarly,feed<strong>in</strong>g n-3 FA from fish oil as Ca-LCFA did notimprove pregnancy at first postpartum AI when comparedwith feed<strong>in</strong>g beef tallow (Juchem 2007) or withCa-LCFA of palm oil (Silvestre, unpublished data),although pregnancy at second postpartum AI wasgreater for cows fed n-3 FA (Silvestre, unpublisheddata). Juchem (2007) evaluated the effect of feed<strong>in</strong>g preandpostpartum cows Ca-LCFA of palm oil or a blendof C18:2 n-6 and trans-octadecenoic FA. Cows fedunsaturated FA were 1.5 times more likely to bepregnant at 27 or 41 days after AI compared with cowsfed palm oil. Improvements <strong>in</strong> pregnancy when cowswere fed Ca salts of a mix of C18:2 n-6 and transoctadecenoicFA were supported by <strong>in</strong>creased fertilizationand embryo quality <strong>in</strong> non-superovulated lactat<strong>in</strong>gdairy cows (Cerri et al. 2004).FatÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


28 JEP Santos, TR Bilby, WW Thatcher, CR Staples and FT SilvestreTable 4. Effect of fatty acid (FA) supplementation on pregnancy per AI <strong>in</strong> lactat<strong>in</strong>g dairy cowsFA source – Pregnancy per AI, %Reference Cows Amount ⁄ daySaturated n-6 FA n-3 FAAmbrose et al. (2006) 121 0.75 kg of fat – 32.2 48.4**Fuentes et al. (2008) 356 0.40 kg of fat – 39.2 38.8Juchem (2007) 699 0.40 kg of FA 40.7 – 35.9Juchem (2007) 323 0.40 kg of FA 22.8 – 24.8Juchem (2007) 344 2% of ration 28.6 37.9** –Petit and Twagiramungu (2006) 110 0.6–0.8 kg of FA 55.9 40.0 44.4Silvestre (unpublished data) – first AI 1055 1.5% of rationTransition 39.0 35.9 –>30 d postpartum 37.3 – 37.6Silvestre (unpublished data) – second AI 604 1.5% of rationTransition 29.0 34.5 –>30 d postpartum 27.2 – 37.0 *Saturated = mostly saturated and monounsaturated FA; n-6 FA = source rich <strong>in</strong> C18:2 n-6; n-3 FA = source rich <strong>in</strong> C18:3 n-3 or C20:5 n-3 + C22:6 n-3.* With<strong>in</strong> a row, effect of source of FA (p < 0.05).** With<strong>in</strong> a row, effect of source of FA (p < 0.07).Table 5. Effect of fatty acid (FA) supplementation on pregnancy losses after first postpartum AI <strong>in</strong> lactat<strong>in</strong>g dairy cowsFA source – Pregnancy loss, %Reference Pregnancies Amount ⁄ daySaturated n-6 FA n-3 FAAmbrose et al. (2006) 77 0.75 kg of fat – 27.3 9.8 *Juchem (2007) 257 0.40 kg of FA 20.4 – 23.5Juchem (2007) 77 0.40 kg of FA 5.4 – 10.0Juchem (2007) 114 2% of the ration 9.8 6.3 –Petit and Twagiramungu (2006) 51 0.6–0.8 kg of FA 21.1 12.5 0 *Silvestre (unpublished data) 388 1.5% of rationTransition 8.3 12.1 –>30 d postpartum 13.6 – 6.3 *Saturated = mostly saturated and monounsaturated FA; n-6 FA = source rich <strong>in</strong> C18:2 n-6; n-3 FA = source rich <strong>in</strong> C18:3 n-3 or C20:5 n-3 + C22:6 n-3.* With<strong>in</strong> a row, effect of source of FA (p < 0.05).Because n-3 FA can suppress uter<strong>in</strong>e secretion ofPGF 2a (Mattos et al. 2002, 2003, 2004), it may improveembryonic survival <strong>in</strong> cattle (Mattos et al. 2000). Inthree of five experiments, feed<strong>in</strong>g the n-3 FA C18:3 n-3(Ambrose et al. 2006; Petit and Twagiramungu 2006) orEPA and DHA (Silvestre, unpublished data) reducedpregnancy losses <strong>in</strong> lactat<strong>in</strong>g dairy cows (Table 5). Onthe other hand, when n-6 FA were fed as Ca-LCFA,pregnancy losses were similar to those observed for cowsfed Ca-LCFA of palm oil (Juchem 2007; Silvestre,unpublished data).Collectively, these data suggest that feed<strong>in</strong>g fat to dairycows generally improves fertility and responses areobserved with the energy <strong>in</strong>crement <strong>in</strong> the diet; also, thesedata suggest that fertility responses to fat feed<strong>in</strong>g arealtered accord<strong>in</strong>g to the type of dietary FA, althoughresponses are not always consistent. Feed<strong>in</strong>g n-3 FA fromflaxseeds or as Ca-LCFA improved pregnancy per AI <strong>in</strong>some, but not all studies. Similarly, feed<strong>in</strong>g Ca-LCFArich <strong>in</strong> n-6 FA improved pregnancy per AI <strong>in</strong> one of twoexperiments with lactat<strong>in</strong>g dairy cows. Although feed<strong>in</strong>gn-3 FA has not consistently <strong>in</strong>creased the risk ofpregnancy, it has reduced pregnancy losses <strong>in</strong> dairy cows.ConclusionsFat is recommended to be <strong>in</strong>corporated <strong>in</strong>to dairy cattlediets at moderate amounts. Feed<strong>in</strong>g fat to cattlegenerally improved establishment and ma<strong>in</strong>tenance ofpregnancy, but benefits to fertility can be negated whenweight losses are exacesbated by fat feed<strong>in</strong>g. Potentialimprovements <strong>in</strong> fertility of cows caused by fat feed<strong>in</strong>ghave generally been associated with enhanced follicledevelopment postpartum, <strong>in</strong>creased diameter of theovulatory follicle, <strong>in</strong>creased progesterone concentrationsdur<strong>in</strong>g the luteal phase of the cycle, altered uter<strong>in</strong>e⁄ embryo cross-talk by modulat<strong>in</strong>g PG synthesis,and improved oocyte and embryo quality. Some ofthese effects have been more <strong>in</strong>fluenced by the type offatty acid than by fat feed<strong>in</strong>g per se. Differentialresponses <strong>in</strong> vivo to FA feed<strong>in</strong>g suggest that unsaturatedFA of the n-6 and n-3 families were most beneficial.AcknowledgementsPart of the research mentioned on this manuscript and conducted bythe authors was supported by the National Research InitiativeCompetitive Grant no. 2004-35203-14137 from the USDA CooperativeState Research, Education, and Extension Service.ReferencesAmbrose DK, Kastelic JP, Corbett R, Pitney PA, Petit HV,Small JA, Zalkovic P, 2006: Lower pregnancy losses <strong>in</strong>lactat<strong>in</strong>g dairy cows fed a diet enriched <strong>in</strong> a-l<strong>in</strong>olenic acid.J Dairy Sci 89, 3066–3074.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Factors Influenc<strong>in</strong>g <strong>Reproduction</strong> <strong>in</strong> Cattle 29Balaguer SA, Persh<strong>in</strong>g RA, Rodriguez-Sallaberry C, ThatcherWW, Bad<strong>in</strong>ga L, 2005: Effects of bov<strong>in</strong>e somatotrop<strong>in</strong> onuter<strong>in</strong>e genes related to the prostagland<strong>in</strong> cascade <strong>in</strong>lactat<strong>in</strong>g dairy cows. J Dairy Sci 88, 543–552.Beam SW, Butler WR, 1997: Energy balance and ovarianfollicle development prior to the first ovulation postpartum<strong>in</strong> dairy cows receiv<strong>in</strong>g three levels of dietary fat. BiolReprod 56, 133–142.Bilby TR, Block J, do Amaral BC, Sa Filho O, Silvestre FT,Hansen PJ, Staples CR, Thatcher WW, 2006a: Effects ofdietary unsaturated fatty acids on oocyte quality andfollicular development <strong>in</strong> lactat<strong>in</strong>g dairy cows <strong>in</strong> summerJ. Dairy Sci 89, 3891–3903.Bilby TR, Guzeloglu A, MacLaren LA, Staples CR, ThatcherWW, 2006b: Pregnancy, bST and omega-3 fatty acids <strong>in</strong>lactat<strong>in</strong>g dairy cows: II. Gene expression related to ma<strong>in</strong>tenanceof pregnancy. J Dairy Sci 89, 3375–3385.Burke JM, Staples CR, Risco CA, De La Sota RL, ThatcherWW, 1996: Effect of rum<strong>in</strong>ant grade menhaden fish meal onreproductive and productive performance of lactat<strong>in</strong>g dairycows. J Dairy Sci 80, 3386–3398.Burns PD, Engle TE, Harris MA, Enns RM, Whittier JC,2003: Effect of fish meal supplementation on plasma andendometrial fatty acid composition <strong>in</strong> nonlactat<strong>in</strong>g beefcows. J Anim Sci 81, 2840–2846.Burr GO, Burr MM, 1929: A new disease produced bythe rigid exclusion of fat from the diet. J Biol Chem 82, 345–367.Burr GO, Burr MM, 1930: The nature and role of the fattyacids essential <strong>in</strong> nutrition. J Biol Chem 86, 587–621.Carroll DJ, Jerred MJ, Grummer RR, Combs DK, PiersonRA, Hauser ER, 1990: Effects of fat supplementation andimmature alfalfa to concentrate ratio on plasma progesterone,energy balance, and reproductive traits of dairy cattle.J Dairy Sci 73, 2855–2863.Cerri RLA, Bruno RGS, Chebel RC, Galva˜ o KN, RutglianoH, Juchem SO, Thatcher WW, Luch<strong>in</strong>i D, Santos JEP, 2004:Effect of fat sources differ<strong>in</strong>g <strong>in</strong> fatty acid profile onfertilization rate and embryo quality <strong>in</strong> lactat<strong>in</strong>g dairy cows.J Dairy Sci 87(Suppl. 1), 297 (abstr).Cullens FM, Staples CR, Bilby TR, Silvestre FT, Bartolome J,Sozzi A, Bad<strong>in</strong>ga L, Thatcher WW, Arth<strong>in</strong>gton JD, 2004:Effect of tim<strong>in</strong>g of <strong>in</strong>itiation of fat supplementation on milkproduction, plasma hormones and metabolites, and conceptionrates of Holste<strong>in</strong> cows <strong>in</strong> summer. J Dairy Sci 86(Suppl.1), 308 (abstr).De Fries CA, Neuendorff DA, Randel RD, 1998: Fatsupplementation <strong>in</strong>fluences postpartum reproductive performance<strong>in</strong> Brahman cows. J Anim Sci 76, 864–870.Drackley JK, 1999: ADSA foundation scholar award. Biologyof dairy cows dur<strong>in</strong>g the transition period: the f<strong>in</strong>al frontier?J Dairy Sci 82, 2259–2273.Ferguson JD, Sklan D, Chalupa WV, Kronfeld DS, 1990:Effects of hard fats on <strong>in</strong> vitro and <strong>in</strong> vivo rumenfermentation, milk production, and reproduction <strong>in</strong> dairycows. J Dairy Sci 73, 2864–2879.Fouladi-Nashta AA, Gutierrez CG, Gong JG, GarnsworthyPC, Webb R, 2007: Impact of dietary fatty acids on oocytequality and development <strong>in</strong> lactat<strong>in</strong>g dairy cows. BiolReprod 77, 9–17.Fuentes MC, Calsamiglia SS, Sa´nchez C, Gonza´lez A,Newbold J, Santos JEP, Rodrı´guez-Alcala´ LM, FontechaJ, 2008: Effect of extruded l<strong>in</strong>seed on productive andreproductive performance of lactat<strong>in</strong>g dairy cows. LivestSci 113, 144–154.Garcia-Bojalil CM, Staples CR, Risco CA, Savio JD, ThatcherWW, 1998: Prote<strong>in</strong> degradability and calcium salts of longcha<strong>in</strong>fatty acids <strong>in</strong> the diets of lactat<strong>in</strong>g dairy cows:productive responses. J Dairy Sci 81, 1374–1384.Grummer RR, Carroll DJ, 1991: Effects of dietary fat onmetabolic disorders and reproductive performance of dairycattle. J Anim Sci 69, 3838–3852.Hawk<strong>in</strong>s DE, Niswender KD, Oss GM, Moeller CL, OddeKG, Sawyer HR, Niswender GD, 1995: An <strong>in</strong>crease <strong>in</strong>serum lipids <strong>in</strong>creases luteal lipid content and alters thedisappearance rate of progesterone <strong>in</strong> cows. J Anim Sci 73,541–545.Hightshoe RB, Cochran RC, Corah LR, Kiracofe GH,Harmon DL, Perry RC, 1991: Effects of calcium soaps offatty acids on postpartum reproductive function <strong>in</strong> beefcows. J Anim Sci 69, 4097–4103.Howard JJ, Scott RG, Britt JH, 1990: Associations amongprogesterone, estradiol-17b, and prostagland<strong>in</strong> <strong>in</strong> cattletreated with hCG dur<strong>in</strong>g diestrus to extend corpus luteumfunction. Prostagland<strong>in</strong>s 40, 51–70.Juchem SO, 2007: Lipid Digestion and Metabolism <strong>in</strong> DairyCows: Effects on Production, <strong>Reproduction</strong> and Health.PhD thesis, University of California Davis.Kim JY, K<strong>in</strong>oshita M, Ohnishi M, Fukui Y, 2001: Lipid andfatty acid analysis of fresh and frozen-thawed immature and<strong>in</strong> vitro matured bov<strong>in</strong>e oocytes. <strong>Reproduction</strong> 122, 131–138.Knickerbocker JJ, Thatcher WW, Foster DB, Wolfenson D,Bartol FF, Caton D, 1986: Uter<strong>in</strong>e prostagland<strong>in</strong> and bloodflow responses to estradiol-17b <strong>in</strong> cyclic cattle. Prostagland<strong>in</strong>s31, 757–776.Lammoglia MA, Willard ST, Oldham JR, Randel RD, 1996:Effects of dietary fat and season on steroid hormonalprofiles before parturition and on hormonal, cholesterol,triglycerides, follicular pattern, and postpartum reproduction<strong>in</strong> Brahman cows. J Anim Sci 74, 2253–2262.Lammoglia MA, Williard ST, Hallford DM, Randel RD,1997: Effects of dietary fat on follicular development andcirculat<strong>in</strong>g concentrations of lipids, <strong>in</strong>sul<strong>in</strong>, progesterone,estradiol-17b, 13,14-dihydro-15-keto-prostagland<strong>in</strong> F2a andgrowth hormone <strong>in</strong> estrous cyclic Brahman cows. J Anim Sci75, 1591–1600.Lim H, Gupta RA, Ma WG, Paria BC, Moller DE, MorrowJD, BuBois RN, Trzaskos JM, Dey SK, 1999: Cyclooxygenase-2-derivedprostacycl<strong>in</strong> mediates embryo implantation<strong>in</strong> the mouse via PPARd. Gene Dev 13, 1561–1574.Lucy MC, Staples CR, Michel FM, Thatcher WW, Bolt DJ,1991: Effect of feed<strong>in</strong>g calcium soaps to early postpartumdairy cows on plasma prostagland<strong>in</strong> F 2a , lute<strong>in</strong>iz<strong>in</strong>g hormone,and follicular growth. J Dairy Sci 74, 483–489.Lucy MC, De La Sota RL, Staples CR, Thatcher WW, 1993:Ovarian follicular populations <strong>in</strong> lactat<strong>in</strong>g dairy cowstreated with recomb<strong>in</strong>ant bov<strong>in</strong>e somatotrop<strong>in</strong> (sometribove)or sal<strong>in</strong>e and fed diets differ<strong>in</strong>g <strong>in</strong> fat content and energy.J Dairy Sci 76, 1014–1027.MacLaren LA, Guzeloglu A, Michel F, Thatcher WW, 2006:Peroxisome proliferator-activated receptor (PPAR) expression<strong>in</strong> cultured bov<strong>in</strong>e endometrial cells and response toomega-3 fatty acid, growth hormone and agonist stimulation<strong>in</strong> relation to series 2 prostagland<strong>in</strong> production. DomAnim Endocr 30, 155–169.Mattos R, Staples CR, Thatcher WW, 2000: Effects of dietaryfatty acids on reproduction <strong>in</strong> rum<strong>in</strong>ants. Rev Reprod 5, 38–45.Mattos R, Staples CR, Williams J, Amorocho A, McGuireMA, Thatcher WW, 2002: Uter<strong>in</strong>e, ovarian, and productionresponses of lactat<strong>in</strong>g dairy cows to <strong>in</strong>creas<strong>in</strong>g dietaryconcentrations of menhaden fish meal. J Dairy Sci 85, 755–764.Mattos R, Guzeloglu A, Bad<strong>in</strong>ga L, Staples CR, ThatcherWW, 2003: Polyunsaturated fatty acids and bov<strong>in</strong>e <strong>in</strong>terferon-smodify phorbol ester-<strong>in</strong>duced secretion of prostagland<strong>in</strong>F2aand expression of prostagland<strong>in</strong> endoperoxideÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


30 JEP Santos, TR Bilby, WW Thatcher, CR Staples and FT Silvestresynthase-2 and phospholipase-A2 <strong>in</strong> bov<strong>in</strong>e endometrialcells. Biol Reprod 69, 780–787.Mattos R, Staples CR, Arteche A, Wiltbank MC, Diaz FJ,Jenk<strong>in</strong>s TC, Thatcher WW, 2004: The effects of feed<strong>in</strong>g fishoil on uter<strong>in</strong>e secretion of PGF 2a , milk composition, andmetabolic status of periparturient Holste<strong>in</strong> cows. J Dairy Sci87, 921–932.McEvoy TG, Coull GD, Broadbent PJ, Hutch<strong>in</strong>son JSM,Speake BK, 2000: Fatty acid composition of lipids <strong>in</strong>immature cattle, pig and sheep oocytes with <strong>in</strong>tact zonapellucida. J Reprod Fertil 118, 163–170.McNamara S, Butler T, Ryan DP, Mee JF, Dillon P, O’MaraFP, Butler ST, Anglese D, Rath M, Murphy JJ, 2003: Effectof offer<strong>in</strong>g rumen-protected fat supplements on fertility andperformance <strong>in</strong> spr<strong>in</strong>g-calv<strong>in</strong>g Holste<strong>in</strong>–Friesian cows.Anim Reprod Sci 79, 45–56.Moussavi AR, Gilbert RO, Overton TR, Bauman DE, ButlerWR, 2007: Effects of feed<strong>in</strong>g fish meal and n-3 fatty acids onovarian and uter<strong>in</strong>e responses <strong>in</strong> early lactat<strong>in</strong>g dairy cows.J Dairy Sci 90, 145–154.Oldick BS, Staples CR, Thatcher WW, Gyawu P, 1997:Abomasal <strong>in</strong>fusion of glucose and fat-effect on digestion,production, and ovarian and uter<strong>in</strong>e function of cows. JDairy Sci 80, 1315–1328.Petit HV, Twagiramungu H, 2006: Conception rate andreproductive function of dairy cows fed different fat sources.Theriogenology 66, 1316–1324.Petit HV, Germiquet C, Lebel D, 2004: Effect of feed<strong>in</strong>gwhole, unprocessed sunflower seeds and flaxseed on milkproduction, milk composition, and prostagland<strong>in</strong> secretion<strong>in</strong> dairy cows. J Dairy Sci 87, 3889–3898.Rob<strong>in</strong>son RS, Pushpakumara PGA, Cheng Z, Peters AR,Abayasekara DEE, Wathes DC, 2002: Effects of dietarypolyunsaturated fatty acids on ovarian and uter<strong>in</strong>e function<strong>in</strong> lactat<strong>in</strong>g dairy cows. <strong>Reproduction</strong> 124, 119–131.Ryan DP, Spoon RA, Williams GL, 1992: Ovarian follicularcharacteristics, embryo recovery, and embryo viability <strong>in</strong>heifers fed high fat diets and treated with follicle-stimulat<strong>in</strong>ghormone. J Anim Sci 70, 3505–3513.Sch<strong>in</strong>goethe DJ, Casper DP, 1991: Total lactational responseto added fat dur<strong>in</strong>g early lactation. J Dairy Sci 74, 2617–2622.Schneider P, Sklan D, Chalupa W, Kronfeld DS, 1988:Feed<strong>in</strong>g calcium salts of fatty acids to lactat<strong>in</strong>g cows. JDairy Sci 71, 2143–2150.Scott TA, Shaver RD, Zepeda L, Yandell B, Smith TR, 1995:Effects of rumen-<strong>in</strong>ert fat on lactation, reproduction, andhealth of high produc<strong>in</strong>g Holste<strong>in</strong> herds. J Dairy Sci 78,2435–2451.Sklan D, Moallem U, Folman Y, 1991: Effect of feed<strong>in</strong>gcalcium soaps of fatty acids on production and reproductiveresponses <strong>in</strong> high produc<strong>in</strong>g lactat<strong>in</strong>g cows. J Dairy Sci 74,510–517.Sklan D, Kaim M, Moallem U, Folman Y, 1994: Effect ofdietary calcium soaps on milk yield, body weight, reproductivehormones, and fertility <strong>in</strong> first parity and older cows.J Dairy Sci 77, 1652–1660.Son J, Grant RJ, Larson LL, 1996: Effects of tallow and escapeprote<strong>in</strong> on lactational and reproductive performance ofdairy cows. J Dairy Sci 79, 822–830.Spicer LJ, Vernon RK, Tucker WB, Wettemann RP, HogueJF, Adams GD, 1993: Effects of <strong>in</strong>ert fat on energy balance,plasma concentrations of hormones, and reproduction <strong>in</strong>dairy cows. J Dairy Sci 76, 2664–2673.Staples CR, Thatcher WW, 2005: Effects of fatty acids onreproduction of dairy cows. In: Garnsworthy PC, WisemanJ(eds), Recent Advances <strong>in</strong> Animal Nutrition. Nott<strong>in</strong>ghamUniversity Press, Nott<strong>in</strong>gham, UK, pp. 229–256.Staples CR, Burke JM, Thatcher WW, 1998: Influence ofsupplemental fats on reproductive tissues and performanceof lactat<strong>in</strong>g cows. J Dairy Sci 81, 856–871.Staples CR, Wiltbank MC, Grummer RR, Guenther J, SartoriR, Diaz FJ, Bertics S, Mattos R, Thatcher WW, 2000: Effectof long cha<strong>in</strong> fatty acids on lactation performance andreproductive tissues of Holste<strong>in</strong> cows. J Dairy Sci 83(Suppl.1), 278 (Abstr.)Thangavelu G, Colazo MG, Ambrose DJ, Oba M, Ok<strong>in</strong>e EK,Dyck MK, 2007: Diets enriched <strong>in</strong> unsaturated fatty acidsenhance early embryonic development <strong>in</strong> lactat<strong>in</strong>g Holste<strong>in</strong>cows. Theriogenology 68, 949–957.Thomas MG, Williams GL, 1996: Metabolic hormone secretionand FSH-<strong>in</strong>duced superovulatory responses of beefheifers fed dietary supplements conta<strong>in</strong><strong>in</strong>g predom<strong>in</strong>antlysaturated or polyunsaturated fatty acids. Theriogenology45, 451–458.Williams GL, 1989: Modulation of luteal activity <strong>in</strong> postpartumbeef cows through changes <strong>in</strong> dietary lipid. J AnimSci 67, 785–793.Zeron Y, Ocheretny A, Kedar O, Borochov A, Sklan D, AravA, 2001: Seasonal changes <strong>in</strong> bov<strong>in</strong>e fertility: relation todevelopmental competence of oocytes, membrane propertiesand fatty acid composition of follicles. <strong>Reproduction</strong> 121,447–454.Zeron Y, Sklan D, Arav A, 2002: Effect of polyunsaturated fattyacid supplementation on biophysical parameters and chill<strong>in</strong>gsensitivity of ewe oocytes. Mol Reprod Dev 61, 271–278.Author’s address (for correspondence): JEP Santos, LE ‘‘Red’’ LassonBuild<strong>in</strong>g, Department of Animal Sciences, University of Florida,Ga<strong>in</strong>esville, FL, 32611 USA. E-mail: jepsantos@ufl.eduConflict of <strong>in</strong>terest: All authors declare no conflict of <strong>in</strong>terests.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 31–39 (2008); doi: 10.1111/j.1439-0531.2008.01140.xISSN 0936-6768Functional Differences <strong>in</strong> the Growth Hormone and Insul<strong>in</strong>-like Growth Factor Axis<strong>in</strong> Cattle and Pigs: Implications for Post-partum Nutrition and <strong>Reproduction</strong>MC LucyDivision of Animal Sciences, University of Missouri, Columbia, MO, USAContentsGrowth hormone (GH) and <strong>in</strong>sul<strong>in</strong>-like growth factor-I(IGF-I) control growth and lactation <strong>in</strong> cattle and sw<strong>in</strong>e.Insul<strong>in</strong> participates <strong>in</strong> the endocr<strong>in</strong>ology of growth andlactation because <strong>in</strong>sul<strong>in</strong> and GH are antagonistic <strong>in</strong> theiractions. Dairy cows experience a period of negative energybalance dur<strong>in</strong>g the first 4–8 weeks post-partum. Dur<strong>in</strong>g thisperiod, their somatotropic axis (comprised of GH, the GHreceptor and IGF-I) becomes uncoupled and there is elevatedGH and dim<strong>in</strong>ished IGF-I <strong>in</strong> the circulation. Blood <strong>in</strong>sul<strong>in</strong>concentrations are low as well. Sows are different from dairycows because their somatotropic axis rema<strong>in</strong>s coupled dur<strong>in</strong>glactation and both GH and IGF-I are elevated. Nonetheless,sows that become catabolic dur<strong>in</strong>g lactation will havereduced IGF-I concentrations. Sows are <strong>in</strong>sem<strong>in</strong>ated afterwean<strong>in</strong>g so their metabolic state is different from post-partumbeef and dairy cows that are <strong>in</strong>sem<strong>in</strong>ated when they arelactat<strong>in</strong>g. Dairy cows are fed ad libitum and naturally havelow IGF-I dur<strong>in</strong>g lactation. Sows have elevated IGF-I whenthey are well-fed. A threshold of IGF-I prote<strong>in</strong> <strong>in</strong> follicularfluid may be met by local ovarian (paracr<strong>in</strong>e ⁄ autocr<strong>in</strong>e) andendocr<strong>in</strong>e sources of IGF-I. Nutritionally <strong>in</strong>duced changes <strong>in</strong><strong>in</strong>sul<strong>in</strong> and <strong>in</strong> liver IGF-I secretion that arise from perturbationsof the somatotropic axis have a direct effect on theovary through the endocr<strong>in</strong>e actions of <strong>in</strong>sul<strong>in</strong> and IGF-I.Sows and cows that are nutritionally compromised have lowconcentrations of <strong>in</strong>sul<strong>in</strong> and IGF-I <strong>in</strong> their blood and thistheoretically reduces ovarian responsiveness to gonadotrop<strong>in</strong>s.Although sows are <strong>in</strong>sem<strong>in</strong>ated after wean<strong>in</strong>g, thereappear to be carry-over effects of the previous lactation onthe ovarian follicular populations that develop after the sowis weaned. Understand<strong>in</strong>g the mechanisms through whichmetabolic hormones control ovarian function may lead toimproved reproductive management of both pigs and cattlebecause lactation and post-partum reproduction are closelytied <strong>in</strong> both species.IntroductionGrowth hormone (GH) is a pituitary hormone thatcontrols growth and lactation. Insul<strong>in</strong>-like growthfactor-I (IGF-I) is released from liver <strong>in</strong> response toGH and is believed to control growth and lactation aswell (Renaville et al. 2002). There are a series of wellstudiedIGF-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s (IGFBP) that <strong>in</strong>hibit IGFactivity (Le Roith et al. 2001). In addition to caus<strong>in</strong>gIGF-I release, GH antagonizes <strong>in</strong>sul<strong>in</strong> action (Ethertonand Bauman 1998). Antagoniz<strong>in</strong>g the actions of <strong>in</strong>sul<strong>in</strong>has a nutrient partition<strong>in</strong>g effect through which thedevelopment of lean tissue and the production of milkare favoured. A complete understand<strong>in</strong>g of nutrientpartition<strong>in</strong>g dur<strong>in</strong>g growth, lactation and reproductiontypically <strong>in</strong>volves an appreciation of GH, IGF-I, IGFBPand <strong>in</strong>sul<strong>in</strong> because collectively they act on the <strong>in</strong>dividualanimal (Chagas et al. 2007).Reproductive events are controlled primarily bygonadotrop<strong>in</strong>s (LH and FSH). Growth hormone,IGF-I and <strong>in</strong>sul<strong>in</strong> can promote gonadotrop<strong>in</strong> actionby potentiat<strong>in</strong>g gonadotrop<strong>in</strong> receptor function (Webbet al. 2004; Scaramuzzi et al. 2006). Each hormone canalso act directly through their respective hormonereceptors on the ovary. Metabolic events such aslactation, therefore, are tied to reproductive eventsthrough a hormonal l<strong>in</strong>k that <strong>in</strong>volves GH, IGF-I and<strong>in</strong>sul<strong>in</strong>. Although not the primary focus of this review,additional metabolic hormones <strong>in</strong>clud<strong>in</strong>g lept<strong>in</strong> andadiponect<strong>in</strong> should be considered when nutrition, reproductionand lactation are discussed.The comparison of cattle and sw<strong>in</strong>e is <strong>in</strong>terest<strong>in</strong>g andworthwhile because it may give <strong>in</strong>sights <strong>in</strong>to underly<strong>in</strong>gbiological mechanisms through which lactation andreproduction are l<strong>in</strong>ked. Beef and dairy cattle aredist<strong>in</strong>ctly different <strong>in</strong> terms of lactation and nutrientpartition<strong>in</strong>g. The underly<strong>in</strong>g biology of GH, IGF-I and<strong>in</strong>sul<strong>in</strong> are different as well. In many respects, sw<strong>in</strong>e mayshare more similarities to beef cattle than dairy cattlebecause beef cattle and sw<strong>in</strong>e nurse their young.Mach<strong>in</strong>e milk<strong>in</strong>g systems for dairy cows were never<strong>in</strong>tended to recapitulate the physiology or lactationcurve of the nursed mother.Cattle and sw<strong>in</strong>e are very different with respect to theco<strong>in</strong>cidence of lactation and post-partum breed<strong>in</strong>g(<strong>in</strong>sem<strong>in</strong>ation). Post-partum beef and dairy cattle are<strong>in</strong>sem<strong>in</strong>ated while they are lactat<strong>in</strong>g. Post-partum sw<strong>in</strong>e,however, are <strong>in</strong>sem<strong>in</strong>ated after wean<strong>in</strong>g. Although bothcattle and sw<strong>in</strong>e share common physiology with regardto the local actions of GH, IGF-I, IGFBP and <strong>in</strong>sul<strong>in</strong>on the ovary, breed<strong>in</strong>g a weaned animal compared witha lactat<strong>in</strong>g animal represents a dist<strong>in</strong>ct physiologicaldifference, particularly when metabolic hormones areconsidered. This review will exam<strong>in</strong>e the somatotropicaxis with<strong>in</strong> cattle and sw<strong>in</strong>e dur<strong>in</strong>g lactation andsubsequent changes <strong>in</strong> the axis after wean<strong>in</strong>g. Implicationsfor reproduction will be exam<strong>in</strong>ed with an eyetoward understand<strong>in</strong>g nutrition and reproduction collectively.The Somatotropic Axis <strong>in</strong> Post-partum CattleDairy cows are well-known for the period of negativeenergy balance that they experience dur<strong>in</strong>g the first 4–8 weeks post-partum (Drackley 1999). This period ofnegative energy balance is caused by the tremendouscapacity of the mammary gland to sequester nutrientsand synthesize milk. Cows cannot consume enough feedto meet the energy demands for milk production so theyenter <strong>in</strong>to a negative energy balance. Negative energyÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


32 MC Lucybalance is associated with adipose tissue mobilizationand elevated blood non-esterified fatty acid (NEFA)concentrations. Dur<strong>in</strong>g negative energy balance, bloodGH concentrations are <strong>in</strong>creased. The <strong>in</strong>crease <strong>in</strong> GHdrives nutrient partition<strong>in</strong>g and supports milk production.Actions of GH dur<strong>in</strong>g lactationMultiple tissues are affected by GH but coord<strong>in</strong>atedevents <strong>in</strong> liver and adipose tissue may be most importantfor lactation (Bell 1995). In liver, the post-partum<strong>in</strong>crease <strong>in</strong> GH stimulates an <strong>in</strong>crease <strong>in</strong> gluconeogenesisthat meets the requirement for mammary lactose synthesis.The <strong>in</strong>crease <strong>in</strong> gluconeogenesis <strong>in</strong>volves a directeffect of GH on the gluconeogenic pathway as well an<strong>in</strong>direct effect of GH through an antagonism of <strong>in</strong>sul<strong>in</strong>action (Etherton and Bauman 1998). In adipose tissue,GH promotes lipolysis while antagoniz<strong>in</strong>g lipogenesisand block<strong>in</strong>g <strong>in</strong>sul<strong>in</strong>-dependent glucose uptake.Lactat<strong>in</strong>g dairy cows suffer from <strong>in</strong>sul<strong>in</strong> resistance(<strong>in</strong>sensitivity to <strong>in</strong>sul<strong>in</strong> that is manifested at the tissuelevel) and this <strong>in</strong>sul<strong>in</strong>-resistant state conserves glucosefor mammary lactose synthesis (Hayirli 2006). After an<strong>in</strong>itial post-partum period of elevated GH, there is asecond period where GH circulates at a lower bloodconcentration but rema<strong>in</strong>s elevated <strong>in</strong> high-produc<strong>in</strong>gdairy cows (Reist et al. 2003). The steady-state bloodGH concentrations may ultimately determ<strong>in</strong>e the relativepropensity for catabolism with<strong>in</strong> the <strong>in</strong>dividual cow.Cows <strong>in</strong> United States herds are supplemented withrecomb<strong>in</strong>ant GH (Bauman 1999). The recomb<strong>in</strong>ant GHacts <strong>in</strong> a manner that is consistent with the normaleffects of GH (antagoniz<strong>in</strong>g <strong>in</strong>sul<strong>in</strong> action, promot<strong>in</strong>ggluconeogenesis and block<strong>in</strong>g lipogenesis). The neteffect on the cow is greater milk production throughthe nutrient partition<strong>in</strong>g effect of GH.The actions of GH are mediated by the GH receptor(GHR). The highest concentrations of GHR are found<strong>in</strong> liver with the second most-abundant location be<strong>in</strong>gadipose tissue (Lucy et al. 1998). Perhaps the mostwidely understood action of GH <strong>in</strong> liver is the <strong>in</strong>crease<strong>in</strong> the synthesis and secretion of IGF-I. The IGF-I actsas an endocr<strong>in</strong>e hormone that controls GH secretionthrough a negative feedback loop (Le Roith et al. 2001).Nutritional mechanisms that control GH secretion maymanifest themselves through this negative feedbackloop. For example, one mechanism through whichnutritional restriction for energy or prote<strong>in</strong> elevatesGH is by block<strong>in</strong>g GHR action and reduc<strong>in</strong>g liver IGF-Isynthesis and secretion (Thissen et al. 1999). LesserIGF-I <strong>in</strong> blood relieves negative feedback and elevatesblood GH dur<strong>in</strong>g undernutrition. In addition to its clearrole <strong>in</strong> GH negative feedback, IGF-I aris<strong>in</strong>g from theliver may also control the growth and function of cellsand tissues throughout the body via an endocr<strong>in</strong>emechanism (Jones and Clemmons 1995). The significanceof this endocr<strong>in</strong>e mechanism of IGF-I action hasbeen questioned recently because conditional knock-outmice that do not synthesize IGF-I <strong>in</strong> liver have normalgrowth (Le Roith et al. 2001).There are three GHR mRNA variants (1A, 1B and1C, respectively) <strong>in</strong> cattle that differ with<strong>in</strong> the 5¢untranslated region (Jiang and Lucy 2001). The GHRprote<strong>in</strong> is the same for each variant because the GHRprote<strong>in</strong> is encoded <strong>in</strong> exons 2 through 10 of the mRNA.The only tissue that expresses GHR 1A mRNA is theliver of adult animals and the GHR 1A comprises thebulk of liver GHR mRNA (Lucy et al. 1998). The GHR1A mRNA is different from the GHR 1B and 1CmRNA because the GHR 1A mRNA is regulated by avariety of biological signals (Butler et al. 2003; Rhoadset al. 2004; Radcliff et al. 2006). The GHR 1B and 1Care found <strong>in</strong> many tissues <strong>in</strong>clud<strong>in</strong>g liver but thesealternative GHR mRNA undergo less biological regulationand appear to be constitutively expressed at a lowlevel.Changes <strong>in</strong> GHR and IGF-I <strong>in</strong> post-partum dairy cowsIn dairy cows, liver GHR 1A expression changesdramatically dur<strong>in</strong>g the period before and immediatelyafter calv<strong>in</strong>g (Lucy et al. 2001a; Radcliff et al. 2003a; b).The amount of GHR 1A mRNA decreases approximately2 days before calv<strong>in</strong>g, rema<strong>in</strong>s low for approximately1 week, and slowly <strong>in</strong>creases dur<strong>in</strong>g the secondweek after calv<strong>in</strong>g. The expression profile for IGF-ImRNA is similar to GHR 1A (decreased after calv<strong>in</strong>g)but the decrease <strong>in</strong> IGF-I post-partum occurs slightlylater than the decl<strong>in</strong>e <strong>in</strong> GHR 1A mRNA. The IGF-Idelay may reflect the dependence of liver IGF-I on GHand GHR 1A. There are post-partum changes <strong>in</strong> bloodIGFBP concentrations associated with the change <strong>in</strong>IGF-I (lesser IGFBP-3 and greater IGFBP-2; Skaaret al. 1991). The changes <strong>in</strong> IGFBP may affect thecapacity of the blood to carry IGF-I and the movementof IGF-I out of the vascular bed (Jones and Clemmons1995; Thissen et al. 1999). Blood <strong>in</strong>sul<strong>in</strong> concentrationsdecrease as well after calv<strong>in</strong>g <strong>in</strong> dairy cows. The decrease<strong>in</strong> <strong>in</strong>sul<strong>in</strong> occurs approximately 2–3 days after thedecrease <strong>in</strong> GHR 1A mRNA and is co<strong>in</strong>cident withthe decrease <strong>in</strong> IGF-I. The decrease <strong>in</strong> post-partumGHR 1A and IGF-I is associated with an <strong>in</strong>crease <strong>in</strong>blood GH and NEFA. There is a high correlationbetween the GHR 1A mRNA and GH b<strong>in</strong>d<strong>in</strong>g <strong>in</strong>periparturient liver (Radcliff et al. 2003a). Thus, thedecrease <strong>in</strong> GHR 1A mRNA co<strong>in</strong>cides with a decrease<strong>in</strong> GH b<strong>in</strong>d<strong>in</strong>g (prote<strong>in</strong> expression) and the loss of GHaction <strong>in</strong> liver. One possibility is that the decrease <strong>in</strong>GHR 1A is <strong>in</strong>directly caus<strong>in</strong>g the changes <strong>in</strong> GH andNEFA because lower GHR 1A may lead to lower liverIGF-I production and lesser GH negative feedback.Liver gluconeogenesis <strong>in</strong>creases dur<strong>in</strong>g this period ofdim<strong>in</strong>ished GH action (Drackley et al. 2006). Onequestion that needs to be addressed is what controlsliver gluconeogenesis when the liver is refractory to GH.Changes <strong>in</strong> GHR <strong>in</strong> other cattle breeds and implicationsfor nutrient partition<strong>in</strong>gThe GHR studies described <strong>in</strong> the previous section weredone <strong>in</strong> Holste<strong>in</strong> dairy cows. We have also studiedGuernsey dairy cows and found that the Guernsey cowsunderwent nearly identical changes <strong>in</strong> GHR 1A whencompared with a contemporary control group of Holste<strong>in</strong>cows (Okamura et al. 2007). Guernsey cattle thatÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


GH and IGF-I <strong>in</strong> Cattle and Pigs 33have been selected for milk production <strong>in</strong>dependently ofHolste<strong>in</strong>, therefore, experience similar changes <strong>in</strong> GHR1A dur<strong>in</strong>g the post-partum period. Angus beef cattle,however, did not undergo a decrease <strong>in</strong> GHR 1A atcalv<strong>in</strong>g (Jiang et al. 2005).One collective <strong>in</strong>terpretation of these studies is thatGHR 1A expression partially coord<strong>in</strong>ates the high milkproduction phenotype that typifies dairy cattle. Aftercalv<strong>in</strong>g there is a decrease <strong>in</strong> liver GHR 1A mRNA thatleads to low post-partum liver GHR expression. Theloss of liver GHR causes a GH refractory state <strong>in</strong> liverwhere liver does not produce IGF-I <strong>in</strong> response to GH.This refractory state represents an uncoupl<strong>in</strong>g of GHand IGF-I that is caused by the <strong>in</strong>activation of theGHR. The subsequent decrease <strong>in</strong> blood IGF-I concentrationleads to dim<strong>in</strong>ished negative feedback andelevated pituitary production of GH. Elevated postpartumGH supports milk production.Beef cattle may not uncouple their somatotropic axisafter calv<strong>in</strong>g. The failure to uncouple this axis leads torelatively higher blood IGF-I concentrations and lowerblood GH concentrations. This endocr<strong>in</strong>e state (higherIGF-I and lesser GH) is relatively anabolic whencompared with the endocr<strong>in</strong>e state of low IGF-I andhigh GH. The uncoupl<strong>in</strong>g of the somatotropic axis <strong>in</strong>post-partum dairy cows cannot be expla<strong>in</strong>ed by a simplenutritional phenomena <strong>in</strong>volv<strong>in</strong>g negative energy balancebecause feed restriction <strong>in</strong> later lactation dairycows did not decrease liver GHR 1A expression whenthe question was tested by two different laboratories(Kobayashi et al. 2002; Rhoads et al. 2007).Recoupl<strong>in</strong>g of the GH axis after calv<strong>in</strong>gThe recoupl<strong>in</strong>g of the GH axis after calv<strong>in</strong>g <strong>in</strong> highproduc<strong>in</strong>gdairy cows has been l<strong>in</strong>ked to post-partumnutrition and energy balance. Insul<strong>in</strong> <strong>in</strong>fusion <strong>in</strong>to earlypost-partum dairy cows <strong>in</strong>creased liver GHR 1AmRNA, liver IGF-I mRNA and blood IGF-I concentrations(Butler et al. 2003; Rhoads et al. 2004). Insul<strong>in</strong>,therefore, controls the recoupl<strong>in</strong>g through its positiveeffects on GHR 1A expression. There was also an effectof <strong>in</strong>sul<strong>in</strong> on GHR <strong>in</strong> adipose tissue (reduced <strong>in</strong>response to <strong>in</strong>sul<strong>in</strong>) but this effect was opposite to whatwas observed <strong>in</strong> liver (Butler et al. 2003). These data foran <strong>in</strong>hibitory effect of <strong>in</strong>sul<strong>in</strong> on GHR mRNA need tobe reconciled with the fact that others have shown astimulatory effect of <strong>in</strong>sul<strong>in</strong> on GHR <strong>in</strong> adipose tissue(Rhoads et al. 2004). Furthermore, GHR prote<strong>in</strong> <strong>in</strong>adipose tissue was reduced <strong>in</strong> low-<strong>in</strong>sul<strong>in</strong> states such asthose found <strong>in</strong> early post-partum dairy cows (Rhoadset al. 2004) and underfed dairy cows (Rhoads et al.2007).Greater nutrient <strong>in</strong>take and improved energy balance<strong>in</strong> later lactation dairy cows leads to elevated <strong>in</strong>sul<strong>in</strong> anda direct effect on the GH axis via the effects of <strong>in</strong>sul<strong>in</strong> onGHR expression (Lucy 2004). The recoupl<strong>in</strong>g of the axiscauses an <strong>in</strong>crease <strong>in</strong> IGF-I that acts negatively on GHsecretion. The <strong>in</strong>crease <strong>in</strong> <strong>in</strong>sul<strong>in</strong> and IGF-I and thedecrease <strong>in</strong> GH switches the post-partum dairy cowfrom a catabolic state (low-<strong>in</strong>sul<strong>in</strong>, low IGF-I andelevated GH) to an anabolic state (high <strong>in</strong>sul<strong>in</strong>, highIGF-I and low GH).Little <strong>in</strong>formation is available on the recoupl<strong>in</strong>g of thesomatotropic axis <strong>in</strong> post-partum beef cows. The failureof post-partum beef cows to undergo a decrease <strong>in</strong> GHR1A at calv<strong>in</strong>g (Jiang et al. 2005) may imply that the axisis either never uncoupled or m<strong>in</strong>imally uncoupled postpartum.Lake et al. (2006) reported that cows <strong>in</strong> lowerbody condition had greater GH and lesser IGF-I whencompared with cows <strong>in</strong> better body condition. Theuncoupl<strong>in</strong>g of the somatotropic axis for a beef cow <strong>in</strong>low body condition, however, is conceptually differentfrom the homeorhetic mechanism that occurs <strong>in</strong> postpartumdairy cows. For beef cows, feed<strong>in</strong>g additionalenergy or prote<strong>in</strong> will typically recouple the somatotropicaxis and elevate IGF-I (Lalman et al. 2000). For thebeef cow, either the mammary gland is too small to usethe additional nutrients or the calf is too small toconsume the additional milk. In the latter case, failure tofully evacuate the gland promotes <strong>in</strong>volution andessentially redirects nutrients back toward the mother.Dairy cows are fed ad libitum post-partum. Despite thisgenerous feed<strong>in</strong>g their somatotropic axis is uncoupleduntil the demand of the mammary gland for nutrients isreduced (<strong>in</strong>volution of the gland) or the capacity of thecow to consume nutrients (feed <strong>in</strong>take) <strong>in</strong>creases. Ananalogous scenario existed when New Zealand andNorth American dairy cows were studied. Feed<strong>in</strong>g 3 kgof concentrate improved body condition <strong>in</strong> post-partumNew Zealand Holste<strong>in</strong>–Friesian cows but the sameamount of concentrate feed<strong>in</strong>g had no effect on bodycondition <strong>in</strong> North American Holste<strong>in</strong> cows (Rocheet al. 2006). In this comparison, the greater capacity ofthe North American (vs New Zealand) cows to producemilk affected the partition<strong>in</strong>g of nutrients to adiposetissue.The Somatotropic Axis <strong>in</strong> Post-partum SowsDifferences <strong>in</strong> animal management for pigs and cattleaffect the somatotropic axis with<strong>in</strong> the respectivespecies. As mentioned above, lactat<strong>in</strong>g dairy cowsare fed ad libitum post-partum and cont<strong>in</strong>ue to lactatethrough the breed<strong>in</strong>g period. For the dairy cow,lactation is typically term<strong>in</strong>ated when she is <strong>in</strong> the lasttrimester of gestation. Likewise for beef, calves areweaned <strong>in</strong> the autumn when cows have a well-establishedpregnancy. In the United States system, dairycows are fed a totally-mixed ration and this ration isbalanced for ad libitum <strong>in</strong>take. For beef cows and dairycows on pasture, pasture growth and quality maydeterm<strong>in</strong>e the overall energetics of the animal (Kolver2003).Obesity <strong>in</strong> sows creates farrow<strong>in</strong>g difficulties andmetabolic problems, and also antagonizes post-partummilk production. Sows, therefore, are fed a ma<strong>in</strong>tenanceration dur<strong>in</strong>g gestation and their body condition ismanaged carefully (Aherne and Williams 1992). Afterfarrow<strong>in</strong>g and dur<strong>in</strong>g lactation, ad libitum feed<strong>in</strong>g ispracticed. Obviously, newborn piglets are small andtheir capacity to consume milk is low so sow milkproduction is lowest immediately after farrow<strong>in</strong>g (Nobletand Etienne 1989). Greatest levels of milk productionare achieved near the time of wean<strong>in</strong>g. The lengthof lactation differs between production systems and canÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


34 MC Lucybe 18–21 days (North American systems) or closer to28 days (European systems). After wean<strong>in</strong>g, sows arereturned to a lower level of <strong>in</strong>take (approximately onehalfto one-third of their lactation ration).The preced<strong>in</strong>g paragraph illustrates the complexity ofthe question when an attempt is made to understand theimpact of nutrition on the somatotropic axis <strong>in</strong> alactat<strong>in</strong>g sow. Feed<strong>in</strong>g and management practices havea major effect on the outcomes of studies of metabolichormones. It is necessary to exam<strong>in</strong>e managementdetails carefully when <strong>in</strong>spect<strong>in</strong>g the scientific literatureon the somatotropic axis <strong>in</strong> sows.Growth hormone, IGF-I and <strong>in</strong>sul<strong>in</strong>Few studies have exam<strong>in</strong>ed <strong>in</strong>tensively the concentrationsof GH, IGF-I, IGFBP and <strong>in</strong>sul<strong>in</strong> <strong>in</strong> the sowdur<strong>in</strong>g lactation. There is an <strong>in</strong>crease <strong>in</strong> blood GHconcentrations after farrow<strong>in</strong>g (Schams et al. 1994;Mejia-Guadarrama et al. 2002; Govoni et al. 2007).The <strong>in</strong>crease <strong>in</strong> blood GH is associated with an <strong>in</strong>crease<strong>in</strong> blood NEFA concentrations dur<strong>in</strong>g lactation. The<strong>in</strong>crease <strong>in</strong> NEFA implies that GH is mediat<strong>in</strong>g lipidcatabolism dur<strong>in</strong>g lactation. The <strong>in</strong>crease <strong>in</strong> GH dur<strong>in</strong>glactation is caused partly by the suckl<strong>in</strong>g stimulus fromthe piglets (Rushen et al. 1993). The suckl<strong>in</strong>g-<strong>in</strong>ducedGH release expla<strong>in</strong>s why a relatively high blood GHconcentration is susta<strong>in</strong>ed throughout lactation even <strong>in</strong>well-fed sows. With regard to GH, the lactat<strong>in</strong>g sow issimilar to the lactat<strong>in</strong>g dairy cow because both specieshave elevated GH dur<strong>in</strong>g lactation. The causes ofelevated GH may be different (lactat<strong>in</strong>g dairy cows arenot suckled) but they may also share some commonmechanisms because catabolic states typically elevateGH.One apparent difference between cows and sows<strong>in</strong>volves the recoupl<strong>in</strong>g of the somatotropic axis postpartum(Fig. 1). Post-partum sows have elevated IGF-Iafter farrow<strong>in</strong>g (Schams et al. 1994; Govoni et al. 2007)and this endocr<strong>in</strong>e state suggests that the <strong>in</strong>crease <strong>in</strong> GHafter farrow<strong>in</strong>g can stimulate the liver to synthesize andsecrete IGF-I. In the dairy cow there is a large decrease<strong>in</strong> IGF-I after calv<strong>in</strong>g that is associated with anuncoupl<strong>in</strong>g of the somatotropic axis (Radcliff et al.2003a; b). Little is known about changes <strong>in</strong> IGF-I <strong>in</strong>beef cattle around calv<strong>in</strong>g but there appears to be atleast some reduction <strong>in</strong> IGF-I on day 3 post-partumrelative to day 6 post-partum (Lake et al. 2006).There are discrepancies with respect to <strong>in</strong>sul<strong>in</strong> concentrations<strong>in</strong> lactat<strong>in</strong>g sows. Some studies reportgreater <strong>in</strong>sul<strong>in</strong> concentrations <strong>in</strong> sows after farrow<strong>in</strong>g(Guedes and Nogueira 2001), whereas others reportlower <strong>in</strong>sul<strong>in</strong> concentrations after farrow<strong>in</strong>g (Revellet al. 1998). The latter case (lower <strong>in</strong>sul<strong>in</strong>) is similar towhat is reported for cattle after calv<strong>in</strong>g and this lower<strong>in</strong>sul<strong>in</strong> appears to be a consequence of metabolic glucoseconsumption dur<strong>in</strong>g lactation. Sows may become <strong>in</strong>sul<strong>in</strong>-resistantdur<strong>in</strong>g lactation (Quesnel et al. 2007).Sows fed more energy have greater blood IGF-I postpartum(de Braganca and Prunier 1999; van den Brandet al. 2001). The <strong>in</strong>crease <strong>in</strong> IGF-I post-partum may bepartly expla<strong>in</strong>ed by the fact that gestat<strong>in</strong>g sows are fedma<strong>in</strong>tenance diets dur<strong>in</strong>g late pregnancy. In the gestat<strong>in</strong>gsow, therefore, feed<strong>in</strong>g level may limit liver IGF-Iproduction. The comb<strong>in</strong>ation of elevated GH andad libitum feed<strong>in</strong>g after farrow<strong>in</strong>g drives the somatotropicaxis and <strong>in</strong>creases liver IGF-I production. The<strong>in</strong>crease <strong>in</strong> IGF-I may be a consequence of elevated GHbut also may arise from feed-dependent mechanismssuch as greater liver GHR concentration, an enhancedcapacity for GH to signal through the GHR orGH-<strong>in</strong>dependent mechanism through which feed<strong>in</strong>g<strong>in</strong>creases IGF-I.Dur<strong>in</strong>g lactation, IGF-I concentration may rema<strong>in</strong>high (well-fed sows) or may decrease over time (underfedsows) (van den Brand et al. 2001). Presumably, <strong>in</strong>the underfed sow, IGF-I concentrations decrease dur<strong>in</strong>glactation because the somatotropic axis becomes uncoupled.The uncoupl<strong>in</strong>g may occur <strong>in</strong> the second and thirdweek of lactation when litter milk consumption and sowmilk production are greater (Noblet and Etienne 1989).The sow enters <strong>in</strong>to negative energy balance because sheFig. 1. Conceptual diagram for changes <strong>in</strong> blood GH, blood IGF-I and liver GHR after parturition (P; farrow<strong>in</strong>g or calv<strong>in</strong>g) <strong>in</strong> sows (leftdiagram) and dairy cows (right diagram). In sows, the somatotropic axis rema<strong>in</strong>s coupled after farrow<strong>in</strong>g and GH concentrations <strong>in</strong>creasebecause of suckl<strong>in</strong>g. This leads to elevated GH and IGF-I dur<strong>in</strong>g lactation. Dur<strong>in</strong>g the latter half of lactation, the somatotropic axis becomesuncoupled perhaps because there is negative energy balance and the amount of GHR <strong>in</strong> liver is less. Wean<strong>in</strong>g (W) causes a decrease <strong>in</strong> GH andIGF-I. The blood IGF-I concentration <strong>in</strong>crease after wean<strong>in</strong>g as the sow approaches breed<strong>in</strong>g (B). In dairy cows, the liver GHR decreases aftercalv<strong>in</strong>g and this causes uncoupl<strong>in</strong>g of the somatotropic axis. Loss of the GHR leads to a decrease <strong>in</strong> blood IGF-I. The decrease <strong>in</strong> blood IGF-Imay alleviate GH negative feedback and cause elevated GH <strong>in</strong> early lactation. Improved energy balance and <strong>in</strong>sul<strong>in</strong> may <strong>in</strong>crease the GHR andIGF-I and reduce GH concentrations as cows approach the breed<strong>in</strong>g period (B).Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


GH and IGF-I <strong>in</strong> Cattle and Pigs 35has <strong>in</strong>adequate feed. In all cases, first parity sows are afocus of metabolic studies because their capacity for feedconsumption is less and they are more likely to becatabolic (van den Brand et al. 2001).Wean<strong>in</strong>g is associated with changes <strong>in</strong> GH, <strong>in</strong>sul<strong>in</strong>,glucose and IGF-I but these changes may be confoundedby the lower feed level after wean<strong>in</strong>g andother management considerations. Wean<strong>in</strong>g is stressfulfor sows and the comb<strong>in</strong>ation of stress and thediscomfort of a fully engorged mammary gland maydepress appetite. Loss of appetite and decreased feed<strong>in</strong>take reduce blood <strong>in</strong>sul<strong>in</strong> and IGF-I. Growthhormone concentrations typically decrease after wean<strong>in</strong>g(Mejia-Guadarrama et al. 2002; Govoni et al.2007) because suckl<strong>in</strong>g stimulates GH release <strong>in</strong> pigs(Rushen et al. 1993). If sows are underfed dur<strong>in</strong>glactation (negative energy balance) then wean<strong>in</strong>g mayimprove <strong>in</strong>sul<strong>in</strong> and IGF-I status (Koketsu et al. 1998;Mejia-Guadarrama et al. 2002). If sows are well-feddur<strong>in</strong>g lactation then wean<strong>in</strong>g may have lesser effectson <strong>in</strong>sul<strong>in</strong> and IGF-I (Koketsu et al. 1998; Mejia-Guadarrama et al. 2002). Regardless of their nutritionalstatus dur<strong>in</strong>g lactation, there appears to be anormalization of IGF-I with<strong>in</strong> 3 days after wean<strong>in</strong>g.Sows will typically undergo a short period of lowIGF-I immediate after wean<strong>in</strong>g (associated with loss ofappetite, lower feed<strong>in</strong>g level and stress) and then haveelevated IGF-I as they progress toward oestrus. The<strong>in</strong>crease <strong>in</strong> IGF-I is not necessarily a metabolicresponse because there are stimulatory effects ofoestradiol on liver IGF-I synthesis and secretion(White et al. 2003).The porc<strong>in</strong>e GHRSurpris<strong>in</strong>g little is known about the porc<strong>in</strong>e GHR. Ina study of pigs from birth to 1 year of age, the liverGHR mRNA <strong>in</strong>creased with <strong>in</strong>creas<strong>in</strong>g age but muscleGHR mRNA failed to change (Schnoebelen-Combeset al. 1996). The tissue-specific pattern of GHRexpression that was identified <strong>in</strong> the grow<strong>in</strong>g pigsuggested that GHR gene expression was controlledby different mechanisms <strong>in</strong> liver and muscle; asituation that is known to occur <strong>in</strong> cattle (see previoussection). Likewise, other <strong>in</strong>vestigators have reportedtissue-specific effects of nutrition and GH on theexpression of GHR <strong>in</strong> liver, muscle and adipose tissuebut a simple theory that expla<strong>in</strong>s the physiologicalresponse has not evolved (Dauncey et al. 1994;Brameld et al. 1996; Combes et al. 1997). To ourknowledge there has been only one study of GHRvariants <strong>in</strong> pigs. In that study, both GHR 1A andGHR 1B mRNA were found <strong>in</strong> pig liver (Liu et al.2000a). The GHR 1B mRNA but not the GHR 1AmRNA was detected <strong>in</strong> muscle. The pig, therefore,was like other species because GHR 1B was found <strong>in</strong>a variety of tissues, whereas GHR 1A was a liverspecificmRNA. Beyond this <strong>in</strong>formation, we knownoth<strong>in</strong>g about the regulation of specific GHR variants<strong>in</strong> pig liver. We do not know if liver GHR expressionchanges dur<strong>in</strong>g lactation or if negative energy balance<strong>in</strong> late lactation uncouples the somatotropic axisthrough a GHR-dependent mechanism.Unique aspects of the somatotropic axis dur<strong>in</strong>g porc<strong>in</strong>elactationSows are typically catabolic dur<strong>in</strong>g lactation and loseweight and body condition. Despite the catabolic natureof lactation, the somatotropic axis rema<strong>in</strong>s coupled <strong>in</strong>lactat<strong>in</strong>g sows and IGF-I is elevated dur<strong>in</strong>g lactation.Greater blood IGF-I concentrations, however, do notsuppress blood GH dur<strong>in</strong>g the sow lactation perhapsbecause nurs<strong>in</strong>g piglets stimulate GH secretion andsupersede the IGF-I negative feedback. In the dairycow, the act of mach<strong>in</strong>e milk<strong>in</strong>g will <strong>in</strong>crease bloodprolact<strong>in</strong> concentration but, based on limited data, thereappears to be little change <strong>in</strong> blood GH <strong>in</strong> response tomach<strong>in</strong>e milk<strong>in</strong>g (Negrao and Marnet 2002). The releaseof GH <strong>in</strong> response to suckl<strong>in</strong>g <strong>in</strong> the beef cow has notbeen studied extensively.Mechanisms that L<strong>in</strong>k the Somatotropic Axis to<strong>Reproduction</strong>Sw<strong>in</strong>e and cattle share many aspects of the underly<strong>in</strong>gbiology controll<strong>in</strong>g ovarian follicular growth. Forexample, <strong>in</strong> both species follicular development iscontrolled by a comb<strong>in</strong>ation of LH and FSH. The<strong>in</strong>hibition of LH secretion by lactation prevents preovulatoryfollicular development <strong>in</strong> sows before wean<strong>in</strong>g(Varley and Foxcroft 1990). After wean<strong>in</strong>g, basal LHconcentrations and numbers of LH peaks <strong>in</strong>crease(Shaw and Foxcroft 1985). The <strong>in</strong>crease <strong>in</strong> LH pulsatilityis believed to drive follicular growth towardovulation. Concentrations of FSH <strong>in</strong>itially <strong>in</strong>creaseafter wean<strong>in</strong>g, but then decl<strong>in</strong>e as preovulatory folliclesdevelop (Shaw and Foxcroft 1985). Although LH isclearly important for follicular growth, a high correlationbetween <strong>in</strong>tensity of LH secretion and <strong>in</strong>terval tooestrus has not been found for sows.In cows, follicular development beg<strong>in</strong>s shortly aftercalv<strong>in</strong>g with a transient <strong>in</strong>crease <strong>in</strong> FSH and thedevelopment of a dom<strong>in</strong>ant follicle. A major componentof the process is the secretion of LH dur<strong>in</strong>g the earlypost-partum period (Lucy 2003). Low LH pulsatility isassociated with anoestrus. The primary causes ofanoestrus are different for beef and dairy cows but theessential features are similar. In beef cows, suckl<strong>in</strong>g<strong>in</strong>hibits LH pulsatility and the lack of LH pulsatilityleads to anoestrus (Williams and Griffith 1995). Thissituation is analogous to the sow (suckl<strong>in</strong>g <strong>in</strong>hibits LHsecretion). Anoestrus <strong>in</strong> dairy cattle is viewed as lesscommon (compared with beef) because the post-partum<strong>in</strong>hibition of LH may be less <strong>in</strong> animals that are notsuckled.Systemic <strong>in</strong>teractions of metabolic hormones with thereproductive axisChanges <strong>in</strong> metabolic hormones like <strong>in</strong>sul<strong>in</strong> and IGF-Iare dynamic <strong>in</strong> post-partum cows and sows. Insul<strong>in</strong> andIGF-I concentrations are <strong>in</strong>itially depressed but gradually<strong>in</strong>crease post-partum <strong>in</strong> cows (Lucy 2003). BloodIGF-I concentrations <strong>in</strong>crease between wean<strong>in</strong>g andoestrus <strong>in</strong> sows (Mejia-Guadarrama et al. 2002). Thefact that sows are weaned and have elevated IGF-IÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


36 MC Lucybefore breed<strong>in</strong>g represents a major physiological differencebetween cows and sows. Cattle <strong>in</strong> poor bodycondition or cows fail<strong>in</strong>g to <strong>in</strong>crease body conditiondur<strong>in</strong>g lactation have low blood IGF-I dur<strong>in</strong>g thebreed<strong>in</strong>g period (Chagas et al. 2007). Sows that areunderfed dur<strong>in</strong>g lactation have low blood IGF-I concentrationsbut blood IGF-I normalizes rapidly afterwean<strong>in</strong>g if sows are fed properly. Underfeed<strong>in</strong>g afterwean<strong>in</strong>g <strong>in</strong> sows will suppress IGF-I, compromisefollicular development (prolonged wean<strong>in</strong>g to oestrus<strong>in</strong>tervals and reduced ovulation rate), and reduceembryonic survival (Zak et al. 1997). Underfeed<strong>in</strong>gafter wean<strong>in</strong>g, however, would be an atypical managementpractice <strong>in</strong> sw<strong>in</strong>e herds. In lactat<strong>in</strong>g dairy cows,low IGF-I would be a typical situation because lowIGF-I and high GH are favourable for milk production(Chagas et al. 2007).Under normal physiological conditions, a thresholdof IGF-I prote<strong>in</strong> <strong>in</strong> follicular fluid may be met by localovarian (paracr<strong>in</strong>e ⁄ autocr<strong>in</strong>e) and endocr<strong>in</strong>e sources ofIGF-I. Accord<strong>in</strong>g to the somatomed<strong>in</strong> hypothesis,nutritionally <strong>in</strong>duced changes <strong>in</strong> liver IGF-I secretionhave a direct effect on the ovary through the endocr<strong>in</strong>eactions of IGF-I. Sows and cows that are nutritionallycompromised have low concentrations of <strong>in</strong>sul<strong>in</strong> andIGF-I <strong>in</strong> their blood. The lower <strong>in</strong>sul<strong>in</strong> and IGF-Iconcentrations theoretically reduce ovarian responsivenessto gonadotrop<strong>in</strong>s (see below). At the same time,post-partum cows and weaned sows have low blood LHconcentrations, <strong>in</strong> part because of the effects of metabolichormones on GnRH secretion from the hypothalamus.Thus, the effects of nutrition on reproduction aremanifested at the ovary and at the hypothalamus andpituitary. Overcom<strong>in</strong>g one limitation will not necessarilyrecover ovarian function. Responses to metabolic hormonesand gonadotrop<strong>in</strong>s typically follow a plateaumodel where further improvement <strong>in</strong> reproductivefunction is not seen once a critical threshold of hormoneconcentration is achieved.Local expression of IGFs and their b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>sInsul<strong>in</strong>-like growth factors and their b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>sare expressed with<strong>in</strong> specific cell layers [granulosa and(or) theca] of develop<strong>in</strong>g follicles. This parar<strong>in</strong>e ⁄ autocr<strong>in</strong>esynthesis of the IGF has been well-studied andwill not be extensively reviewed here. The reader isreferred to previous reviews on this topic for pigs andcattle (Liu et al. 2000b; Lucy 2000, 2007; Prunier andQuesnel 2000; Lucy et al. 2001b; Webb et al. 2004).There are two types of IGF, IGF-I and IGF-II, butIGF-I is the primary IGF under metabolic control. Theporc<strong>in</strong>e follicle expresses IGF-I (granulosa cell layer)and IGF-II (theca cell layer). Likewise for the cow,theca cells express IGF-II. Expression of IGF-I <strong>in</strong>bov<strong>in</strong>e granulosa cells has been reported by some butnot all laboratories (Webb et al. 2004). The IGF-I and-II b<strong>in</strong>d to specific receptors (type I and type II IGFreceptors). The type I IGF receptor is closely related tothe <strong>in</strong>sul<strong>in</strong> receptor. Both <strong>in</strong>sul<strong>in</strong> and type I IGFreceptors are receptor tyros<strong>in</strong>e k<strong>in</strong>ases and bothreceptors signal through related <strong>in</strong>tracellular pathways.The type II IGF receptor is an unusual receptorbecause its primary function is to b<strong>in</strong>d, <strong>in</strong>ternalize anddegrade IGF-II. In the pig, granulosa and theca cellsexpress <strong>in</strong>sul<strong>in</strong> receptors as well as type I and type IIIGF receptors. Thus, <strong>in</strong>sul<strong>in</strong> and IGF-I can act directlyon ovarian cells <strong>in</strong> pigs. For the cow, there is type IIGF receptor expression <strong>in</strong> the granulosa cells and typeII IGF receptor expression <strong>in</strong> granulosa and thecacells. The IGFs (but not <strong>in</strong>sul<strong>in</strong>) are bound to at leastsix different b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s (IGFBP). Each b<strong>in</strong>d<strong>in</strong>gprote<strong>in</strong> has its own unique physiology. With<strong>in</strong> thebov<strong>in</strong>e and porc<strong>in</strong>e ovary, IGFBP-2 and -4 areexpressed and are regulators of IGF function (Fortuneet al. 2004).Ovarian follicles conta<strong>in</strong> GHR mRNA and prote<strong>in</strong>(Lucy 2000). In liver, GH acts on the GHR and causesan <strong>in</strong>crease <strong>in</strong> hepatic IGF-I synthesis and secretion. If asimilar mechanism exists <strong>in</strong> the ovary of cows and sowsthen GH, act<strong>in</strong>g directly on ovarian follicles, couldaffect ovarian function by simulat<strong>in</strong>g the local productionof IGF-I. Growth hormone-dependent, ovarianIGF-I synthesis has been shown <strong>in</strong> the pig (Hsu andHammond 1987). In cattle, however, GH failed to<strong>in</strong>crease ovarian IGF-I synthesis (Kirby et al. 1996).The bov<strong>in</strong>e study, however, measured IGF-I mRNA <strong>in</strong>whole ovaries. Subtle changes <strong>in</strong> IGF-I gene expressionthat may have occurred <strong>in</strong> ovarian follicles may not havebeen detected.Synergism between <strong>in</strong>sul<strong>in</strong>-like growth factors andgonadotrop<strong>in</strong>sAt the ovarian level, follicular growth <strong>in</strong> both cows andsows depends on LH and FSH and also <strong>in</strong>sul<strong>in</strong> andIGF-I. The <strong>in</strong>dependent contribution of each hormoneis difficult to establish, however, because there arecoord<strong>in</strong>ated changes <strong>in</strong> each of these hormones whennutrition is improved. Insul<strong>in</strong> and IGF-I can act at thelevel of the hypothalamus to stimulate GnRH secretionand therefore control the release of LH and FSH (Lucy2003; Daftary and Gore 2005). Hypothetically, mechanismsthat <strong>in</strong>crease LH pulsatility through their actionson the hypothalamus and pituitary also affect theresponsiveness of the ovary to gonadotrop<strong>in</strong>s. Forexample, LH pulsatility <strong>in</strong>creases <strong>in</strong> cows dur<strong>in</strong>g thepost-partum period. Both <strong>in</strong>sul<strong>in</strong> and IGF-I concentrations<strong>in</strong> blood <strong>in</strong>crease as well. The collective effects ofLH, <strong>in</strong>sul<strong>in</strong> and IGF-I act<strong>in</strong>g on the ovary promotefollicular development and ovulation.The location of specific components of the IGFsystem corresponds to the location of LH and FSHreceptors with<strong>in</strong> the develop<strong>in</strong>g follicles. The co-localizationof IGF and gonadotrop<strong>in</strong> receptor genes suggestsa coord<strong>in</strong>ation of gonadotrop<strong>in</strong> and IGF actionwith<strong>in</strong> ovary. There is a synergistic relationship betweenthe IGFs and <strong>in</strong>sul<strong>in</strong> with gonadotrop<strong>in</strong>s for a variety ofcellular functions <strong>in</strong>clud<strong>in</strong>g mitogenesis and steroidogenesis(Lucy 2000, 2007; Webb et al. 2004). Thesynergism is caused by the ability of IGFs and <strong>in</strong>sul<strong>in</strong>to <strong>in</strong>crease gonadotrop<strong>in</strong> receptor numbers and <strong>in</strong>creasethe activity of gonadotrop<strong>in</strong> receptor second messengersystems. At the same time, gonadotrop<strong>in</strong>s <strong>in</strong>crease type IIGF receptor expression and may <strong>in</strong>crease IGF-Isynthesis <strong>in</strong> granulosa cells.Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


GH and IGF-I <strong>in</strong> Cattle and Pigs 37Both <strong>in</strong>sul<strong>in</strong> and IGF-I may play a role <strong>in</strong> folliculardevelopment dur<strong>in</strong>g periods of under-feed<strong>in</strong>g or weightloss because undernutrition causes a decrease <strong>in</strong> bloodconcentrations of IGF-I and <strong>in</strong>sul<strong>in</strong>. Accord<strong>in</strong>g to awidely held hypothesis, blood <strong>in</strong>sul<strong>in</strong> and IGF-I act <strong>in</strong>an endocr<strong>in</strong>e manner to affect ovarian cells. Thedecrease <strong>in</strong> blood IGF-I and <strong>in</strong>sul<strong>in</strong> concentrationsdur<strong>in</strong>g undernutrition decreases the responsiveness ofthe ovary to gonadotrop<strong>in</strong>s which ultimately leads to adecrease <strong>in</strong> follicular growth. Cosgrove et al. (1992)found that re-alimentation of feed-restricted gilts could<strong>in</strong>crease follicular growth <strong>in</strong> the absence of a change <strong>in</strong>LH secretion. The change <strong>in</strong> follicular development wasassociated with greater plasma IGF-I and <strong>in</strong>sul<strong>in</strong> <strong>in</strong> there-alimented group. Thus, feed<strong>in</strong>g <strong>in</strong>creases the responsivenessof the ovary to LH through its effects on <strong>in</strong>sul<strong>in</strong>and IGF-I.Carry-over effects of lactation <strong>in</strong> sowsBeef and dairy cows are <strong>in</strong>sem<strong>in</strong>ated while they arelactat<strong>in</strong>g so there may not be appreciable changes <strong>in</strong>blood <strong>in</strong>sul<strong>in</strong> and IGF-I before the breed<strong>in</strong>g period.Sows are weaned approximately 1 week before breed<strong>in</strong>gand wean<strong>in</strong>g has a large effect on the metabolism of thesow and blood concentrations of <strong>in</strong>sul<strong>in</strong> and IGF-I(discussed <strong>in</strong> previous section). The blood IGF-I concentrationsare normalized with<strong>in</strong> approximately 3 daysafter wean<strong>in</strong>g. Nonetheless, there appear to be carryovereffects of lactation. Thus, a compromised state ofovarian follicular development that develops dur<strong>in</strong>glactation can potentially <strong>in</strong>fluence follicular growth andthe time of oestrus after wean<strong>in</strong>g. Evidence for thisevolved from a variety of studies <strong>in</strong>clud<strong>in</strong>g those offollicular development before and after wean<strong>in</strong>g <strong>in</strong> sows(Lucy et al. 2001b). Sows with the shortest <strong>in</strong>tervals toovulation had larger follicles shortly after wean<strong>in</strong>g. Inthese same sows (those with the shortest <strong>in</strong>terval toovulation), there was also greater estrogenic activity offollicles before wean<strong>in</strong>g and an earlier rise <strong>in</strong> preovulatoryoestradiol after wean<strong>in</strong>g. Based on these data, itappears that some component of the follicular developmentafter wean<strong>in</strong>g is controlled by physiologicalprocesses dur<strong>in</strong>g lactation that affect the ovarian follicularpopulations. These physiological processes probably<strong>in</strong>volve gonadotrop<strong>in</strong>s as well as key metabolichormones like GH, <strong>in</strong>sul<strong>in</strong> and IGF-I that affectgonadotrop<strong>in</strong> action at the level of the ovary.Summary and ConclusionsGrowth hormone, IGF-I and <strong>in</strong>sul<strong>in</strong> are metabolichormones that control growth and lactation <strong>in</strong> cattleand sw<strong>in</strong>e. Cows and sows typically enter <strong>in</strong>to negativeenergy balance dur<strong>in</strong>g lactation and this negative energybalance is associated with low concentrations of <strong>in</strong>sul<strong>in</strong>and IGF-I <strong>in</strong> the blood. Insul<strong>in</strong> and IGF-I affect thereproductive axis through direct effects on ovarian cellsand also through their effects on gonadotrop<strong>in</strong> secretionand gonadotrop<strong>in</strong> receptor function. Understand<strong>in</strong>g themechanisms through which metabolic hormones controlovarian function may lead to improved reproductivemanagement of both cattle and pigs because lactationand post-partum reproduction are closely tied <strong>in</strong> bothspecies.AcknowledgementsThe author would like to thank Dr Tim Safranski and AmandaWilliams of the University of Missouri for their helpful suggestionsdur<strong>in</strong>g the preparation of this manuscript.ReferencesAherne FX, Williams IH, 1992: Nutrition for optimiz<strong>in</strong>gbreed<strong>in</strong>g herd performance. Vet Cl<strong>in</strong> N Am Food AnimPract 8, 589–608.Bauman DE, 1999: Bov<strong>in</strong>e somatotrop<strong>in</strong> and lactation: frombasic science to commercial application. Domest AnimEndocr<strong>in</strong>ol 17, 101–116.Bell AW, 1995: Regulation of organic nutrient metabolismdur<strong>in</strong>g transition from late pregnancy to early lactation. JAnim Sci 73, 2804–2819.de Braganca MM, Prunier A, 1999: Effects of low feed <strong>in</strong>takeand hot environment on plasma profiles of glucose, nonesterifiedfatty acids, <strong>in</strong>sul<strong>in</strong>, glucagon, and IGF-I <strong>in</strong> lactat<strong>in</strong>gsows. Domest Anim Endocr<strong>in</strong>ol 16, 89–101.Brameld JM, Atk<strong>in</strong>son JL, Saunders JC, Pell JM, Buttery PJ,Gilmour RS, 1996: Effects of growth hormone adm<strong>in</strong>istrationand dietary prote<strong>in</strong> <strong>in</strong>take on <strong>in</strong>sul<strong>in</strong>-like growth factorI and growth hormone receptor mRNA expression <strong>in</strong>porc<strong>in</strong>e liver, skeletal muscle, and adipose tissue. J AnimSci 74, 1832–1841.van den Brand H, Prunier A, Soede NM, Kemp B, 2001: Inprimiparous sows, plasma <strong>in</strong>sul<strong>in</strong>-like growth factor-I canbe affected by lactational feed <strong>in</strong>take and dietary energysource and is associated with lute<strong>in</strong>iz<strong>in</strong>g hormone. ReprodNutr Dev 41, 27–39.Butler ST, Marr AL, Pelton SH, Radcliff RP, Lucy MC, ButlerWR, 2003: Insul<strong>in</strong> restores GH responsiveness dur<strong>in</strong>glactation-<strong>in</strong>duced negative energy balance <strong>in</strong> dairy cattle:effects on expression of IGF-I and GH receptor 1A.J Endocr<strong>in</strong>ol 176, 205–217.Chagas LM, Bass JJ, Blache D, Burke CR, Kay JK, L<strong>in</strong>dsayDR, Lucy MC, Mart<strong>in</strong> GB, Meier S, Rhodes FM, RocheJR, Thatcher WW, Webb R, 2007: Invited review: newperspectives on the roles of nutrition and metabolic priorities<strong>in</strong> the subfertility of high-produc<strong>in</strong>g dairy cows. J DairySci 90, 4022–4032.Combes S, Louveau I, Bonneau M, 1997: Moderate foodrestriction affects skeletal muscle and liver growth hormonereceptors differently <strong>in</strong> pigs. J Nutr 127, 1944–1949.Cosgrove JR, Tilton JE, Hunter MG, Foxcroft GR, 1992:Gonadotrop<strong>in</strong>-<strong>in</strong>dependent mechanisms participate <strong>in</strong> ovarianresponses to realimentation <strong>in</strong> feed-restricted prepubertalgilts. Biol Reprod 47, 736–745.Daftary SS, Gore AC, 2005: IGF-1 <strong>in</strong> the bra<strong>in</strong> as a regulatorof reproductive neuroendocr<strong>in</strong>e function. Exp Biol Med(Maywood) 230, 292–306.Dauncey MJ, Burton KA, White P, Harrison AP, Gilmour RS,Duchamp C, Cattaneo D, 1994: Nutritional regulation ofgrowth hormone receptor gene expression. FASEB J 8, 81–88.Drackley JK, 1999: ADSA Foundation Scholar Award.Biology of dairy cows dur<strong>in</strong>g the transition period: the f<strong>in</strong>alfrontier?. J Dairy Sci 82, 2259–2273.Drackley JK, Donk<strong>in</strong> SS, Reynolds CK, 2006: Major advances<strong>in</strong> fundamental dairy cattle nutrition. J Dairy Sci 89, 1324–1336.Etherton TD, Bauman DE, 1998: Biology of somatotrop<strong>in</strong> <strong>in</strong>growth and lactation of domestic animals. Physiol Rev 78,745–761.Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


38 MC LucyFortune JE, Rivera GM, Yang MY, 2004: Follicular development:the role of the follicular microenvironment <strong>in</strong>selection of the dom<strong>in</strong>ant follicle. Anim Reprod Sci 82–83,109–126.Govoni N, Parmeggiani A, Galeati G, Penazzi P, De Iasio R,Pagotto U, Pasquali R, Taman<strong>in</strong>i C, Seren E, 2007: Acylghrel<strong>in</strong> and metabolic hormones <strong>in</strong> pregnant and lactat<strong>in</strong>gsows. Reprod Domest Anim 42, 39–43.Guedes RM, Nogueira RH, 2001: The <strong>in</strong>fluence of parity orderand body condition and serum hormones on wean<strong>in</strong>g-toestrus<strong>in</strong>terval of sows. Anim Reprod Sci 67, 91–99.Hayirli A, 2006: The role of exogenous <strong>in</strong>sul<strong>in</strong> <strong>in</strong> the complexof hepatic lipidosis and ketosis associated with <strong>in</strong>sul<strong>in</strong>resistance phenomenon <strong>in</strong> postpartum dairy cattle. Vet ResCommun 30, 749–774.Hsu CJ, Hammond JM, 1987: Concomitant effects of growthhormone on secretion of <strong>in</strong>sul<strong>in</strong>-like growth factor I andprogesterone by cultured porc<strong>in</strong>e granulosa cells. Endocr<strong>in</strong>ology121, 1343–1348.Jiang H, Lucy MC, 2001: Variants of the 5¢-untranslatedregion of the bov<strong>in</strong>e growth hormone receptor mRNA:isolation, expression and effects on translational efficiency.Gene 265, 45–53.Jiang H, Lucy MC, Crooker BA, Beal WE, 2005: Expressionof growth hormone receptor 1A mRNA is decreased <strong>in</strong>dairy cows but not <strong>in</strong> beef cows at parturition. J Dairy Sci88, 1370–1377.Jones JI, Clemmons DR, 1995: Insul<strong>in</strong>-like growth factors andtheir b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s: biological actions. Endocr Rev 16, 3–34.Kirby CJ, Thatcher WW, Collier RJ, Simmen FA, Lucy MC,1996: Effects of growth hormone and pregnancy on expressionof growth hormone receptor, <strong>in</strong>sul<strong>in</strong>-like growth factor-I, and <strong>in</strong>sul<strong>in</strong>-like growth factor b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>-2 and -3genes <strong>in</strong> bov<strong>in</strong>e uterus, ovary, and oviduct. Biol Reprod 55,996–1002.Kobayashi Y, Boyd CK, McCormack BL, Lucy MC, 2002:Reduced <strong>in</strong>sul<strong>in</strong>-like growth factor-I after acute feedrestriction <strong>in</strong> lactat<strong>in</strong>g dairy cows is <strong>in</strong>dependent of changes<strong>in</strong> growth hormone receptor 1A mRNA. J Dairy Sci 85,748–754.Koketsu Y, Dial GD, Pettigrew JE, Xue J, Yang H, Lucia T,1998: Influence of lactation length and feed <strong>in</strong>take onreproductive performance and blood concentrations ofglucose, <strong>in</strong>sul<strong>in</strong> and lute<strong>in</strong>iz<strong>in</strong>g hormone <strong>in</strong> primiparoussows. Anim Reprod Sci 52, 153–163.Kolver ES, 2003: Nutritional limitations to <strong>in</strong>creasedproduction on pasture-based systems. Proc Nutr Soc 62,291–300.Lake SL, Scholljegerdes EJ, Hallford DM, Moss GE, RuleDC, Hess BW, 2006: Effects of body condition score atparturition and postpartum supplemental fat on metaboliteand hormone concentrations of beef cows and their suckl<strong>in</strong>gcalves. J Anim Sci 84, 1038–1047.Lalman DL, Williams JE, Hess BW, Thomas MG, KeislerDH, 2000: Effect of dietary energy on milk production andmetabolic hormones <strong>in</strong> th<strong>in</strong>, primiparous beef heifers.J Anim Sci 78, 530–538.Le Roith D, Bondy C, Yakar S, Liu JL, Butler A, 2001: Thesomatomed<strong>in</strong> hypothesis: 2001. Endocr Rev 22, 53–74.Liu J, Carroll JA, Matteri RL, Lucy MC, 2000a: Expression oftwo variants of growth hormone receptor messenger ribonucleicacid <strong>in</strong> porc<strong>in</strong>e liver. J Anim Sci 78, 306–317.Liu J, Koenigsfeld AT, Cantley TC, Boyd CK, Kobayashi Y,Lucy MC, 2000b: Growth and the <strong>in</strong>itiation of steroidogenesis<strong>in</strong> porc<strong>in</strong>e follicles are associated with uniquepatterns of gene expression for <strong>in</strong>dividual components ofthe ovarian <strong>in</strong>sul<strong>in</strong>-like growth factor system. Biol Reprod63, 942–952.Lucy MC, 2000: Regulation of ovarian follicular growth bysomatotrop<strong>in</strong> and <strong>in</strong>sul<strong>in</strong>-like growth factors <strong>in</strong> cattle. JDairy Sci 83, 1635–1647.Lucy MC, 2003: Mechanisms l<strong>in</strong>k<strong>in</strong>g nutrition and reproduction<strong>in</strong> postpartum cows. Reprod Suppl 61, 415–427.Lucy MC, 2004: Mechanisms l<strong>in</strong>k<strong>in</strong>g the somatotropic axiswith <strong>in</strong>sul<strong>in</strong>: Lessons from the postpartum dairy cow. ProcN Z Soc Anim Prod 64, 19–23.Lucy MC, 2007: The bov<strong>in</strong>e dom<strong>in</strong>ant ovarian follicle. J AnimSci 85, E89–E99.Lucy MC, Boyd CK, Koenigsfeld AT, Okamura CS, 1998:Expression of somatotrop<strong>in</strong> receptor messenger ribonucleicacid <strong>in</strong> bov<strong>in</strong>e tissues. J Dairy Sci 81, 1889–1895.Lucy MC, Jiang H, Kobayashi Y, 2001a: Changes <strong>in</strong> thesomatotrop<strong>in</strong> axis associated with the <strong>in</strong>itiation of lactation.J Dairy Sci 84, E113–E119.Lucy MC, Liu J, Boyd CK, Bracken CJ, 2001b: Ovarianfollicular growth <strong>in</strong> sows. Reprod Suppl 58, 31–45.Mejia-Guadarrama CA, Pasquier A, Dourmad JY, Prunier A,Quesnel H, 2002: Prote<strong>in</strong> (lys<strong>in</strong>e) restriction <strong>in</strong> primiparouslactat<strong>in</strong>g sows: effects on metabolic state, somatotropic axis,and reproductive performance after wean<strong>in</strong>g. J Anim Sci 80,3286–3300.Negrao JA, Marnet PG, 2002: Effect of calf suckl<strong>in</strong>g onoxytoc<strong>in</strong>, prolact<strong>in</strong>, growth hormone and milk yield <strong>in</strong>crossbred Gir · Holste<strong>in</strong> cows dur<strong>in</strong>g milk<strong>in</strong>g. Reprod NutrDev 42, 373–380.Noblet J, Etienne M, 1989: Estimation of sow milk nutrientoutput. J Anim Sci 67, 3352–3359.Okamura CA, Bader JF, Cantley TC, Lucy MC, 2007: Growthhormone receptor expression <strong>in</strong> two dairy breeds dur<strong>in</strong>g theperiparturient period. J Dairy Sci 90, 8–4 (abstract).Prunier A, Quesnel H, 2000: Influence of the nutritional statuson ovarian development <strong>in</strong> female pigs. Anim Reprod Sci60–61, 185–197.Quesnel H, Etienne M, Pere MC, 2007: Influence of litter sizeon metabolic status and reproductive axis <strong>in</strong> primiparoussows. J Anim Sci 85, 118–128.Radcliff RP, McCormack BL, Crooker BA, Lucy MC, 2003a:Growth hormone (GH) b<strong>in</strong>d<strong>in</strong>g and expression of GHreceptor 1A mRNA <strong>in</strong> hepatic tissue of periparturient dairycows. J Dairy Sci 86, 3933–3940.Radcliff RP, McCormack BL, Crooker BA, Lucy MC, 2003b:Plasma hormones and expression of growth hormonereceptor and <strong>in</strong>sul<strong>in</strong>-like growth factor-I mRNA <strong>in</strong> hepatictissue of periparturient dairy cows. J Dairy Sci 86, 3920–3926.Radcliff RP, McCormack BL, Keisler DH, Crooker BA, LucyMC, 2006: Partial feed restriction decreases growth hormonereceptor 1A mRNA expression <strong>in</strong> postpartum dairycows. J Dairy Sci 89, 611–619.Reist M, Erd<strong>in</strong> D, von Euw D, Tschuemperl<strong>in</strong> K, LeuenbergerH, Delavaud C, Chilliard Y, Hammon HM, Kuenzi N,Blum JW, 2003: Concentrate feed<strong>in</strong>g strategy <strong>in</strong> lactat<strong>in</strong>gdairy cows: metabolic and endocr<strong>in</strong>e changes with emphasison lept<strong>in</strong>. J Dairy Sci 86, 1690–1706.Renaville R, Hammadi M, Portetelle D, 2002: Role of thesomatotropic axis <strong>in</strong> the mammalian metabolism. DomestAnim Endocr<strong>in</strong>ol 23, 351–360.Revell DK, Williams IH, Mullan BP, Ranford JL, Smits RJ,1998: Body composition at farrow<strong>in</strong>g and nutrition dur<strong>in</strong>glactation affect the performance of primiparous sows. I:Voluntary feed <strong>in</strong>take, weight loss, and plasma metabolites.J Anim Sci 76, 1729–1737.Rhoads RP, Kim JW, Leury BJ, Baumgard LH, Segoale N,Frank SJ, Bauman DE, Boisclair YR, 2004: Insul<strong>in</strong><strong>in</strong>creases the abundance of the growth hormone receptor<strong>in</strong> liver and adipose tissue of periparturient dairy cows. JNutr 134, 1020–1027.Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


GH and IGF-I <strong>in</strong> Cattle and Pigs 39Rhoads RP, Kim JW, Van Amburgh ME, Ehrhardt RA,Frank SJ, Boisclair YR, 2007: Effect of nutrition on the GHresponsiveness of liver and adipose tissue <strong>in</strong> dairy cows. JEndocr<strong>in</strong>ol 195, 49–58.Roche JR, Berry DP, Kolver ES, 2006: Holste<strong>in</strong>–Friesianstra<strong>in</strong> and feed effects on milk production, body weight, andbody condition score profiles <strong>in</strong> graz<strong>in</strong>g dairy cows. J DairySci 89, 3532–3543.Rushen J, Foxcroft G, De Passille AM, 1993: Nurs<strong>in</strong>g-<strong>in</strong>ducedchanges <strong>in</strong> pa<strong>in</strong> sensitivity, prolact<strong>in</strong>, and somatotrop<strong>in</strong> <strong>in</strong>the pig. Physiol Behav 53, 265–270.Scaramuzzi RJ, Campbell BK, Down<strong>in</strong>g JA, Kendall NR,Khalid M, Munoz-Gutierrez M, Somchit A, 2006: A reviewof the effects of supplementary nutrition <strong>in</strong> the ewe on theconcentrations of reproductive and metabolic hormones andthe mechanisms that regulate folliculogenesis and ovulationrate. Reprod Nutr Dev 46, 339–354.Schams D, Kraetzl WD, Brem G, Graf F, 1994: Secretorypattern of metabolic hormones <strong>in</strong> the lactat<strong>in</strong>g sow. ExpCl<strong>in</strong> Endocr<strong>in</strong>ol 102, 439–447.Schnoebelen-Combes S, Louveau I, Postel-V<strong>in</strong>ay MC, BonneauM, 1996: Ontogeny of GH receptor and GH-b<strong>in</strong>d<strong>in</strong>gprote<strong>in</strong> <strong>in</strong> the pig. J Endocr<strong>in</strong>ol 148, 249–255.Shaw HJ, Foxcroft GR, 1985: Relationships between LH,FSH and prolact<strong>in</strong> secretion and reproductive activity <strong>in</strong> theweaned sow. J Reprod Fertil 75, 17–28.Skaar TC, Vega JR, Pyke SN, Baumrucker CR, 1991: Changes<strong>in</strong> <strong>in</strong>sul<strong>in</strong>-like growth factor-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s <strong>in</strong> bov<strong>in</strong>emammary secretions associated with pregnancy and parturition.J Endocr<strong>in</strong>ol 131, 127–133.Thissen JP, Underwood LE, Ketelslegers JM, 1999: Regulationof <strong>in</strong>sul<strong>in</strong>-like growth factor-I <strong>in</strong> starvation and <strong>in</strong>jury.Nutr Rev 57, 167–176.Varley MA, Foxcroft GR, 1990: Endocr<strong>in</strong>ology of thelactat<strong>in</strong>g and weaned sow. J Reprod Fertil Suppl 40, 47–61.Webb R, Garnsworthy PC, Gong JG, Armstrong DG, 2004:Control of follicular growth: local <strong>in</strong>teractions and nutritional<strong>in</strong>fluences. J Anim Sci 82, E63–E74.White ME, Johnson BJ, Hathaway MR, Dayton WR, 2003:Growth factor messenger RNA levels <strong>in</strong> muscle and liver ofsteroid-implanted and nonimplanted steers. J Anim Sci 81,965–972.Williams GL, Griffith MK, 1995: Sensory and behaviouralcontrol of gonadotroph<strong>in</strong> secretion dur<strong>in</strong>g suckl<strong>in</strong>g-mediatedanovulation <strong>in</strong> cows. J Reprod Fertil Suppl 49, 463–475.Zak LJ, Cosgrove JR, Aherne FX, Foxcroft GR, 1997: Patternof feed <strong>in</strong>take and associated metabolic and endocr<strong>in</strong>echanges differentially affect post-wean<strong>in</strong>g fertility <strong>in</strong> primiparouslactat<strong>in</strong>g sows. J Anim Sci 75, 208–216.Author’s address (for correspondence): MC Lucy, Division of AnimalSciences, University of Missouri, Columbia, MO 65211, USA. E-mail:lucym@missouri.eduConflict of <strong>in</strong>terest: The author declares no conflict of <strong>in</strong>terest.Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Seasonality of <strong>Reproduction</strong> <strong>in</strong> Mammals 4140302010Harvest (10 6 liters)PricePrice (Euros/liter)Harvest0.610.530.460.38% females show<strong>in</strong>g at least one ovulation/oestrus per month1008060402000246OvulationsOestrus81012141618202224262830320MonthsJ F M A M J J A S O N D0.30Oct. Feb. Oct. Feb. Oct. Feb.MonthsFig. 1. Seasonal variations <strong>in</strong> the harvest and farm price of goat milk<strong>in</strong> France (year 1992; adapted from Chem<strong>in</strong>eau et al. 1996)post-partum ovulatory activity <strong>in</strong> beef cattle characterizedby a limited period of time for ovulations (Ingrandet al. 2003; Agabriel et al. 2004).In rams and he-goats, although spermatogenic activityand sexual behaviour do not stop, they deeply varywith season. In Soay rams, a primitive breed from NorthScotland, testicular size (which reflects spermatogenicactivity), plasma FSH and testosterone concentrationsas well as sexual ‘flush’ and aggressive behaviour reachtheir maximum between August and November, the‘rut’ season <strong>in</strong> this breed (L<strong>in</strong>coln 1979). Ile-de-Francerams show testicular weight and sperm production pertestis (directly measured at its output) vary<strong>in</strong>g from lessthan 200 g and 1 billion per day <strong>in</strong> March, to more than300 g and 5 billion per day <strong>in</strong> September, respectively(Ortavant et al. 1985). Alp<strong>in</strong>e bucks also display dramaticvariations <strong>in</strong> sexual behaviour (0–1.5 mat<strong>in</strong>gs <strong>in</strong>10 m<strong>in</strong>), sperm <strong>in</strong>dividual motility (2.5–3.5 over 5) andfertiliz<strong>in</strong>g ability (20%–70% of kidd<strong>in</strong>gs after AI)between the spr<strong>in</strong>g-summer and autumn-w<strong>in</strong>ter periods(Delgadillo et al. 1992). In this last breed, changes <strong>in</strong>ejaculate volume and sperm concentration occur seasonally,which, <strong>in</strong> this species show<strong>in</strong>g a deleteriouseffect of sem<strong>in</strong>al plasma on <strong>in</strong> vitro sperm survival, hasimportant implications on semen technology. F<strong>in</strong>ally,Fig. 3. Seasonal variations of the occurrence of ovulations andoestrous behaviour <strong>in</strong> Alp<strong>in</strong>e goats (adapted from Chem<strong>in</strong>eau et al.1992)stallions also show seasonal variations <strong>in</strong> sexual behaviourand sperm quality, the lowest season be<strong>in</strong>g <strong>in</strong>w<strong>in</strong>ter and the highest <strong>in</strong> spr<strong>in</strong>g-summer (Magistr<strong>in</strong>iet al. 1987).With<strong>in</strong> a given species, the various breeds may expressvariable degrees of seasonality. For example, Soay andTexel ewes are highly seasonal, while Mer<strong>in</strong>o andManchega ewes present more discrete expression ofseasonality (Hafez 1952; Santiago-Moreno et al. 2000).Breeds raised <strong>in</strong> the Subtropics and <strong>in</strong> the Tropicsgenerally present a low seasonality or cycle all the yearround with no anovulatory period (Gonzalez-Stagnaro1983; Khaldi 1984; Yenikoye 1984; Chem<strong>in</strong>eau 1986;Mahieu et al. 1989; Arroyo et al. 2007). This is an<strong>in</strong>terest<strong>in</strong>g trait of these breeds for local farmers whocan then organize the breed<strong>in</strong>g season of their flock allthe year round, without expensive hormonal treatments.Unfortunately, a marked seasonality is expressed <strong>in</strong>these breeds when subjected to the large photoperiodicvariations and temperate climates of northern countries(Chem<strong>in</strong>eau et al. 2004), prevent<strong>in</strong>g the possible use ofthese breeds <strong>in</strong> flocks under temperate latitude. However,<strong>in</strong> temperate breeds ma<strong>in</strong>ta<strong>in</strong>ed under environmentalconditions similar from which they orig<strong>in</strong>ate,<strong>in</strong>tra-breed variability also exists. Some reproductiveFig. 2. Tim<strong>in</strong>g of the annualreproductive cycle of some seasonalbreeders. Breed<strong>in</strong>g season is<strong>in</strong>dicated as a grey box and deliver<strong>in</strong>gseason as a black box(adapted from Ortavant et al.1985)HorseFeral horseFeral cattleGoatTexel sheepSoay sheepMouflonCalifornian mounta<strong>in</strong> sheepAlaska mounta<strong>in</strong> sheepWild pigM<strong>in</strong>kSpr<strong>in</strong>gSummerAutumn W<strong>in</strong>terBreed<strong>in</strong>g seasonBirth seasonJ F M A M J J A S O N D J F M A M J J A S O N DMonthsÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Seasonality of <strong>Reproduction</strong> <strong>in</strong> Mammals 43reproduction (Lark<strong>in</strong> et al. 2002; Lehman et al. 2002;Paul et al. 2008) or pelage moult (Paul et al. 2008). Thisendogenous rhythm is timed, <strong>in</strong> ewes, by discrete signalsgiven by external changes <strong>in</strong> photoperiod (Barrell et al.2000). However, the specific physiological mechanismsunderly<strong>in</strong>g this circannual system rema<strong>in</strong> largely unknown.The specific role of short days (SD) and long days(LD) days <strong>in</strong> tim<strong>in</strong>g reproductive activity varies amongspecies. In long-day breeders (i.e. animals which arefully sexually active dur<strong>in</strong>g LD) like horses, LD arestimulatory and SD <strong>in</strong>hibitory of sexual activity. Incontrast, LD <strong>in</strong>hibit while SD stimulate sexual activity<strong>in</strong> short-day breeders such as sheep and goats. However<strong>in</strong> mammals subjected to a constant photoperiod, thedaylength-specific effect is not permanent. <strong>Animals</strong>‘escape’ and become ’refractory’ to the prevail<strong>in</strong>g photoperiod:LD are no longer stimulatory <strong>in</strong> mares or<strong>in</strong>hibitory <strong>in</strong> sheep and goats, SD are no longer<strong>in</strong>hibitory <strong>in</strong> mares or stimulatory <strong>in</strong> sheep and goats.This refractor<strong>in</strong>ess could conceptually be considered asmerely the first step of the expression of the circannualendogenous rhythm. It can be ‘broken’ by transferr<strong>in</strong>ganimals <strong>in</strong>to the opposite photoperiod: refractor<strong>in</strong>ess toSD, which occurs naturally <strong>in</strong> sheep <strong>in</strong> late w<strong>in</strong>ter, isbroken by 2 months of exposure to LD <strong>in</strong> December–January, allow<strong>in</strong>g the efficiency of stimulatory SD to berestored. Thus, by subject<strong>in</strong>g animals to oppositephotoperiods, it is possible to control seasonality ofreproduction. This property is now commonly used <strong>in</strong>photoperiodic treatments applied on farms and ⁄ or <strong>in</strong>AI centres (see below). The def<strong>in</strong>ition of what is reallyLD and SD is not straightforward: it is possible todef<strong>in</strong>e a threshold of photosensitivity based on thenumber of light hours per day, under which LD arestimulatory and below which SD are <strong>in</strong>hibitory (seereviews Chem<strong>in</strong>eau et al. 1996 and Malpaux et al.1996). The ‘photoperiodic history’ of each <strong>in</strong>dividualshould also be taken <strong>in</strong>to account. Thus, it is nowcommonly accepted that LD are days longer than thepreced<strong>in</strong>g ones, and that SD are days shorter than thepreced<strong>in</strong>g ones. This property is <strong>in</strong>terest<strong>in</strong>g under fieldconditions: follow<strong>in</strong>g a period of artificial long days,animals perceive SD even though natural daylength islonger than 12 h of light per day. Another <strong>in</strong>terest<strong>in</strong>gproperty can be used under farm conditions to applylong days: the illum<strong>in</strong>ation of a specific phase of thenight, the so-called ‘photosensitive phase’, generallysituated 14–16 h after dawn <strong>in</strong> sheep and 9.5 h afterdusk <strong>in</strong> mares, allows animals to perceive LD eventhough real LD are not applied (see reviews of Malpauxet al. 1996; Chem<strong>in</strong>eau et al. 1996 <strong>in</strong> sheep and goats,and Guillaume 1996 <strong>in</strong> mares).More generally, photoperiod, which entra<strong>in</strong>s theendogenous circannual rhythms of reproduction, exertsits action through two different but complementary anddependent pathways by (1) adjust<strong>in</strong>g the phases ofgonadal development with external natural conditionsand (2) by synchroniz<strong>in</strong>g the reproductive periodbetween <strong>in</strong>dividuals of the same species. In mammals,all the photoperiodic <strong>in</strong>put is perceived exclusivelythrough the eyes then transmitted via a multi-synapticpathway to the p<strong>in</strong>eal gland, which transduces thephotic signal <strong>in</strong>to a chemical one by synthesiz<strong>in</strong>g andsecret<strong>in</strong>g melaton<strong>in</strong>. Synthesized dur<strong>in</strong>g the night by thep<strong>in</strong>eal gland, it is thought that melaton<strong>in</strong> is delivered tothe bra<strong>in</strong> via the cerebrosp<strong>in</strong>al fluid, and to peripheraltissues by general circulation. To control reproductiveactivity <strong>in</strong> sheep, melaton<strong>in</strong> acts on the pre-mammillaryhypothalamus where specific receptors transcripts areexpressed (Migaud et al. 2005) and the membranereceptor is present, which stimulates, approximately45 days after the onset of daily impregnation (Viguie´et al. 1995), the pulsatile activity of LHRH-LH which <strong>in</strong>turn will drive gonadal and behavioural sexual activities(review by Malpaux 2006; Malpaux et al. 2001). Externalmelaton<strong>in</strong> can be given to ‘mimic’ short days and hasbeen of practical use <strong>in</strong> sheep and goats to stimulatereproduction <strong>in</strong> spr<strong>in</strong>g. Details of the neuroendocr<strong>in</strong>emechanisms responsible for the seasonal and lightcontrol of LHRH pulsatility could not be discussedextensively here (see Malpaux 2006 for an extensivereview), especially what differs between short-day breeders(sheep and goats) and long-day breeders (horses andhamsters) to expla<strong>in</strong> their opposite change <strong>in</strong> LHRHneuron pulsatility to the same melaton<strong>in</strong> signal. However,it is likely that the difference lies <strong>in</strong> the neuralnetwork connect<strong>in</strong>g hypothalamic melaton<strong>in</strong> receptorbear<strong>in</strong>g cells and LHRH neurones, a network that isalso responsible for the long delay of action of melaton<strong>in</strong>on LHRH pulsatility (Viguié et al. 1995). Forexample, dopam<strong>in</strong>ergic activity <strong>in</strong>hibits the LHRHpulsatility at the end of the neural network dur<strong>in</strong>g longdays <strong>in</strong> sheep (Thiery et al. 1989). Interest<strong>in</strong>gly, dopam<strong>in</strong>efrom the median em<strong>in</strong>ence is also correlated withthe <strong>in</strong>hibition of gonadotrop<strong>in</strong> release, which takes placeunder short days <strong>in</strong> these species (Steger et al. 1985;Glass et al. 1988).Although molecular mechanisms underly<strong>in</strong>g the centralcontrol of gonadotrope system by melaton<strong>in</strong> stillconstitute a ‘black box’, numerous experiments us<strong>in</strong>gonly photoperiodic variations have lead to proposals tofarmers and artificial <strong>in</strong>sem<strong>in</strong>ation centres of specificlight devices, which may be able to control seasonalityof reproduction of their animals.Us<strong>in</strong>g Artificial Photoperiodic Treatments toControl Seasonality of <strong>Reproduction</strong> <strong>in</strong> Farm<strong>Animals</strong>Photoperiodic treatments have been of practical <strong>in</strong>terestfor controll<strong>in</strong>g seasonal reproduction essentially <strong>in</strong>sheep, goats and horses. Sheep and goat AI centres,equipped with dark hous<strong>in</strong>g, use alternate light regimeswith 1 month LD and 1 month SD which allowpermanent high semen production <strong>in</strong> rams and bucks,with no seasonal variations <strong>in</strong> sperm quality. Currently,<strong>in</strong> the French national genetic improvement scheme, allbucks (approximately 70 per year) are permanentlytreated with rapid alternation LD–SD, which <strong>in</strong>creasesthe AI dose production (+40%) per buck and per year(Delgadillo et al. 1993) and reduces duration of thebreed<strong>in</strong>g period of males (slaughtered after 18 monthsof production, or approximately 18 months earlier thatthose ma<strong>in</strong>ta<strong>in</strong>ed under natural photoperiod). In otherAI centers which do not require all the year-roundÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


44 P Chem<strong>in</strong>eau, D Guillaume, M Migaud, JC Thie´ry, MT Pellicer-Rubio and B Malpauxproduction of semen doses, rams raised <strong>in</strong> open barnsare subjected to 2–3 months extra ‘LD’ (from Decemberto February) followed either by return to naturalphotoperiod, or by subcutaneous treatment with melaton<strong>in</strong>implants which mimics SD (‘SD’) (Fig. 5). Thissuccession ‘LD’–‘SD’ stimulates semen output <strong>in</strong> quantityand quality <strong>in</strong> spr<strong>in</strong>g, thus mimick<strong>in</strong>g the normalsexual season which itself normally lasts 2–3 months(Malpaux et al. 1995).In goat farms, (always <strong>in</strong> open barns), males andfemales are subjected to the second type of treatmentperformed <strong>in</strong> AI centres (succession ‘LD’–‘SD’). Thistreatment should be associated with a ‘buck effect’(<strong>in</strong>troduction of treated bucks for 45 days, after 35–75‘SD’) <strong>in</strong> order to <strong>in</strong>duce ovulations and oestrousbehaviour and to get high fertility rate. Under suchconditions, out-of-season fertility and prolificacy can bekept at high levels (>75% kidd<strong>in</strong>g rate with approximatelytwo kids per kidd<strong>in</strong>g) (Chem<strong>in</strong>eau et al. 1996).This type of treatment could be satisfactorily associatedwith AI on fixed-time basis with fertility results equivalentto ‘classical’ hormonal treatment with FGAsponge and eCG (Pellicer-Rubio et al. 2007, 2008).For local breeds <strong>in</strong> subtropical conditions where seasonalityis less marked than those raised under temperatelatitudes, the treatment of females is not necessary.When the LD treatment is applied only <strong>in</strong> bucks usedfor the ‘buck-effect’, the percentages of ovulat<strong>in</strong>gfemales and their fertility after natural mat<strong>in</strong>g are high(Delgadillo et al. 2002, 2004).In ewes, a majority of out-of-season lamb<strong>in</strong>gs are stillobta<strong>in</strong>ed us<strong>in</strong>g ‘classical’ hormonal treatments (FGAsponges + eCG), but the frequency of utilization ofmelaton<strong>in</strong> implants to get out-of-season breed<strong>in</strong>g is<strong>in</strong>creas<strong>in</strong>g, especially <strong>in</strong> Mediterranean countries. Theuse of implants also <strong>in</strong>creases fecundity: + 0.20lamb ⁄ ewe treated ⁄ year has been almost always observed,ma<strong>in</strong>ly due to <strong>in</strong>creased proportions of tw<strong>in</strong>srather than triplets (Chem<strong>in</strong>eau et al. 1996).Photoperiodic treatments are also applied <strong>in</strong> mares toadvance the annual breed<strong>in</strong>g season and to provide foalswith a decisive age-related advance when compet<strong>in</strong>gwith their contemporaries born the same year (seeabove). This is generally performed by expos<strong>in</strong>g maresto LD or pseudo LD dur<strong>in</strong>g autumn and w<strong>in</strong>ter. Suchtreatments allow mares to be fertilized approximately2–3 months earlier than females kept under a naturalphotoperiod (Guillaume 1996).Thus, photoperiodic treatments are now used <strong>in</strong> bothsexes of nearly all farm species to control seasonalreproduction. Whatever the species, they use commonproperties of alternations between <strong>in</strong>hibitory and stimulatoryphotoperiods, where durations are adapted tothe species and sex. When us<strong>in</strong>g pure light treatments(without melaton<strong>in</strong>), especially when applied <strong>in</strong> openbarns, they could be considered as non-<strong>in</strong>vasive whichfully respects animal welfare considerations. It is veryprobable that these photoperiodic treatments will beused more extensively <strong>in</strong> the future as livestock productionsystems strive to be more susta<strong>in</strong>able.(a)Light-proof build<strong>in</strong>gs:17h 16DuskDawn16LProgressivedecrease8L0 60JanuaryMarch150 daysJune(b)17h 16(c)(d)DuskDawn0 60JanuaryMarch17h 16DuskDawn0 60JanuaryMarchOpen barns:2420"Long days"2nd period of suppl. lightShort daysMelaton<strong>in</strong>"Short days"Natural dusk150 daysJune150 daysJunehours16128401st period of suppl. lightJanuary February March April May June"Long days"Natural lightorMelaton<strong>in</strong>"Short days"Natural dawnFig. 5. Photoperiodic treatmentsto control sexual activity <strong>in</strong> smallrum<strong>in</strong>ants raised <strong>in</strong> closed (a–c) oropen (d) barns (adapted fromChem<strong>in</strong>eau et al. 1996)Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Seasonality of <strong>Reproduction</strong> <strong>in</strong> Mammals 45ConclusionThe seasonality of animal products release is more theconsequence of <strong>in</strong>teractions between the naturalendogenous rhythmicity of animals and environmentalconstra<strong>in</strong>ts than a deliberate choice of the farmer toproduce at a def<strong>in</strong>ite season. In most seasonal speciesand breeds <strong>in</strong>volved <strong>in</strong> these productions, specificphotoperiodic treatments derived, at least <strong>in</strong> part,from the cumulative knowledge of physiological mechanisms<strong>in</strong>volved <strong>in</strong> the control of the reproductivefunction, have progressively been proposed to overcomethe problem raised by the seasonal availability ofreproduction-derived products. In mammals, suchtreatments should be taken as an <strong>in</strong>terest<strong>in</strong>g alternativeto classical hormonal treatments <strong>in</strong> a general contextof reduction of hormonal utilization <strong>in</strong> more susta<strong>in</strong>ableanimal production systems. The existence ofstrong genetic bases for seasonality of reproductiveactivity <strong>in</strong> the ma<strong>in</strong> farm animal species should befurther explored to propose selection criteria and ⁄ orgene markers accessible to primary breeder and producerorganizations will<strong>in</strong>g to reduce seasonality <strong>in</strong>their flocks.ReferencesAdams VL, Goodman RL, Salm AK, Coolen LM, Karsch FJ,Lehman MN, 2006: Morphological plasticity <strong>in</strong> the neuralcircuitry responsible for seasonal breed<strong>in</strong>g <strong>in</strong> the ewe.Endocr<strong>in</strong>ology 147, 4843–4851.Agabriel J, Blanc F, Egal D, Dhour P, 2004: Influencescomb<strong>in</strong>ées de la saison de mise bas et de l’exposition autaureau sur la venue en cyclicite´ de vaches Charolaises.Rencontres Recherches Rum<strong>in</strong>ants Paris 8-9 de´c 11, 398.Al-Shorepy SR, Notter DR, 1997: Response to selection forfertility <strong>in</strong> a fall-lamb<strong>in</strong>g sheep flock. J Anim Sci 75, 2033–2040.Anderson GM, Connors JM, Hardy SL, Valent M, GoodmanRL, 2001: Oestradiol microimplants <strong>in</strong> the ventromedialpreoptic area <strong>in</strong>hibit secretion of lute<strong>in</strong>iz<strong>in</strong>g hormone viadopam<strong>in</strong>e neurones <strong>in</strong> anoestrous ewes. J Neuroendocr<strong>in</strong>ol13, 1051–1058.Arroyo LJ, Gallegos-Sanchez J, Villa-Godoy A, BerruecosJM, Perera G, Valencia J, 2007: Reproductive activity ofPelibuey and Suffolk ewes at 19° north latitude. AnimReprod Sci 102, 24–30.Barrell GK, Thrun LA, Brown ME, Viguie C, Karsch FJ,2000: Importance of photoperiodic signal quality to entra<strong>in</strong>mentof the circannual reproductive rhythm of the ewe. BiolReprod 63, 769–774.Bill<strong>in</strong>gs HJ, Viguie C, Karsch FJ, Goodman RL, Connors JM,Anderson GM, 2002: Temporal requirements of thyroidhormones for seasonal changes <strong>in</strong> LH secretion. Endocr<strong>in</strong>ology143, 2618–2625.Bronson FH, 1989: Mammalian Reproductive Biology. TheUniversity of Chicago Press Ltd, Chicago and London.Chem<strong>in</strong>eau P, 1986: Sexual behaviour and gonadal activitydur<strong>in</strong>g the year <strong>in</strong> the tropical Creole meat goat. I. Femaleoestrous behaviour and ovarian activity. Reprod Nutr Dev26, 441–452.Chem<strong>in</strong>eau P, Daveau A, Maurice F, Delgadillo JA, 1992:Seasonality of oestrus and ovulation is not deeply modifiedby submitt<strong>in</strong>g Alp<strong>in</strong>e goats to a tropical photoperiod. SmallRum<strong>in</strong>ant Res 8, 299–312.Chem<strong>in</strong>eau P, Malpaux B, Pelletier J, Leboeuf B, DelgadilloJA, Deletang F, Pobel T, Brice G, 1996: Emploi desimplants de me´laton<strong>in</strong>e et des traitements photope´riodiquespour maîtriser la reproduction saisonnie` re chez les ov<strong>in</strong>s etles capr<strong>in</strong>s. Prod Anim 9, 45–60.Chem<strong>in</strong>eau P, Daveau A, Pelletier J, Malpaux B, Karsch FJ,Viguie C, 2003: Changes <strong>in</strong> the 5-HT 2A receptor system <strong>in</strong>the pre-mammillary hypothalamus of the ewe are related toregulation of LH pulsatile secretion by an endogenouscircannual rhythm. BioMedCentral Neurosci 4, 1 (http://www.biomedcentral.com/1471-2202/4/1).Chem<strong>in</strong>eau P, Daveau A, Cognie Y, Aumont G, Chesneau D,2004: Seasonal ovulatory activity exists <strong>in</strong> tropical Creolefemale goats and Black Belly ewes subjected to a temperatephotoperiod. BioMedCentral Physiol 4, 12 (http://www.biomedcentral.com/1472-6793/4/12).Delgadillo JA, Leboeuf B, Chem<strong>in</strong>eau P, 1992: Abolition ofseasonal variations <strong>in</strong> semen quality and ma<strong>in</strong>tenance ofsperm fertiliz<strong>in</strong>g ability by short photoperiodic cycles <strong>in</strong>he-goats. Small Rum<strong>in</strong>ant Res 9, 47–59.Delgadillo JA, Leboeuf B, Chem<strong>in</strong>eau P, 1993: Ma<strong>in</strong>tenanceof sperm production <strong>in</strong> bucks dur<strong>in</strong>g a third year of shortphotoperiodic cycles. Reprod Nutr Dev 33, 609–617.Delgadillo JA, Flores JA, Veliz FG, Hernandez HF, Duarte G,Vielma J, Po<strong>in</strong>dron P, Chem<strong>in</strong>eau P, Malpaux B, 2002:Induction of sexual activity <strong>in</strong> lactat<strong>in</strong>g anovulatory femalegoats us<strong>in</strong>g male goats treated only with artificially longdays. J Anim Sci 80, 2780–2786.Delgadillo JA, Fitz-Rodriguez G, Duarte G, Veliz FG,Carrillo E, Flores JA, Vielma J, Hernandez H, MalpauxB, 2004: Management of photoperiod to control capr<strong>in</strong>ereproduction <strong>in</strong> the subtropics. Reprod Fertil Dev 16, 471–478.Gallegos-Sa´nchez J, Delaleu B, Caraty A, Malpaux B,Thie´ry JC, 1997: Estradiol acts locally with<strong>in</strong> the retrochiasmaticarea to <strong>in</strong>hibit pulsatile lute<strong>in</strong>iz<strong>in</strong>g-hormonerelease <strong>in</strong> the female sheep dur<strong>in</strong>g anestrus. Biol Reprod56, 1544–1549.Glass JD, Ferrera S, Deaver DR, 1988: Photoperiodic adjustment<strong>in</strong> hypothalamic am<strong>in</strong>es gonadotroph<strong>in</strong>-relesa<strong>in</strong>g hormoneand b-endorph<strong>in</strong> <strong>in</strong> white-footed mouse.Endocr<strong>in</strong>ology 123, 1119–1127.Gonzalez-Stagnaro C, 1983: Comportamiento reproductivo delas razas locales de rumiantes en el tropico americano. In:Chem<strong>in</strong>eau P, Gauthier D, Thimonier J (eds), <strong>Reproduction</strong>des Rum<strong>in</strong>ants en Zone Tropicale, Vol. 20. Inra Publications,Versailles, France, pp. 1–84.Guillaume D, 1996: Action de la photope´riode sur la reproductiondes équidés. Prod Anim 9, 61–69.Hafez ESE, 1952: Studies on the breed<strong>in</strong>g season andreproduction of the ewe. J Agric Sci 42, 189–265.Hanocq E, Bod<strong>in</strong> L, Thimonier J, Teyssier J, Malpaux B,Chem<strong>in</strong>eau P, 1999: Genetics parameters of spontaneousspr<strong>in</strong>g ovulatory activity <strong>in</strong> Mer<strong>in</strong>os d’Arles sheep. GenetSel Evol 31, 77–90.Hanrahan JP, 1987: Genetic variation <strong>in</strong> seasonal reproduction<strong>in</strong> sheep. Proceed<strong>in</strong>gs 38th Annual Meet<strong>in</strong>g of theEuropean Association for Animal Production, Lisbon,Portugal, 28 September–1 October, 14 pp.Ingrand S, Cournut S, Dedieu B, Antheaume F, 2003: Laconduite de la reproduction du troupeau de vaches allaitantes:mode´lisation des prises de décision. Prod Anim 16,263–270.Jansen HT, Jackson GL, 1993: Circannual rhythms <strong>in</strong> theewe: patterns of ovarian cycles and prolact<strong>in</strong> secretionunder two different constant photoperiods. Biol Reprod49, 627–634.Karsch FJ, Bittman EL, Foster DL, Goodman RL, Legan SJ,Rob<strong>in</strong>son JE, 1984: Neuroendocr<strong>in</strong>e basis of seasonalreproduction. Recent Progress Hormone Res 40, 185–232.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


46 P Chem<strong>in</strong>eau, D Guillaume, M Migaud, JC Thie´ry, MT Pellicer-Rubio and B MalpauxKhaldi G, 1984: Variations saisonnie` res de l’activite´ ovarienne,du comportement d’oestrus et de la dure´e del’anoestrus post-partum des femelles ov<strong>in</strong>es de raceBarbar<strong>in</strong>e: <strong>in</strong>fluence du niveau alimentaire et de lapre´sence du mâle. PhD Thesis, University of Montpellier,France, 168 pp.Langlois B, Blou<strong>in</strong> C, 1997: Effect of a horse’s month of birthon its future sport performance. I. Effect on annualphenoypte <strong>in</strong>dices. Ann Zootech 46, 393–398.Langlois B, Blou<strong>in</strong> C, 1998: Effect of a horse’s month ofbirth on its future sport performance. II. Effect on annualearn<strong>in</strong>g and annual earn<strong>in</strong>g per start. Ann Zootech 47,67–74.Langlois B, Blou<strong>in</strong> C, 2004: Statistical analysis of some factorsaffect<strong>in</strong>g the number of horse births <strong>in</strong> France. Reprod NutrDev 44, 583–596.Lark<strong>in</strong> JE, Jones J, Zucker I, 2002: Temperature dependenceof gonadal regression <strong>in</strong> Syrian hamsters exposed to shortday lengths. Am J Physiol-Regul Integr Comp Physiol 282,R744–R752.Le Corre S, Segu L, Caldani M, Chem<strong>in</strong>eau P, 1994:Differences <strong>in</strong> Ketanser<strong>in</strong> b<strong>in</strong>d<strong>in</strong>g <strong>in</strong> the ventromedialhypothalamus of ewes responsive or refractory to shortdays. Neuroendocr<strong>in</strong>ology 60, 589–600.Legan SJ, Karsch FJ, 1980: Photoperiodic control of seasonalbreed<strong>in</strong>g <strong>in</strong> ewes: modulation of the negative feedbackaction of estradiol. Biol Reprod 23, 1061–1068.Lehman MN, Coolen LM, Goodman RL, Viguie C, Bill<strong>in</strong>gsHJ, Karsch FJ, 2002: Seasonal plasticity <strong>in</strong> the bra<strong>in</strong>: the useof large animal models for neuroanatomical research.<strong>Reproduction</strong> Suppl 59, 149–165.L<strong>in</strong>coln G, 1979: Photoperiodic control of seasonal breed<strong>in</strong>g <strong>in</strong>the ram: participation of the cranial sympathetic nervoussystem. J Endocr<strong>in</strong>ol 82, 135–147.Magistr<strong>in</strong>i M, Chanteloube P, Palmer E, 1987: Influence ofseason and frequency of ejaculation on production ofstallion semen for freez<strong>in</strong>g. J Reprod Fertil Suppl 35, 127–133.Mahieu M, Jego Y, Driancourt MA, Chem<strong>in</strong>eau P, 1989:Reproductive performances of Creole and Black-Belly ewes<strong>in</strong> the West Indies. A new major gene controll<strong>in</strong>g ovulationrate? Anim Reprod Sci 19, 235–243.Malpaux B, 2006: Seasonal regulation of reproduction <strong>in</strong>mammals. In: Neill JD (ed.), Knobil and Neill’s Physiologyof <strong>Reproduction</strong>, 3rd edn. Elsevier, Amsterdam, pp. 2231–2281.Malpaux B, Maurice–Mandon F, Daveau A, Chem<strong>in</strong>eau P,1995: Utilisation de la lumie` re et de la me´laton<strong>in</strong>e pour lamaıˆ trise de la reproduction des ov<strong>in</strong>s et des capr<strong>in</strong>s.Rencontres Recherches Rum<strong>in</strong>ants Paris 13–14 de´c 2, 379–386.Malpaux B, Viguie C, Thiery JC, Chem<strong>in</strong>eau P, 1996:Controˆ le photope´riodique de la reproduction. Prod Anim9, 9–23.Malpaux B, Migaud M, Tricoire H, Chem<strong>in</strong>eau P, 2001:Biology of mammalian photoperiodism and the critical roleof the p<strong>in</strong>eal gland and melaton<strong>in</strong>. J Biol Rhythms 16, 336–347.Migaud M, Daveau A, Malpaux B, 2005: MTNR1A melaton<strong>in</strong>receptors <strong>in</strong> the ov<strong>in</strong>e premammillary hypothalamus:day–night variation <strong>in</strong> the expression of the transcripts. BiolReprod 72, 393–398.Ortavant R, Pelletier J, Ravault JP, Thimonier J, Volland P,1985: Photoperiod: ma<strong>in</strong> proximal and distal factor of thecircannual cycle of reproduction <strong>in</strong> farm mammals. OxfordRev Reprod Biol 7, 305–345.Palmer E, Driancourt MA, 1983: Some <strong>in</strong>teractions of seasonof foal<strong>in</strong>g, photoperiod and ovarian activity <strong>in</strong> the equ<strong>in</strong>e.Livest Prod Sci 10, 197–210.Paul MJ, Zucker I, Schwartz WJ, 2008: Track<strong>in</strong>g the seasons:the <strong>in</strong>ternal calendars of vertebrates. Philos Trans R SocLond B Biol Sci 363, 341–361.Pelletier J, Ortavant R, 1975: Photoperiodic control of LHrelease <strong>in</strong> the ram. II. Light-androgens <strong>in</strong>teraction. ActaEndocr<strong>in</strong>ol (Copenhagen) 78, 442–450.Pellicer-Rubio MT, Leboeuf B, Bernelas D, Forgerit Y,Pougnard JL, Bonne JL, Senty E, Chem<strong>in</strong>eau P, 2007:Highly synchronous and fertile reproductive activity <strong>in</strong>ducedby the male effect dur<strong>in</strong>g deep anoestrus <strong>in</strong> lactat<strong>in</strong>ggoats subjected to treatment with artificially long daysfollowed by a natural photoperiod. Anim Reprod Sci 98,241–258.Pellicer-Rubio MT, Leboeuf B, Bernelas D, Forgerit Y,Pougnard JL, Bonne JL, Senty E, Breton S, Brun F,Chem<strong>in</strong>eau P, 2008: High fertility us<strong>in</strong>g artificial <strong>in</strong>sem<strong>in</strong>ationdur<strong>in</strong>g deep anoestrus after <strong>in</strong>duction and synchronisationof ovulatory activity by the ‘male effect’<strong>in</strong> lactat<strong>in</strong>ggoats subjected to treatment with artificial long days andprogestagens. Anim Reprod Sci (<strong>in</strong> press).Quirke JF, Hanrahan JP, Loughnane W, Triggs R, 1986:Components of the breed<strong>in</strong>g and non-breed<strong>in</strong>g seasons <strong>in</strong>sheep: breed effects and repeatability. Irish J Agr Food Res25, 167–172.Ricordeau G, 1982: Selection for reduced seasonality and postpartumanoestrus. In: Second World Congress of GeneticsApplied to Livestock Production, Madrid, Spa<strong>in</strong>, 5, 338–347.Salazar-Ortiz J, 2006: Bilan des effets du niveau d’alimentationsur la reproduction de la jument. PhD Thesis, University ofTours, France, 343 pp.Santiago-Moreno J, Lopez-Sebastian A, Gonzalez-Bulnes A,Gomez-Brunet A, Chem<strong>in</strong>eau P, 2000: Seasonal changes<strong>in</strong> ovulatory activity, plasma prolact<strong>in</strong>, and melaton<strong>in</strong>concentrations, <strong>in</strong> Mouflon (Ovis gmel<strong>in</strong>i musimon) andManchega (Ovis aries) ewes. Reprod Nutr Dev 40, 421–430.Smith JF, Johnson DL, Reid TC, 1992: Genetic parametersand performance of flocks selected for advancedlamb<strong>in</strong>g date. Proc New Zealand Soc Anim Prod 52, 50(Abstr).Steger RW, Matt KS, Klemke HG, Bartke A, 1985: Interactionsof photoperiod and ectopic graphs on hypothalamicand pituitary function <strong>in</strong> male hamsters. Neuroendocr<strong>in</strong>ology41, 89–96.Thiery JC, Malpaux B, 2003: Seasonal regulation ofreproductive activity <strong>in</strong> sheep: modulation of access ofsex steroids to the bra<strong>in</strong>. Ann N Y Acad Sci 1007, 169–175.Thiery JC, Mart<strong>in</strong> GB, Tillet Y, Caldani M, Quent<strong>in</strong> M,Jama<strong>in</strong> C, Ravault JP, 1989: Role of hypothalamic catecholam<strong>in</strong>es<strong>in</strong> the regulation of lute<strong>in</strong>iz<strong>in</strong>g hormone andprolact<strong>in</strong> secretion <strong>in</strong> the ewe dur<strong>in</strong>g seasonal anestrus.Neuroendocr<strong>in</strong>ology 49, 80–87.Thiery JC, Gayrard V, Le Corre S, Viguie C, Mart<strong>in</strong> GB,Chem<strong>in</strong>eau P, Malpaux B, 1995: Dopam<strong>in</strong>ergic control ofLH secretion by the A15 nucleus <strong>in</strong> anoestrous ewes. JReprod Fertil Suppl 49, 285–296.Thiery JC, Lomet D, Schumacher M, Liere P, Tricoire H,Locatelli A, Delagrange P, Malpaux B, 2006: Concentrationsof estradiol <strong>in</strong> ewe cerebrosp<strong>in</strong>al fluid are modulatedby photoperiod through p<strong>in</strong>eal-dependent mechanisms. JP<strong>in</strong>eal Res 41, 306–312.Thimonier J, Mauléon P, 1969: Variations saisonnie` res ducomportement d’oestrus et des activite´s ovariennes ethypophysaires chez les ov<strong>in</strong>s. Ann Biol Anim Bioch Biophys9, 233–250.Viguie´ C, Caraty A, Locatelli A, Malpaux B, 1995: Regulationof lute<strong>in</strong>iz<strong>in</strong>g hormone-releas<strong>in</strong>g hormone (LHRH)Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Seasonality of <strong>Reproduction</strong> <strong>in</strong> Mammals 47secretion by melaton<strong>in</strong> <strong>in</strong> the ewe. I. Simultaneous delayed<strong>in</strong>crease <strong>in</strong> LHRH and lute<strong>in</strong>iz<strong>in</strong>g hormone pulsatile secretion.Biol Reprod 52, 1114–1120.Yenikoye A, 1984: Annual variations <strong>in</strong> estrual behavior, rateand possibilities for ovulation <strong>in</strong> Peulh ewes from Niger.Reprod Nutr Dev 24, 11–19.Author’s address (for correspondence): P Chem<strong>in</strong>eau, UMR Physiologiede la <strong>Reproduction</strong> et des Comportements, INRA, CNRS,Université F. Rabelais, Haras Nationaux, 37380 Nouzilly, France.E-mail: philippe.chem<strong>in</strong>eau@tours.<strong>in</strong>ra.frConflict of <strong>in</strong>terest: P Chem<strong>in</strong>eau has received a research grant fromCEVA Animal Health and has no potential conflicts to declare.J-C Thiéry has received a research grant from ‘‘Institut de RechercheServier’’ and has no potential conflicts to declare.D Guillaume has no potential conflicts to declare.M Migaud has received a research grant from ‘‘Institut de RechercheServier’’ and has no potential conflicts to declare.M-T Pellicer-Rubio has no potential conflicts-to declare.B Malpaux has received a research grant from ‘‘Institut de RechercheServier’’, from ‘‘CEVA Animal Health’’ and has no potential conflictsto declare.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 48–56 (2008); doi: 10.1111/j.1439-0531.2008.01142.xISSN 0936-6768Mechanisms for Dom<strong>in</strong>ant Follicle Selection <strong>in</strong> Monovulatory Species: AComparison of Morphological, Endocr<strong>in</strong>e and Intraovarian Events <strong>in</strong> Cows, Maresand WomenM Mihm 1 and ACO Evans 21 Division of Cell Sciences, Faculty of Veter<strong>in</strong>ary Medic<strong>in</strong>e, University of Glasgow, Glasgow, UK; 2 School of Agriculture, Food Science andVeter<strong>in</strong>ary Medic<strong>in</strong>e and the Conway Institute, University College Dubl<strong>in</strong>, Belfield, Dubl<strong>in</strong> 4, IrelandContentsThe selection of a s<strong>in</strong>gle ovarian follicle for further differentiationand f<strong>in</strong>ally ovulation is a shared phenomenon <strong>in</strong>monovulatory species from different phylogenetic classes. Thecommonality of dom<strong>in</strong>ant follicle (DF) development leads usto hypothesize that mechanisms for DF selection are conserved.This review highlights similarities and differences <strong>in</strong>follicular wave growth between cows, mares and women,addresses the commonality of the transient rises <strong>in</strong> FSHconcentrations, and discusses the follicular secretions oestradioland <strong>in</strong>hib<strong>in</strong> with their regulatory roles for FSH. In allthree species, ris<strong>in</strong>g FSH concentrations <strong>in</strong>duce the emergenceof a follicle wave and cohort attrition occurs dur<strong>in</strong>g decl<strong>in</strong><strong>in</strong>gFSH concentrations, culm<strong>in</strong>at<strong>in</strong>g <strong>in</strong> DF selection. Cohortsecretions are <strong>in</strong>itially responsible for decl<strong>in</strong><strong>in</strong>g FSH, which issubsequently suppressed by the selected DF lower<strong>in</strong>g it belowthe threshold of FSH requirements of all other cohort follicles.The DF acquires relative FSH-<strong>in</strong>dependence <strong>in</strong> order tocont<strong>in</strong>ue growth and differentiation dur<strong>in</strong>g low (cow, mare)or further decl<strong>in</strong><strong>in</strong>g FSH concentrations (women), and thusmay be the one cohort follicle with the lowest FSH requirementdue to enhanced FSH signall<strong>in</strong>g. In all three monovulatoryspecies a transition from FSH- to LH-dependence ispostulated as the mechanism for the cont<strong>in</strong>ued development ofthe selected DF. In addition, FSH and IGF enhance eachother’s ability to stimulate follicle cell function and access ofIGF-I and -II to the type 1 receptor is regulated by IGFb<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s that are <strong>in</strong> turn regulated by specificproteases; all of which have been ascribed a role <strong>in</strong> DFdevelopment. No fundamental differences <strong>in</strong> DF selectionmechanisms have been identified between the different speciesstudied. Thus functional studies of the selection of DFs <strong>in</strong>cattle and mares are also valuable for identify<strong>in</strong>g genes andpathways regulat<strong>in</strong>g DF development <strong>in</strong> women.IntroductionLarge antral follicle growth <strong>in</strong> ovaries does not occurrandomly. For example <strong>in</strong> cattle, detailed endocr<strong>in</strong>e andquantitative morphological studies followed by thosewhich allowed the <strong>in</strong>dividual monitor<strong>in</strong>g of antralfollicles from 3 mm <strong>in</strong> diameter [surgical (<strong>in</strong>k mark<strong>in</strong>gof follicles) and ultrasound studies] showed that growthis wave-like and coord<strong>in</strong>ated, and is reflected <strong>in</strong> regularfluctuations of follicular hormone secretions (Evans2003; Mihm and Bleach 2003). In adults from monovulatoryspecies f<strong>in</strong>al antral follicle growth is characterizedby a shared phenomenon whereby a s<strong>in</strong>gle follicle1 Please note that <strong>in</strong> this review the general term ‘cows’is used to <strong>in</strong>clude heifers and parous cows.[the dom<strong>in</strong>ant follicle (DF)] is selected from multipleand simultaneously grow<strong>in</strong>g cohort follicles to undergof<strong>in</strong>al differentiation and ultimately ovulate, or at leastacquire the ability for ovulation (G<strong>in</strong>ther et al. 2001a).Clearly, the development of a mechanism to guarantees<strong>in</strong>gle offspr<strong>in</strong>g <strong>in</strong>creased the chances of its pre- andpostnatal survival <strong>in</strong> these species.There is commonality <strong>in</strong> follicle wave growth <strong>in</strong> verydifferent species from different classes, orders andfamilies of the phylogenetic tree. This leads us tohypothesize that conserved mechanisms for DF selectionexist. Identification of such shared mechanisms isof great importance to veter<strong>in</strong>ary medic<strong>in</strong>e and science,as well as human reproductive medic<strong>in</strong>e. In all studiedmonovulatory species disruption of the selection phenomenonoccurs; for example, when tw<strong>in</strong> or multipleovulations occur, or <strong>in</strong> polycystic ovary syndrome,which is diagnosed <strong>in</strong> more than 75% of women withanovulatory <strong>in</strong>fertility (Franks et al. 2006). This canresult <strong>in</strong> temporary or long-term <strong>in</strong>fertility due to cycleabnormalities, abortions, <strong>in</strong>creased foetal and neonataldisease and mortality, or sub(<strong>in</strong>)fertility follow<strong>in</strong>gdystocia and post-partum disease when multiples aredelivered. Thus it is essential to develop a betterunderstand<strong>in</strong>g and improved methods for diagnosisand treatment of any exist<strong>in</strong>g pathology.Conversely, controlled <strong>in</strong>terference with DF selectionto improve the reproductive efficiency or fertility <strong>in</strong> thedifferent species (which <strong>in</strong>clude treatments for tw<strong>in</strong>n<strong>in</strong>g,or superovulatory treatments for embryo transfer) mayhave enormous economic (agriculturally important species)or psychological (women) benefits. Interest<strong>in</strong>gly,however, the likelihood of double ovulations ⁄ tw<strong>in</strong>n<strong>in</strong>gdecreases even among known monovulatory speciesfrom mares (20% <strong>in</strong> thoroughbred mares, G<strong>in</strong>ther 1992)to cows 1 (


Dom<strong>in</strong>ant Follicle Selection <strong>in</strong> Cows, Mares and Women 49been determ<strong>in</strong>ed <strong>in</strong> the three species. Subsequently, twoimportant <strong>in</strong>trafollicular functions regulated dur<strong>in</strong>g DFselection (FSH and LH-responsiveness, <strong>in</strong>sul<strong>in</strong>-likegrowth factor (IGF) b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> production) areproposed as examples of conserved DF selection mechanisms.F<strong>in</strong>ally, new candidate genes for atta<strong>in</strong>ment ofdom<strong>in</strong>ance are presented from genomic bov<strong>in</strong>e studies.Dom<strong>in</strong>ant Follicle Selection: Follicle WaveMorphologyAt specific times dur<strong>in</strong>g the luteal phase of the oestrouscycle <strong>in</strong> cows, but also at <strong>in</strong>tervals of 7–10 days <strong>in</strong> mostother reproductive states (and this <strong>in</strong>cludes the prepubertal,post-partum and early pregnant anoestrousstate), rapid growth of a cohort of small (from largerthan 1 mm <strong>in</strong> diameter) antral follicles to 3–5 mm andbeyond is observed us<strong>in</strong>g daily ovarian ultrasound(Roche et al. 1998; Ireland et al. 2000). This observationhas been termed ‘emergence of a follicular wave’ and isfollowed by cont<strong>in</strong>ued growth of a constantly reduc<strong>in</strong>gnumber of cohort follicles over the next 3 days, whereonly the m<strong>in</strong>ority will reach diameters of 6 mm, two orthree follicles will reach 7–8 mm, and generally only onefollicle is selected to be the DF and grows beyond 8 mm<strong>in</strong> diameter (Fig. 1). For a large number of <strong>in</strong> vivostudies, the time of DF selection <strong>in</strong> the bov<strong>in</strong>e has beenrout<strong>in</strong>ely def<strong>in</strong>ed us<strong>in</strong>g ultrasound criteria such as thetime of divergence between the DFs cont<strong>in</strong>ued rapidgrowth vs the reduced growth shown by its closestcompetitor, the largest subord<strong>in</strong>ate follicle (SF1) (onsetof deviation; G<strong>in</strong>ther et al. 1997), or a m<strong>in</strong>imum size of8.5 mm and m<strong>in</strong>imum diameter difference of 1 to>2 mm to the SF1 (G<strong>in</strong>ther et al. 1997; Mihm et al.1997, 2008). All other cohort (subord<strong>in</strong>ate) folliclescease growth at different times after wave emergence,decrease <strong>in</strong> size, and eventually disappear. In general,two or three follicle waves emerge dur<strong>in</strong>g the lutealFig. 1. The relationship between the transient rise <strong>in</strong> FSH, follicle wavegrowth, which culm<strong>in</strong>ates <strong>in</strong> selection of the DF, and the follicularsecretions oestradiol and <strong>in</strong>hib<strong>in</strong> <strong>in</strong> cows, mares and women. Note that<strong>in</strong> women this is most likely the ovulatory follicle wave which emergesat luteolysis and grows dur<strong>in</strong>g the early follicular phase, <strong>in</strong> mares theovulatory follicle wave emerges <strong>in</strong> the second half of the luteal phaseand the DF is selected at luteolysis, and <strong>in</strong> cows the most studied folliclewave is the first wave of the cycle, which is anovulatory and emerges atovulation, with DF selection occurr<strong>in</strong>g <strong>in</strong> the early luteal phase.Cohort, dom<strong>in</strong>ant and subord<strong>in</strong>ate follicles <strong>in</strong> mares are generally twicethe size of follicles <strong>in</strong> the other two species. Note the longer FSH decl<strong>in</strong>e<strong>in</strong> mares compared with cows, and the particularly long FSH decl<strong>in</strong>e <strong>in</strong>women. DF, dom<strong>in</strong>ant follicle, SF, subord<strong>in</strong>ate folliclesphase of the 21-day oestrous cycle <strong>in</strong> cattle, eachselect<strong>in</strong>g a DF, and with the f<strong>in</strong>al DF selected atluteolysis or shortly after and thus becom<strong>in</strong>g theovulatory DF <strong>in</strong> the relatively short follicular phase(Savio et al. 1988; G<strong>in</strong>ther et al. 1989). Very rarely theemergence of a wave has been detected which may growfor one day but does not lead to selection of a new DF(Mihm et al. 1999). There are several <strong>in</strong>terest<strong>in</strong>g featuresof DF selection <strong>in</strong> the bov<strong>in</strong>e, apparent when study<strong>in</strong>gthis process closely by ultrasound: (1) the future DFgenerally seems to emerge first, 6–7 h before the largestSFI, and the size advantage is ma<strong>in</strong>ta<strong>in</strong>ed throughoutthe common parallel growth phase until deviation(G<strong>in</strong>ther et al. 1997); (2) the number of cohort folliclesemerg<strong>in</strong>g over a 48–72 h period is highly variablebetween animals and can also vary with<strong>in</strong> the cycle,rang<strong>in</strong>g from only 2 to more than 50 small antralfollicles per wave (Burns et al. 2005); (3) follicle numbersper wave <strong>in</strong> animals represent<strong>in</strong>g the two extremes, thatis <strong>in</strong> animals with very low (25) numbers of follicles per wave are consistentwith<strong>in</strong> the cycle and from cycle to cycle (Burns et al.2005); (4) follicle numbers per wave do not usually<strong>in</strong>fluence the process of select<strong>in</strong>g one s<strong>in</strong>gle DF, as nohigher <strong>in</strong>cidence of co-dom<strong>in</strong>ant follicles has beenreported <strong>in</strong> animals show<strong>in</strong>g very high follicle numbers(Burns et al. 2005), or <strong>in</strong> animals treated exogenouslywith bov<strong>in</strong>e growth hormone which <strong>in</strong>creases thenumber of small antral follicles (cohort follicles) peranimal (Gong et al. 1991). The exception are cows fromherds selected for tw<strong>in</strong>n<strong>in</strong>g such as those described <strong>in</strong>Echternkamp 2000; (5) the diameter of the DF at thetime of selection (approximately 8.5 mm) is very consistentacross all (oestrous and anoestrous) reproductivestates, and across different breeds and types of cattle,and <strong>in</strong> cyclic animals is <strong>in</strong>dependent of when dur<strong>in</strong>g theluteal phase (beg<strong>in</strong>n<strong>in</strong>g, mid, end) the DF emerges(Roche et al. 1998; Ireland et al. 2000; G<strong>in</strong>ther et al.2001a); and (6) co-dom<strong>in</strong>ance <strong>in</strong> animals not geneticallyselected for tw<strong>in</strong>n<strong>in</strong>g is generally rare (approximately5%) but can occur either temporarily with late selectionof a s<strong>in</strong>gle follicle, or throughout the complete dom<strong>in</strong>anceperiod, lead<strong>in</strong>g to tw<strong>in</strong> ovulations if the last waveof the cycle is affected (Echternkamp 2000; Kulick et al.2001) Overall, these features <strong>in</strong>dicate that the DFselection mechanism <strong>in</strong> the bov<strong>in</strong>e is very robust and<strong>in</strong> unselected cows can only be <strong>in</strong>hibited by pharmacological<strong>in</strong>terference which elevates FSH or abolishesGnRH pulsatility (Prendiville et al. 1995; Gong et al.1996; Mihm et al. 1997).In contrast to cows, <strong>in</strong> mares and women not allfollicle waves detected dur<strong>in</strong>g an <strong>in</strong>terovulatory <strong>in</strong>tervalwill result <strong>in</strong> the selection of a DF (these are termed‘m<strong>in</strong>or waves’ where cohort follicles never reach the sizeof a DF; details of major and m<strong>in</strong>or wave follicledynamics <strong>in</strong> both species are reported <strong>in</strong> G<strong>in</strong>ther 1993;Baerwald et al. 2003; G<strong>in</strong>ther et al. 2004a). The ovulatorywave <strong>in</strong> mares, which is the only follicle waveguaranteed to result <strong>in</strong> selection of a DF, emerges <strong>in</strong> thesecond half of the luteal phase lead<strong>in</strong>g to selection of theDF co<strong>in</strong>cident with luteal regression. In women, however,the ovulatory wave only emerges dur<strong>in</strong>g the firsthalf of the follicular phase, and aga<strong>in</strong> is the only waveÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


50 M Mihm and ACO Evansguaranteed to result <strong>in</strong> a DF. Thus, mares and womenshare very similar wave dynamics: (1) The occurrence ofmajor (lead<strong>in</strong>g to selection of a DF) anovulatory wavesdur<strong>in</strong>g the luteal phase <strong>in</strong> 20% (mares) or 30% (women)of cycles. (2) The occurrence of m<strong>in</strong>or waves (whichdescribes the emergence and growth of cohort folliclesfollowed by their regression without DF selection)dur<strong>in</strong>g both the luteal and follicular phases of the cycle,albeit the percentage differs between mares (almost100%) and women (only 57%). (3) The high percentageof pre-deviation follicles (mares 37%, women 48%)which are follicles that emerge with or ahead of the DF<strong>in</strong> ovulatory waves, reach DF size 1 day before theovulatory DF but beg<strong>in</strong> to regress at the time ofdeviation of the ovulatory DF. At emergence anddeviation, respectively, cows and women share verysimilar cohort (3–6 mm), largest subord<strong>in</strong>ate (7–8 mm)and DF diameters (8.5–10 mm) determ<strong>in</strong>ed by ultrasound.Only women show an enhanced growth rate ofthe DF and regression rate of SF follow<strong>in</strong>g deviation,while <strong>in</strong> mares and cows growth rates of DF arema<strong>in</strong>ta<strong>in</strong>ed at rates seen before deviation (G<strong>in</strong>ther et al.2001b, 2004a). While ‘local’ effects of DF on antralfollicles from the ipsi- vs the contralateral ovary havenot been described <strong>in</strong> cows or mares, <strong>in</strong> women the DFaffects subord<strong>in</strong>ate growth and atresia on the ispilateralovary follow<strong>in</strong>g its selection, lead<strong>in</strong>g to ‘corona formation’around the DF towards the end of the follicularphase (Gore et al. 1997). Compared to women, tw<strong>in</strong>ovulations are frequent <strong>in</strong> mares (20%, G<strong>in</strong>ther 1992;G<strong>in</strong>ther et al. 2004a) and have recently been shown toreach similar levels <strong>in</strong> high-yield<strong>in</strong>g dairy cows(Wiltbank et al. 2000). Similar to cows, follicle waveshave also been observed dur<strong>in</strong>g the later half of theanovulatory season (Donadeu and G<strong>in</strong>ther 2002a;Watson and Al-zi’abi 2002) and dur<strong>in</strong>g early pregnancy<strong>in</strong> mares (G<strong>in</strong>ther and Bergfelt 1992). However, theoccurrence of follicular waves dur<strong>in</strong>g anovulatory states(such as dur<strong>in</strong>g pregnancy or follow<strong>in</strong>g weight loss) <strong>in</strong>women does not appear to have been <strong>in</strong>vestigated.The FSH Rise and Decl<strong>in</strong>eIn all three monovulatory species, ris<strong>in</strong>g FSH concentrations<strong>in</strong> systemic circulation <strong>in</strong>duce the emergence ofa follicle wave which is usually detected at the time ofthe FSH peak (Aust<strong>in</strong> et al. 2001; G<strong>in</strong>ther et al. 2005)(Fig. 1). Cohort attrition occurs dur<strong>in</strong>g decl<strong>in</strong><strong>in</strong>g FSHconcentrations, and culm<strong>in</strong>ates <strong>in</strong> DF selection (cows:Adams et al. 1992; Sunderland et al. 1994; mares:Bergfelt and G<strong>in</strong>ther 1993; women: Van Santbr<strong>in</strong>ket al. 1995). All grow<strong>in</strong>g cohort follicles contribute tothe FSH suppression, lead<strong>in</strong>g to a direct relationshipbetween the number of cohort follicles grow<strong>in</strong>g <strong>in</strong>response to the FSH rise and the degree of FSHsuppression (Gibbons et al. 1997; Burns et al. 2005;G<strong>in</strong>ther et al. 2005). The FSH decl<strong>in</strong>e causes DFselection and this has been shown functionally byprevent<strong>in</strong>g the decl<strong>in</strong>e or elevat<strong>in</strong>g FSH <strong>in</strong> cows(Adams et al. 1993; Mihm et al. 1997), mares (Donadeuand G<strong>in</strong>ther 2001) and women (Schipper et al.1998; Hohmann et al. 2001). Therefore, DF selection isa systemic process simultaneously affect<strong>in</strong>g cohortgrowth on both ovaries via reductions <strong>in</strong> FSH follow<strong>in</strong>ga transient rise. However, the <strong>in</strong>cidence of transientFSH rises, the magnitude of each rise, and the lengthof the decl<strong>in</strong>e differ between the species. In cows,sequential FSH rises of similar magnitude as thegonadotroph<strong>in</strong> surge are associated with new folliclewaves dur<strong>in</strong>g the luteal phase of the oestrous cycle(Adams et al. 1992; Sunderland et al. 1994), <strong>in</strong> thepost-partum period (Crowe et al. 1998; Stagg et al.1998), dur<strong>in</strong>g pregnancy (G<strong>in</strong>ther et al. 1996) andbefore puberty (Evans et al. 1994). Functional <strong>in</strong> vivostudies showed that suppress<strong>in</strong>g the first or second riseof FSH dur<strong>in</strong>g the cycle or long-term suppression ofGnRH pulsatility will <strong>in</strong>hibit wave emergence, whichcan aga<strong>in</strong> be <strong>in</strong>duced us<strong>in</strong>g exogenous FSH (Mihm andBleach 2003).Similar to cows, several transient FSH rises have beendemonstrated dur<strong>in</strong>g the luteal phase <strong>in</strong> the mares (200–400% of basal levels), but <strong>in</strong>terest<strong>in</strong>gly, luteal rises arenot always associated with emergence of a follicle waveas detected by ultrasound (Gastal et al. 1997; G<strong>in</strong>theret al. 2005). Three features are different <strong>in</strong> womencompared to cows and mares: (1) no m<strong>in</strong>or or majorwaves emerge dur<strong>in</strong>g the early to mid luteal phase(Baerwald et al. 2003); (2) very small magnitude FSHrises can <strong>in</strong>duce emergence of the ovulatory wave<strong>in</strong>dicat<strong>in</strong>g a relatively high sensitivity of the populationof small antral follicles to FSH (Baird 1987; VanSantbr<strong>in</strong>k et al. 1995); and (3) DF selection occurs alsodur<strong>in</strong>g the <strong>in</strong>itial decl<strong>in</strong>e <strong>in</strong> FSH, but FSH cont<strong>in</strong>ues todecl<strong>in</strong>e dur<strong>in</strong>g the long dom<strong>in</strong>ance period of the DF.Thus FSH is elevated for more than 10 days <strong>in</strong> thefollicular phase (Van Santbr<strong>in</strong>k et al. 1995; Baerwaldet al. 2003), which is <strong>in</strong> contrast to cows and mareswhere a f<strong>in</strong>al reduction of FSH to nadir levels occursjust after deviation (and this f<strong>in</strong>al decl<strong>in</strong>e is hypothesizedto <strong>in</strong>duce SF1 atresia at deviation, G<strong>in</strong>ther et al. 1999;Bergfelt et al. 2001).The Follicular Secretions Oestradiol and Inhib<strong>in</strong>The FSH decl<strong>in</strong>e causes DF selection, and <strong>in</strong>itialsuppression of FSH concentrations follow<strong>in</strong>g the riseis contributed to the cohort follicles grow<strong>in</strong>g after waveemergence <strong>in</strong> all three monovulatory species (Fig. 1)(G<strong>in</strong>ther et al. 2001a). Thus, cohort secretions <strong>in</strong>to thesystemic circulation must <strong>in</strong>duce the decl<strong>in</strong>e <strong>in</strong> FSH.Cohort follicles, therefore, <strong>in</strong>directly cause their ownatresia except for the DF which must acquire relativeFSH-<strong>in</strong>dependence <strong>in</strong> order to grow and differentiatedur<strong>in</strong>g low (cows, mares) or further decl<strong>in</strong><strong>in</strong>g (women)FSH concentrations. Dom<strong>in</strong>ant follicle secretions thencont<strong>in</strong>ue to suppress FSH to prevent further waveemergence. Cohort and DFs <strong>in</strong> cows, mares and womenproduce the FSH-<strong>in</strong>hibitors oestradiol and <strong>in</strong>hib<strong>in</strong>-A(cows: Sunderland et al. 1996; Mihm et al. 1997; Bleachet al. 2001; mares: Bergfelt et al. 2001; Watson et al.2002; women: Schneyer et al. 2000; Laven and Fauser2004), and <strong>in</strong> cows and women cohort follicles produce<strong>in</strong>hib<strong>in</strong>-B (Beg et al. 2002; Laven and Fauser 2004). Inheifers and cows dur<strong>in</strong>g growth of the first cohort of theoestrous cycle, <strong>in</strong>hib<strong>in</strong>-A <strong>in</strong>creases <strong>in</strong> circulation co<strong>in</strong>cidentwith small rises <strong>in</strong> systemic oestradiol reach<strong>in</strong>g anÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Dom<strong>in</strong>ant Follicle Selection <strong>in</strong> Cows, Mares and Women 51early plateau before DF selection (<strong>in</strong>hib<strong>in</strong>-A) or a peakon the day of DF selection (oestradiol) while FSHconcentrations decl<strong>in</strong>e (Bleach et al. 2001; Kaneko et al.2002; Parker et al. 2003). Follicular <strong>in</strong>hib<strong>in</strong>-A secretionappears to be the most important regulator of FSHconcentrations dur<strong>in</strong>g <strong>in</strong>itial follicle wave growth <strong>in</strong>cattle and is aided by oestradiol secreted from the DF atthe onset of dom<strong>in</strong>ance (G<strong>in</strong>ther et al. 2000, 2001a;Mihm and Bleach 2003): when atresia of grow<strong>in</strong>g cohortfollicles is caused us<strong>in</strong>g exogenous steroid treatments<strong>in</strong>hib<strong>in</strong>-A decl<strong>in</strong>es rapidly followed by a rise <strong>in</strong> FSHconcentrations despite high systemic oestradiol (Mihmet al. 2001). Similarly, FSH only rises aga<strong>in</strong> dur<strong>in</strong>g lossof dom<strong>in</strong>ance of the first DF of the cycle when <strong>in</strong>hib<strong>in</strong>-Aconcentrations reach their nadir, despite oestradiolhav<strong>in</strong>g decl<strong>in</strong>ed two days previously (Bleach et al.2001). However, other follicular secretions may also be<strong>in</strong>volved <strong>in</strong> the regulation of FSH dur<strong>in</strong>g cohort growthsuch as follistat<strong>in</strong> (Mihm and Bleach 2003).In mares oestradiol concentrations rise <strong>in</strong> circulationapproximately 1 day before selection of the DF andluteolysis (Bergfelt et al. 2001). As FSH concentrationsdecl<strong>in</strong>e immediately after wave emergence which isseveral days from deviation, other follicular secretionsthan oestradiol must be responsible for the <strong>in</strong>itial FSHsuppression. Inhib<strong>in</strong>-A is synthesized by cohort andDFs <strong>in</strong> the mares (Watson et al. 2002) and rises <strong>in</strong> total<strong>in</strong>hib<strong>in</strong> have been observed associated with DF growth(Bergfelt et al. 2001). In women, similar to cows,oestradiol concentrations show a small but significant<strong>in</strong>crease after wave emergence. Very importantly, however,<strong>in</strong>hib<strong>in</strong>-B concentrations rise dur<strong>in</strong>g cohort growthand appear to peak just before DF selection, decl<strong>in</strong><strong>in</strong>gdur<strong>in</strong>g the long dom<strong>in</strong>ance period (Muttukrishna et al.2000). Thus, <strong>in</strong>hib<strong>in</strong>-B secretion from cohort folliclescontrols the <strong>in</strong>itial FSH decl<strong>in</strong>e responsible for DFselection <strong>in</strong> women (Schneyer et al. 2000). Follow<strong>in</strong>gselection of the ovulatory DF <strong>in</strong> the follicular phase,oestradiol and <strong>in</strong>hib<strong>in</strong>-A concentrations rise rapidly,and together control the rema<strong>in</strong><strong>in</strong>g protracted FSHdecl<strong>in</strong>e (Muttukrishna et al. 1994, 2000). In contrast tocattle, where oestradiol only exerts a transient <strong>in</strong>hibitionon FSH (O’Rourke et al. 2000), oestradiol is clearlyrequired for cont<strong>in</strong>ued FSH suppression <strong>in</strong> women, asanti-oestradiol treatment (passive immunization orreceptor antagonist adm<strong>in</strong>istration) dur<strong>in</strong>g the midfollicularand the end-follicular phase will cause a rise <strong>in</strong>FSH and abolish DF selection or cause DF atresia(Zeleznik 2001).Intraovarian (Cellular) Mechanisms for DFSelection: Gonadotroph<strong>in</strong> Responsiveness andInsul<strong>in</strong>-like Growth Factor B<strong>in</strong>d<strong>in</strong>g Prote<strong>in</strong>(IGFBP) ExpressionAntral follicles of 2–5 mm (cows, women) or 10–13 mm(mares) are clearly dependent on elevated FSH forcont<strong>in</strong>ued development, firstly because they undergoatresia <strong>in</strong> the absence of FSH rises, and secondly, cohortfollicles undergo atresia after their emergence when FSHdecl<strong>in</strong>es aga<strong>in</strong> follow<strong>in</strong>g its transient rise. At deviationbov<strong>in</strong>e DFs can ma<strong>in</strong>ta<strong>in</strong> follicular cell proliferation andenhance oestradiol production despite decl<strong>in</strong><strong>in</strong>g FSHconcentrations, and the DF may be the one cohortfollicle with the lowest FSH requirement due to<strong>in</strong>creased or ma<strong>in</strong>ta<strong>in</strong>ed high FSH receptor mRNAexpression and FSH-b<strong>in</strong>d<strong>in</strong>g allow<strong>in</strong>g it to pass the8.5 mm diameter threshold (Ireland and Roche 1983;Evans and Fortune 1997; Bao and Garverick 1998;Rozell et al. 2005). Interest<strong>in</strong>gly, the follicle with highest<strong>in</strong>trafollicular oestradiol 33 h after the FSH peak (themost successful cohort follicle <strong>in</strong> the wave) also hashighest activ<strong>in</strong>-A concentrations <strong>in</strong> follicular fluid(Aust<strong>in</strong> et al. 2001), which is known to <strong>in</strong>crease FSHreceptor expression <strong>in</strong> granulosa cells (Knight andGlister 2006). This <strong>in</strong>formation is extended to women,where it is postulated that the follicle which becomes theDF must first develop ‘activ<strong>in</strong>-dom<strong>in</strong>ance’ before the‘<strong>in</strong>hib<strong>in</strong>-dom<strong>in</strong>ance’ characteristic for the DF (Schneyeret al. 2000).In all three monovulatory species a transition fromFSH- to LH-dependence is postulated as the mechanismwhich allows cont<strong>in</strong>ued development of theselected DF (G<strong>in</strong>ther et al. 2001a), and <strong>in</strong> the animalspecies this is based on functional studies abolish<strong>in</strong>g oradd<strong>in</strong>g LH pulses dur<strong>in</strong>g follicle wave growth and afterDF selection (Gastal et al. 1999; Mihm and Bleach2003). Granulosa cells from newly selected bov<strong>in</strong>e DFhave acquired an enhanced ability to b<strong>in</strong>d LHcompared with SF1s and show <strong>in</strong>creased mRNAexpression for the LH receptor dur<strong>in</strong>g their earlygrowth phase (Ireland and Roche 1983; Evans andFortune 1997; Bao and Garverick 1998; Evans et al.2004; Mihm et al. 2006). Similarly, <strong>in</strong> mares theselected DF is characterized by its absolute dependenceon <strong>in</strong>creased LH for enhanced oestradiol synthesis andcont<strong>in</strong>ued growth (Bergfelt et al. 2001). This alsoapplies <strong>in</strong> women, as it is suggested that LH-<strong>in</strong>ducedcAMP production <strong>in</strong> granulosa cells from DF replacesthe FSH-stimulated cAMP production seen <strong>in</strong> grow<strong>in</strong>gcohort follicles (Zeleznik 2001). However, at this po<strong>in</strong>tit is not clear <strong>in</strong> any species whether the identified<strong>in</strong>crease <strong>in</strong> LH receptor expression <strong>in</strong> the DF is cause(will the first follicle that acquires LH receptor activitybecome selected?) or consequence of the selectionprocess. Recently a slight <strong>in</strong>crease <strong>in</strong> LH receptormRNA expression was detected <strong>in</strong> granulosa cells fromthe largest bov<strong>in</strong>e follicle a few hours before the onsetof deviation and the appearance of morphological andhormonal (follicular fluid oestradiol) differences, comparedwith the second largest follicle (Beg et al. 2001).This may <strong>in</strong>dicate that acquisition of <strong>in</strong>creased LHreceptor expression characterizes the follicle with thedevelopmental advantage even before other differencesto the cohort become apparent. As IGF1 has beenshown to enhance FSH-<strong>in</strong>duced granulosa cell differentiation,particularly LH receptor acquisition (Hirakawaet al. 1999), the <strong>in</strong>creased free IGF concentrationspostulated for the future DF dur<strong>in</strong>g cohort growth (seebelow) may be responsible for such a developmentaladvantage.Because FSH is anti-apoptotic, proliferative anddifferentiative (it stimulates aromatase production andLH receptor acquisition), enhanced FSH-responsivenessor the ability to amplify FSH-stimulated granulosacell functions <strong>in</strong> one cohort follicle may lead toÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


52 M Mihm and ACO Evansits selection. FSH and IGF enhance each other’sability to stimulate proliferation, steroid, activ<strong>in</strong> and<strong>in</strong>hib<strong>in</strong> production <strong>in</strong> bov<strong>in</strong>e granulosa cells (Glisteret al. 2001). Access of IGF1 and 2 to the type 1receptor which is the ma<strong>in</strong> receptor convey<strong>in</strong>g theirproliferative and steroidogenic activities (Spicer andAad 2007) is regulated by a group of differentmolecular weight IGF b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s with differ<strong>in</strong>gligand aff<strong>in</strong>ity and differential expression patterns <strong>in</strong>healthy and atretic follicles (San Roman and Magoff<strong>in</strong>1993; Monget et al. 2002; Roberts and Echternkamp2003). In all three monovulatory species characteristicdifferences <strong>in</strong> the expression pattern of IGF b<strong>in</strong>d<strong>in</strong>gprote<strong>in</strong>s have been observed between follicles grow<strong>in</strong>gwith<strong>in</strong> a wave, or between dom<strong>in</strong>ant and subord<strong>in</strong>ateor cohort follicles (Schuller et al. 1993; Stewart et al.1996; Mihm et al. 1997). This has been attributed tothe ability of FSH-stimulated cohort follicles or theselected DF to downregulate expression of specificIGF b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s (e.g. IGF b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> 2) or<strong>in</strong>crease their proteolytic process<strong>in</strong>g (for example, IGFb<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s 4 and 5) by express<strong>in</strong>g a proteasesuch as PAPP-A, thus ‘free<strong>in</strong>g’ IGF for stimulation ofcellular proliferation and steroidogenesis dur<strong>in</strong>gdecl<strong>in</strong><strong>in</strong>g or low FSH (Conover et al. 2001; Mongetet al. 2002). In vivo experiments <strong>in</strong> heifers have showna reduction <strong>in</strong> the lower molecular weight IGFb<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s 2, 4 and 5 <strong>in</strong> the largest cohortfollicles and the selected dom<strong>in</strong>ant compared withSF1s (Mihm et al. 1997; Aust<strong>in</strong> et al. 2001); a specificreduction <strong>in</strong> IGF-BP4 (the non-glycosylated 24 kDaform) <strong>in</strong> the cohort follicle most likely to become theDF (Mihm et al. 2000); and <strong>in</strong>creased proteolyticactivity ⁄ PAPP-A <strong>in</strong> the DF at deviation which isspecific for the two lowest molecular weight IGFb<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s 4 and 5 (Rivera et al. 2001; Riveraand Fortune 2003). Insul<strong>in</strong>-like growth factor b<strong>in</strong>d<strong>in</strong>gprote<strong>in</strong> 2 is <strong>in</strong>creased <strong>in</strong> SF1 <strong>in</strong> mares at deviation(Donadeu and G<strong>in</strong>ther 2002b). In women amounts ofIGF b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> 3 (the highest molecular weightform), the 28 kDa and the 24 kDa (most likely IGFb<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> 4) <strong>in</strong> follicular fluid are reduced <strong>in</strong>dom<strong>in</strong>ant compared with normal cohort or atreticfollicles, while IGF b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> 2 <strong>in</strong>creases <strong>in</strong>atretic follicles; <strong>in</strong>creased PAPP-A is also considered amarker for DF selection (San Roman and Magoff<strong>in</strong>1993; Schuller et al. 1993; Amato et al. 1998; Conoveret al. 2001). Therefore, the ability to differentiallyreduce the expression of specific IGF b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>sor <strong>in</strong>duce their proteolysis dur<strong>in</strong>g decl<strong>in</strong><strong>in</strong>g FSHappears to be associated with DF selection <strong>in</strong> allthree monovulatory species. Recent <strong>in</strong> vivo studies <strong>in</strong>heifers and mares whereby IGF-1 and IGF b<strong>in</strong>d<strong>in</strong>gprote<strong>in</strong> 3 were adm<strong>in</strong>istered <strong>in</strong>to the SF1 and DF,respectively, confirm the differentiative and steroidogenicroles of IGF dur<strong>in</strong>g the selection process(G<strong>in</strong>ther et al. 2004b; c). In addition, actions ofFSH and IGF are mediated (at least <strong>in</strong> part) viaAkt (prote<strong>in</strong> k<strong>in</strong>ase B) and Erk (extracellular regulatedk<strong>in</strong>ase) <strong>in</strong> bov<strong>in</strong>e granulosa and theca cells, anda greater activity of these pathways <strong>in</strong> newly selectedDFs compared to contemporary SF1s has beendemonstrated (Ryan et al. 2007). Thus a difference<strong>in</strong> these pathways may mark the future DF from SF1sbefore differences <strong>in</strong> follicle diameters or follicularfluid oestradiol and IGFBP concentrations appear.Because antral follicle growth and differentiationclearly depends on nutrient, hormone and growth factorsupply to the <strong>in</strong>dividual follicle, one other sharedmechanism recently proposed for DF selection <strong>in</strong> allthree monovulatory species is the possible differentialregulation of blood vessel formation and permeability <strong>in</strong>the theca layers of cohort follicles. This hypothesis issupported by several studies, which have shown<strong>in</strong>creased expression of angiogenic factors such asvascular endothelial growth factor (VEGF) with <strong>in</strong>creasedfollicle differentiation, <strong>in</strong>creased blood flow <strong>in</strong>the selected DF, and follicular arrest and atresiafollow<strong>in</strong>g the experimental reduction of VEGF signall<strong>in</strong>g(Acosta et al. 2004, 2005; Hunter et al. 2004; Fraserand Duncan 2005). Further functional studies are nowneeded to elucidate whether angiogenic factors arecausatively <strong>in</strong>volved <strong>in</strong> DF selection.Candidate Genes for Regulation of Dom<strong>in</strong>antFollicle Selection Identified Us<strong>in</strong>g the Bov<strong>in</strong>eModelThe bov<strong>in</strong>e follicle wave model has so far been used forfour studies us<strong>in</strong>g genomic approaches to identify genesexpressed by granulosa and theca cells which are regulatedat the time of DF selection, thus provid<strong>in</strong>g candidatesfor further functional studies of follicle survival anddifferentiation <strong>in</strong> monovulatory species (suppressionsubtractive hybridization, Sisco et al. 2003; Fayad et al.2004; custom cDNA microarray, Evans et al. 2004; serialanalysis of gene expression, SAGE, Mihm et al. 2008).Additional molecular studies have <strong>in</strong>vestigated transcriptionof candidate genes identified through genomicapproaches dur<strong>in</strong>g growth of the follicle wave (Siscoet al. 2003; Zielak et al. 2007; Forde et al. 2008). Forexample, comparisons of granulosa cell mRNA expressionprofiles between the cohort follicle most likely fatedto become the DF, the newly selected DF and the SF1have led to the identification of a number of knowndifferentiation genes, but also of genes not previouslyassociated with bov<strong>in</strong>e DF development or ovarianfunction, which are upregulated at dom<strong>in</strong>ance and are<strong>in</strong>volved <strong>in</strong> oestradiol synthesis, anti-oxidant activity,LH-responsiveness, cell proliferation, anti-apoptoticactivity, and RNA splic<strong>in</strong>g (Table 1; Mihm et al. 2008).Similar studies have not been carried out <strong>in</strong> mares andwomen. Because <strong>in</strong> these two species <strong>in</strong>duction of higharomatase activity and <strong>in</strong>creased LH responsiveness arealso functions of the selected DF (G<strong>in</strong>ther et al. 2001a;Zeleznik 2001), other cellular functions or pathwaysassociated with enhanced mRNA expression for thearomatase and LH receptor genes and identified us<strong>in</strong>gthe bov<strong>in</strong>e follicle wave model may be relevant for DFselection <strong>in</strong> all three monovulatory species, and thusneed to be tested further us<strong>in</strong>g functional <strong>in</strong> vivo and <strong>in</strong>vitro approaches. For this the bov<strong>in</strong>e DF selectionmodel is particularly suited, as difficulties <strong>in</strong> obta<strong>in</strong><strong>in</strong>gtissues or suitable technological platforms will prohibitthe study of follicle development <strong>in</strong> mares and women ascompletely as <strong>in</strong> cattle.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Dom<strong>in</strong>ant Follicle Selection <strong>in</strong> Cows, Mares and Women 53Table 1. Summary table of granulosa cell genes expressed at highest levels <strong>in</strong> the largest cohort follicle follow<strong>in</strong>g emergence, or <strong>in</strong> the newlyselected dom<strong>in</strong>ant follicle compared to the most oestrogenic cohort follicle before selection and(or) the largest subord<strong>in</strong>ate follicle (Evans et al.2004; Forde et al. 2008; Mihm et al. 2008; Zielak et al. 2007). Such identified genes are proposed as gene candidates for the regulation of DFselection <strong>in</strong> cowsBov<strong>in</strong>e candidate genesfor DF selection Intracellular role Molecular approach ReferenceCell differentiationCYP19A1 a Estradiol synthesis, cell differentiation Custom microarray andqRT-PCR, SAGE andqRT-PCRDQ004742Uncharacterized, splice variant foraromataseGADD45BCell differentiation, response to cellularstress, apoptosisEvans et al. (2004),Mihm et al. (2008)SAGE and qRT-PCR Mihm et al. (2008)SAGE and qRT-PCR Mihm et al. (2008)GSTA2 Steroid synthesis, antioxidant SAGE and qRT-PCR Mihm et al. (2008)INHA Cell differentiation, <strong>in</strong>hib<strong>in</strong> synthesis SAGE and qRT-PCR Mihm et al. (2008)LHCGRGonadotroph<strong>in</strong> receptor, cell differentiationCustom microarray andEvans et al. (2004)qRT-PCRTBC1D1 Cell differentiation, proliferation Custom microarray andZielak et al. (2007)qRT-PCROvary-specific acidic prote<strong>in</strong>Uncharacterized but l<strong>in</strong>ked to highSAGE and qRT-PCR Mihm et al. (2008)oestradiol synthesis <strong>in</strong> pre-ovulatoryfolliclesCell proliferation, apoptosisCCND2 Cell proliferation SAGE and qRT-PCR Mihm et al. (2008)MCL-1 Cell proliferation, antiapoptotic factor Custom microarray andEvans et al. (2004)qRT-PCRMIF Cell proliferation, antiapoptotic factor SAGE and qRT-PCR Mihm et al. (2008)Signal transductionANXA2 Tissue development, signal transduction SAGE and qRT-PCR Mihm et al. (2008)BCAR1 Signal transduction, cell proliferation Custom microarray andForde et al. (2008)qRT-PCRCALM2 Signal transduction SAGE and qRT-PCR Mihm et al. (2008)CLIC1 Signal transduction, chloride transport SAGE and qRT-PCR Mihm et al. (2008)Prote<strong>in</strong>, DNA and RNA synthesisDICE-1Nuclear processes (DNA repair, transcription,Custom microarray andEvans et al. (2004)RNA splic<strong>in</strong>g)qRT-PCRRFC4 Nuclear processes (DNA repair) SAGE and qRT-PCR Mihm et al. (2008)SFRS9 Nuclear processes (RNA splic<strong>in</strong>g) SAGE and qRT-PCR Mihm et al. (2008)SLC22A17 Prote<strong>in</strong> synthesis, ion transport SAGE and qRT-PCR Mihm et al. (2008)a Gene symbols are presented.ConclusionsThe similarities <strong>in</strong> follicle wave dynamics, the transientFSH rise and its regulation, follicular acquisition ofFSH and LH-responsiveness, and <strong>in</strong> the differentialpattern of IGF b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> expression <strong>in</strong> cohort andDFs are strik<strong>in</strong>g between the three monovulatoryspecies. Differences exist ma<strong>in</strong>ly between cows andwomen <strong>in</strong> the follicle wave pattern, between womenand the two animal species <strong>in</strong> the extent of the FSHdecl<strong>in</strong>e, and <strong>in</strong> the <strong>in</strong>hib<strong>in</strong> forms secreted by cohortfollicles, and possibly also <strong>in</strong> the specific form of IGFb<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> reduced <strong>in</strong> DF. Because of the considerableadvantage of large, relatively easily accessiblefollicles, mares present themselves as well-suited forfunctional experiments <strong>in</strong>volv<strong>in</strong>g <strong>in</strong> vivo sampl<strong>in</strong>g oradm<strong>in</strong>istration of candidate substances. However, thebov<strong>in</strong>e model is most easily monitored, extremely wellcharacterized,and has the advantage that its genomehas been sequenced allow<strong>in</strong>g comparative molecularstudies with human or rodent models of follicle differentiationnot possible so far <strong>in</strong> other animal species. Inaddition, the bov<strong>in</strong>e model has recently been used todeterm<strong>in</strong>e the relationship between low and high cohortfollicle numbers, the primordial follicle pool, andresponse to superovulatory treatment (Ireland et al.2007a,b). Thus, us<strong>in</strong>g selected heifers or cows as a modelof low antral follicle numbers may represent an excit<strong>in</strong>gopportunity to study effects of reduced primordial poolsize on future cohort follicle or DF function simulat<strong>in</strong>gevents <strong>in</strong> pre-menopausal women.ReferencesAcosta TJ, Gastal EL, Gastal MO, G<strong>in</strong>ther OJ, 2004:Differential blood flow changes between the future dom<strong>in</strong>antand subord<strong>in</strong>ate follicles precede diameter changesdur<strong>in</strong>g follicle selection <strong>in</strong> mares. Biol Reprod 71, 502–507.Acosta TJ, Hayashi K-G, Matsui M, Miyamoto A, 2005:Changes <strong>in</strong> follicular vascularity dur<strong>in</strong>g the first follicularwave <strong>in</strong> lactat<strong>in</strong>g cows. J Reprod Dev 51, 273–280.Adams GP, Matteri RL, Kastelic JP, Ko JCH, G<strong>in</strong>ther OJ,1992: Association between surges of follicle-stimulat<strong>in</strong>ghormone and the emergence of follicular waves <strong>in</strong> heifers.J Reprod Fertil 94, 177–188.Adams GP, Kot K, Smith CA, G<strong>in</strong>ther OJ, 1993: Selection ofa dom<strong>in</strong>ant follicle and suppression of follicular growth <strong>in</strong>heifers. Anim Reprod Sci 30, 259–271.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


54 M Mihm and ACO EvansAmato G, Izzo A, Tucker A, Bellastella A, 1998: Insul<strong>in</strong>-likegrowth factor b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>-3 reduction <strong>in</strong> follicular fluid <strong>in</strong>spontaneous and stimulated cycles. Fertil Steril 70, 141–144.Aust<strong>in</strong> EJ, Mihm M, Evans ACO, Knight PG, Ireland JLH,Ireland JJ, Roche JF, 2001: Alterations <strong>in</strong> <strong>in</strong>trafollicularregulatory factors and apoptosis dur<strong>in</strong>g selection of follicles<strong>in</strong> the first follicular wave of the bov<strong>in</strong>e estrous cycle. BiolReprod 64, 839–848.Baerwald AR, Adams GP, Pierson RA, 2003: Characterizationof ovarian follicular wave dynamics <strong>in</strong> women. Biol Reprod69, 1023–1031.Baird DT, 1987: A model for follicular selection and ovulation– lessons from superovulation. J Steroid Biochem Mol Biol27, 15–23.Bao B, Garverick HA, 1998: Expression of steroidogenicenzyme and gonadotrop<strong>in</strong> receptor genes <strong>in</strong> bov<strong>in</strong>e folliclesdur<strong>in</strong>g ovarian follicular waves: a review. J Anim Sci 76,1903–1921.Beg MA, Bergfelt DR, Kot K, Wiltbank MC, G<strong>in</strong>ther OJ,2001: Follicular-fluid factors and granulosa-cell gene expressionassociated with follicle deviation <strong>in</strong> cattle. Biol Reprod64, 432–441.Beg MA, Bergfelt DR, Kot K, G<strong>in</strong>ther OJ, 2002: Follicleselection <strong>in</strong> cattle: dynamics of follicular fluid factors dur<strong>in</strong>gdevelopment of follicle dom<strong>in</strong>ance. Biol Reprod 66, 120–126.Bergfelt DR, G<strong>in</strong>ther OJ, 1993: Relationships between FSHsurges and follicular waves dur<strong>in</strong>g the estrous-cycle <strong>in</strong>mares. Theriogenology 39, 781–796.Bergfelt DR, Gastal EL, G<strong>in</strong>ther OJ, 2001: Response ofestradiol and <strong>in</strong>hib<strong>in</strong> to experimentally reduced lute<strong>in</strong>iz<strong>in</strong>ghormone dur<strong>in</strong>g follicle deviation <strong>in</strong> mares. Biol Reprod 65,426–432.Bleach ECL, Glencross RG, Feist SA, Groome NP, KnightPG, 2001: Plasma <strong>in</strong>hib<strong>in</strong> A <strong>in</strong> heifers: relationship withfollicle dynamics, gonadotrop<strong>in</strong>s, and steroids dur<strong>in</strong>g theestrous cycle and after treatment with bov<strong>in</strong>e follicular fluid.Biol Reprod 64, 743–752.Burns DS, Jimenez-Krassel F, Ireland JLH, Knight PG,Ireland JJ, 2005: Numbers of antral follicles dur<strong>in</strong>g follicularwaves <strong>in</strong> cattle: evidence for high variation among animals,very high repeatability <strong>in</strong> <strong>in</strong>dividuals, and an <strong>in</strong>verseassociation with serum follicle-stimulat<strong>in</strong>g hormone concentrations.Biol Reprod 73, 54–62.Conover CA, Faessen GF, Ilg KE, Chandrasekher YA,Christiansen M, Overgaard MT, Oxvig C, Giudice LC,2001: Pregnancy-associated plasma prote<strong>in</strong>-A is the <strong>in</strong>sul<strong>in</strong>likegrowth factor b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>-4 protease secreted byhuman ovarian granulosa cells and is a marker of dom<strong>in</strong>antfollicle selection and the corpus luteum. Endocr<strong>in</strong>ology 142,2155–2158.Crowe MA, Padmanabhan V, Mihm M, Beit<strong>in</strong>s IZ, Roche JF,1998: Resumption of follicular waves <strong>in</strong> beef cows is notassociated with periparturient changes <strong>in</strong> follicle-stimulat<strong>in</strong>ghormone heterogeneity despite major changes <strong>in</strong> steroid andgonadotrop<strong>in</strong> concentrations. Biol Reprod 58, 1445–1450.Donadeu FX, G<strong>in</strong>ther OJ, 2001: Effect of number anddiameter of follicles on plasma concentrations of <strong>in</strong>hib<strong>in</strong>and FSH <strong>in</strong> mares. <strong>Reproduction</strong> 121, 897–903.Donadeu FX, G<strong>in</strong>ther OJ, 2002a: Follicular waves andcirculat<strong>in</strong>g concentrations of gonadotroph<strong>in</strong>s, <strong>in</strong>hib<strong>in</strong> andoestradiol dur<strong>in</strong>g the anovulatory season <strong>in</strong> mares. <strong>Reproduction</strong>124, 875–885.Donadeu FX, G<strong>in</strong>ther OJ, 2002b: Changes <strong>in</strong> concentrationsof follicular-fluid factors dur<strong>in</strong>g follicle selection <strong>in</strong> mares.Biol Reprod 66, 1111–1118.Echternkamp SE, Roberts AJ, Lunstra DD, Wise T, Spicer LJ,2004: Ovarian follicular development <strong>in</strong> cattle selected fortw<strong>in</strong> ovulations and births. J Anim Sci 82, 459–471.Echternkamp SE, 2000: Endocr<strong>in</strong>ology of <strong>in</strong>creased ovarianfolliculogenesis <strong>in</strong> cattle selected for tw<strong>in</strong> births. Proc AmSoc Anim Sci. Available at http://www.asas.org/jas/symposia/proceed<strong>in</strong>gs/0935.pdf.Evans ACO, 2003: Characteristics of ovarian follicle development<strong>in</strong> domestic animals. Reprod Domest Anim 38, 240–246.Evans ACO, Fortune JE, 1997: Selection of the dom<strong>in</strong>antfollicle <strong>in</strong> cattle occurs <strong>in</strong> the absence of differences <strong>in</strong> theexpression of messenger ribonucleic acid for gonadotrop<strong>in</strong>receptors. Endocr<strong>in</strong>ology 138, 2963–2971.Evans ACO, Adams GP, Rawl<strong>in</strong>gs NC, 1994: Endocr<strong>in</strong>e andovarian follicular changes lead<strong>in</strong>g up to the first ovulation <strong>in</strong>prepubertal heifers. J Reprod Fertil 100, 187–194.Evans ACO, Ireland JLH, W<strong>in</strong>n ME, Lonergan P, Smith GW,Coussens PM, Ireland JJ, 2004: Identification of genes<strong>in</strong>volved <strong>in</strong> apoptosis and dom<strong>in</strong>ant follicle developmentdur<strong>in</strong>g follicular waves <strong>in</strong> cattle. Biol Reprod 70,1475–1484.Fayad T, Levesque V, Sirois J, Silversides DW, Lussier JG,2004: Gene expression profil<strong>in</strong>g of differentially expressedgenes <strong>in</strong> granulosa cells of bov<strong>in</strong>e dom<strong>in</strong>ant follicles us<strong>in</strong>gsuppression subtractive hybridization. Biol Reprod 70, 523–533.Forde N, Mihm M, Canty MJ, Zielak AE, Baker PJ, Park SD,Lonergan P, Smith GW, Coussens PM, Ireland JJ, EvansAC, 2008: Differential expression of signal transductionfactors <strong>in</strong> ovarian follicle development; a role for betaglycanFIBP <strong>in</strong> granulosa cells <strong>in</strong> cattle. Physiol Genomics 33, 193–204.Franks S, McCarthy MI, Hardy K, 2006: Development ofpolycystic ovary syndrome: <strong>in</strong>volvement of genetic andenvironmental factors. Int J Androl 29, 278–285.Fraser HM, Duncan WC, 2005: Vascular morphogenesis <strong>in</strong> theprimate ovary. Angiogenesis 8, 101–116.Gastal EL, Bergfelt DR, Nogueira GP, Gastal MO, G<strong>in</strong>therOJ, 1999: Role of lute<strong>in</strong>iz<strong>in</strong>g hormone <strong>in</strong> follicle deviationbased on manipulat<strong>in</strong>g progesterone concentrations <strong>in</strong>mares. Biol Reprod 61, 1492–1498.Gibbons JR, Wiltbank MC, G<strong>in</strong>ther OJ, 1997: Functional<strong>in</strong>terrelationships between follicles greater than 4 mm andthe follicle-stimulat<strong>in</strong>g hormone surge <strong>in</strong> heifers. BiolReprod 57, 1066–1073.G<strong>in</strong>ther OJ, 1992: Reproductive Biology of the Mare, Basicand Applied Aspects, 2nd edn. Equiservices Publish<strong>in</strong>g,Cross Pla<strong>in</strong>s, WI, USA.G<strong>in</strong>ther OJ, 1993: Major and m<strong>in</strong>or follicular waves dur<strong>in</strong>g theequ<strong>in</strong>e estrous-cycle. J Equ Vet Sci 13, 18–25.G<strong>in</strong>ther OJ, Bergfelt DR, 1992: Associations between FSHconcentrations and major and m<strong>in</strong>or follicular waves <strong>in</strong>pregnant mares. Theriogenology 38, 807–821.G<strong>in</strong>ther OJ, Knopf L, Kastelic JP, 1989: Temporal associationsamong ovarian events <strong>in</strong> cattle dur<strong>in</strong>g oestrous cycleswith two or three follicular waves. J Reprod Fertil 87, 223–230.G<strong>in</strong>ther OJ, Kot K, Kulick LJ, Mart<strong>in</strong> S, Wiltbank MC, 1996:Relationships between FSH and ovarian follicular wavesdur<strong>in</strong>g the last six months of pregnancy <strong>in</strong> cattle. J ReprodFert 108, 271–279.G<strong>in</strong>ther OJ, Kot K, Kulick LJ, Wiltbank MC, 1997: Emergenceand deviation of follicles dur<strong>in</strong>g the development offollicular waves <strong>in</strong> cattle. Theriogenology 48, 75–87.G<strong>in</strong>ther OJ, Bergfelt DR, Kulick LJ, Kot K, 1999: Selection ofthe dom<strong>in</strong>ant follicle <strong>in</strong> cattle: establishment of follicledeviation <strong>in</strong> less than 8 hours through depression of FSHconcentrations. Theriogenology 52, 1079–1093.G<strong>in</strong>ther OJ, Bergfelt DR, Kulick LJ, Kot K, 2000: Selection ofthe dom<strong>in</strong>ant follicle <strong>in</strong> cattle: role of estradiol. Biol Reprod63, 383–389.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Dom<strong>in</strong>ant Follicle Selection <strong>in</strong> Cows, Mares and Women 55G<strong>in</strong>ther OJ, Beg MA, Bergfelt DR, Donadeu FX, Kot K,2001a: Follicle selection <strong>in</strong> monovular species. Biol Reprod65, 638–647.G<strong>in</strong>ther OJ, Bergfelt DR, Beg MA, Kot K, 2001b: Follicleselection <strong>in</strong> cattle: relationships among growth rate, diameterrank<strong>in</strong>g, and capacity for dom<strong>in</strong>ance. Biol Reprod 65,345–350.G<strong>in</strong>ther OJ, Gastal EL, Gastal MO, Bergfelt DR, BaerwaldAR, Pierson RA, 2004a: Comparative study of the dynamicsof follicular waves <strong>in</strong> mares and women. Biol Reprod 71,1195–1201.G<strong>in</strong>ther OJ, Bergfelt DR, Beg MA, Meira C, Kot K, 2004b: Invivo effects of an <strong>in</strong>trafollicular <strong>in</strong>jection of <strong>in</strong>sul<strong>in</strong>-likegrowth factor 1 on the mechanism of follicle deviation <strong>in</strong>heifers and mares. Biol Reprod 70, 99–105.G<strong>in</strong>ther OJ, Gastal EL, Gastal MO, Beg MA, 2004c: Criticalrole of <strong>in</strong>sul<strong>in</strong>-like growth factor system <strong>in</strong> follicle selectionand dom<strong>in</strong>ance <strong>in</strong> mares. Biol Reprod 70, 1374–1379.G<strong>in</strong>ther OJ, Beg MA, Gastal EL, Gastal MO, Baerwald AR,Pierson RA, 2005: Systemic concentrations of hormonesdur<strong>in</strong>g the development of follicular waves <strong>in</strong> mares andwomen: a comparative study. <strong>Reproduction</strong> 130, 379–388.Glister C, Tannetta DS, Groome NP, Knight PG, 2001:Interactions between follicle-stimulat<strong>in</strong>g hormone andgrowth factors <strong>in</strong> modulat<strong>in</strong>g secretion of steroids and<strong>in</strong>hib<strong>in</strong>-related peptides by nonlute<strong>in</strong>ized bov<strong>in</strong>e granulosacells. Biol Reprod 65, 1020–1028.Gong JG, Bramley T, Webb R, 1991: The effect ofrecomb<strong>in</strong>ant bov<strong>in</strong>e somatotrop<strong>in</strong> on ovarian function <strong>in</strong>heifers – follicular populations and peripheral hormones.Biol Reprod 45, 941–949.Gong JG, Campbell BK, Bramley TA, Gutierrez CG, PetersAR, Webb R, 1996: Suppression <strong>in</strong> the secretion of folliclestimulat<strong>in</strong>ghormone and lute<strong>in</strong>iz<strong>in</strong>g hormone, and ovarianfollicle development <strong>in</strong> heifers cont<strong>in</strong>uously <strong>in</strong>fused with agonadotrop<strong>in</strong>-releas<strong>in</strong>g hormone agonist. Biol Reprod 55,68–74.Gore MA, Nayudu PL, Vlaisavljevic V, 1997: Atta<strong>in</strong><strong>in</strong>gdom<strong>in</strong>ance <strong>in</strong> vivo: dist<strong>in</strong>guish<strong>in</strong>g dom<strong>in</strong>ant from challengerfollicles <strong>in</strong> humans. Hum Reprod 12, 2741–2747.Hirakawa T, M<strong>in</strong>egishi T, abe K, Kishi H, Ibuki Y, MiyamotoK, 1999: A role of <strong>in</strong>sul<strong>in</strong>-like growth factor I <strong>in</strong> lute<strong>in</strong>iz<strong>in</strong>ghormone receptor expression <strong>in</strong> granulosa cells. Endocr<strong>in</strong>ology140, 4965–4971.Hohmann FP, Laven JSE, de Jong FH, Eijkemans MJC,Fauser BCJM, 2001: Low-dose exogenous FSH <strong>in</strong>itiateddur<strong>in</strong>g the early, mid or late follicular phase can <strong>in</strong>ducemultiple dom<strong>in</strong>ant follicle development. Hum Reprod 16,846–854.Hunter MG, Rob<strong>in</strong>son RS, Mann GE, Webb R, 2004:Endocr<strong>in</strong>e and paracr<strong>in</strong>e control of follicular developmentand ovulation rate <strong>in</strong> farm species. Anim Reprod Sci 82–83,461–477.Ireland JJ, Roche JF, 1983: Development of non-ovulatoryantral follicles <strong>in</strong> heifers: changes <strong>in</strong> steroids <strong>in</strong> follicularfluid and receptors for gonadotrop<strong>in</strong>s. Endocr<strong>in</strong>ology 112,150–156.Ireland JJ, Mihm M, Aust<strong>in</strong> EA, Disk<strong>in</strong> MG, Roche JF, 2000:Dom<strong>in</strong>ant follicle turnover <strong>in</strong> cattle: key concepts, studiesand terms. J Dairy Sci 83, 1648–1658.Ireland JJ, Ward F, Jimenez-Krassel F, Ireland JL, Smith GW,Lonergan P, Evans AC, 2007a: Follicle numbers are highlyrepeatable with<strong>in</strong> animal but <strong>in</strong>versely correlated with FSHconcentrations and the proportion of good quality embryosafter superstimulation <strong>in</strong> cattle. Hum Reprod 22, 1687–1695.Ireland JLH, Jimenez-Krassel F, Smith G, Lonergan P, EvansA, Ireland J, 2007b: Variation <strong>in</strong> number of follicles grow<strong>in</strong>gdur<strong>in</strong>g follicular waves reflects size of the ovarian reserve <strong>in</strong>cattle. Biol Reprod, Special Issue 128–129 (Abstract 218).Kaneko H, Noguchi J, Kikuchi K, Todoroki J, Hasegawa Y,2002: Alterations <strong>in</strong> peripheral concentrations of <strong>in</strong>hib<strong>in</strong> A<strong>in</strong> cattle studied us<strong>in</strong>g a time-resolved immunofluorometricassay: relationship with estradiol and follicle-stimulat<strong>in</strong>ghormone <strong>in</strong> various reproductive conditions. Biol Reprod67, 38–45.Knight PG, Glister C, 2006: TGF-b superfamily members andovarian follicle development. <strong>Reproduction</strong> 132, 191–206.Kulick LJ, Bergfelt DR, Kot K, G<strong>in</strong>ther OJ, 2001: Follicleselection <strong>in</strong> cattle: follicle deviation and codom<strong>in</strong>ance with<strong>in</strong>sequential waves. Biol Reprod 65, 839–846.Laven JSE, Fauser BCJM, 2004: Inhib<strong>in</strong>s and adult ovarianfunction. Mol Cell Endocr<strong>in</strong>ol 225, 37–44.Mihm M, Bleach ECL, 2003: Endocr<strong>in</strong>e regulation of ovarianantral follicle development <strong>in</strong> cattle. Anim Reprod Sci 78,217–237.Mihm M, Good TEM, Ireland JLH, Ireland JJ, Knight PG,Roche JF, 1997: The decl<strong>in</strong>e <strong>in</strong> serum FSH concentrationsalters key <strong>in</strong>trafollicular growth factors <strong>in</strong>volved <strong>in</strong> selectionof the dom<strong>in</strong>ant follicle <strong>in</strong> heifers. Biol Reprod 57, 1328–1337.Mihm M, Curran N, Hyttel P, Knight PG, Boland MP, RocheJF, 1999: Dom<strong>in</strong>ant follicle persistence <strong>in</strong> beef heifers results<strong>in</strong> alterations <strong>in</strong> follicular fluid estradiol and <strong>in</strong>hib<strong>in</strong> anddeviations <strong>in</strong> oocyte maturation. J Reprod Fertil 116, 293–304.Mihm M, Aust<strong>in</strong> EJ, Good TEM, Ireland JLH, Knight PG,Roche JF, Ireland JJ, 2000: Identification of potential<strong>in</strong>trafollicular markers <strong>in</strong>volved <strong>in</strong> selection of the dom<strong>in</strong>antfollicle <strong>in</strong> heifers. Biol Reprod 63, 811–819.Mihm M, Khan A, Bleach E, Knight PG, 2001: Health andatresia <strong>in</strong> first wave follicles is reflected <strong>in</strong> chang<strong>in</strong>g serumdimeric <strong>in</strong>hib<strong>in</strong>-A concentrations <strong>in</strong> dairy heifers. Mol CellEndocr<strong>in</strong>ol 180, 200 (Abstract).Mihm M, Baker PJ, Ireland JLH, Smith GW, Coussens PM,Evans ACO, Ireland JJ, 2006: Molecular evidence thatgrowth of dom<strong>in</strong>ant follicles <strong>in</strong>volves a reduction <strong>in</strong> folliclestimulat<strong>in</strong>g hormone – dependence and an <strong>in</strong>crease <strong>in</strong>lute<strong>in</strong>iz<strong>in</strong>g hormone – dependence <strong>in</strong> cattle. Biol Reprod74, 1051–1059.Mihm M, Baker PJ, Flem<strong>in</strong>g LM, Monteiro AM, O’ShaughnessyPJ, 2008: Differentiation of the bov<strong>in</strong>e dom<strong>in</strong>antfollicle from the cohort upregulates mRNA expression fornew tissue development genes. <strong>Reproduction</strong> 135, 253–265.Monget P, Fabre S, Mulsant P, Lecerf F, Elsen JM, MazerbourgS, Pisselet C, Monniaux D, 2002: Regulation ofovarian folliculogenesis by IGF and BMP system <strong>in</strong>domestic animals. Domest Anim Endocr<strong>in</strong>ol 23, 139–154.Muttukrishna S, Fowler PA, Groome NP, Mitchell GG,Robertson WR, Knight PG, 1994: Serum concentrations ofdimeric <strong>in</strong>hib<strong>in</strong> dur<strong>in</strong>g the spontaneous human menstrualcycleand after treatment with exogenous gonadotrop<strong>in</strong>.Hum Reprod 9, 1634–1642.Muttukrishna S, Child T, Lockwood GM, Groome NP,Barlow DH, Ledger WL, 2000: Serum concentrations ofdimeric <strong>in</strong>hib<strong>in</strong>s, activ<strong>in</strong> A, gonadotroph<strong>in</strong>s and ovariansteroids dur<strong>in</strong>g the menstrual cycle <strong>in</strong> older women. HumReprod 15, 549–556.O’Rourke M, Disk<strong>in</strong> MG, Sreenan JM, Roche JF, 2000: Theeffect of dose and route of oestradiol benzoate adm<strong>in</strong>istrationon plasma concentrations of oestradiol and FSH <strong>in</strong>long term ovariectomised heifers. Anim Reprod Sci 59,1–12.Parker KI, Robertson DM, Groome NP, Macmillan KL,2003: Plasma concentrations of <strong>in</strong>hib<strong>in</strong> A and folliclestimulat<strong>in</strong>ghormone differ between cows with two or threeÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


56 M Mihm and ACO Evanswaves of ovarian follicular development <strong>in</strong> a s<strong>in</strong>gle estrouscycle. Biol Reprod 68, 822–828.Prendiville DJ, Enright WJ, Crowe MA, F<strong>in</strong>nerty M, HynesN, Roche JF, 1995: Immunization of heifers aga<strong>in</strong>stgonadotrop<strong>in</strong> releas<strong>in</strong>g hormone: antibody titres, ovarianfunction, body growth and carcass characteristics. J AnimSci 73, 2382–2389.Rivera GM, Fortune JE, 2003: Selection of the dom<strong>in</strong>antfollicle and <strong>in</strong>sul<strong>in</strong>-like growth factor (IGF)-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s:evidence that pregnancy-associated plasma prote<strong>in</strong> Acontributes to proteolysis of IGF-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> 5 <strong>in</strong>bov<strong>in</strong>e follicular fluid. Endocr<strong>in</strong>ology 144, 437–446.Rivera GM, Chandrasekher YA, Giudice LC, Evans ACO,Fortune JE, 2001: A potential role for <strong>in</strong>sul<strong>in</strong>-like growthfactor b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>-4 proteolysis <strong>in</strong> the establishment ofovarian follicular dom<strong>in</strong>ance <strong>in</strong> cattle. Biol Reprod 65, 102–111.Roberts AJ, Echternkamp SE, 2003: Insul<strong>in</strong>-like growth factorb<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s <strong>in</strong> granulosa and thecal cells from bov<strong>in</strong>eovarian follicles at different stages of development. J AnimSci 81, 2826–2839.Roche JF, Mihm M, Disk<strong>in</strong> M, Ireland JJ, 1998: A reviewof regulation of follicle growth <strong>in</strong> cattle. J Anim Sci76(Suppl. 3), 16–29.Rozell TG, Monteiro AM, Baker PJ, Mihm M, 2005: Evidencefor differential expression of FSH receptor exons 10 and 11dur<strong>in</strong>g follicular wave development <strong>in</strong> the cow. Biol Reprod,Special Issue, 179 (Abstract M445).Ryan KE, Casey SM, Canty MJ, Crowe MA, Mart<strong>in</strong> F, EvansACO, 2007: Akt and Erk signal transduction pathways areearly markers of differentiation <strong>in</strong> dom<strong>in</strong>ant and subord<strong>in</strong>ateovarian follicles <strong>in</strong> cattle. <strong>Reproduction</strong> 133, 617–626.San Roman GA, Magoff<strong>in</strong> DA, 1993: Insul<strong>in</strong>-like growthfactor-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s <strong>in</strong> healthy and atretic follicles dur<strong>in</strong>gnatural menstrual cycles. J Cl<strong>in</strong> Endocr<strong>in</strong>ol Metab 76, 625–632.Savio JD, Keenan L, Boland MP, Roche JF, 1988: Pattern ofgrowth of dom<strong>in</strong>ant follicles dur<strong>in</strong>g the oestrous cycle ofheifers. J Reprod Fertil 83, 663–671.Schipper I, Hop WCJ, Fauser BCJM, 1998: The folliclestimulat<strong>in</strong>ghormone (FSH) threshold ⁄ w<strong>in</strong>dow conceptexam<strong>in</strong>ed by different <strong>in</strong>terventions with exogenous FSHdur<strong>in</strong>g the follicular phase of the normal menstrual cycle:duration, rather than magnitude, of FSH <strong>in</strong>crease affectsfollicle development. J Cl<strong>in</strong> Endocr<strong>in</strong>ol Metab 83, 1292–1298.Schneyer AL, Fujiwara T, Fox J, Welt CK, Adams J,Messerlian GM, Taylor AE, 2000: Dynamic changes <strong>in</strong> the<strong>in</strong>trafollicular <strong>in</strong>hib<strong>in</strong> ⁄ activ<strong>in</strong> ⁄ follistat<strong>in</strong> axis dur<strong>in</strong>g humanfollicular development: relationship to circulat<strong>in</strong>g hormoneconcentrations. J Cl<strong>in</strong> Endocr<strong>in</strong>ol Metab 85, 3319–3330.Schuller AGP, L<strong>in</strong>denberghkortleve DJ, Pache TD, ZwarthoffEC, Fauser BCJM, Drop SLS, 1993: Insul<strong>in</strong>-like growthfactorb<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>-2, 28-kDa and 24-kDa <strong>in</strong>sul<strong>in</strong>-likegrowth-factor b<strong>in</strong>d<strong>in</strong>g-prote<strong>in</strong> levels are decreased <strong>in</strong> fluid ofdom<strong>in</strong>ant follicles, obta<strong>in</strong>ed from normal and polycysticovaries. Regul Pept 48, 157–163.Sisco B, Hagemann LJ, Shell<strong>in</strong>g AN, Pfeffer PL, 2003:Isolation of genes differentially expressed <strong>in</strong> dom<strong>in</strong>ant andsubord<strong>in</strong>ate bov<strong>in</strong>e follicles. Endocr<strong>in</strong>ology 144, 3904–3913.Spicer LJ, Aad PY, 2007: Insul<strong>in</strong>-like growth factor (IGF) 2stimulates steroidogenesis and mitosis of bov<strong>in</strong>e granulosacells through the IGF1 receptor: role of follicle-stimulat<strong>in</strong>ghormone and IGF2 receptor. Biol Reprod 77, 18–27.Stagg K, Spicer LJ, Disk<strong>in</strong> MG, Sreenan JM, Roche JF, 1998:Effect of calf isolation on follicular wave dynamics, gonadotrop<strong>in</strong>and metabolic hormone changes, and <strong>in</strong>terval tofirst ovulation <strong>in</strong> beef cows fed either of two energy levelspostpartum. Biol Reprod 59, 777–783.Stewart RE, Spicer LJ, Hamilton TD, Keefer BE, Dawson LJ,Morgan GL, Echternkamp SE, 1996: Levels of <strong>in</strong>sul<strong>in</strong>-likegrowth factor (IGF) b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s, lute<strong>in</strong>iz<strong>in</strong>g hormoneand IGF-I receptors, and steroids <strong>in</strong> dom<strong>in</strong>ant folliclesdur<strong>in</strong>g the first follicular wave <strong>in</strong> cattle exhibit<strong>in</strong>g regularestrous cycles. Endocr<strong>in</strong>ology 137, 2842–2850.Sunderland SJ, Crowe MA, Boland MP, Roche JF, Ireland JJ,1994: Selection, dom<strong>in</strong>ance and atresia of follicles dur<strong>in</strong>g theoestrous cycle of heifers. J Reprod Fertil 101, 547–555.Sunderland SJ, Knight PG, Boland MP, Roche JF, Ireland JJ,1996: Alterations <strong>in</strong> <strong>in</strong>trafollicular levels of different molecularmass forms of <strong>in</strong>hib<strong>in</strong> dur<strong>in</strong>g development of follicularandluteal-phase dom<strong>in</strong>ant follicles <strong>in</strong> heifers. Biol Reprod54, 453–462.Van Santbr<strong>in</strong>k EJP, Hop WC, Van Dessel TJHM, De JongFH, Fauser BCJM, 1995: Decremental follicle-stimulat<strong>in</strong>ghormoneand dom<strong>in</strong>ant follicle development dur<strong>in</strong>g thenormal menstrual-cycle. Fertil Steril 64, 37–43.Watson ED, Al-zi’abi MO, 2002: Characterization of morphologyand angiogenesis <strong>in</strong> follicles of mares dur<strong>in</strong>g spr<strong>in</strong>gtransition and the breed<strong>in</strong>g season. <strong>Reproduction</strong> 124, 227–234.Watson ED, Thomassen R, Steele M, Heald M, Leask R,Groome NP, Riley SC, 2002: Concentrations of <strong>in</strong>hib<strong>in</strong>,progesterone and oestradiol <strong>in</strong> fluid from dom<strong>in</strong>ant andsubord<strong>in</strong>ate follicles from mares dur<strong>in</strong>g spr<strong>in</strong>g transitionand the breed<strong>in</strong>g season. Anim Reprod Sci 74, 55–67.Wiltbank MC, Fricke PM, Sangsritavong S, Sartori R,G<strong>in</strong>ther OJ, 2000: Mechanisms that prevent and producedouble ovulations <strong>in</strong> dairy cattle. J Dairy Sci 83, 2998–3007.Zeleznik AJ, 2001: Follicle selection <strong>in</strong> primates: ‘many arecalled but few are chosen’. Biol Reprod 65, 655–659.Zielak AE, Forde N, Park SD, Doohan F, Coussens PM,Smith GW, Ireland JJ, Lonergan P, Evans AC, 2007:Identification of novel genes associated with dom<strong>in</strong>ant follicledevelopment <strong>in</strong> cattle. Reprod Fertil Dev 19, 967–975.Author’s address (for correspondence): M Mihm, Division of CellSciences, Faculty of Veter<strong>in</strong>ary Medic<strong>in</strong>e, University of Glasgow,Bearsden Road, Glasgow, G61 1QH, UK. E-mail: m.mihm@vet.gla.ac.ukConflict of <strong>in</strong>terest: The authors declare no conflict of <strong>in</strong>terests.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 57–65 (2008); doi: 10.1111/j.1439-0531.2008.01143.xISSN 0936-6768Regulation of Luteal Function and Corpus Luteum Regression <strong>in</strong> Cows: HormonalControl, Immune Mechanisms and Intercellular CommunicationDJ Skarzynski 1 , G Ferreira-Dias 2 and K Okuda 31 Department of Reproductive Immunology, Institute of Animal <strong>Reproduction</strong> and Food Research of PAS, Olsztyn, Poland; 2 C.I.I.S.A., Facultyof Veter<strong>in</strong>ary Medic<strong>in</strong>e, TU Lisbon, Portugal; 3 Laboratory of Reproductive Endocr<strong>in</strong>ology, Graduate School of Natural Science andTechnology, Okayama University, Okayama, JapanContentsThe ma<strong>in</strong> function of the corpus luteum (CL) is production ofprogesterone (P4). Adequate luteal function to secrete P4 iscrucial for determ<strong>in</strong><strong>in</strong>g the physiological duration of theoestrous cycle and for achiev<strong>in</strong>g a successful pregnancy. Thebov<strong>in</strong>e CL grows very fast and regresses with<strong>in</strong> a few days atluteolysis. Mechanisms controll<strong>in</strong>g development and secretoryfunction of the bov<strong>in</strong>e CL may <strong>in</strong>volve many factors that areproduced both with<strong>in</strong> and outside the CL. Some of theseregulators seem to be prostagland<strong>in</strong>s (PGs), oxytoc<strong>in</strong>, growthand adrenergic factors. Moreover, there is evidence that P4acts with<strong>in</strong> the CL as an autocr<strong>in</strong>e or paracr<strong>in</strong>e regulator. Eachof these factors may act on the CL <strong>in</strong>dependently or maymodify the actions of others. Although uter<strong>in</strong>e PGF 2a isknown to be a pr<strong>in</strong>cipal luteolytic factor, its direct action onthe CL is mediated by local factors: cytok<strong>in</strong>es, endothel<strong>in</strong>-1,nitric oxide. The changes <strong>in</strong> ovarian blood flow have also beensuggested to have some role <strong>in</strong> regulation of CL development,ma<strong>in</strong>tenance and regression.IntroductionThe corpus luteum (CL) is a transient ovarian organthat is established by cells of a follicle after ovulation.The mammalian CL is composed of a heterogeneousmixture of cell types. There are at least two types ofsteroidogenic cells, large and small luteal cells, whichorig<strong>in</strong>ate from the granulosa and thecal cells of thefollicle ruptured at ovulation, respectively. The CLconsists of not only steroidogenic luteal cells but alsonon-steroidogenic cells, i.e. vascular endothelial cells,fibroblasts, pericytes and immune cells such as lymphocytes,leucocytes and macrophages (Lei et al. 1991).Macrophages and endothelial cells <strong>in</strong>filtrate <strong>in</strong>to thenewly formed CL concomitant with vascular angiogenesis(Reynolds and Redmer 1999). Most of the cellsproliferat<strong>in</strong>g dur<strong>in</strong>g growth of the CL are endothelialcells under hypoxic conditions, although leucocytes havebeen shown to proliferate <strong>in</strong> CL as well (Reynolds andRedmer 1999; Towson et al. 2002). There is evidencethat steroids, prote<strong>in</strong> hormones, growth factors, eicosanoidsand cytok<strong>in</strong>es produced by the component cells ofthe CL play roles <strong>in</strong> establish<strong>in</strong>g CL (Reynolds andRedmer 1999; Berisha and Schams 2005).Progesterone (P4), the primary product of the CL, isrequired for establishment and ma<strong>in</strong>tenance of pregnancy.P4 concentration <strong>in</strong> blood <strong>in</strong>creases dur<strong>in</strong>g thedevelop<strong>in</strong>g luteal stage. The CL cont<strong>in</strong>ues to secrete ahigh level of P4 until late luteal stage, and then its abilityrapidly decreases <strong>in</strong> the regress<strong>in</strong>g luteal stage. Indomestic animals, lute<strong>in</strong>iz<strong>in</strong>g hormone (LH) released<strong>in</strong> a pulsatile fashion from the anterior pituitary is oneof the most potent regulators of synthesis and secretionof P4 <strong>in</strong> the CL (Niswender et al. 2007). Lute<strong>in</strong>iz<strong>in</strong>ghormone stimulates P4 production <strong>in</strong> small luteal cellsvia the LH receptor. In addition to pituitary hormones,<strong>in</strong>traluteal substances produced by the component cellsof the CL play important roles <strong>in</strong> regulat<strong>in</strong>g P4production dur<strong>in</strong>g the luteal stages <strong>in</strong> bov<strong>in</strong>e CL.Moreover, the changes <strong>in</strong> P4 concentration throughoutthe luteal phase are closely associated with blood flow aswell as steroidogenic capacity of luteal cells. Dur<strong>in</strong>gluteal regression, the decrease <strong>in</strong> P4 is concerned withreduced blood flow and loss of LH receptors <strong>in</strong> the CL(Niswender et al. 1976).When animals do not become pregnant, regression ofthe CL, termed luteolysis, is essential for normalcyclicity as it allows development of a new ovulatoryfollicle. In the cow, luteolysis is <strong>in</strong>itiated by prostagland<strong>in</strong>(PG)F 2a released from the uterus at the late lutealstage. Luteolysis consists of two phases, functionalluteolysis and structural luteolysis <strong>in</strong> mammals (McCrackenet al. 1999). A rapid functional regression of CLis characterized by a decrease of P4 production,followed by a phase of structural regression (McCrackenet al. 1999). Dur<strong>in</strong>g structural luteolysis, CL cellsundergo apoptosis, a process that has been describedby morphological and biochemical parameters <strong>in</strong> rum<strong>in</strong>ants(Juengel et al. 1993; Rueda et al. 1995, 1997). It isgenerally accepted that the immune response playscentral roles <strong>in</strong> apoptosis of several tissues and celltypes (Nagata 1997). Recently, <strong>in</strong>traluteal mediators<strong>in</strong>clud<strong>in</strong>g cytok<strong>in</strong>es and nitric oxide (NO) are thought toplay some roles as pro-apoptotic and anti-apoptoticfactors <strong>in</strong> the bov<strong>in</strong>e CL, respectively (Petroff et al.2001; Skarzynski et al. 2005).This review focuses on mediators for regulat<strong>in</strong>gbov<strong>in</strong>e CL function throughout the oestrous cycle.Furthermore, some of our and others’ data on mediators(EDN1, cytok<strong>in</strong>es, NO) of uter<strong>in</strong>e PGF 2a action onbov<strong>in</strong>e CL as well cell-to-cell contact and <strong>in</strong>teractionsdur<strong>in</strong>g luteolysis have also been reviewed. Moreover,<strong>in</strong>tra-luteal mechanisms controll<strong>in</strong>g sensitivity of the CLto extragonadal PGF 2a are discussed.Development and Ma<strong>in</strong>tenance of the CorpusLuteumThe mechanism controll<strong>in</strong>g development, ma<strong>in</strong>tenanceand secretory function of the CL may <strong>in</strong>volve factorsthat are produced both with<strong>in</strong> the CL and outside theovary (Fig. 1). Some of these regulators, that act asÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


58 DJ Skarzynski, G Ferreira-Dias and K Okuda(+)Prostagland<strong>in</strong>s(PGE 2, PGI 2, PGF 2a )LH/gonadotrop<strong>in</strong>s(+)P4Endothelial/luteal cellsdifferentiation and/orproliferationautocr<strong>in</strong>e and ⁄ or paracr<strong>in</strong>e factors, seem to be PGs andother arachidonic acid metabolites [PGE 2 , PGF 2a ,leucotrienes (LT)], neuropeptides [noradrenal<strong>in</strong>e (NA)],peptide hormones [i.e. oxytoc<strong>in</strong> (OT)], NO, growthfactors and hormones [vascular endothelial growthfactor (VEGF), fibroblastic growth factors (FGF),epidermal growth factor (EGF), growth hormone(GH), prolact<strong>in</strong> (PRL)] and steroids [P4 and 17boestradiol(E 2 )].Corpus luteum developmentThe physiological processes of CL growth and formationmight be regulated by many different factors, stillnot completely understood (Fig. 1) (Reynolds andRedmer 1999; Acosta and Miyamoto 2004; Schamsand Berisha 2004; Meidan et al. 2005; Ferreira-Dias andSkarzynski 2008). After ovulation, as the CL formsfrom the wall of the ruptured follicle, it grows andvascularizes rapidly. In fact, the rates of tissue growthand angiogenesis <strong>in</strong> the CL rival those of even thefastest grow<strong>in</strong>g tumours. The CL is a complex tissuecomposed of paranchymal (small and large steroidogenic)and non-parenchymal (fibroblast, vascularsmooth muscle, pericytes and endothelial) cells (Reynoldsand Redmer 1999; Lei et al. 2000; Towson et al.2002). In agreement with data from other tissues,VEGF seem to be a major angiogenic factor responsiblefor vascularization of the develop<strong>in</strong>g CL. Recent datasuggest that luteal expression of VEGF occurs primarily<strong>in</strong> steroidogenic cells (granulose-lute<strong>in</strong> cells) and less <strong>in</strong>endothelial cells (specific perivascular cells, <strong>in</strong>clud<strong>in</strong>garteriolar smooth muscle and capillary pericytes), and isregulated primarily by oxygen levels (Berisha andSchams 2005; Ferreira-Dias and Skarzynski 2008).Soon after ovulation, pericytes derived from the thecalcompartment appear to be the first vascular cells to<strong>in</strong>vade the develop<strong>in</strong>g luteal parenchyma. The granulosa-derivedcells produce a factor that stimulatespericytes migration. Moreover, NO, which is a potentvasodilator and stimulates VEGF production andangiogenesis, is produced by endothelial cells of lutealarterioles and capillaries, often <strong>in</strong> association withexpression of VEGF by luteal perivascular cells (Berishaand Schams 2005).(+)Growth factors, cytok<strong>in</strong>es(VEGF, FGF, IGF, EGF,TNFa)Fig. 1. Hypothetical model of the regulation of the CL development(see text for the details; adapted from Ferreira-Dias and Skarzynski2008)Basic FGF mRNA and prote<strong>in</strong> and its receptors arepresent <strong>in</strong> the CL and may stimulate proliferation ofluteal endothelial cells (Reynolds and Redmer 1999;Berisha and Schams 2005). The IGF system plays a role<strong>in</strong> CL development and may <strong>in</strong>directly affect angiogenesis<strong>in</strong> the early CL by stimulat<strong>in</strong>g VEGF production(Schams and Berisha 2004; Berisha and Schams 2005).Thus, several growth factors act as auto-paracr<strong>in</strong>eregulators affect<strong>in</strong>g proliferation and differentiation ofthe develop<strong>in</strong>g CL cells (Reynolds and Redmer 1999;Acosta and Miyamoto 2004; Berisha and Schams 2005).Moreover, the luteotropic paracr<strong>in</strong>e and ⁄ or autocr<strong>in</strong>eeffects of OT, NO, PGF 2a and other metabolites ofarachidonic acid (PGE 2 , PGI 2 , leucotrienes) suggest thateikosanoids assume different and perhaps oppos<strong>in</strong>groles depend<strong>in</strong>g on the cellular and hormonal milieu(Skarzynski et al. 2000, 2001; Weems et al. 2004; Meidanet al. 2005). Prostagland<strong>in</strong>s and leucotrienes mayregulate cell proliferation and stimulate P4 productionregulat<strong>in</strong>g ⁄ stimulat<strong>in</strong>g CL development and formation.Just after ovulation, the basement membrane breaksdown, and blood vessels from the theca <strong>in</strong>terna <strong>in</strong>vadethe avascular granulose-lute<strong>in</strong> cell layers (Acosta andMiyamoto 2004; Berisha and Schams 2005). Thesephenomena are thought to occur under hypoxic conditions.Thus, we have proposed a model for the <strong>in</strong>itialprocess of luteal vascularization <strong>in</strong> which hypoxia playsa major role. In this model, a paracr<strong>in</strong>e loop existsbetween the vascular endothelial cells which produceNO, and granulose-lute<strong>in</strong> cells which ma<strong>in</strong>ly produceVEGF, to ensure coord<strong>in</strong>ated regulation of lutealvasodilation and angiogenesis. Dur<strong>in</strong>g this decade,hypoxia-<strong>in</strong>ducible factor 1 (HIF1) has been recognizedto have critical roles <strong>in</strong> angiogenesis via transcriptionalregulation of angiogenic factors, such as VEGF. Ourrecent study <strong>in</strong>dicates that HIF1 is essential for theVEGF-<strong>in</strong>duced angiogenesis dur<strong>in</strong>g luteal development,and suggests that formation of luteal vasculature <strong>in</strong>cows is regulated by hypoxic condition follow<strong>in</strong>g folliclerupture (Nishimura R and Okuda K, unpublished data).Ma<strong>in</strong>tenance of the corpus luteum: mandatory role ofprogesteroneIn cows, LH and GH are the primary hormones whichsupport the development and function of the CL (Hanseland Dowd 1986; Niswender et al. 2007). However, thelocal angiogenic growth factors (VEGD, EGF, FGF),PGs and peptide hormones (OT) should be considerednot only the potent regulators of luteal development, butalso important factors regulat<strong>in</strong>g P4 secretion andlifespan of CL (Skarzynski et al. 2001; Berisha andSchams 2005; Meidan et al. 2005; Fig. 1). Moreover, P4also has an effect on function of the bov<strong>in</strong>e early and midCL <strong>in</strong> an autocr<strong>in</strong>e and paracr<strong>in</strong>e fashion (Skarzynskiand Okuda 1999; Duras et al. 2005). In order to removethe <strong>in</strong>fluence of P4 produced by cultured bov<strong>in</strong>e lutealcells, we treated cells with a specific P4 antagonist(onapristone; Skarzynski and Okuda 1999; Okuda et al.2004). In early luteal cells, secretions of P4 OT, PGF 2aand PGE 2 were reduced by onapristone. Moreover, theP4 antagonist <strong>in</strong>hibited OT secretion by mid-cycle lutealcells, although it stimulated PGF 2a secretion (SkarzynskiÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Regulation of Luteal Function 59and Okuda 1999). These f<strong>in</strong>d<strong>in</strong>gs suggest that P4stimulates P4, OT and PGs secretion by early CL, butat mid-cycle CL, P4 <strong>in</strong>hibits PGF 2a secretion. Pate (1988,1996) also reported that P4 regulates PGF 2a secretion <strong>in</strong>the mid-cycle CL, but not <strong>in</strong> the late CL. However,dur<strong>in</strong>g pregnancy P4 did not affect its own secretion bybov<strong>in</strong>e (Weems et al. 2002) and ov<strong>in</strong>e luteal slices (Kimet al. 2001). Therefore, P4 may affect the secretoryfunction of the bov<strong>in</strong>e CL <strong>in</strong> a stage-dependent fashionand P4 effects may depend on cell-to-cell contact andcomposition (Pate 1996; Skarzynski and Okuda 1999;Skarzynski et al. 2001; Weems et al. 2002).Recent studies have demonstrated that <strong>in</strong>tra-luteal P4is one of the most important factors support<strong>in</strong>g ma<strong>in</strong>tenanceof the CL (Fig. 2). It has been shown that P4may suppress apoptosis <strong>in</strong> bov<strong>in</strong>e luteal cells throughthe <strong>in</strong>hibition of Fas and caspase-3 mRNA expressionand <strong>in</strong>hibition of caspase-3 activation (Rueda et al.2000; Okuda et al. 2004). In addition to our previousf<strong>in</strong>d<strong>in</strong>g that P4 may stimulate its own production bybov<strong>in</strong>e CL (Skarzynski and Okuda 1999), we havedemonstrated that blockage of the autocr<strong>in</strong>e and ⁄ orparacr<strong>in</strong>e action of P4 (by a specific antagonist –onapristone) reduced viability of the luteal cells(Fig. 2; Okuda et al. 2004). In conclusion, the overallresults showed that P4 may <strong>in</strong>hibit Fas L-mediatedapoptosis via a decrease of Fas and caspase-3 expressionand caspase-3 activity by bov<strong>in</strong>e luteal cells. Themechanisms or signall<strong>in</strong>g pathways by which P4 exertsits action on the bov<strong>in</strong>e CL are unknown. P4 seems toact on the secretory function of bov<strong>in</strong>e CL (Bogackiet al. 2000) non-genomically, i.e. through membraneb<strong>in</strong>d<strong>in</strong>g sites (Rae et al. 1998) rather than throughnuclear mediation. In support of this idea, Cannon et al.(2003) have shown that P4 suppresses luteal cell-stimulatedT lymphocyte proliferation. However,classical-genomic P4 receptors were not expressed <strong>in</strong> Tlymphocytes show<strong>in</strong>g that P4 may act non-genomicallyon the cells (Sug<strong>in</strong>o et al. 1997). On the other hand, ithas been suggested that P4 plays an active role <strong>in</strong><strong>in</strong>hibition of luteal regression by a direct effect onclassical P4 genomic b<strong>in</strong>d<strong>in</strong>g sites <strong>in</strong> bov<strong>in</strong>e luteal cells(Rueda et al. 2000). Moreover, P4 promotes survival ofbov<strong>in</strong>e luteal cells by <strong>in</strong>hibit<strong>in</strong>g apoptosis via theglucocorticoid receptors (GR; Rueda et al. 2000).All these f<strong>in</strong>d<strong>in</strong>gs confirmed the previous suppositionthat P4 is an universal autocr<strong>in</strong>e and paracr<strong>in</strong>e regulatorof luteal function <strong>in</strong> cows and plays one or more role(s)<strong>in</strong> the regulation of development and ma<strong>in</strong>tenance of thebov<strong>in</strong>e CL (Pate 1996; Rae et al. 1998; Skarzynski andOkuda 1999; Rueda et al. 2000; Okuda et al. 2004).Thus, it could be assumed that <strong>in</strong>tra-luteal P4 isimplicated <strong>in</strong> a survival pathway <strong>in</strong> the CL by stimulationPGs, OT and its own production as well as by the<strong>in</strong>hibition of apoptosis of the bov<strong>in</strong>e CL.Intraluteal Factors Mediate Luteolytic Actionof PGF 2aIn the absence of an embryo(s) <strong>in</strong> the uterus, the processof CL regression beg<strong>in</strong>s on days 17–19 of the oestrouscycle <strong>in</strong> cows (McCracken et al. 1999). This process ischaracterized not only by functional but also structural(a)Cell viability (% of control)0(b)500Progesterone (ng/2x10 5 cells)(c)Caspase-3 activity(µmol pNA/m<strong>in</strong>/2x10 5 cells)12510075502540030020010003020100aaaaabchanges that occur <strong>in</strong> the luteal (steroidogenic) and nonluteal(accessory) cells of the CL (Juengel et al. 1993;Meidan et al. 1999). In most species, uter<strong>in</strong>e PGF 2a isknown to be a pr<strong>in</strong>cipal luteolytic factor, but its directaction with<strong>in</strong> the CL is still debated (Skarzynski andOkuda 1999; Pate 2003; Schams and Berisha 2004;Meidan et al. 2005; Skarzynski et al. 2005). The actionof PGF 2a on bov<strong>in</strong>e CL is mediated by local factors:EDN1, cytok<strong>in</strong>es and NO (Fig. 3).Endothel<strong>in</strong>-1 and vasoactive peptidesA pivotal role for the endothelial cell product – EDN1<strong>in</strong> PGF 2a -<strong>in</strong>duced luteal regression <strong>in</strong> cows has been welldocumented by Meidan’s group (Girsh et al. 1996;bbControl Fas L OP Fas L/OP TNF a/IFNg(6 nmol/l) (100 µmol/M) (by 3 nmol/l)Fig. 2. Effects of Fas L (6 nM; 05-351; Upstate Biotechnology, LakePlacid, NY, USA) and a specific progesterone antagonist (OP;100 lM; Scher<strong>in</strong>g AG, Berl<strong>in</strong>, Germany; #ZK98.299) on cell viability(a) progesterone secretion (b) and activity of caspase-3 (c) <strong>in</strong> luteal cellsobta<strong>in</strong>ed from the cows at the mid-luteal phase of the oestrous cycle(days 8–12 of the cycle) and cultured <strong>in</strong> medium supplemented with0.1% of BSA (#735078; Roche Diagnostics GmbH, Mannheim,Germany). Cytok<strong>in</strong>es (TNF-a and INFc; both 3 nM; gift of Da<strong>in</strong>ipponSumitomo Pharma Co., Ltd, Osaka, Japan) were added as apositive control (adapted from Okuda et al. 2004). The data are shownas the mean ± SEM of values obta<strong>in</strong>ed <strong>in</strong> four separate experimentseach performed <strong>in</strong> triplicatebcccdbcÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


60 DJ Skarzynski, G Ferreira-Dias and K Okuda(+)(+)(+)EDN1(–)PGF 2aNO(–)(–)P4Apoptosis of steroidogenic andendothelial cells of the CLCytok<strong>in</strong>es(TNFa,FAS-L,IFNg)Fig. 3. Hypothetical model of the structural and functional regressionof the CL (see text for the details; adapted from Ferreira-Dias andSkarzynski 2008)Mamluk et al. 1999; Meidan et al. 1999, 2005; Levyet al. 2000). EDN1 <strong>in</strong>hibited P4 secretion, <strong>in</strong> a dosedependent manner via selective EDN1 b<strong>in</strong>d<strong>in</strong>gs sites(EBN A ; Girsh et al. 1996). The expression of membersof the EDN1 system (EDN1, EDN convert<strong>in</strong>g enzymes,and EDN A and EDN B receptors) <strong>in</strong>creases dur<strong>in</strong>g lutealregression (Ohtani et al. 1998; Klipper et al. 2004;Rosiansky-Sultan et al. 2006). Moreover, PGF 2a upregulatesEDN1 and EDN A expression with<strong>in</strong> the CL(Mamluk et al. 1999). Besides EDN1, other vasoactivepeptides [i.e. angiothens<strong>in</strong> II (ANG-II), atrial natriureticpeptide (ANP)] are considered important factors <strong>in</strong>mediat<strong>in</strong>g PGF 2a luteolytic action (Schams and Berisha2004; Berisha and Schams 2005). These vasoactivepeptides decreased blood flow and triggered the luteolyticcascade and consequently <strong>in</strong>hibited P4 secretion(Kobayashi et al. 2002; Shirasuna et al. 2004). However,Flores with colleagues showed that the EDN1 system(EDN1, EDN A and EDN convert<strong>in</strong>g enzymes) exists <strong>in</strong>the bov<strong>in</strong>e CL through the oestrous cycle and PGF 2a<strong>in</strong>creased EDN1 mRNA expression <strong>in</strong> vivo only at 10 hafter treatment (Wright et al. 2001). In fact, Schamset al. (2003) showed that up-regulation of EDN1 andANG-2 occurred ma<strong>in</strong>ly dur<strong>in</strong>g structural CL regression.Therefore, it has been suggested that EDN1 is<strong>in</strong>volved <strong>in</strong> the process of structural CL regression bypromot<strong>in</strong>g leucocyte migration and stimulat<strong>in</strong>g macrophagesto release cytok<strong>in</strong>es [i.e. tumour necrosis factor a(TNF-a), <strong>in</strong>terferon c (IFN-c); Meidan et al. 1999,2005].Cytok<strong>in</strong>esImmune cells <strong>in</strong>filtrat<strong>in</strong>g the bov<strong>in</strong>e CL play a centralrole <strong>in</strong> structural luteolysis of both steroidogenic andendothelial CL cells (Friedman et al. 2000; Pate andLandis Keyes 2001; Pate 2003). The number of leucocytes(i.e. T lymphocytes, macrophages) <strong>in</strong>creased at thetime of structural luteolysis (Penny et al. 1999; Towsonet al. 2002). Shaw and Britt (1995) us<strong>in</strong>g a CL microdialysissystem showed that TNF-a is released dur<strong>in</strong>gspontaneous and PGF 2a -<strong>in</strong>duced luteolysis <strong>in</strong> cows.Then, it was shown that the mRNAs for TNF-a and itsspecific receptors (TNFR type-I) are present <strong>in</strong> thebov<strong>in</strong>e CL dur<strong>in</strong>g luteolysis (Sakumoto et al. 2000;Neuvians et al. 2004). Tumour necrosis factor a <strong>in</strong>comb<strong>in</strong>ation with IFN-c reduced P4 production and<strong>in</strong>duced apoptosis and PGF 2a production by luteal cells<strong>in</strong> vitro (Sakumoto et al. 2000; Petroff et al. 2001;Korzekwa et al. 2006). Specific b<strong>in</strong>d<strong>in</strong>g sites for TNFaare also present <strong>in</strong> endothelial cells derived frombov<strong>in</strong>e CL (Okuda et al. 1999). Furthermore, TNF-a<strong>in</strong>duces EDN1 production by endothelial cells that maylead to structural regression of the CL (Okuda et al.1999; Friedman et al. 2000). Tumour necrosis factor aact<strong>in</strong>g via TNFR type-I <strong>in</strong>duces apoptotic death ofsteroidogenic (Petroff et al. 2001) and endothelial cells(Friedman et al. 2000) of bov<strong>in</strong>e CL. However, some <strong>in</strong>vitro studies <strong>in</strong>dicated that TNF-a <strong>in</strong>duces luteolysisonly <strong>in</strong> comb<strong>in</strong>ation with INFc or other factors (i.e.EDN1; Petroff et al. 2001; Korzekwa et al. 2006).Therefore, we tested whether TNF-a acts as a luteolyticfactor <strong>in</strong> vivo, and whether it changes the lifespan ofbov<strong>in</strong>e CL (Skarzynski et al. 2003a). Lower doses ofTNF-a <strong>in</strong>creased PGF 2a and nitrite ⁄ nitrate (stablemetabolites of NO), decreased P4 level and consequentlyresulted <strong>in</strong> shorten<strong>in</strong>g of the oestrous cycle. Surpris<strong>in</strong>gly,higher doses of TNF-a stimulated the synthesis of P4and PGE 2 and consequently resulted <strong>in</strong> prolongation ofthe oestrous cycle (Skarzynski et al. 2003a). However,<strong>in</strong>hibition of PG synthesis by <strong>in</strong>domethac<strong>in</strong>, a cyclooxygenase(COX; PTGS) <strong>in</strong>hibitor, <strong>in</strong>jected <strong>in</strong>to the aortaabdom<strong>in</strong>alis, blocked the actions of TNF-a <strong>in</strong>dicat<strong>in</strong>gthat TNF-a acts ma<strong>in</strong>ly through mediation by arachidonicacid metabolites (Skarzynski et al. 2007). Inaddition to TNFR, other cytok<strong>in</strong>e membrane receptors,second messengers, <strong>in</strong>clud<strong>in</strong>g calcium ions [Ca 2+ ] i andregulatory prote<strong>in</strong>s are <strong>in</strong>volved <strong>in</strong> apoptosis of steroidogenicand endothelial CL cells (Meidan et al. 1999;Petroff et al. 2001; Taniguchi et al. 2002). Fas ligand, amember of the TNF super family, primarily engages itsreceptors (Fas) to <strong>in</strong>duce apoptosis (Taniguchi et al.2002; Okuda et al. 2004). The expression of Fas mRNAwas <strong>in</strong>creased by IFN-c, and TNF-a augmented thestimulatory action of IFN-c on Fas expression (Taniguchiet al. 2002). Moreover, apoptotic bodies wereobserved <strong>in</strong> luteal cells treated with Fas L <strong>in</strong> thepresence of IFN-c and ⁄ or TNF-a, show<strong>in</strong>g that leucocyte-derivedTNF-a and IFN-c play important roles <strong>in</strong>Fas L-Fas-mediated luteal cell death <strong>in</strong> the bov<strong>in</strong>e CL.Nitric oxideNADPH-d localization [a marker for nitric oxidesynthase (NOS)] and immunosta<strong>in</strong><strong>in</strong>g of both isoformsof NOS [<strong>in</strong>ducible (iNOS) and endothelial (eNOS)] weredetected <strong>in</strong> steroidogenic cells and <strong>in</strong> blood vessels of thebov<strong>in</strong>e CL dur<strong>in</strong>g the entire oestrous cycle with<strong>in</strong>creas<strong>in</strong>g activity from the early to the late lutealphases (Skarzynski et al. 2003b). However, Rosiansky-Sultan et al. (2006) presented the highest level of eNOSand iNOS mRNA and prote<strong>in</strong> expression <strong>in</strong> the earlyÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Regulation of Luteal Function 61bov<strong>in</strong>e CL. These data suggest that NO produced bytwo NOS isoforms is <strong>in</strong>volved <strong>in</strong> structural and functionalchanges that occur <strong>in</strong> the bov<strong>in</strong>e CL through thewhole oestrous cycle. Luteolytic or luteotropic actionsof NO on the bov<strong>in</strong>e CL is strictly dependent on thestage of CL, and cell <strong>in</strong>teractions (cell-to-cell contact)and composition (Jaroszewski et al. 2003a; Klipperet al. 2004; Weems et al. 2004; Rosiansky-Sultan et al.2006). Nitric oxide donor (S-NAP) stimulated PGE 2secretion by steroidogenic CL cells <strong>in</strong> the early and midlutealphases (Skarzynski et al. 2000). Dur<strong>in</strong>g developmentand ma<strong>in</strong>tenance of the CL, PGE 2, which is bothluteotropic and antiluteolytic, stimulated NO production(Boiti et al. 2000). Nitric oxide donors and ET-1<strong>in</strong>creased PGE 2 secretion by bov<strong>in</strong>e CL slices <strong>in</strong> vitro ondays 13–14 of the cycle without any effect on P4secretion (Weems et al. 2004). Therefore, <strong>in</strong>creased NOproduction dur<strong>in</strong>g early stages of the cycle and pregnancyis likely to play a role <strong>in</strong> CL development andangiogenesis (Skarzynski et al. 2000; Weems et al. 2004;Vonnahme et al. 2005; Rosiansky-Sultan et al. 2006).In the late luteal phase PGF 2a might simulate a shearstress-like reaction of endothelial CL cells result<strong>in</strong>g <strong>in</strong>compensative NO release dur<strong>in</strong>g the first steps ofluteolysis – up to 2–4 h after PGF 2a treatment (Li et al.2002; Skarzynski et al. 2003b; Acosta et al. 2007). Intralutealadm<strong>in</strong>istration of a NOS <strong>in</strong>hibitor (L-NAME)dur<strong>in</strong>g the late luteal phase <strong>in</strong>creased P4 secretion andprolonged the functional lifespan of bov<strong>in</strong>e CL (Jaroszewskiand Hansel 2000; Sasahara et al. 2007). Whenan analogue of PGF 2a (aPGF 2a , cloprostenol) was<strong>in</strong>jected on day 15 of the cycle <strong>in</strong> comb<strong>in</strong>ation withL-NAME, the luteolytic effect of aPGF 2a was counteractedby the NOS <strong>in</strong>hibitor (Fig. 4; Jaroszewski et al.2003b; Skarzynski et al. 2003b). Nitric oxide has beenfound as the most potent <strong>in</strong>hibitor of P4 secretion<strong>in</strong> vitro (Skarzynski et al. 2003b; Korzekwa et al. 2004,2006) and <strong>in</strong> vivo (Sasahara et al. 2007). Moreover, aNO donor (Sperm<strong>in</strong>e NONOate) strongly stimulatedproduction of PGF 2a and LTC 4 by bov<strong>in</strong>e CL both<strong>in</strong> vitro and <strong>in</strong> vivo, show<strong>in</strong>g that NO is <strong>in</strong>volved <strong>in</strong> theprocess of luteal regression by bov<strong>in</strong>e CL (Fig. 3;Progesterone (ng/ml)129630L-NAME/aPGF (n = 5)Sal<strong>in</strong>e/aPGF(n = 6)L-NAME/Sal<strong>in</strong>e (n = 4)Sal<strong>in</strong>e/sal<strong>in</strong>e (n = 4)aPGF0 3 6 9 12 15 18 0 4Day of the estrous cycle– EstrusEstrusFig. 4. The effect of 2 h <strong>in</strong>fusion of sal<strong>in</strong>e or nitric oxide synthase<strong>in</strong>hibitor-L-NAME (400 mg ⁄ h) and <strong>in</strong>jection of sal<strong>in</strong>e or PGF 2aanalogue, cloprostenol (aPGF; 100 lg; Bioestrophan, Biowet, GorzowWielkopolski, Poland) at 30 m<strong>in</strong> of <strong>in</strong>fusion on progesterone concentrations<strong>in</strong> peripheral blood plasma of heifers on day 15 of the oestrouscycle. Different subscript letters <strong>in</strong>dicate significant differences(p


62 DJ Skarzynski, G Ferreira-Dias and K Okudato a luteolytic dose of aPGF 2a and suggested that this<strong>in</strong>crease <strong>in</strong> blood flow is related to early luteolytic events(Acosta et al. 2002; Miyamoto et al. 2005). This <strong>in</strong>crease<strong>in</strong> blood flow is thought to be caused by well-knownvasodilators, PGE 2 and ⁄ or NO (Miyamoto et al. 2005).In support, it has been recently shown that <strong>in</strong>tralutealapplication of a NO donor drastically <strong>in</strong>crease lutealblood flow (Sasahara et al. 2007).However, CL blood flow was reduced 8 h after oncePGF 2a i.m. <strong>in</strong>jection <strong>in</strong> cows (Acosta et al. 2002) andwith<strong>in</strong> 4 h after two <strong>in</strong>jections of PGF 2a (at 0 and 4 h)<strong>in</strong>to the uter<strong>in</strong>e lumen <strong>in</strong> ewes (Nett et al. 1976). Thus,PGF 2a may cause CL regression by depriv<strong>in</strong>g the gland ofnutrients, substrates for steroidogenesis, and luteotropicsupport (Nett et al. 1976; Niswender et al. 2000). Therefore,we hypothesized that hypoxic (low oxygen) conditions<strong>in</strong> the CL <strong>in</strong>duced by a decreased blood supply isrelated to the luteolytic cascade <strong>in</strong> cows. We exam<strong>in</strong>edthe <strong>in</strong>fluence of hypoxia on the luteal P4 generat<strong>in</strong>gsystem and luteal cell apoptosis us<strong>in</strong>g bov<strong>in</strong>e luteal cellculture, and found that hypoxia decreased P4 synthesisby suppress<strong>in</strong>g a steroidogenic enzyme P450scc activity(Nishimura et al. 2007) and also <strong>in</strong>duced apoptotic celldeath by enhanc<strong>in</strong>g the expression of pro-apoptoticprote<strong>in</strong> BNIP3 and by activat<strong>in</strong>g caspase-3 (Nishimuraet al. 2008). S<strong>in</strong>ce P4 treatment under hypoxia decreasedluteal cell death, the <strong>in</strong>duction of apoptosis by hypoxiaseems to be partially caused by a decrease <strong>in</strong> P4production (Nishimura et al. 2008). However, <strong>in</strong> ewesafter a s<strong>in</strong>gle PGF 2a <strong>in</strong>jection, levels of P4 decreased by12 h after PGF 2a , but total ovarian blood flow did notdecreased significantly until 30 h (Nett et al. 1976).Blood flow to ov<strong>in</strong>e CL and luteal LH receptors didnot decrease until P4 decl<strong>in</strong>ed <strong>in</strong> the peripheral blood andfunctional lutelysis was almost completed (Nett et al.1976; Niswender et al. 1976). Moreover, Watanebe et al.(2006) showed that P4 decreased even when blood flowrema<strong>in</strong>ed high <strong>in</strong> bov<strong>in</strong>e CL. Therefore, oxygen deficiencydue to decreased blood flow is not mandatory forthe <strong>in</strong>duction of both functional and structural luteolysis<strong>in</strong> cows, but may play such a support<strong>in</strong>g role <strong>in</strong> the f<strong>in</strong>alsteps of bov<strong>in</strong>e CL regression.Resistance to PGF 2a Action Dur<strong>in</strong>g the LutealPhaseThe newly formed CL is resistant to exogenous PGF 2atreatment and action (Henricks et al. 1974; Beal et al.1980). Moreover, the sensitivity of CL to the luteolyticaction of extragonadal PGF 2a seems to <strong>in</strong>crease progressivelytoward the end of the luteal phase (Skarzynskiet al. 1997; Pate 2003; Meidan et al. 2005). S<strong>in</strong>ce a s<strong>in</strong>gleclass of high-aff<strong>in</strong>ity PGF 2a receptors is present with<strong>in</strong>the bov<strong>in</strong>e CL by the second day after ovulation(Sakamoto et al. 1995; Wiltbank et al. 1995), neitherthe lack of responsiveness to PGF 2a <strong>in</strong> the early CL(Henricks et al. 1974; Beal et al. 1980) nor the lowersensitivity to PGF 2a <strong>in</strong> the mid-luteal CL (Skarzynskiet al. 1997) can be attributed to a deficiency of highaff<strong>in</strong>ityPGF 2a receptors. Wiltbank et al. (1990) suggestedthat <strong>in</strong>complete vascularization or <strong>in</strong>completedifferentiation of degenerative mechanisms <strong>in</strong> the earlybov<strong>in</strong>e CL are responsible for the lack of luteolyticcapacity. In recent years, Meidan et al. have expla<strong>in</strong>edhow the products of endothelial cells are <strong>in</strong>volved <strong>in</strong> theacquisition of luteolytic capacity by the bov<strong>in</strong>e CL(Girsh et al. 1996; Mamluk et al. 1999; Meidan et al.1999, 2005; Levy et al. 2000). They showed that the lackof ET-1 synthesis and response to EDN-1 <strong>in</strong> the earlyand mid-luteal CL may make the CL unresponsive tothe luteolytic action of PGF 2a (Mamluk et al. 1999;Levy et al. 2000; Rosiansky-Sultan et al. 2006). In fact,PGF 2a upregulated the amount of mRNA encod<strong>in</strong>gEDN-1 convert<strong>in</strong>g enzymes dur<strong>in</strong>g the mid- and latelutealphase of the bov<strong>in</strong>e CL and this action of PGF2awas cycle-phase specific, because it was not observed <strong>in</strong>the early CL. Additionally, the steroidogenic cells of theCL may also possess autocr<strong>in</strong>e mechanisms that protectbov<strong>in</strong>e CL from premature luteolysis. PGF 2a was foundto <strong>in</strong>duce mRNA for PTGS-2 on day 11 but not on day4 of the oestrous cycle, suggest<strong>in</strong>g that such autoamplificationof luteal PGF 2a is also a key component ofluteolytic capacity by the bov<strong>in</strong>e CL (Tsai and Wiltbank1998). Moreover, Silva et al. (2000) showed that upregulationof 15-hydroxyPG dehydrogenases <strong>in</strong> ov<strong>in</strong>e CLdur<strong>in</strong>g early pregnancy may deactivate PGF 2a as amechanism for maternal recognition of pregnancy.We showed that the lack of response to PGF 2a by theearly bov<strong>in</strong>e CL and the lower reaction of bov<strong>in</strong>e CL toPGF 2a dur<strong>in</strong>g the mid-luteal phase depended uponlocally produced PGs, OT and P4 (Skarzynski andOkuda 1999), NA and NO (Skarzynski et al. 2000). Thelack of response to PGF 2a <strong>in</strong> the early CL could be aconsequence of receptor desensitization and the biologicalwanes over time (Lohse 1993; Skarzynski et al.2001). Based on our f<strong>in</strong>d<strong>in</strong>gs, one could assume thatluteal neuropeptide, NO, OT, PGs and P4 are componentsof an auto ⁄ paracr<strong>in</strong>e positive feedback cascade <strong>in</strong>bov<strong>in</strong>e CL, and that they also play roles <strong>in</strong> regulat<strong>in</strong>gthe function of PGF 2a receptors and the PGF 2a -<strong>in</strong>tracellularcalcium ([Ca 2+ ] i )-prote<strong>in</strong> k<strong>in</strong>ase C cascade(Skarzynski and Okuda 1999; Skarzynski et al. 2000,2001). The auto ⁄ paracr<strong>in</strong>e positive feedback loop can bea mechanism for protection aga<strong>in</strong>st premature luteolysisdur<strong>in</strong>g the early and mid-luteal phase. Furthermore,each of the factors that directly affect PGs, OT and P4secretion by luteal cells may be <strong>in</strong>directly <strong>in</strong>volved <strong>in</strong>regulat<strong>in</strong>g function of PGF 2a receptors, and may beresponsible for the vary<strong>in</strong>g sensitivity of bov<strong>in</strong>e lutealcells to PGF 2a . In contrast, locally produced NO maysensitize bov<strong>in</strong>e CL to luteolytic action of PGF 2a(Skarzynski et al. 2000, 2003b). The amplified effectsof PGF 2a on luteal cells by pre-exposure to a NO donor(S-NAP) suggest that prim<strong>in</strong>g of bov<strong>in</strong>e CL by NO isneeded for complete luteal regression (Skarzynski et al.2000). In support, NO up-regulated PGF 2a receptors <strong>in</strong>bov<strong>in</strong>e CL endothelial cells (Rosiansky-Sultan et al.2006). Based on the overall f<strong>in</strong>d<strong>in</strong>gs, the differentresponse by CL to PGF 2a dur<strong>in</strong>g the luteal phase andpregnancy should be expla<strong>in</strong>ed not only by changes <strong>in</strong>the concentration of PGF 2a receptors but also bychanges <strong>in</strong> their sensitivity as well as on the maturationof ET-1 and NO systems. Therefore, a number ofdifferent pathways may account for the lack of PGF 2a -<strong>in</strong>duced luteolysis dur<strong>in</strong>g the early luteal phase as well asdur<strong>in</strong>g early pregnancy.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Regulation of Luteal Function 63(+/–)Growth factorsEGFIGF-1(+)TGF(+/–)ET-1AVPOTConclusionThe mechanism controll<strong>in</strong>g development, ma<strong>in</strong>tenanceand secretory function of the CL may <strong>in</strong>volve factorsthat are produced both with<strong>in</strong> the CL and outside theovary (Fig. 5). Some of these regulators seem to be PGsand other arachidonic acid metabolites (PGE 2 , PGF 2a ,LT), neuropeptides (NA), peptide hormones (OT, EDN-1), growth factors and hormones (VEGF, FGF, GH,PRL) and steroids (P4 and E2) that act as autocr<strong>in</strong>eand ⁄ or paracr<strong>in</strong>e factors. Although PGF 2a is known tobe the pr<strong>in</strong>cipal luteolytic factor, its action on the CL ismediated by other <strong>in</strong>tra-ovarian factors: cytok<strong>in</strong>es, NO,EDN-1. Nitric oxide, TNF-a <strong>in</strong> comb<strong>in</strong>ation with IFN-creduced P4 secretion, <strong>in</strong>creased luteal PGF 2a production,and <strong>in</strong>duced apoptosis of the luteal cells.AcknowledgementsbFGF(?)(+/–)(+)(?)(+/–)PeptidesLHP4Neuropeptides(+)Cytok<strong>in</strong>esIL-1IFNEicosanoidsTNFPGI2PGE2PGF2aCLThe work was supported by the grants-<strong>in</strong>-aid: from the Polish M<strong>in</strong>istryof Sciences and Higher Education (Scientific net; D.J. Skarzynski),<strong>in</strong>ternational grant PORTUGALIA 78 ⁄ 2007 (G. Ferreira-Dias), andfrom the Japanese-Polish Jo<strong>in</strong>t Research Project under the agreementbetween Japan Society for Promotion of Sciences and Polish Academyof Sciences (K. Okuda).ReferencesAcosta TJ, Miyamoto A, 2004: Vascular control of ovarianfunction: ovulation, corpus luteum formation and regression.Anim Reprod Sci 82, 127–140.Acosta TJ, Yoshizawa N, Ohtani M, Miyamoto A, 2002:Local changes <strong>in</strong> blood flow with<strong>in</strong> the early and midcyclecorpus luteum after PGF 2a <strong>in</strong>jection <strong>in</strong> the cow. Biol Reprod66, 651–658.Acosta T, Bah M, Jaroszewski J, Skarzynski DJ, Okuda K,2007: Acute changes <strong>in</strong> circulat<strong>in</strong>g concentrations of oxygen,nitric oxide and progesterone. Biol Reprod 77 (Sp. Iss.),121–122.Beal WE, Milvae RA, Hansel W, 1980: Oestrous cycle lengthand plasma progesterone concentrations follow<strong>in</strong>g adm<strong>in</strong>istrationof PGF 2a early <strong>in</strong> the bov<strong>in</strong>e oestrous cycle. JReprod Fertil 59, 393–396.Berisha B, Schams D, 2005: Ovarian function <strong>in</strong> rum<strong>in</strong>ants.Domest Anim Endocr<strong>in</strong>ol 29, 305–217.Bogacki M, Skarzynski DJ, Kotwica J, 2000: Non genomicaction of progesterone <strong>in</strong> luteal and endometrial epithelialcells <strong>in</strong> cattle. ESDAR Newslett 5, 23.(+)(–) (–)(–)(+)(+)(+)(+)Fig. 5. Schematic presentation of factors <strong>in</strong>volved <strong>in</strong> the growth,ma<strong>in</strong>tenance and regression of the CL (see text for the details)(+)Boiti C, Zerani M, Zamp<strong>in</strong>i D, Gobbetti A, 2000: Nitric oxidesynthase activity and progesterone release by isolatedcorpora lutea of rabbits <strong>in</strong> the early and mid-luteal phasesof pseudopregnancy are modulated differently by PGE 2 andPGF 2a via AC and PKC. J Endocr<strong>in</strong>ol 164, 179–186.Cannon MJ, Petroff MG, Pate JL, 2003: Effects of PGF 2a andprogesterone on the ability of bov<strong>in</strong>e luteal cells to stimulateT lymphocyte proliferation. Biol Reprod 69, 695–700.Duras M, Brzosko E, Kotwica J, 2005: Influence of progesterone,pregnenolone and 17b-hydroxy-progesterone on thefunction of bov<strong>in</strong>e luteal cells treated with lute<strong>in</strong>iz<strong>in</strong>ghormone, noradrenal<strong>in</strong>e and PGE 2 . Pol J Vet Sci 8, 113–119.Ferreira-Dias GM, Skarzynski DJ, 2008: Some aspects ofregulation of luteal function and luteolysis <strong>in</strong> equ<strong>in</strong>e corporalutea. Pferdeheikunde 24, 10–14.Ford SP, Chenault JR, 1981: Blood flow to the corpus luteumbear<strong>in</strong>govary and ipsilateral uter<strong>in</strong>e horn of cows dur<strong>in</strong>goestrous cycle and early pregnancy. J Reprod Fertil 62, 555–562.Friedman A, Weiss S, Levy N, Meidan R, 2000: Role of TNFaand its type-1 receptor <strong>in</strong> luteal regression: <strong>in</strong>duction ofprogrammed cell death <strong>in</strong> bov<strong>in</strong>e corpus luteum-derivedendothelial cells. Biol Reprod 63, 1905–1915.G<strong>in</strong>ther OJ, Silva LA, Araujo RR, Beg MA, 2007: Temporalassociations among pulses of 13,14-dihydro-15-keto-PGF 2a ,luteal blood flow, and luteolysis <strong>in</strong> cattle. Biol Reprod 76,506–513.Girsh E., Milvae RA, Wang W, Meidan R, 1996: Effect ofendothel<strong>in</strong>-1 on bov<strong>in</strong>e luteal cell function: role <strong>in</strong> PGF 2a -<strong>in</strong>duced antisteroidogenic action. Endocr<strong>in</strong>ology 137, 1306–1312.Hansel W, Dowd JP, 1986: New concepts of the control ofcorpus luteum function. J Reprod Fert 78, 755–768.Henricks DM, Long JT, Hill JR, 1974: The effect of PGF 2adur<strong>in</strong>g various stages of the ooestrous cycle of beef heifers.J Reprod Fertil 41, 113–120.Jaroszewski JJ, Hansel W, 2000: Intraluteal adm<strong>in</strong>istration ofa nitric oxide synthase blocker stimulates progesterone andoxytoc<strong>in</strong> secretion and prolongs the life span of the bov<strong>in</strong>ecorpus luteum. Proc Soc Exp Biol Med 224, 50–55.Jaroszewski JJ, Skarzynski DJ, Blair RM, Hansel W, 2003a:Influence of nitric oxide on the secretory function of thebov<strong>in</strong>e corpus luteum: dependence on cell composition andcell-to-cell communication. Exp Biol Med 228, 741–748.Jaroszewski JJ, Skarzynski DJ, Hansel W, 2003b: Nitric oxideas a local mediator of prostagland<strong>in</strong> F 2a -<strong>in</strong>duced regression<strong>in</strong> bov<strong>in</strong>e corpus luteum: an <strong>in</strong> vivo study. Exp Biol Med228, 1057–1062.Juengel JL, Garverick HA, Johnson AL, Youngquist RS,Smith MF, 1993: Apoptosis dur<strong>in</strong>g luteal regression <strong>in</strong>cattle. Endocr<strong>in</strong>ology 132, 249–254.Kim L, Weems YS, Bridges PJ, LeaMaster BR, Ch<strong>in</strong>g L,V<strong>in</strong>cent DL, Weems CW, 2001: Effects of <strong>in</strong>domethac<strong>in</strong>,lute<strong>in</strong>iz<strong>in</strong>g hormone, PGE 2 , trilostane, mifepristone, ethamoxytriphetolon secretion of PGE 2 , PGF 2a and progesteroneby ov<strong>in</strong>e corpora lutea of pregnancy or the estrous cycle.Prostagland<strong>in</strong>s 63, 189–203.Klipper E, Gilboa T, Levy N, Kisliouk T, Soanel-Borowski K,Meidan R, 2004: Characterization of endothel<strong>in</strong>-1 and nitricoxide generat<strong>in</strong>g systems <strong>in</strong> corpus luteum-derived endothelialcells. <strong>Reproduction</strong> 128, 463–473.Kobayashi S, Acosta TJ, Ozawa T, Hayashi K, Berisha B,Ohtani M, Schams D, Miyamoto A, 2002: Intraluteal releaseof angiotens<strong>in</strong> II and progesterone <strong>in</strong> vivo dur<strong>in</strong>g corporalutea development <strong>in</strong> the cow: effect of vasoactive peptides.Biol Reprod 66, 174–179.Korzekwa A, Jaroszewski JJ, Bogacki M, Deptua KM,Mas´lanka TS, Acosta TJ, Okuda K, Skar_zyn´ski DJ, 2004:Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


64 DJ Skarzynski, G Ferreira-Dias and K OkudaEffects of prostagland<strong>in</strong> F 2a and nitric oxide on the secretoryfunction of bov<strong>in</strong>e luteal cells. J Reprod Dev 50, 411–417.Korzekwa A, Shuko M, Jaroszewski J, Izabela Wocawek-Potocka, Okuda K, Skarzynski DJ, 2006: Nitric oxide<strong>in</strong>duces programmed cell dead <strong>in</strong> the bov<strong>in</strong>e corpus luteum:mechanism of action. J Reprod Dev 52, 353–361.Lei ZM, Cheg<strong>in</strong>i N, Rao Ch V, 1991: Quantitative cellcomposition of human and bov<strong>in</strong>e corpora lutea fromvarious reproductive states. Biol Reprod 44, 1148–1156.Levy N, Kobayashi S, Roth Z, Wolfenson D, Miyamoto A,Meidan R, 2000: Adm<strong>in</strong>istration of PGF 2a dur<strong>in</strong>g the earlybov<strong>in</strong>e luteal phase does not alter the expression of ET-1and its type A receptors: a possible cause for corpus luteumrefractor<strong>in</strong>ess. Biol Reprod 63, 377–382.Li H, Wallerath T, Forstermann U, 2002: Physiologicalmechanisms regulat<strong>in</strong>g the expression of endothelial-typeNO synthase. Nitric Oxide 7, 132–147.Lohse MJ, 1993: Molecular mechanisms of membrane receptordesensitization. Biochim Biophys Acta 1179, 171–188.Mamluk R, Levy N, Ruead B, Davis JS, Meidan R, 1999:Characterization and regulation of type A endothel<strong>in</strong>receptor gene expression <strong>in</strong> bov<strong>in</strong>e luteal cells types.Endocr<strong>in</strong>ology 40, 2110–2116.McCracken JA, Custer EE, Lamsa JC, 1999: Luteolysis: aneuroendocr<strong>in</strong>e-mediated event. Physiol Rev 79, 263–323.Meidan R, Milvae RA, Weiss S, Levy N, Friedman A, 1999:Intra-ovarian regulation of luteolysis. J Reprod Fertil Suppl54, 217–228.Meidan R, Levy N, Kisliouk T, Podlovny L, Rusiansky M,Klipper E, 2005: The y<strong>in</strong> and yang of corpus luteum-derivedendothelial cells: Balanc<strong>in</strong>g life and death. Domest AnimEndocr<strong>in</strong>ol 29, 318–328.Miyamoto A, Shirasuna K, Wijayagunawardane MP, WatanabeS, Hayashi M, Yamamoto D, Matsui M, Acosta TJ,2005: Blood flow: a key regulatory component of corpusluteum function <strong>in</strong> the cow. Domest Anim Endocr<strong>in</strong>ol 29,329–339.Nagata S, 1997: Apoptosis by death factor. Cell 88, 355–365.Nett TM, McClellan MC, Niswender GD, 1976: Effects ofprostagland<strong>in</strong>s on the ov<strong>in</strong>e corpus luteum: blood flow,secretion of progesterone and morphology. Biol Reprod 15,66–78.Neuvians TP, Schams D, Berisha B, Pfaffl MW, 2004:Involvement of pro-<strong>in</strong>flammatory cytok<strong>in</strong>es, mediators of<strong>in</strong>flammation, and basic fibroblast growth factor <strong>in</strong> PGF 2a -<strong>in</strong>duced luteolysis <strong>in</strong> bov<strong>in</strong>e corpus luteum. Biol Reprod 70,473–480.Nishimura R, Sakumoto R, Tatsukawa Y, Acosta T, OkudaK, 2007: Oxygen concentration is an important factor formodulat<strong>in</strong>g progesterone synthesis <strong>in</strong> bov<strong>in</strong>e corpus luteum.Endocr<strong>in</strong>ology 147, 4273–4280.Nishimura R, Komiyama J, Tasaki Y, Acosta T, Okuda K,2008: Hypoxia promotes luteal cell death <strong>in</strong> bov<strong>in</strong>e corpusluteum. Biol Reprod 78, <strong>in</strong> press.Niswender GD, Reimers TJ, Diekman MA, Nett TM, 1976:Blood flow: a mediator of ovarian function. Biol Reprod 14,64–81.Niswender GD, Juengel JL, Silva PJ, Rollyson MK, McIntushEW, 2000: Mechanisms controll<strong>in</strong>g the function and lifespan of the corpus luteum. Physiol Rev 80, 1–29.Niswender GD, Davies TL, Griffith RJ, Bogan RL, Monser K,Bott RC, Bruemmer JE, Nett TM, 2007: Judge, jury andexecutioner: the auto-regulation of luteal function. ReprodDom Rum<strong>in</strong>ants VII; Soc Reprod Fertil 64, 191–206.Ohtani M, Kobayashi S, Miyamoto A, Hayashi K, Fukui Y,1998: Real-time relationships between <strong>in</strong>traluteal andplasma concentrations of endothel<strong>in</strong>, oxytoc<strong>in</strong>, and progesteronedur<strong>in</strong>g PGF 2a -<strong>in</strong>duced luteolysis <strong>in</strong> the cow. BiolReprod 58, 103–108.Okuda K, Sakumoto R, Uenoyama Y, Berisha B, MiyamotoA, Schams D, 1999: TNFa receptors <strong>in</strong> microvascularendothelial cells from bov<strong>in</strong>e corpus luteum. Biol Reprod61, 1017–1022.Okuda K, Korzekwa A, Shibaya M, Murakami S, NishimuraR, Tsubouchi M, Woclawek-Potocka I, Skarzynski DJ,2004: Progesterone is a suppressor of apoptosis <strong>in</strong> bov<strong>in</strong>eluteal cells. Biol Reprod 71, 2065–2071.Pate JL, 1988: Regulation of prostagland<strong>in</strong>s synthesis byprogesterone <strong>in</strong> the bov<strong>in</strong>e corpus luteum. Prostagland<strong>in</strong>s36, 303–315.Pate JL, 1996: Intercellular communication <strong>in</strong> the bov<strong>in</strong>ecorpus luteum. Theriogenology 45, 1381–1397.Pate JL, 2003: Lives <strong>in</strong> the balance: responsiveness of thecorpus luteum to uter<strong>in</strong>e and embryonic signals. <strong>Reproduction</strong>61(Suppl), 207–217.Pate JL, Landis Keyes P, 2001: Immune cells <strong>in</strong> the corpusluteum: friends or foes? <strong>Reproduction</strong> 122, 665–676.Penny LA, Armstrong D, Bramley TA, Webb R, Coll<strong>in</strong>s RA,Watson ED, 1999: Immune cells and cytok<strong>in</strong>e production <strong>in</strong>the bov<strong>in</strong>e corpus luteum throughout the ooestrous cycleand after <strong>in</strong>duced luteolysis. J Reprod Fertil 115, 87–96.Petroff MG, Petroff BK, Pate JL, 2001: Mechanisms ofcytok<strong>in</strong>e-<strong>in</strong>duced death of cultured bov<strong>in</strong>e luteal cells.<strong>Reproduction</strong> 121, 753–760.Rae MT, Menzis GS, McNeilly AS, Woad K, Webb R,Bramley TA, 1998: Specific non-genomic, membrane-localizedb<strong>in</strong>d<strong>in</strong>g sites for progesterone <strong>in</strong> the bov<strong>in</strong>e corpusluteum. Biol Reprod 58, 1394–1406.Reynolds L, Redmer D, 1999: Growth and development of thecorpus luteum. J Reprod Fertil Suppl 54, 181–191.Rosiansky-Sultan M, Klipper E, Spanel-Borowski K, MeidanR., 2006: Inverse relationship between nitric oxide synthasesand endothel<strong>in</strong>-1 synthesis <strong>in</strong> bov<strong>in</strong>e corpus luteum: <strong>in</strong>teractionsat the level of luteal endothelial cell. Endocr<strong>in</strong>ology147, 5228–5235.Rueda BR, Tilly KI, Hansen TR, Hoyer PB, Tilly JL, 1995:Expression of superoxide dismutase, catalase and glutathioneperoxidase <strong>in</strong> the bov<strong>in</strong>e corpus luteum: evidencesupport<strong>in</strong>g a role for oxidative stress <strong>in</strong> luteolysis. Endocr3, 227–232.Rueda BR, Tilly KI, Hansen TR, Jolly PD, Hoyer PB, TillyJL, 1997: Increased Bax and <strong>in</strong>terleuk<strong>in</strong>-1b-convert<strong>in</strong>genzyme messenger RNA levels co<strong>in</strong>cide with apoptosis <strong>in</strong>the corpus luteum dur<strong>in</strong>g structural regression. Biol Reprod56, 186–193.Rueda BR, Hendry IR, Hendry WJ III, Fong HW, StormashakF, Slaydenlayden OD, Davis JS, 2000: Decreasedprogesterone level and progesterone receptors antagonistspromote apoptotic cell death <strong>in</strong> bov<strong>in</strong>e luteal cells. BiolReprod 62, 269–276.Sakamoto K, Miwa K, Ezashi T, Okuda-Ashitaka E, OkudaK, Houtani T, Sugimoto T, Ito S, Hayaishi O, 1995:Expression of mRNA encod<strong>in</strong>g the PGF 2a receptor <strong>in</strong>bov<strong>in</strong>e corpora lutea throughout the ooestrous cycle andpregnancy. J Reprod Fertil 103, 99–105.Sakumoto R, Berisha B, Kawate N, Schams D, Okuda K,2000: TNFa and its receptors <strong>in</strong> bov<strong>in</strong>e corpus luteumthroughout the oestrous cycle. Biol Reprod 62, 192–199.Sasahara K, Shirasuna K, Watanabe S, Asahi T, Nagai K,Miyamoto A, 2007: Nitric oxide stimulated by PGF 2a<strong>in</strong>duces the drastic <strong>in</strong>crease of luteal blood flow and has arole <strong>in</strong> functional and structural luteolysis. Biol Reprod 77(Sp. Iss.), 225–225 (Abstract 578).Schams D, Berisha B, 2004: Regulation of corpus luteumfunction <strong>in</strong> cattle – an overview. Reprod Dom Anim 39,241–251.Schams D, Berisha B, Neuvians T, Amselgruber W, KraetzlWD, 2003: Real-time changes of the local vasoactive peptideÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Regulation of Luteal Function 65systems (angiotens<strong>in</strong>, endothel<strong>in</strong>) <strong>in</strong> the bov<strong>in</strong>e corpusluteum after <strong>in</strong>duced luteal regression. Mol Reprod Dev65, 57–66.Shaw DW, Britt JH, 1995: Concentrations of TNFa and P4with<strong>in</strong> the bov<strong>in</strong>e corpus luteum sampled by cont<strong>in</strong>uousflowmicrodialysis dur<strong>in</strong>g luteolysis <strong>in</strong> vivo. Biol Reprod 53,847–854.Shirasuna K, Asaoka H, Acosta TJ, WijayagunawardaneMPB, Ohtani M, Hayashi M, Matsui M, Miyamoto A,2004: Real-time relationships <strong>in</strong> <strong>in</strong>traluteal release amongPGF 2a , endothel<strong>in</strong>-1, and angiotens<strong>in</strong> II dur<strong>in</strong>g spontaneousluteolysis <strong>in</strong> the cow. Biol Reprod 71, 1706–1711.Silva PJ, Juengel JL, Rollyson MK, Niswender GD, 2000:Prostagland<strong>in</strong> metabolism <strong>in</strong> the ov<strong>in</strong>e corpus luteum:catabolism of PGF 2a co<strong>in</strong>cides with resistance of the CLto PGF 2a . Biol Reprod 63, 1229–1236.Skarzynski DJ, Okuda K, 1999: Sensitivity of bov<strong>in</strong>e corporalutea to PGF 2a is dependent on progesterone, oxytoc<strong>in</strong> andprostagland<strong>in</strong>s. Biol Reprod 60, 1292–1298.Skarzynski DJ, Bogacki M, Kotwica J, 1997: Changes <strong>in</strong>ovarian OT secretion as an <strong>in</strong>dicator of corpus luteumresponse to PGF 2a treatment <strong>in</strong> cattle. Theriogenology 48,733–742.Skarzynski DJ, Kobayashi S, Okuda K, 2000: Influence ofnitric oxide and noradrenal<strong>in</strong>e on PGF 2a -<strong>in</strong>duced oxytoc<strong>in</strong>secretion and <strong>in</strong>tracellular calcium mobilization <strong>in</strong> culturedbov<strong>in</strong>e luteal cells. Biol Reprod 63, 1000–1005.Skarzynski DJ, Jaroszewski JJ, Okuda K, 2001: Luteotropicmechanisms <strong>in</strong> the bov<strong>in</strong>e corpus luteum: role of oxytoc<strong>in</strong>,PGF 2a , progesterone and noradrenal<strong>in</strong>e. J Reprod Dev 47,125–137.Skarzynski DJ, Bah MM, Wocawek-Potocka I, Deptua K,Korzekwa A, Shibaya M, Pilawski W, Okuda K, 2003a:Roles of TNFa <strong>in</strong> the regulation of the oestrous cycle <strong>in</strong>cattle: an <strong>in</strong> vivo study. Biol Reprod 69, 1907–1913.Skarzynski DJ, Jaroszewski JJ, Bah MM, Deptua KM, BarszczewskaB, Gawronska B, Hansel W, 2003b: Adm<strong>in</strong>istrationof a nitric oxide synthase <strong>in</strong>hibitor counteracts PGFF 2a -<strong>in</strong>duced luteolysis <strong>in</strong> cattle. Biol Reprod 68, 1674–1681.Skarzynski DJ, Jaroszewski JJ, Okuda K, 2005: Role of TNFaand nitric oxide <strong>in</strong> luteolysis <strong>in</strong> cattle. Domest AnimEndocr<strong>in</strong>ol 29, 340–346.Skarzynski DJ, Woclawek-Potocka I, Korzekwa A, Bah MM,Piotrowska KK, Barszczewska B, Okuda K, 2007: Infusionof exogenous TNF dose dependently alters the length ofthe luteal phase <strong>in</strong> cattle: differential responses to treatmentwith <strong>in</strong>domethac<strong>in</strong> and a nitric oxide synthase <strong>in</strong>hibitor(L-NAME). Biol Reprod 76, 619–627.Sug<strong>in</strong>o N, Telleria CM., Gibori G, 1997: Progesterone <strong>in</strong>hibits20a-HSD expression <strong>in</strong> the rat corpus luteum through theglucocorticoid receptor. Endocr<strong>in</strong>ology 138, 1497–1500.Taniguchi H, Yokomizo Y, Okuda K, 2002: Fas-fas ligandsystem mediates luteal cell death <strong>in</strong> bov<strong>in</strong>e corpus luteum.Biol Reprod 66, 754–759.Towson DH, O’Connor CL, Pru JK, 2002: Expression ofMCP-1 and distribution of immune cells populations <strong>in</strong> thebov<strong>in</strong>e corpus luteum through the oestrous cycle. BiolReprod 66, 361–366.Tsai S-J, Wiltbank MC, 1998: PGF 2a regulates dist<strong>in</strong>ctphysiological changes <strong>in</strong> early and mid-cycle bov<strong>in</strong>e corporalutea. Biol Reprod 58, 346–352.Vonnahme KA, Wilson ME, Li Y, Rupnow HL, PhernettonTM, Ford SP, Magness RR, 2005: Circulat<strong>in</strong>g levels ofnitric oxide and vascular endothelial growth factor throughoutov<strong>in</strong>e pregnancy. J Physiol-Lond 565, 101–109.Watanebe S, Shirasuna K, Matsui M, Yamamoto D, BerishaB, Schams D, Miyamoto A, 2006: Effect of <strong>in</strong>traluteal<strong>in</strong>jection of endothel<strong>in</strong> type A receptor antagonist onPGF 2a -<strong>in</strong>duced luteolysis <strong>in</strong> the cow. J Reprod Dev 52,551–559.Weems YS, Lewis AW, Randel RD, Weems CW, 2002: Effectsof PGE 2 and PGF 2a , trilostane, mifepristone, palmitic acid(PA), <strong>in</strong>domethac<strong>in</strong>, ethamoxytriphetol, PGE 2 + PA, orPGF 2a + PA on PGE 2 , PGF 2a , and progesterone secretionby bov<strong>in</strong>e corpora lutea of mid-pregnancy <strong>in</strong> vitro. Ch<strong>in</strong> JPhysiol 45, 163–168.Weems YS, Randel RD, Tatman S, Lewis AW, NeuendorffDA, Weems CW, 2004: Effects of estrous synchronizationon the response to nitric oxide donors, nitric oxide synthase<strong>in</strong>hibitors, and endothel<strong>in</strong>-1 <strong>in</strong> vitro. Prostagland<strong>in</strong> OtherLipid Med 74, 45–59.Wiltbank MC, Gallagher KP, Christensen AK, Brabec RK,Keyes PL, 1990: Physiological and immunocytochemicalevidence for new concept of blood flow regulation <strong>in</strong> corpusluteum. Biol Reprod 42, 139–149.Wiltbank MC, Shiao TF, Bergfelt DR, G<strong>in</strong>ther OJ, 1995:PGF 2a receptors <strong>in</strong> the early bov<strong>in</strong>e corpus luteum. BiolReprod 52, 74–78.Wright MF, Sayre B, Keith Inskeep EK, Flores JA, 2001:PGF 2a regulation of the bov<strong>in</strong>e corpus luteum endothel<strong>in</strong>system dur<strong>in</strong>g the early and midluteal phase. Biol Reprod65, 1710–1717.Author’s address (for correspondence): DJ Skarzynski, Department ofReproductive Immunology, Institute of Animal <strong>Reproduction</strong> andFood Research of PAS, 10-747 Olsztyn, Poland. E-mail: skadar@pan.olsztyn.plConflict of <strong>in</strong>terest: All authors declare no conflict of <strong>in</strong>terests.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 66–73 (2008); doi: 10.1111/j.1439-0531.2008.01144.xISSN 0936-6768Captive Breed<strong>in</strong>g of Cheetahs <strong>in</strong> South Africa – 30 Years of Data from the de WildtCheetah and Wildlife CentreHJ Bertsch<strong>in</strong>ger 1 , DGA Meltzer 2 and A van Dyk 31 Section of <strong>Reproduction</strong>, Department of Production Animal Studies, Faculty of Veter<strong>in</strong>ary Science, University of Pretoria, Pretoria, South Africa;2 Emeritus Prof., Brooklyn, Pretoria, Gauteng Prov<strong>in</strong>ce, South Africa; 3 de Wildt Cheetah and Wildlife Trust, de Wildt, North West Prov<strong>in</strong>ce, SouthAfricaContentsThe de Wildt Cheetah and Wildlife Centre was established <strong>in</strong>1971 and the first cheetah cubs were born <strong>in</strong> 1975. Dur<strong>in</strong>g theperiod 1975–2005, 242 litters were born with a total of 785cubs. Mean cub survival from 1 to 12 months and greater than12 months of age was 71.3 and 66.2%, respectively. Themajority of losses (84.9%) occurred dur<strong>in</strong>g the first monthpostpartum whereas only 15.1% deaths took place between 1and 12 months of age. Females were first bred at an age ofapproximately 3 years, reached maximum reproductive age at6–8 years, where after fertility decl<strong>in</strong>ed. Males reached peakreproduction at 6 and ma<strong>in</strong>ta<strong>in</strong>ed this for up to 12 years ofage. Male fertility was best correlated with sperm morphology.Dur<strong>in</strong>g recent years, for practical purposes, males wereallocated to ‘good’ (‡70% normal), ‘fair’ (40–70% normal)and ‘poor’ (


Captive Breed<strong>in</strong>g of Cheetahs <strong>in</strong> South Africa 67Fig. 1. Layout of the camps at thede Wildt Cheetah and WildlifeCentre. C = quarant<strong>in</strong>e; D =walkway ⁄ service road; E =camps for weaned cubs; G =camps for subadults; F = breed<strong>in</strong>gcamps for females; M =maternity section with<strong>in</strong> eachbreed<strong>in</strong>g camp. Insert shows anexample of a maternity sectionwith den for cubs and triangularfeed<strong>in</strong>g area with crush for catch<strong>in</strong>gand restra<strong>in</strong><strong>in</strong>g cheetahs andperform<strong>in</strong>g m<strong>in</strong>or procedures suchas collection of blood samplesAnnual selection of breed<strong>in</strong>g animalsSelection based on pedigree and genetic markersEvery year, prior to the ma<strong>in</strong> breed<strong>in</strong>g period (November–June),potential males and females are selectedaccord<strong>in</strong>g to pedigree and, from 2007 onwards, based onarea of orig<strong>in</strong> and an unique breed<strong>in</strong>g value calledWEDS (Oliehoek et al. 2006) that maximizes diversity.In addition, microsatellite genetic marker-based analysisof the de Wildt population has allowed us to select malesand females that represent the major breed<strong>in</strong>g l<strong>in</strong>es andclarify orig<strong>in</strong> and relatedness between selected breeders.This has been done based on cluster analysis (Pritchardet al. 2000) and genetic marker-based relatedness(Lynch and Ritland 1999; Wang 2002) thus m<strong>in</strong>imiz<strong>in</strong>g<strong>in</strong>breed<strong>in</strong>g levels as much as possible.Selection of males on the basis of their fertilityThe males that have been selected as described above arethen submitted to a breed<strong>in</strong>g soundness exam<strong>in</strong>ation. Acomplete spermiogram is performed us<strong>in</strong>g phase contrastmicroscopy (Meltzer et al. 1998). In addition tothis, the animal’s behaviour towards females is established.Males show<strong>in</strong>g aggressive behaviour towardsfemales are not suitable. Dur<strong>in</strong>g the last 12 years, asimplified classification for the selection of breed<strong>in</strong>gmales has been used at de Wildt. Males are classified<strong>in</strong>to ‘good’ (‡70% normal), ‘fair’ (40–70% normal) and‘poor’ (


68 HJ Bertsch<strong>in</strong>ger, DGA Meltzer and A van Dykcollected from six cheetah females two to three times perweek. The blood samples were assayed for progesteroneand 17b-oestradiol (Bertsch<strong>in</strong>ger et al. 1984).ResultsLitters and cubs produced 1975–2005Dur<strong>in</strong>g the period 1975–2005, 242 litters were born witha total of 785 cubs. It should be noted that <strong>in</strong> some yearssmall numbers of females were bred because of a limiteddemand. In 1982 as a result of a bush fire on theproperty no breed<strong>in</strong>g was attempted. Mean gestationperiod was 93 days. Pseudopregnancy, which is shorterthan pregnancy, occurred sporadically. Mean cub survivalfrom 1 to 12 months and greater than 12 monthsof age was 71.3 and 66.2%, respectively. The majority oflosses (84.9%) occurred dur<strong>in</strong>g the first month postpartumwhereas only 15.1% of deaths took placebetween 1 and 12 months of age.Female effectsBreed<strong>in</strong>g ageFigure 2 shows the age specific birth rate of 55 litters <strong>in</strong>cheetah females at de Wildt for the years between 1975and 1987 (Meltzer 1987). From this it is evident thatcheetah females started breed<strong>in</strong>g when 2.5–3 years ofage but only reached maximum reproductive capabilitywhen 6 years old. First and even second litter femaleswere commonly poor mothers and often abandoned orate their newly born cubs. Maximum reproductive ratewas ma<strong>in</strong>ta<strong>in</strong>ed until the age of eight after which itdecl<strong>in</strong>ed. This was the result of <strong>in</strong>creased morbidity rate<strong>in</strong> females with diseases such as chronic renal failure andgastritis.Seasonal distribution of mat<strong>in</strong>g at de WildtAt de Wildt the practice was to breed cheetahs dur<strong>in</strong>gthe months November–February (Fig. 3). This maycreate the impression that the species is a seasonalbreeder. Births occurred from the end of March andcont<strong>in</strong>ued until the beg<strong>in</strong>n<strong>in</strong>g of June reflect<strong>in</strong>g theduration of the breed<strong>in</strong>g period.Fig. 3. Monthly distribution of conceptions of cheetah at de Wildtthat resulted <strong>in</strong> births »93 days later (Meltzer 1987)Oestrus cycleThe presence of males <strong>in</strong> the walk-way between thefemale camps appeared to stimulate oestrous cycles <strong>in</strong>females. Oestrus was suspected when males accumulatedat the female’s enclosure. Females abandoned theirsecretive behaviour and moved towards males <strong>in</strong> thewalk-way and ur<strong>in</strong>ated close to the fence. Interactionbetween animals gives rise to overt signs of oestrus.They made chirp<strong>in</strong>g sounds <strong>in</strong> response to the stuttercalls of the males; tail lift<strong>in</strong>g and roll<strong>in</strong>g was seen. Theoestrus cycle length <strong>in</strong> six cheetahs that were allowed tocycle before they were bred ranged from 10 to 21 days.A typical progesterone and oestradiol profile observed<strong>in</strong> one of these females that was mated dur<strong>in</strong>g the secondoestrus of the observation period is shown <strong>in</strong> Fig. 4(Bertsch<strong>in</strong>ger et al. 1998). Though difficult to determ<strong>in</strong>eother than by the period of receptivity, the length ofoestrus varied from 1 to 3 days. These figures agreefavourably with those reported for the domestic cat(Wildt et al. 1981). Oestradiol plasma concentrationsshowed cyclical peaks, which lasted approximately12 days. Progesterone plasma concentration only rosesubsequent to mat<strong>in</strong>g mean<strong>in</strong>g that cheetahs, like someof the other members of the cat family, is an <strong>in</strong>ducedFig. 2. Number of litters (total = 55) produced accord<strong>in</strong>g to age offemales <strong>in</strong> years from 1975–1987 at de Wildt (Meltzer 1987)Fig. 4. Oestradiol and progesterone blood profiles dur<strong>in</strong>g two oestrouscycles (cheetah Jean) also show<strong>in</strong>g laparoscopic appearance of afollicle (F) and two corpora lutea (CLs). She was mated dur<strong>in</strong>g thesecond oestrus after which a sharp rise <strong>in</strong> progesterone can be seen(Bertsch<strong>in</strong>ger et al. 1998).Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Captive Breed<strong>in</strong>g of Cheetahs <strong>in</strong> South Africa 69ovulator (Wildt et al. 1980). Plasma progesterone concentrationsare now used rout<strong>in</strong>ely to confirm mat<strong>in</strong>g.The laparoscopic exam<strong>in</strong>ations supported the f<strong>in</strong>d<strong>in</strong>gsof the oestrus observations as well as the hormonalpatterns (Fig. 4; Bertsch<strong>in</strong>ger et al. 1984).Induction of oestrusSome females that did not show spontaneous heatdur<strong>in</strong>g the breed<strong>in</strong>g period were <strong>in</strong>duced for naturalmat<strong>in</strong>g us<strong>in</strong>g a modification of the regimen described byHoward et al. (1992). Serum progesterone concentrationswere measured to ensure basal levels before<strong>in</strong>ductions were attempted. On day 0, 200 IU eCG[Folligon, Intervet SA (Pty) Ltd, Isando, South Africa]was <strong>in</strong>jected and followed by 100 IU hCG (Chorulon,Folligon, Intervet SA (Pty) Ltd., Isando, South Africa)72 h later. A male was <strong>in</strong>troduced <strong>in</strong>to the females’enclosures soon after the hCG-<strong>in</strong>jection. Of the 11females treated 5 (46%) were mated and showed a rise <strong>in</strong>plasma progesterone. Four (36%) gave birth to livecubs. Progesterone concentrations rema<strong>in</strong>ed basal <strong>in</strong> the6 (56%) unmated females.Heterozygosity of de Wildt cheetahs and heritability offemale fertility and cub mortalityThe de Wildt captive population had a mix of ancestryfrom Namibia and South Africa and had lower levels of<strong>in</strong>breed<strong>in</strong>g than free-rang<strong>in</strong>g cheetahs analysed(Wright’s Fst values – 0. 019 vs 0.13, respectively; SPSasidharan, unpublished data). These f<strong>in</strong>d<strong>in</strong>gs wouldseem to be logical, as <strong>in</strong> the wild, with very lowpopulation densities, the degree of <strong>in</strong>breed<strong>in</strong>g should begreater due to the restricted availability of potentialpartners. The heritability for litter size was high at0.5848 (SE 0.078) <strong>in</strong> the de Wildt pedigree (532 progeny,1975–2007; Sasidharan, unpublished data). The maternalheritability for cub mortality was estimated to be0.596 (SE 0.131). As <strong>in</strong> the wild (Caro 1994), cubsurvival was not affected by season.Male effectsBreed<strong>in</strong>g agePotential breed<strong>in</strong>g males were first exam<strong>in</strong>ed at an ageof 3.5–4 years. The proportion of males found to haveacceptable semen quality (‘good’ and ‘fair’ categories)<strong>in</strong>creased from 55.6% (n = 18) to 71.5% (n = 14) <strong>in</strong>the age categories 3.5 to 4.5 to


70 HJ Bertsch<strong>in</strong>ger, DGA Meltzer and A van DykTable 1. Summary characteristics of fresh cheetah semen samples(n = 160)pHVolume(ml)% Livesperm% Sperm withl<strong>in</strong>ear motilitySpermcount · 10 6 ⁄ ml% NormalspermRange 6.4–8.0 0.3–2.1 10–95 8–90 1–211 0–86Mean 0.7 65.2 58.1 32.7 40.3SD 0.5 18.5 18.6 36.13 17.5Table 2 lists the more common sperm defects ofcheetahs. Abnormal heads, knobbed acrosomes(Fig. 7e, f), mid-piece and flagellar defects were themost common morphological abnormalities of cheetahsperm. It is accepted that a normal population ofcheetah sperm may vary moderately as far as head size isconcerned (pleiomorphism; Wildt et al. 1983, 1987).Compared to the sperm of species such as bulls, ramsand boars, cheetah sperm heads are very small and notas flat (Fig. 7b,c). Orientation on the slide probablyexacerbates the pleiomorphic appearance. Flagellardefects, (Fig. 7d,h,i), depend<strong>in</strong>g on their <strong>in</strong>cidence, hada marked affect on l<strong>in</strong>ear sperm motility and generallyspeak<strong>in</strong>g good motility was associated with at least fairmorphology. Where testicular function was severelyimpaired, the <strong>in</strong>cidence of spermatogenic cells was often<strong>in</strong>creased (Fig. 7j).Figure 8 shows the relationship between sperm morphologyand number of litters born at de Wildt(Bertsch<strong>in</strong>ger and Meltzer 1998). As the % normalsperm <strong>in</strong>creased so did the number of litters. As can beexpected, compared to % normal sperm, an <strong>in</strong>verserelationship between % major sperm defects and littersborn was seen. The low number of births on the lefthandside for % major defects and on the right for %normal sperm reflects the small numbers of male cheetahwith good sperm morphology. Approximately 30% ofall males tested fell <strong>in</strong>to the ‘poor’ category (see below).Semen quality of wild-caught males appeared to be nobetter than that of captive-bred animals. Outbred freerang<strong>in</strong>gsouthern African lions and leopards, by comparison,display good sperm morphology and the sizeand shape of sperm heads, which are flatter, areconsistent (Fig. 7k).Dur<strong>in</strong>g the period 1998–2007, 113 semen exam<strong>in</strong>ationswere carried out. The number and percentage ofmales fall<strong>in</strong>g <strong>in</strong>to the ‘good’, ‘fair’ and ‘poor’ categorieswere 41 (36.3%), 27 (23.9%) and 45 (39.8%), respectively.Twenty-six males were exam<strong>in</strong>ed over 2 years ormore dur<strong>in</strong>g the period of 1998–2006. Of these, n<strong>in</strong>eshowed an improvement by one or more categories, n<strong>in</strong>erema<strong>in</strong>ed the same and seven showed a decrease <strong>in</strong>semen quality. Deterioration was often associated withdiseases such as gastritis. We analysed the litter datafrom 1999 to 2007 (exclud<strong>in</strong>g 2002) <strong>in</strong> terms of spermmorphology categories we use at de Wildt. The numberof litters produced and average litter sizes for categories‘good’, ‘fair’ and ‘poor’ males were 21 and 3.44, 18 and3.14 and 18 and 2.28, respectively. The differences <strong>in</strong>litter size were not significant (p = 0.1449; Kruskall–Wallis test).DiscussionThe breed<strong>in</strong>g results achieved at de Wildt over the last31 years prove that cheetah can be bred successfully <strong>in</strong>captivity. The success, however, is dependent on thecorrect hous<strong>in</strong>g, day to day management and breed<strong>in</strong>gmanagement. Other centres such as the Cheetah Projectat Kapama and the Wassenaar Wildlife Breed<strong>in</strong>g Centre<strong>in</strong> The Netherlands have also bred captive cheetahssuccessfully. The breed<strong>in</strong>g strategy at Wassenaar, whichhas resulted <strong>in</strong> 210 cubs born from 62 litters, is worthdescrib<strong>in</strong>g as it differs a little from the one practiced atde Wildt. Briefly, male and female utilize the same camp<strong>in</strong> turns which means that they pick up the scents of theopposite sex (Beekman et al. 1997; Louwman andLouwman 2005). The behaviour of the cheetahs isobserved carefully and once the male and female show<strong>in</strong>tense <strong>in</strong>terest <strong>in</strong> the scents left beh<strong>in</strong>d contact throughthe fence of adjo<strong>in</strong><strong>in</strong>g enclosures is allowed. When thefemale displays signs of oestrus she is given access to themale enclosure and left with him for approximately 48 hdur<strong>in</strong>g which two to five mat<strong>in</strong>gs occur.From our earlier work (Meltzer 1987; Bertsch<strong>in</strong>gerand Meltzer 1998) a relationship was seen betweensperm morphology and number of litters born at deWildt. Despite poor semen quality, we showed morerecently (1999–2007) that some males are capable ofproduc<strong>in</strong>g litters albeit with a smaller average litter size(2.28 vs 3.14 and 3.44). A number of factors, however,may affect semen quality. Semen collected by means ofelectro-stimulation does not necessarily reflect the truepotential of a particular male. Firstly, ur<strong>in</strong>e contam<strong>in</strong>ationaffects especially motility but maybe even tailHead defects Mid-piece defects Tail defectsTable 2. Common defects of cheetahspermKnobbed ⁄ lipped acrosome Aplasia of mitochondrial helix Mid-piece reflexLoose acrosome a Pseudodroplet Bent mid-pieceMacrocephalic with s<strong>in</strong>gle or double tail Stump-tail Coiled tail – coiled belowor on top of the headMicrocephalicProximal dropletImmature headDistal dropletDegenerated or vacuolated headFree tail forms with aplasiaof the mitochondrial helix bPyriformDeformed headsLoose heada This defect, if the sperm is otherwise normal, was not added to the % abnormal sperm.b This defect should merely be noted and not added to the % abnormal sperm. It arises dur<strong>in</strong>g meiosis and is associatedwith macrocephalic heads <strong>in</strong> the ejaculate.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Captive Breed<strong>in</strong>g of Cheetahs <strong>in</strong> South Africa 71(a)(b)(c) (d) (e) (f)(g)(h)(i)Fig. 7. (a) Morphologically normalcheetah sperm (SEM) – MP =mitochondrial sheath, PP = pr<strong>in</strong>cipalpiece, E P = end piece; (b)lateral view of cheetah sperm head(SEM) – A = acrosome, CP =connect<strong>in</strong>g piece, MS = mitochondrialsheath; (c) planar view ofcheetah sperm head (SEM); (d)SEM show<strong>in</strong>g cheetah sperm withcoiled tails and bent midpiece; (e)lateral view of cheetah sperm withacrosomal defect (TEM) – A =acrosome, AC = acrosomal cyst,PC = proximal centriol; (f) lateralview of cheetah sperm with acrosomaldefect (SEM) – AC =acrosomal cyst, MP = mitochondrialsheath; (g) phase contrastmicrograph (PCM) of an underdevelopedcheetah sperm with anucleus and cytoplasm; (h) twocoiledtails and a ‘Dag’ defect <strong>in</strong>cheetah sperm (PCM); (i) Cheetahsperm with bent midpieces (PCM);(j) three (arrows) spermatogeniccells with vacuoles <strong>in</strong> a semensmear; (k) typical head morphologyof normal lion sperm (PCM)(a–f: Coubrough et al. 1976, 1978;Coubrough and Soley 1982)(j)(k)morphology. Secondly, semen evaluation reflects aw<strong>in</strong>dow <strong>in</strong> time and may take place as many as4 months prior to actual mat<strong>in</strong>g of a female. Thirdly,semen quality, especially sperm count, is likely to beaffected by prior sexual activity. Accord<strong>in</strong>g to Amannand Almquist (1976) the daily sperm production <strong>in</strong> dairybulls is 11 million ⁄ g testicular tissue. Cheetahs havesmall testes with a mean comb<strong>in</strong>ed volume of 8 ml(n = 28; »8 g), mean<strong>in</strong>g that the daily sperm productionis limited to approximately 88 million per day. Inaddition the epididymal tails are small with a limitedcapacity for sperm storage. The mean number of spermper cheetah ejaculate was found to be 22.9 million,which is approximately 26% of daily estimated production.Nevertheless, semen evaluation rema<strong>in</strong>s the mostvaluable criterium for selection of males. An <strong>in</strong>vestigation<strong>in</strong>to poor fertility <strong>in</strong> a pair of Gir forest lions at theZurich Zoological gardens illustrates the importance ofsemen quality and fertility (Bertsch<strong>in</strong>ger et al. 2006a).Asian lions are known to less heterozygous than mostfree-rang<strong>in</strong>g African lions (Gaur et al. 2006) and Wildtet al. (1987b) showed that <strong>in</strong>breed<strong>in</strong>g of large catscauses poor semen quality. The Asian lioness <strong>in</strong> questionproduced her last cubs <strong>in</strong> July 1998 and then had eightpresumed pseudopregnant cycles (35–74 days <strong>in</strong> length)and one anovulatory cycle (16 days long) before sheproduced cubs 27 months later. The male was exam<strong>in</strong>eddur<strong>in</strong>g the same month that she conceived and found toÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


72 HJ Bertsch<strong>in</strong>ger, DGA Meltzer and A van DykFig. 8. Number of litters sired <strong>in</strong> relation to % normal sperm andmajor sperm defects (Bertsch<strong>in</strong>ger and Meltzer 1998)have only 12% normal sperm (80.9% major defects) butwith good motility (75% l<strong>in</strong>ear) and total sperm countof 390 million. Another factor that needs to be evaluatedis the heritability of male fertility or semen quality<strong>in</strong> cheetah.Given the fact that natural breed<strong>in</strong>g can be highlysuccessful, <strong>in</strong> our op<strong>in</strong>ion, there seems to be little<strong>in</strong>dication for the use of assisted reproductive techniques<strong>in</strong> captive cheetahs. In any case, reports of success <strong>in</strong>cheetahs and other large cats with assisted reproductivetechniques are scant, despite the fact that the firstreports date back to 1992 (Howard et al. 1992). Artificial<strong>in</strong>sem<strong>in</strong>ation and <strong>in</strong> vitro fertilization have notimproved fertility and certa<strong>in</strong>ly cannot <strong>in</strong>crease the rateof reproduction or reduce the generation <strong>in</strong>terval. Whilethey may be of use for exchange of genetic material andcontrol of spread of diseases, their role for cheetahconservation is limited.The future of cheetah conservation, as for most if notall wildlife species, will depend on availability of habitatand control of illegal hunt<strong>in</strong>g and trad<strong>in</strong>g with wildcaughtcheetahs. Accord<strong>in</strong>g to Marnewick et al. (2007)there were 650 cheetahs <strong>in</strong> 44 captive facilities <strong>in</strong> SouthAfrica <strong>in</strong> 2006. Eleven of these facilities were activelybreed<strong>in</strong>g cheetahs. Exclud<strong>in</strong>g 2000, the average annualnumber of cheetah exported legally from South Africafrom 1996 to 2002 was 30.5. In 2000, 129 cheetahs wereexported. The most likely source for the majority of theanimals exported <strong>in</strong> 2000 is from the wild. As far asillegal hunt<strong>in</strong>g or kill<strong>in</strong>g of cheetahs is concerned thereport <strong>in</strong>dicates that <strong>in</strong> the Thabazimbi district of theLimpopo Prov<strong>in</strong>ce alone, 26 cheetahs were killed over a3-year period from 1999 to 2001. The ma<strong>in</strong> reason forthe kill<strong>in</strong>g of cheetahs is that farmers perceive them as athreat to their stock - both wild and domestic stock. Inorder to protect or save free-rang<strong>in</strong>g cheetahs <strong>in</strong> nonprotectedareas, the de Wildt Cheetah and Wildlife Trustestablished the de Wildt Wild Cheetah Project <strong>in</strong> 2000.The goals were to establish numbers of free-rang<strong>in</strong>gcheetahs outside of protected areas, assist farmers <strong>in</strong>trapp<strong>in</strong>g cheetahs <strong>in</strong> areas where they were not wanted,relocate such animals to fenced game reserves and toeducate farmers and communities about the need forcarnivore conservation <strong>in</strong> general. The project has beenmost successful and many farmers now see cheetahs asan asset rather than a liability. Accord<strong>in</strong>g to Marnewicket al. (2007), the first cheetahs were caught <strong>in</strong> 2000 andby December 2006, 137 had been removed fromfarmlands. Sixteen of these could not be relocatedbecause of serious <strong>in</strong>juries susta<strong>in</strong>ed before or at thetime of capture. N<strong>in</strong>ety-two animals could be f<strong>in</strong>allyreleased (58 males and 33 females) <strong>in</strong>to areas rang<strong>in</strong>gfrom 1500 to 70 000 ha <strong>in</strong> size. The first cubs were born<strong>in</strong> 2002 and by August 2007 94 cubs (average litter size3.9 cubs) had been born to 23 females. The data showthat, given the habitat and opportunity, cheetah reproducewell <strong>in</strong> the wild.The ma<strong>in</strong> role of captive breed<strong>in</strong>g of cheetahs <strong>in</strong>South Africa should be to curtail illegal trade <strong>in</strong> wildcheetahs. Captive-bred animals from de Wildt have alsobeen successfully released <strong>in</strong>to the wild (Pettifer 1981).Three 5-year-old males were released onto a game farm<strong>in</strong> the Hoedspruit area of the Limpopo Prov<strong>in</strong>ce andhunted spontaneously despite be<strong>in</strong>g born <strong>in</strong> captivityand not hav<strong>in</strong>g been ‘taught’ to hunt. They wereeventually recaptured because of their lack of fear forhumans. This latter problem can be overcome if cubs areraised out of contact with humansConclusionsThe de Wildt Cheetah and Wildlife Centre and othershave shown that cheetahs can be bred successfully and<strong>in</strong> a susta<strong>in</strong>ed manner <strong>in</strong> captivity. In order to achievethis, the correct breed<strong>in</strong>g management needs to beapplied. In some smaller South Africa reserves it hasalso become necessary to contracept cheetahs (eithermales or females) to prevent <strong>in</strong>breed<strong>in</strong>g. Deslorel<strong>in</strong>implants (Suprelor<strong>in</strong> Ò , Peptech Animal Health, Sydney,NSW, Australia) provide a safe and reversible methodof contraception for both sexes (Betsch<strong>in</strong>ger et al.2002a; Bertsch<strong>in</strong>ger et al. 2006b). Zoos or sanctuariesthat do not have the facilities, capacity or the necessarycritical mass <strong>in</strong> terms of cheetah numbers, should rathernot attempt breed<strong>in</strong>g. Live animal displays are animportant part of public education and can make avaluable contribution to conservation of a species likethe cheetah. It is advisable to consider tak<strong>in</strong>g animals onloan for display purposes <strong>in</strong> zoos and safari parks. Thesecould be pre-breed<strong>in</strong>g age animals, males with poorsemen quality or animals that are past their prime. It isalso clear that populations <strong>in</strong> zoos and smaller gamereserves need to be managed genetically <strong>in</strong> order toreduce the risks of <strong>in</strong>breed<strong>in</strong>g depression. In order to dothis, a standardized approach should be adopted so that,<strong>in</strong>ternationally, valid comparisons can be made.ReferencesAmann RP, Almquist JO, 1976: Bull management to maximizesperm output. Proceed<strong>in</strong>gs of 6th Conference on ArtificialInsem<strong>in</strong>ation and <strong>Reproduction</strong>, Columbia, pp. 1–10.Beekman SPA, de Wit M, Louman J, Louman H, 1997:Breed<strong>in</strong>g and observations on the behaviour of cheetah(Ac<strong>in</strong>onyx jubatus) at the Wassenaar Wildlife Breed<strong>in</strong>gCentre. Int Zoo YB 35, 43–50.Bertsch<strong>in</strong>ger HJ, Meltzer DGA, 1998: <strong>Reproduction</strong> <strong>in</strong> malecheetahs. 2: Sperm morphology. In: Penzhorn BL(ed.),Proceed<strong>in</strong>gs of a Symposium on Cheetahs as Game RanchÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Captive Breed<strong>in</strong>g of Cheetahs <strong>in</strong> South Africa 73<strong>Animals</strong>. Onderstepoort, Republic of South Africa, WildlifeGroup, South African Veter<strong>in</strong>ary Association, Onderstepoort,23–24 October, pp. 153–158.Bertsch<strong>in</strong>ger HJ, Meltzer DGA, van Dyk A, Coubrough RI,Soley JT, Collett FA, 1984: Cheetah life-l<strong>in</strong>e. Nuclear Active30, 2–7.Bertsch<strong>in</strong>ger HJ, Meltzer DGA, van Dyk A, Strachan A, 1998:Breed<strong>in</strong>g female cheetahs <strong>in</strong> captivity. In: Penzhorn BL(ed.),Proceed<strong>in</strong>gs of a Symposium on Cheetahs as Game Ranch<strong>Animals</strong>. Onderstepoort, 23–24 October, pp. 168–174.Bertsch<strong>in</strong>ger HJ, Isenbu¨ gel E, Jannett F, Ossent P, Wild P,2006a: Infertility <strong>in</strong> a pair of Indian lions <strong>in</strong> Zurich Zoo.Proceed<strong>in</strong>gs of the Hungarian Society of Animal <strong>Reproduction</strong>,Budapest, 26–27 June, pp. 44–46.Bertsch<strong>in</strong>ger HJ, Jago M, No¨ thl<strong>in</strong>g JO, Human A, 2006b:Repeated use of the GnRH analogue deslorel<strong>in</strong> to downregulatereproduction <strong>in</strong> male cheetahs (Ac<strong>in</strong>onyx jubatus).Theriogenology 66, 1762–1767.Bertsch<strong>in</strong>ger HJ, No¨ thl<strong>in</strong>g JO, Nard<strong>in</strong>i RM, Hemmelder S,Broekhuisen MH, 2002b: Collection of semen <strong>in</strong> cheetahs(Ac<strong>in</strong>onyx jubatus) us<strong>in</strong>g electro-ejaculation: attempts toavoid ur<strong>in</strong>e contam<strong>in</strong>ation. Adv Ethol 37, 122.Betsch<strong>in</strong>ger HJ, Trigg TE, Jo¨ chle W, Human A, 2002a:Induction of contraception <strong>in</strong> some African wild carnivoresby down-regulation of LH and FSH secretion us<strong>in</strong>g theGnRH analogue deslorel<strong>in</strong>. <strong>Reproduction</strong> Suppl 60, 41–52.Brown JL, Wildt DE, Wielebnowski N, Goodrowe KL,Graham LH, Wells S, Howard JG, 1996: Reproductiveactivity <strong>in</strong> captive female cheetahs (Ac<strong>in</strong>onyx jubatus)assessed by faecal steroids. J Reprod Fertil 106, 337–346.Caro TM, 1994: Cheetahs of the Serengeti Pla<strong>in</strong>s: GroupLiv<strong>in</strong>g <strong>in</strong> and Asocial Species. The University of ChicagoPress, Chicago and London.Coubrough RI, Bertsch<strong>in</strong>ger HJ, Soley JT, Meltzer DGA,1976: Some aspects of normal and abnormal spermatozoa <strong>in</strong>cheetah (Ac<strong>in</strong>onyx jubatus). Proc Elect Microscopy SocSouthern Africa 6, 5–6.Coubrough RI, Bertsch<strong>in</strong>ger HJ, Soley JT, 1978: Scann<strong>in</strong>gelectron microscopic studies on cheetah spermatozoa. ProcElectron Microscopy Soc Southern Africa 8, 57–58.Eaton RL, 1970: Notes on the reproductive biology of thecheetah. Int Zoo YB 10, 86–89.Gaur A, Shailaja K, S<strong>in</strong>gh A, 2006: Twenty polymorphicmicrosatellite markers <strong>in</strong> the Asiatic lion (Panthera leopersica). Conserv Gene 7, 1005–1008.Howard JG, Donoghue AM, Barone MA, Goodrowe KL,Blumer ES, Snodgrass K, Starnes D, Tucker M, Bush M,Wildt DE, 1992: Successful <strong>in</strong>duction of ovarian activity andlaparoscopic <strong>in</strong>trauter<strong>in</strong>e artificial <strong>in</strong>sem<strong>in</strong>ation <strong>in</strong> the cheetah(Ac<strong>in</strong>onux jubatus). J Zoo Wildl Med 23, 288–300.Kelly MJ, Laurenson MK, FitzGibbon CD, Coll<strong>in</strong>s DA,Durant SM, Frame GW, Bertram BCR, Caro TM, 1998:Demography of the Serengeti cheetah (Ac<strong>in</strong>onyx jubatus)population: the first 25 years. J Zoo London 244, 473–488.Labuschagne W, 1979: ‘n Bio-ekologiese en gedragstudie vandie jagluiperd, Ac<strong>in</strong>onyx jubatus jubatus (Schreber, 1776).MSc Thesis, University of Pretoria.Louwman JWW, Louwman JCM, 2005: Cheetah breed<strong>in</strong>gprogram at Wassenaar Wildlife Breed<strong>in</strong>g Centre. AnimKeeper’s Forum 7 ⁄ 8, 368–369.Lynch M, Ritland K, 1999: Estimation of pairwise relatednesswith molecular markers. Genetics 152, 1753.Marker-Kraus L, Grisham J, 1993: Captive breed<strong>in</strong>g of cheetahs<strong>in</strong> North American zoos: 1987–1991. Zoo Biol 12, 5–18.Marnewick K, Beckhell<strong>in</strong>g A, Cilliers D, Lane E, Mills G,Herr<strong>in</strong>g K, Caldwell P, 2007: The status of the cheetah <strong>in</strong>South Africa. In: Breitenmoser C and Durant S (eds.), Thestatus and conservation needs of the Cheetah <strong>in</strong> SouthernAfrica. Cat News Special Edition, 22–31.Meltzer DGA, 1987: <strong>Reproduction</strong> <strong>in</strong> male cheetah, Ac<strong>in</strong>onyxjubatus jubatus (Schreber, 1776). MSc Dissertation (Pretoria),p. 118.Meltzer DGA, Mu¨ lders MS, 1998: <strong>Reproduction</strong> <strong>in</strong> malecheetahs: III Reproductive hormones. In: Penzhorn BL(ed.), Proceed<strong>in</strong>gs of a Symposium on Cheetahs as GameRanch <strong>Animals</strong>. Onderstepoort, 23–24 October, pp. 159–167.Meltzer DGA, Bertsch<strong>in</strong>ger HJ, van Dyk A, 1998: <strong>Reproduction</strong><strong>in</strong> male cheetahs: I. Breed<strong>in</strong>g management and semenevaluation. In: Penzhorn BL (ed.), Proceed<strong>in</strong>gs of a Symposiumon Cheetahs as Game Ranch <strong>Animals</strong>. Onderstepoort,23–24 October, pp. 145–152.O’Brien SJ, Roelke ME, Marker L, Newman A, W<strong>in</strong>kler CA,Meltzer DA, Colly L, Everman JF, Bush M, Wildt DE,1985: Genetic basis for species vulnerability <strong>in</strong> the cheetah.Science 227, 1428–1434.O’Brien SJ, Wildt DE, Bush M, 1986: The cheetah <strong>in</strong> geneticperil. Sci Am 254, 84–92.Oliehoek PA, W<strong>in</strong>dig JJ, van Arendonk JA, Bijma P, 2006:Estimat<strong>in</strong>g relatedness between <strong>in</strong>dividuals <strong>in</strong> general populationswith a focus on their use <strong>in</strong> conservation programs.Genetics 173, 483–496.Pettifer HHL, 1981: Experimental release of captive-bredcheetah (Ac<strong>in</strong>onyx jubatus) <strong>in</strong>to the natural environment.In: Chapman JA, Punsman P(eds), Worldwide FurbearerConference Proceed<strong>in</strong>gs, Donnelly, VA, pp. 1–9.Pritchard JK, Stephens M, Donnelly P, 2000: Inference ofpopulation structure us<strong>in</strong>g multilocus genotype data. Genetics155, 945–959.Sarri KJ, 1994: Estrous behaviour of the female cheetah(Ac<strong>in</strong>onyx jubatus) and the male cheetahs’ response to anestrous female. In: Caro TM (ed.), Cheetahs of the SerengetiPla<strong>in</strong>s: Group Liv<strong>in</strong>g <strong>in</strong> and Asocial Species. The Universityof Chicago Press, Chicago and London.Ulmer FA, 1957: Cheetahs are born. America’s First Zoo 9, 7.Wang J, 2002: An estimator for pairwise relatedness us<strong>in</strong>gmolecular markers. Genetics 160, 1203–1215.Wildt DE, Seager SWJ, Chakraborty PK, 1980: Effect ofcopulatory stimuli on <strong>in</strong>cidence of ovualtion and serumlute<strong>in</strong>iz<strong>in</strong>g hormone <strong>in</strong> the cat. Endocr<strong>in</strong>ology 107, 1212–1217.Wildt DE, Chan SY, Seager SWJ, Chakraborty PK, 1981:Ovarian activity, circulat<strong>in</strong>g hormones and sexual behaviour<strong>in</strong> the cat. I. Relationships dur<strong>in</strong>g coitus-<strong>in</strong>duced lutealphase and the oestrus period without mat<strong>in</strong>g. Biol Reprod25, 15–28.Wildt DE, Bush M, Howard JG, O’Brien SJ, Meltzer D, VanDijk A, Ebedes H, Brand DJ, 1983: Unique sem<strong>in</strong>al quality<strong>in</strong> the South African cheetah and a comparative evaluation<strong>in</strong> the domestic cat. Biol Reprod 29, 1019–1025.Wildt DE, O’Brien SJ, Howard JG, Caro TM, Roelke ME,Brown JL, Bush M, 1987a: Similarity <strong>in</strong> ejaculate-endocr<strong>in</strong>echaracteristics <strong>in</strong> captive versus free-rang<strong>in</strong>g cheetahs of twosubspecies. Biol Reprod 36, 351–360.Wildt DE, Bush M, Goodrowe KL, Packer C, Pusey AE,Brown JL, Josl<strong>in</strong> P, O’Brien SJ, 1987b: Reproductive andgenetic consequences of found<strong>in</strong>g isolated lion populations.Nature 329, 328–31.Author’s address (for correspondence): HJ Bertsch<strong>in</strong>ger, Section of<strong>Reproduction</strong>, Department of Production Animal Studies, Faculty ofVeter<strong>in</strong>ary Science, University of Pretoria, Private Bag X04, SouthAfrica. E-mail: henk.bertsch<strong>in</strong>ger@up.ac.zaConflict of <strong>in</strong>terest: All authors declare no conflict of <strong>in</strong>terests.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 74–82 (2008); doi: 10.1111/j.1439-0531.2008.01145.xISSN 0936-6768Non-<strong>in</strong>vasive Monitor<strong>in</strong>g of Hormones: A Tool to Improve <strong>Reproduction</strong> <strong>in</strong>Captive Breed<strong>in</strong>g of the Eurasian LynxHormone Monitor<strong>in</strong>g <strong>in</strong> Breed<strong>in</strong>g Programmes of MammalsM Dehnhard 1 , S Naidenko 2 , A Frank 1 , B Braun 1 ,FGo¨ritz 1 and K Jewgenow 11 Leibniz-Institute for Zoo Biology and Wildlife Research, Berl<strong>in</strong>, Germany; 2 A.N. Severtzov Institute of Ecology and Evolution, Moscow, RussiaContentsThe survival of many critical endangered mammal species isoften depend<strong>in</strong>g on successful captive breed<strong>in</strong>g programmeswhich <strong>in</strong>clude the future option of re<strong>in</strong>troduction to the wild.Breed<strong>in</strong>g <strong>in</strong> captivity also demands the application of modernassisted reproductive techniques to ensure maximal biodiversity,but knowledge on reproductive physiology is oftenlimited. Therefore, non-<strong>in</strong>vasive monitor<strong>in</strong>g of ur<strong>in</strong>ary andfaecal hormones has become an important tool for reproductivemanagement. To exemplify the importance of non<strong>in</strong>vasivehormone monitor<strong>in</strong>g, we choose the Eurasian lynxas a model for the world’s most endangered felid species, theIberian lynx. We analysed faecal samples of pregnant andpseudo-pregnant female Eurasian lynxes dur<strong>in</strong>g a 3-year studyperiod. Compared to pre-mat<strong>in</strong>g levels faecal progesteronemetabolite profiles revealed a tendency towards higher levels <strong>in</strong>pregnant and pseudo-pregnant females with no differencebetween both categories. Oestrogen levels raised <strong>in</strong> bothpregnant and pseudo-pregnant females with a tendency to bemore elevated and prolonged <strong>in</strong> pregnant females. Surpris<strong>in</strong>glyboth E2 and P4 metabolites were highly correlated (r 2 =0.8131, p < 0.0001) show<strong>in</strong>g a postpartum <strong>in</strong>crease both <strong>in</strong>pregnant and pseudo-pregnant females. The results from theEurasian lynx revealed that the measurement of faecalprogesterone metabolites led to profiles dissimilar to profilesshown <strong>in</strong> other felid species, but similar to those from faecalgestagen metabolite analysis <strong>in</strong> the Iberian lynx. To identifyfaecal gestagen and oestrogen metabolites a radio-metabolismstudy was performed. Us<strong>in</strong>g the progesterone immunoassaytwo major progesterone metabolites were detected demonstrat<strong>in</strong>gthat the assay <strong>in</strong>deed tracks the relevant metabolites.The oestrogen assay measured authentic 17b-oestradiol andoestrone, and their conjugates. The analysis of the faecalmetabolite composition <strong>in</strong> samples from early and latepregnancy and lactation particularly revealed a dist<strong>in</strong>ct shift<strong>in</strong> the relation between 17b-oestradiol and oestrone thatchanged <strong>in</strong> favour of oestrone. This might <strong>in</strong>dicate differenthormone sources dur<strong>in</strong>g and after pregnancy (corpus luteum,placenta). We hypothesize, that placental steroid analysis <strong>in</strong>comb<strong>in</strong>ation with other highly sophisticated analytical techniques,like liquid chromatography mass spectrometry orur<strong>in</strong>ary relax<strong>in</strong> analysis may led to analytical options toconfirm pregnancy and to differentiate this from pseudopregnancy<strong>in</strong> lynx species.IntroductionThe ma<strong>in</strong> threat of mammalian species is human activityand many <strong>in</strong> situ populations are critically endangeredbecause of landscape development has reduced andfragmented their habitat (Ceballos et al. 2005; Cardilloet al. 2006). Due to the rapid decl<strong>in</strong>e of the species, it isapparent that some mammal populations will alreadynot be able to recover on its own. Therefore, successfulcaptive breed<strong>in</strong>g programmes of managed zoo populationsmight result <strong>in</strong> animals that could be returned topart of their orig<strong>in</strong>al range under protected regimes asdescribed for several species; notably the Arabian andscimitar horned oryx (Mesoch<strong>in</strong>a et al. 2003) and thePrzewalski horse (Ryder 1993). To achieve these objectivesnew approaches to improve the success of mammalconservation is urgently required and captive populationshave become of prime importance for the conservationof species <strong>in</strong> terms of research and education.Concomitantly there is a high priority <strong>in</strong> ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g or<strong>in</strong>creas<strong>in</strong>g genetic diversity <strong>in</strong> small populations to avoid<strong>in</strong>breed<strong>in</strong>g (Earnhardt et al. 2004). Thus, successfulcaptive breed<strong>in</strong>g programmes of endangered speciesmust be performed on scientific basis and depend moreand more on modern assisted reproduction techniques(ART). The last 20 years are characterized by sporadicapplication of artificial <strong>in</strong>sem<strong>in</strong>ation, gamete bank<strong>in</strong>gand ⁄ or embryo production <strong>in</strong> non-domestic species(Masui et al. 1989; Hermes et al. 2001, 2007; Penfoldet al. 2005; Pukazhenthi et al. 2006; Swanson 2006;Hildebrandt et al. 2007; Stoops et al. 2007; Swansonet al. 2007). However, the most strik<strong>in</strong>g limitation ofus<strong>in</strong>g ART <strong>in</strong> breed<strong>in</strong>g programmes is limited knowledgeon reproduction biology, <strong>in</strong> particular knowledgeon hormone regulation of female reproductive cycle andpregnancy. Therefore, the purpose of the first part of thepaper is to give a brief outl<strong>in</strong>e on the current state of theart regard<strong>in</strong>g non-<strong>in</strong>vasive hormone monitor<strong>in</strong>g techniques.Hormones control reproductive success. Know<strong>in</strong>g ananimal’s hormone patterns allows understand<strong>in</strong>g itsbiology, and diagnos<strong>in</strong>g and correct<strong>in</strong>g <strong>in</strong>fertility. Whilemany hormones <strong>in</strong>teract <strong>in</strong> the reproductive process, thesteroid hormones oestradiol, progesterone and testosteroneare the most important regulators of reproductivebehaviours and functions across vertebrates. They areproduced by ovaries and testes, and their concentration<strong>in</strong> blood serum is used to validate reproductive activityof mammals. S<strong>in</strong>gle blood samples, which are usuallyavailable from captive animals, however, can hardly beused to assess reproductive status of an exotic species.Hormone values depend on many factors, like seasonality,reproductive silence, reproductive suppression,pseudo-pregnancy, which are simply not known orstudied before <strong>in</strong> several exotic species.Presum<strong>in</strong>g the knowledge of a particular reproductivepattern of a mammalian species, the analysis ofÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Non-<strong>in</strong>vasive Monitor<strong>in</strong>g of Hormones 75hormones is essential to assist husbandry of animals <strong>in</strong>captivity. These <strong>in</strong>formation’s contributed to the reproductivesuccess by <strong>in</strong>creas<strong>in</strong>g the understand<strong>in</strong>g ofreproductive cycl<strong>in</strong>g and breed<strong>in</strong>g behaviour (Moreiraet al. 2001; Paris et al. 2002; Cavigelli et al. 2003;Pereira et al. 2006), <strong>in</strong>vestigations on reproductiveseasonality (Scheibe et al. 1999; Moor<strong>in</strong>g et al. 2004;Jewgenow et al. 2006), the establishment of successfulbreed<strong>in</strong>g pairs <strong>in</strong> captivity (Heistermann et al. 2004),improvement of oestrous synchronization (Penfold et al.2005), and ovulation <strong>in</strong>duction protocols with gonadotrophicreleas<strong>in</strong>g hormone (GnRH) agonists to achievethe goals of successful artificial <strong>in</strong>sem<strong>in</strong>ation and tocorrect <strong>in</strong>fertility (Asa et al. 2006; Graham et al. 2006).Another important factor to be considered is theknowledge of hormone produc<strong>in</strong>g organs <strong>in</strong> a particularspecies. Beside gonads, the placenta <strong>in</strong> pregnant females,and also the adrenal glands are capable of produc<strong>in</strong>gsteroids. Stress caused by catch<strong>in</strong>g and restra<strong>in</strong><strong>in</strong>gbefore blood sampl<strong>in</strong>g, together with venipuncturemight activate the HPA axis and thus confound theeffects of the experimental treatment. Repeated bloodsampl<strong>in</strong>g might also <strong>in</strong>duce anticipatory stress reactionsof the animals <strong>in</strong>volved. To avoid this, dur<strong>in</strong>g the lastdecades methods have been developed to measuresteroid hormones non-<strong>in</strong>vasively. Collected ur<strong>in</strong>e andfaecal samples have become a substitute for analys<strong>in</strong>ghormones <strong>in</strong> the serum or plasma (see review Schwarzenberger2007). Steroid hormones are extensivelymetabolized <strong>in</strong> various organs, ma<strong>in</strong>ly <strong>in</strong> the liver andare excreted via the bile <strong>in</strong>to the gut and via the kidney<strong>in</strong>to the ur<strong>in</strong>e. These metabolites are the source for non<strong>in</strong>vasivemethods of measur<strong>in</strong>g an animal’s endocr<strong>in</strong>estatus. Therefore, what was once considered a daily‘waste product’ now has become a valuable ‘researchresource’, a virtual pool of biological <strong>in</strong>formation onreproductive cyclicity, the time of ovulation, pregnancyand impend<strong>in</strong>g parturition. Particularly excit<strong>in</strong>g arerecent advances to measure glucocorticoid metabolitesas <strong>in</strong>dicators of stress, which can be used to help enrichzoo environments, thereby improv<strong>in</strong>g reproduction andanimal well-be<strong>in</strong>g.Despite the enormous progress of us<strong>in</strong>g faecal andur<strong>in</strong>e hormone analysis for research on reproductionbiology of exotic species, each analytical method needsto be validated each time for a particular species and aparticular hormone. Steroid metabolism by the liver aswell as microbial impact dur<strong>in</strong>g the <strong>in</strong>test<strong>in</strong>al passage,and re-absorbtion <strong>in</strong>to the enterohepatic circulationgenerate a vast number of faecal steroid metabolites, yetdifferent <strong>in</strong> even closely related species (Palme et al.1996; Heistermann et al. 2006).This necessary species-specific approach depends onthe determ<strong>in</strong>ation of the major faecal hormone metabolite(s)of a steroid. The easiest way for identification ofthese metabolites is a radio-metabolism study, which<strong>in</strong>clude the <strong>in</strong>fusion of radiolabelled steroids and detectionof radioactivity with<strong>in</strong> faecal or ur<strong>in</strong>e hormoneextracts. The disadvantage of radio<strong>in</strong>fusion studies isthat they typically require special permits and equipmentregard<strong>in</strong>g animal care facilities and the generationof radioactive waste, but it should be performed whenever possible. S<strong>in</strong>ce animals <strong>in</strong> captivity are idealresearch subjects and such a study is considered to besafe (level of radioactivity is clearly below the maximumpermitted level), zoos had to be encouraged to performradiometabolism studies as often as possible to contributeto the <strong>in</strong>creas<strong>in</strong>g data base of steroid hormonemetabolites applicable for monitor<strong>in</strong>g reproductivephysiology of exotic species.Currently, different techniques are applied fromresearchers worldwide lead<strong>in</strong>g to diverse concentrationsbetween studies regard<strong>in</strong>g their absolute concentrationsreferred to faecal wet and dry weight, respectively. Thema<strong>in</strong> reason is that different antibodies often generatedaga<strong>in</strong>st the parent steroid were used with unknowncross-reactivities for the relevant steroid metabolites.Even commercially available antibodies advertised ashormone specific have been used with success for faecalsteroid analysis <strong>in</strong> several studies (Brown et al. 1996;Wasser et al. 2000). Nevertheless, comparability existsbetween their physiologic outcome, demonstrat<strong>in</strong>g thatfaecal and ur<strong>in</strong>ary steroid metabolites are reliable<strong>in</strong>dicators of gonadal and adrenal activity whilst themolecular structures of the metabolites rema<strong>in</strong>edunknown <strong>in</strong> most cases. This so called biologicalvalidation of a non-<strong>in</strong>vasive hormone measurementmust be performed before a particular assay can beapplied for monitor<strong>in</strong>g reproductive activity <strong>in</strong> a valuableanimal.The purpose of the second part of this paper is todemonstrate how non-<strong>in</strong>vasive hormone techniques canbe used to monitor reproduction <strong>in</strong> a non-domesticmammal species and which steps had to be performedbefore it can be successfully applied for breed<strong>in</strong>grout<strong>in</strong>e. We choose the Eurasian lynx as a model forbasic research and assay development. Although hormonemetabolites are widely described and validated formonitor<strong>in</strong>g reproduction and pregnancies <strong>in</strong> female wildcats (Brown et al. 1994, 2001; Graham et al. 2005), theexample of the Eurasian lynx will show that validationof hormone metabolites must be performed for each<strong>in</strong>dividual species.Materials and Methods<strong>Animals</strong>Female Eurasian lynx were housed at the scientific fieldstation ‘Tchernogolovka’ of the A.N. Severtzov Institute,situated 50 km north-east of Moscow. Faecalsamples were collected once a month from <strong>in</strong>dividualfemales and stored at )20°C with<strong>in</strong> 1 h after defecationuntil analyses. From February to April (prospectivemat<strong>in</strong>g season) the frequency of collection was <strong>in</strong>creasedto one to two times a week.Faecal samples (0.5 g) were extracted for 30 m<strong>in</strong> byshak<strong>in</strong>g with 4.5 ml of 90% methanol. After centrifugation(15 m<strong>in</strong> at 1200 · g) the supernatant was transferred<strong>in</strong>to a new tube. Aliquots of the faecal extractswere subjected either to high-performance liquid chromatography(HPLC) analysis, or diluted 1 + 1 withwater and added directly to the respective steroid enzymeimmune assay (EIA). All hormone measurements werecarried out <strong>in</strong> duplicates to assess a coefficient ofvariation. The results were expressed as immunoreactivesteroid metabolites <strong>in</strong> lg ⁄ g of wet faeces.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


76 M Dehnhard, S Naidenko, A Frank, B Braun, F Go¨ ritz and K JewgenowRadiometabolism studyTo identify relevant progesterone metabolites, whichreflect the corpus luteum (CL) activity, a radiometabolismstudy was performed: for that purpose a solution(0.25 ml) conta<strong>in</strong><strong>in</strong>g 250 lCi [ 3 H] progesterone(70–110 Ci ⁄ mmol, TRK413; Amersham Bioscience,Freiburg, Germany) <strong>in</strong> ethanol was used. Sterile 0.9%NaCl solution (2.25 ml) was added to the radiolabelledsolution and the total volume was <strong>in</strong>jected <strong>in</strong>to thecephalic ve<strong>in</strong> of a 15-year-old female lynx. Prior to<strong>in</strong>jection, the animal was sedated by an i.m. <strong>in</strong>jectionwith a mixture of 3 ml of Rometar (2% solution ofXylaz<strong>in</strong>e hydrochloride) and 1 ml of 10% Ketam<strong>in</strong>ehydrochloride (both from Spofa, Prague, Czech Republic)correspond<strong>in</strong>g to a dosage 2.4 and 4 mg ⁄ kg bodyweight for Xylaz<strong>in</strong>e and Ketam<strong>in</strong>e, respectively.Follow<strong>in</strong>g radiolabel <strong>in</strong>jection, all voided faecal sampleswere collected separately for 4 days <strong>in</strong> plastic bagsfrom the cage and the floor of the enclosure immediatelyafter defecation and stored at )20°C. Aliquots of eachsample were extracted for progesterone and oestrogendeterm<strong>in</strong>ation and for radioactivity count<strong>in</strong>g. The secondsample, collected on day 2 after <strong>in</strong>jection, conta<strong>in</strong>edthe highest amount of radioactivity (89%) and it wasused for HPLC analyses. All radioactive count<strong>in</strong>g wasconducted <strong>in</strong> a Perk<strong>in</strong> Elmer MicroBeta Trilux counter(Perk<strong>in</strong> Elmer, Rodgau, Germany).Tritiated steroids are usually of very low radiotoxicitybecause of very low energy beta emission. The specificradio activity of <strong>in</strong>jected 3 H-steroids, however, is quitehigh, so the additional mass of these radioactivehormones will not substantially <strong>in</strong>crease the totalconcentration of hormone <strong>in</strong> the body. Furthermore,the amount of radiolabel used for this study is far belowmaximum permitted level. Nevertheless, <strong>in</strong> Germanyradiometabolism studies <strong>in</strong> animals require a priorpermission of the local Animal Experiment EthicalCommittee.HPLC analysis of metabolitesFor separation and characterization of faecal steroidmetabolites, 50 ll portions of faecal extracts were used.For gestagen metabolite analysis a reversed-phase UltrasepES100 ⁄ RP – 18 ⁄ 6 lm HPLC column (4 · 250 mm;Sepserv, Berl<strong>in</strong>, Germany) was used. Metabolites wereseparated with a methanol + water mixture (78 + 22)at a flow rate of 1 ml ⁄ m<strong>in</strong>. Fractions of 0.33 ml werecollected at 20 s <strong>in</strong>tervals and diluted with one volumeof water, before 20 ll of the fractions were added <strong>in</strong>tothe assay systems. The elution positions of authenticprogesterone (4-pregnen-3,20-dione; P4), 5a-pregnan-3,20-dione (DHP), 5a-pregnan-3b-ol-20-one (5a-P), 5bpregnan-3a,20a-diol(pregnanediol, PD) on this columnhad been previously determ<strong>in</strong>ed <strong>in</strong> separate HPLC runs.For faecal oestrogen metabolite separation an AllureBiphenyl 5 lm HPLC column (3.2 · 150 mm; Restek,Bad Homburg, Germany) was used. Metabolites wereseparated with a acetonitrile + water mixture (43 + 57)at a flow rate of 1 ml ⁄ m<strong>in</strong>. Fractions were collected andadded to the assay as described above. The elutionpositions of authentic 1,3,5(10)-oestratrien-3,17-one(oestrone), 1,3,5(10)-oestratrien-3,17a-diol (17a-oestradiol),and 1,3,5(10)-oestratrien-3,17b-diol (17b-oestradiol)on this column had been determ<strong>in</strong>ed <strong>in</strong> separateHPLC runs after their <strong>in</strong>jection.ProgesteroneProgesterone (P4) analyses were carried out with an<strong>in</strong>-house microtitre plate enzyme immunoassay asdescribed earlier (Go¨ ritz et al. 2001) us<strong>in</strong>g a commercialP4 antibody (Sigma P1922, raised <strong>in</strong> rats to progesterone)and 4-pregnen-3,20-dione-3-CMO-peroxidaselabel. The cross-reactivities to progest<strong>in</strong>s were asfollows: 4-pregnen-3,20-dione (progesterone), 100%;5a-pregnan-3,20-dione, 31%; 5a-pregnan-3b-ol-20-one,18%; 5-pregnen-3b-ol-20-one, 12%; 4-pregnen-3aol-20-one,4.2%; 0.05). Intra- and <strong>in</strong>ter-assay coefficients of variationfor two biological samples with low and highconcentrations were 5.0 and 5.1% (n = 10) and 13.7 and22.8% (n = 10), respectively.OestrogensFaecal oestrogen analyses were carried out also with an<strong>in</strong>-house microtitre plate enzyme immunoassay us<strong>in</strong>g apolyclonal antibody raised <strong>in</strong> rabbits to 1,3,5(10)-oestratrien-3,17b-diol-17-HS-BSA and 1,3,5(10)-oestratrien-3,17b-diol-17-HS-peroxidaselabel (Meyer et al.1997). The cross-reactivities to oestrogens were asfollows: 1,3,5(10)-oestratrien-3,17b-diol (17b-oestradiol)100%, 1,3,5(10)-oestratrien-3,17-one (oestrone) 100%,1,3,5(10)-oestratrien-3,17a-diol (17a-oestradiol) 66%,1,3,5(10)-oestratrien-3,16a,17b-triol (oestriol) 1.5%, and 0.05).Intra- and <strong>in</strong>ter-assay coefficients of variation for twobiological samples with low and high concentrationswere 5.8 and 12.3% (n = 10) and 17.0 and 9.7% (n =11), respectively.Results and DiscussionMonitor<strong>in</strong>g of faecal steroid hormonesWe analysed faecal samples of pregnant (n = 15),pseudo-pregnant (n = 7) female Eurasian lynxes dur<strong>in</strong>ga 3-year study period. Figure 1a,b shows the faecalprogesterone and oestrogen metabolite profiles <strong>in</strong> apregnant and a pseudopregnant female. There is atendency towards higher gestagen and oestrogen metaboliteconcentration dur<strong>in</strong>g pregnancy. However, nodist<strong>in</strong>ct difference between profiles from the pregnant(a) and the pseudo-pregnant (b) female were obta<strong>in</strong>ed.In addition, both steroid metabolites showed a postpartum<strong>in</strong>crease with no difference between the pregnantand the pseudo-pregnant female. Surpris<strong>in</strong>gly a highlyÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Non-<strong>in</strong>vasive Monitor<strong>in</strong>g of Hormones 77Fig. 1. Course of fecal progesteroneand oestrogen metabolites <strong>in</strong> apregnant (a) and pseudopregnant(b) Eurasian lynx. Samples werecollected over one year beg<strong>in</strong>n<strong>in</strong>gwith October (M: mat<strong>in</strong>g; P: parturition)significant (n = 310, r 2 = 0.8131, p < 0.0001)correlation between E2 and P4 metabolites wasobta<strong>in</strong>ed, when calculated for all females <strong>in</strong>volved <strong>in</strong>the study. Altogether, the faecal steroid metaboliteprofiles obta<strong>in</strong>ed here are untypical compared to temporalpatterns measured <strong>in</strong> other felids. In pregnantcheetahs and clouded leopards P4 metabolite concentrationsdropped to basel<strong>in</strong>e co<strong>in</strong>cident with parturitionwhereas the duration of elevated faecal P4 metabolitesdur<strong>in</strong>g pseudopregnancy was approximately half thatobserved dur<strong>in</strong>g pregnancy (Brown et al. 1994). Inaddition, as shown for both female lynxes, a dist<strong>in</strong>ctoestrogen peak at mat<strong>in</strong>g time was absent.The results from the Eurasian lynx revealed thatfaecal progesterone metabolites are a poor <strong>in</strong>dicator ofreproductive status <strong>in</strong> this species as they fail to showclear elevations follow<strong>in</strong>g copulation or dur<strong>in</strong>g pregnancy.Such unusual endocr<strong>in</strong>e profile conflicts withcourses reported from other felid species (Brown et al.1994, 2001), but is similar to faecal gestagen metaboliteanalysis <strong>in</strong> the Iberian lynx (Pelican et al. 2006).Faecal oestrogen and gestagen metabolites wereunreliable for oestrus and pregnancy diagnosis <strong>in</strong>Eurasian lynx, wherefore a radiometabolism study wasnecessary to determ<strong>in</strong>e whether our enzyme immunoassaywould ‘catch’ the relevant metabolites reflect<strong>in</strong>g thebiological active hormone. In addition a biologicalvalidation of the presumed luteal activity wasdemanded. It <strong>in</strong>cludes the characterization of immunoreactivemetabolites dur<strong>in</strong>g and after pregnancy, thedeterm<strong>in</strong>ation of blood serum hormones and, f<strong>in</strong>ally, anultrasound exam<strong>in</strong>ation to verify the functional activityof CL outside breed<strong>in</strong>g season.Radiometabolism studyTo identify the relevant progesterone metabolites, whichreflect CL activity <strong>in</strong> the Eurasian lynx, a radiometabolismstudy was performed. For the development oftechniques for faecal steroid analysis, experiments onthe metabolism of radiolabelled steroids have provided avaluable <strong>in</strong>sight <strong>in</strong>to the metabolism and the excretionof hormone metabolites via faeces and ur<strong>in</strong>e. After<strong>in</strong>jection of 3 H (tritiated) labelled hormone, the excretedmetabolites of a particular steroid can be analysed byHPLC.Figure 2a shows the distribution of radiolabelledprogesterone metabolites <strong>in</strong> a faecal extract of a femaleÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


78 M Dehnhard, S Naidenko, A Frank, B Braun, F Go¨ ritz and K JewgenowFig. 2. Elution profile of radiolabelledprogesterone metabolitesand immunoreactive progesteronemetabolites <strong>in</strong> a female Eurasianlynx. Faecal extracts chosen fromdays 18 and 36 of pregnancy and14 days p.p. were subjected toHPLC separation. Arrows <strong>in</strong>dicatethe elution positions of progesterone(4-pregnen-3,20-dione, P4),5a-pregnane-3,20-dione (DHP),5-pregnen-3b-ol-20-one (pregnenolon,PD), and 5a-pregnane-3bol-20-one(5a-P). For methodologicaldetails see Jewgenow et al.(2006). Radiometabolism studyand detection of immunoreactivitieswere carried out <strong>in</strong> differentanimalsEurasian lynx. When pass<strong>in</strong>g through the non-polar(reversed phase) column the metabolites are reta<strong>in</strong>edand separated based on differences <strong>in</strong> their polarity. Theextract of female lynx faeces is composed of four majorpolar radiolabelled gestagen metabolites were detectable<strong>in</strong> fractions 6–8, 10, 12–13, and 16–17. Only m<strong>in</strong>oramounts of radiolabelled progesterone metabolites weredetectable at positions of non-polar substances correspond<strong>in</strong>gto progesterone and one of the other gestagenstandards. This suggests that the circulat<strong>in</strong>g hormoneitself is merely present <strong>in</strong> m<strong>in</strong>or quantities <strong>in</strong> faeces.Usually the pattern of radiolabelled metabolites <strong>in</strong>HPLC-analysis and those of immunoreactive hormonemetabolites does not co<strong>in</strong>cide due to different immunologicalcross-reactivities of used commercial,custom-made or <strong>in</strong>-house antibodies, which are directedaga<strong>in</strong>st orig<strong>in</strong>al hormones or known derivates. It isextremely important to consider that the peak seen <strong>in</strong> anHPLC immunogram does not reflect the quantity offaecal hormone metabolites but is a result of %crossreactivity of the antibody together with the amountof metabolite <strong>in</strong> a particular HPLC fraction. In contrastto that, the radioactivity with<strong>in</strong> a fraction directlyreflects the quantitative amount, but cannot be relatedto any chemical structure.The progesterone immunoassay demonstrated fivedifferent immunoreactive progesterone metabolites(Fig. 2b–d). Two of them were consistent with the twomajor radiolabelled metabolites (fractions 6–8 and12–13), whereas three immunoreactive metabolitesÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Non-<strong>in</strong>vasive Monitor<strong>in</strong>g of Hormones 79elut<strong>in</strong>g later than fraction 20 corresponded to progesterone,DHP and 5a-P, respectively. The broad-shoulderedpeak at fractions 7–9 <strong>in</strong>dicates a cluster of polarconjugated steroid metabolites. Enzymatic hydrolysis(Jewgenow et al. 2006) of samples changed the elutionpattern result<strong>in</strong>g <strong>in</strong> the disappearance of conjugatedmetabolites towards an <strong>in</strong>crease of free steroids (datanot shown).Figure 3a–c shows the elution profile of immunoreactiveoestrogen metabolites from the different reproductivephases (same samples as <strong>in</strong> Fig. 2). Theoestradiol assay detected four different immunoreactivemetabolites. As described for progesterone, the firstpeak at fractions 4–5 <strong>in</strong>dicates polar conjugated steroidmetabolites followed by a peak of unknown metabolites.Two additional metabolites co-elute with authentic17b-oestradiol and oestrone. Due to the peak broaden<strong>in</strong>gon their basis both a proportion of 17a-oestradioland an unknown metabolite elut<strong>in</strong>g close to oestronecan be assumed.The partial identity of immunoreactive and radiolabelledgestagen metabolites and the detection of dist<strong>in</strong>ctproportions of oestrone and oestradiol allows theconclusion, that the assay systems used are able todetect the relevant faecal steroid hormone metabolitesreflect<strong>in</strong>g ovarian steroid secretion. A chemical identificationof faecal metabolites is not possible, s<strong>in</strong>ce noneof the assays used is able to identify novel steroidmetabolites. However, to design a more specific assay(antibody), particularly the identification of majormetabolites will be atta<strong>in</strong>able us<strong>in</strong>g the sensitivity andthe selectivity of modern LC-MS <strong>in</strong>strumentation.Fig. 3. Elution profile of immunoreactiveoestrogen metabolites <strong>in</strong> afemale Eurasian lynx. Faecalextracts chosen from days 18 and36 of pregnancy and 14 days p.p.were subjected to HPLC separation(see also fig. 2). Arrows <strong>in</strong>dicatethe elution positions of17b-oestradiol, 17a-oestradiol,and oestroneÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


80 M Dehnhard, S Naidenko, A Frank, B Braun, F Go¨ ritz and K JewgenowBiological validationBiological validations are aimed to demonstrate thathormone metabolite measurements reflect the physiologicalevent <strong>in</strong> question. One possibility to validate anon-<strong>in</strong>vasive hormone assay is to compare and correlatefaecal metabolite levels with hormone levels <strong>in</strong> blood(Heistermann et al. 1993; Barrett et al. 2002; Capezzutoet al. 2008). Even if blood sampl<strong>in</strong>g contradicts the non<strong>in</strong>vasiveapproach, veter<strong>in</strong>ary check-ups and treatmentsshould be used to collect blood and fresh faecal samplessimultaneously.Appropriate techniques to physiologically validatenon-<strong>in</strong>vasive methods are pharmacological stimulationsor <strong>in</strong>hibitions of steroid hormone release. These methodstypically <strong>in</strong>volve the adm<strong>in</strong>istration of high doses ofreleas<strong>in</strong>g hormones such as GnRH to stimulate theproduction of gonadal sex steroids (Kretzschmar et al.2004) and adrenocorticotrophic hormone to stimulatethe adrenal gland to produce corticosteroids (Wasseret al. 2000).A biological validation can also be based on dataanalysis. The measured hormone pattern <strong>in</strong> the lynx(Fig. 1) should mirror the reproductive events of afemale. The predicted hormone pattern (derived fromother related felid species) should <strong>in</strong>clude an oestradiolpeak around mat<strong>in</strong>g, elevated progesterone levels dur<strong>in</strong>gpregnancy, followed by a decrease towards basal after4 weeks <strong>in</strong> pseudopregnant females and a more or lessimmediate decrease to basel<strong>in</strong>e prior to parturition <strong>in</strong>pregnant females.Our results, however, revealed differences from thesepredicted patterns: oestrogens did not reflect follicularactivity peak<strong>in</strong>g around ovulation (mat<strong>in</strong>g), but werestrongly correlated to the excretion of gestagens.Therefore, we assume that faecal oestradiol immunoreactivityreflect the activity of corpora lutea. Theprogesterone profiles of Eurasian lynxes were also not<strong>in</strong> accordance to typical felid hormone patterns. Wefound elevated progesterone (and oestradiol) metabolitelevels throughout pregnancy and thereafter. However,the composition of hormone metabolites after parturitionwas different from those dur<strong>in</strong>g pregnancy. Particularlythe relation between the progesterone metaboliteselut<strong>in</strong>g between fractions 7–9 and 13–14 differed markedlydur<strong>in</strong>g pregnancy and the postpartum (p.p.) period(Fig. 2b–d). Compar<strong>in</strong>g the oestrogen profiles from thedifferent reproductive phases (Fig. 3a–c) the relation ofconjugates vs the fractions <strong>in</strong>clud<strong>in</strong>g the unpolarmetabolites rema<strong>in</strong>ed relatively constant whereas therelation between 17b-oestradiol and oestrone changeddramatically <strong>in</strong> the p.p. period <strong>in</strong> favour of oestrone(Fig. 3c).This might be contributed by different hormonesources dur<strong>in</strong>g and after pregnancy (CL, placenta, oradrenals). To confirm this, an additional validation forCL function had to be performed. In case of theEurasian lynx, we performed a transrectal ultrasound<strong>in</strong>vestigation by which we found corpora lutea. This is <strong>in</strong>agreement with the above mentioned high p.p. progesteronevalues, altogether support<strong>in</strong>g the hypothesis of ap.p. luteal activity. At present it is uncerta<strong>in</strong> whether theCL are the result from reta<strong>in</strong>ed CL of pregnancy orfrom a p.p. ovulation. In conclusion there is still noanalytical parameter available <strong>in</strong> faeces which can beused to monitor luteal activity <strong>in</strong> the lynx. Onealternative might be an extensive metabolite screen<strong>in</strong>gus<strong>in</strong>g LC-MS to detect suitable pregnancy markers <strong>in</strong>the lynx. Then, these specific markers can be measuredcomb<strong>in</strong><strong>in</strong>g the selectivity of high pressure liquid chromatographywith mass spectrometric detection, <strong>in</strong> orderto be able to dist<strong>in</strong>guish between pregnancy andpseudopregnancy.Currently the only pregnancy-specific method is thetransabdom<strong>in</strong>al ultrasonographic or radiographic imag<strong>in</strong>gof the uterus (Davidson et al. 1986), but thistechnique usually requires handl<strong>in</strong>g and anesthesia ofthe female, potentially stress<strong>in</strong>g both the queen anddevelop<strong>in</strong>g offspr<strong>in</strong>g.A second option are proteohormones produced by theplacenta, like relax<strong>in</strong>. The cat placenta produces largequantities of relax<strong>in</strong>, beg<strong>in</strong>n<strong>in</strong>g approximately 20 daysof gestation (Addiego et al. 1987). As with dogs, relax<strong>in</strong>has not been detected <strong>in</strong> the serum of cycl<strong>in</strong>g orpseudopregnant cats (Stewart and Stabenfeldt 1985).However, the development of a ur<strong>in</strong>e-based relax<strong>in</strong>pregnancy test would prove extremely useful for breed<strong>in</strong>gmanagement of wildlife species. A radioimmuneassay (RIA) for relax<strong>in</strong> was recently validated for domescat ur<strong>in</strong>e. Ur<strong>in</strong>ary relax<strong>in</strong> was first detected betweendays 14 and 21 of gestation, whereas the levels peaked at42–49 days, followed by a decl<strong>in</strong>e dur<strong>in</strong>g the last2 weeks prior to parturition (de Haas van Dorsser et al.2006). A similar ur<strong>in</strong>ary relax<strong>in</strong> profile was demonstrated<strong>in</strong> the leopard (de Haas van Dorsser et al. 2006).These results <strong>in</strong>dicate that measurement of ur<strong>in</strong>aryrelax<strong>in</strong> might turn out to be a reliable method forpregnancy determ<strong>in</strong>ation <strong>in</strong> felids from as early as 3–4 weeks of gestation. First results analys<strong>in</strong>g ur<strong>in</strong>aryrelax<strong>in</strong> <strong>in</strong> the Iberian lynx showed that relax<strong>in</strong> is anappropriate analyte to differentiate between pregnantand pseudopregnant females particularly dur<strong>in</strong>g thesecond half of pregnancy (BC Braun et al., unpublisheddata). Ur<strong>in</strong>ary relax<strong>in</strong>-based pregnancy diagnosis mayprove useful <strong>in</strong> the breed<strong>in</strong>g management of other felidspecies, and provides a foundation for future studies onpregnancy <strong>in</strong> captive exotic felids.Perhaps one of the greatest uses of steroid metabolitemonitor<strong>in</strong>g will be <strong>in</strong> assist<strong>in</strong>g reproductive managementby identification of ovarian cycle, oestrous, andpregnancy of females. Early pregnancy diagnosis, however,is complicated by the phenomenon of pseudopregnacny<strong>in</strong> carnivoes. Particularly <strong>in</strong> felids, steroidanalyses did not provide an early pregnancy-specificdiagnosis. In the lynx, it is even impossible to use faecalP4 and estrogen metabolite analyses as an <strong>in</strong>dex ofpregnancy. However, there are evidences that theplacenta might contribute to steroid biosynthesis.Therefore, LC-MS based metabolite screen<strong>in</strong>gs compar<strong>in</strong>gpregnant and pseudopregnant females offer anoption to identify pregnancy specific placental steroids.Their identification might allow the development of aspecific assay whose cross-species applicability had to beproved for practice <strong>in</strong> other felid species.Alternatively, sensitive ur<strong>in</strong>ary relax<strong>in</strong> assays mightbe used to confirm pregnancy <strong>in</strong> the lynx. For theÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Non-<strong>in</strong>vasive Monitor<strong>in</strong>g of Hormones 81management of felids <strong>in</strong> zoos, ur<strong>in</strong>ary relax<strong>in</strong> analysiswill be an important research tool supposed ur<strong>in</strong>ecollections could be arranged. Due to species specificbarriers further research is necessary to assess whetherur<strong>in</strong>ary relax<strong>in</strong> could be used to diagnose pregnancy<strong>in</strong> other felids. For free-rang<strong>in</strong>g populations,however, techniques based on faecal samples areessential for study<strong>in</strong>g animals with<strong>in</strong> their naturalhabitat.ReferencesAddiego LA, Tsutsui T, Stewart DR, Stabenfeldt GH, 1987:Determ<strong>in</strong>ation of the source of immunoreactive relax<strong>in</strong> <strong>in</strong>the cat. Biol Reprod 37, 1165–1169.Asa CS, Bauman K, Callahan P, Bauman J, Volkmann DH,Jo¨ chle W, 2006: GnRH agonist <strong>in</strong>duction of fertile estruswith either natural mat<strong>in</strong>g or artificial <strong>in</strong>sem<strong>in</strong>ation, followedby birth of pups <strong>in</strong> gray wolves (Canis lupus).Theriogenology 66, 1778–1782.Barrett GM, Shimizu K, Bardi M, Mori A, 2002: Fecaltestosterone immunoreactivity as a non-<strong>in</strong>vasive <strong>in</strong>dex offunct<strong>in</strong>al testosterone dynamics <strong>in</strong> male Japanese macaques(Macaca fuscata). Primates 43, 29–39.Brown JL, Wasser SK, Wildt DE, Graham LH, 1994:Comparative aspects of steroid hormone metabolism andovarian activity <strong>in</strong> felids, measured non<strong>in</strong>vasively <strong>in</strong> faeces.Biol Reprod 51, 776–786.Brown JL, Terio KE, Graham LH, 1996: Faecal androgenmetabolite analysis for non <strong>in</strong>vasive of testicular steroidogenicactivity <strong>in</strong> felids. Zoo Biol 15, 425–434.Brown JL, Graham LG, Wielebnowski N, Swanson WF,Wildt DE, Howard JG, 2001: Understand<strong>in</strong>g the basicreproductive biology of wild felids by monitor<strong>in</strong>g of faecalsteroids. J Reprod Fertil Suppl 57, 71–82.Capezzuto A, Chel<strong>in</strong>i MOM, Felippe ECG, Oliveira CA, 2008:Correlation between serum and fecal concentrations ofreproductive steroids throughout gestation <strong>in</strong> goats. AnimReprod Sci 103, 78–86.Cardillo M, Mace GM, Gittleman JL, Purvis A, 2006: Latentext<strong>in</strong>ction risk and the future battlegrounds of mammalconservation. Proc Natl Acad Sci U S A 103, 4157–4161.Cavigelli SA, Dubovick T, Levash W, Jolly A, Pitts A, 2003:Female dom<strong>in</strong>ance status and faecal corticoids <strong>in</strong> a cooperativebreeder with low reproductive skew: r<strong>in</strong>g-tailedlemurs (Lemur catta). Horm Behav 43, 166–179.Ceballos G, Ehrlich PR, Sobero´ n J, Salazar I, Fay JP, 2005:Global mammal conservation: what must we manage?Science 309, 546–547.Davidson AP, Nyland TG, Tsutsui T, 1986: Pregnancydiagnosis with ultrasound <strong>in</strong> the domestic cat. Vet Radiol27, 109–114.Earnhardt JM, Thompson SD, Schad K, 2004: Strategicplann<strong>in</strong>g for captive populations: project<strong>in</strong>g changes <strong>in</strong>genetic diversity. Anim Conserv 7, 9–16.Go¨ ritz F, Quest M, Hildebrandt TB, Meyer HHD, Kolter L,Jewgenow K, 2001: Antiprogest<strong>in</strong>s – a new approach tocontrol reproduction <strong>in</strong> captive bears. J Reprod Fertil Suppl57, 249–254.Graham LH, Goodrowe KL, Raeside JI, Liptrap RM, 2005:Non-<strong>in</strong>vasive monitor<strong>in</strong>g of ovarian function <strong>in</strong> several felidspecies by measurement of faecal estradiol-17 and progest<strong>in</strong>s.Zoo Biol 14, 223–237.Graham LH, Byers AP, Armstrong DL, Loskutoff NM,Swanson WF, Wildt DE, Brown JL, 2006: Natural andgonadotrop<strong>in</strong>-<strong>in</strong>duced ovarian activity <strong>in</strong> tigers (Pantheratigris) assessed by fecal steroid analyses. Gen Comp Endocr<strong>in</strong>ol147, 362–370.de Haas van Dorsser FJ, Swanson WF, Lasano S, Ste<strong>in</strong>etz BG,2006: Development, validation, and application of a ur<strong>in</strong>aryrelax<strong>in</strong> radioimmunoassay for the diagnosis and monitor<strong>in</strong>gof pregnancy <strong>in</strong> felids. Biol Reprod 74, 1090–1095.Heistermann M, Tari S, Hodges JK, 1993: Measurement offaecal steroids for monitor<strong>in</strong>g ovarian function <strong>in</strong> NewWorld primates, Callitrichidae. J Reprod Fertil 99, 243–251.Heistermann M, Ademmer C, Kaumanns W, 2004: Ovariancycle and effect of social changes on adrenal and ovarianfunction <strong>in</strong> Pygathrix nemaeus. Int J Primatol 25, 689–708.Heistermann M, Palme R, Gansw<strong>in</strong>dt A, 2006: Comparison ofdifferent enzyme immunoassays for assessment of adrenocorticalactivity <strong>in</strong> primates based on fecal analysis. Am JPrimatol 68, 257–273.Hermes R, Go¨ ritz F, Maltzan J, Blottner S, Proudfoot J,Fritsch G, Fassbender M, Quest M, Hildebrandt TB, 2001:Establishment of assisted reproduction technologies <strong>in</strong>female and male African wild dogs (Lycaon pictus). JReprod Fertil Suppl 57, 315–321.Hermes R, Go¨ ritz F, Streich WJ, Hildebrandt TB, 2007:Assisted reproduction <strong>in</strong> female rh<strong>in</strong>oceros and elephants –current status and future perspective. Reprod Domest Anim42(Suppl. 2), 33–44.Hildebrandt TB, Hermes R, Walzer C, So´ s E, Molnar V,Mezo¨ si L, Schnorrenberg A, Sil<strong>in</strong>ski S, Streich J, SchwarzenbergerF, Go¨ ritz F, 2007: Artificial <strong>in</strong>sem<strong>in</strong>ation <strong>in</strong> theanoestrous and the postpartum white rh<strong>in</strong>oceros us<strong>in</strong>gGnRH analogue to <strong>in</strong>duce ovulation. Theriogenology 67,1473–1484.Jewgenow K, Naidenko SV, Goeritz F, Vargas A, DehnhardM, 2006: Monitor<strong>in</strong>g testicular activity of male Eurasian(Lynx lynx) and Iberian (Lynx pard<strong>in</strong>us) lynx by fecaltestosterone metabolite measurement. Gen Comp Endocr<strong>in</strong>ol149, 151–158.Kretzschmar P, Gansloßer U, Dehnhard M, 2004: Relationshipbetween androgens, environmental factors and reproductivebehavior <strong>in</strong> male white rh<strong>in</strong>oceros (Ceratotheriumsimum simum). Horm Behav 45, 1–9.Masui M, Hiramatsu H, Nose N, Nakasato R, Sagawa Y,Tajima H, Saito K, 1989: Successful artificial <strong>in</strong>sem<strong>in</strong>ation<strong>in</strong> the giant panda (Ailuropoda melanoleuca) at Ueno Zoo.Zoo Biol 8, 17–26.Mesoch<strong>in</strong>a P, Bed<strong>in</strong> E, Ostrowski S, 2003: Re<strong>in</strong>troduc<strong>in</strong>gantelopes <strong>in</strong>to arid areas: lessons learnt from the oryx <strong>in</strong>Saudi Arabia. CR Biol 326(Suppl. 1), S158–S165.Meyer HHD, Rohleder M, Streich WJ, Go¨ ltenboth R, Ochs A,1997: Sexualsteroidprofile und Ovaraktivita¨ ten des PandaweibchensYAN YAN im Berl<strong>in</strong>er Zoo. Berl Mu¨ nchTiera¨ rztl Wschr 110, 143–147.Moor<strong>in</strong>g MS, Patton ML, Lance VA, Hall BM, Schaad EW,Fort<strong>in</strong> SS, Jella JE, McPeak KM, 2004: Fecal androgens ofbison bulls dur<strong>in</strong>g the rut. Horm Behav 46, 392–398.Moreira N, Monteiro-Filho ELA, Moraes W, Swanson WF,Graham LH, Pasquali OL, Gomes MLF, Morais RN, WildtDE, Brown JL, 2001: Reproductive steroid hormones andovarian activity <strong>in</strong> felids of the Leopardus genus. Zoo Biol20, 103–116.Palme R, Fischer P, Schildorfer H, Ismail MN, 1996:Excretion of <strong>in</strong>fused 14C-steroid hormones via faeces andur<strong>in</strong>e <strong>in</strong> domestic livestock. Anim Reprod Sci 43, 43–63.Paris MCJ, White A, Reiss A, West M, Schwarzenberger F,2002: Faecal progesterone metabolites and behaviouralobservations for the non-<strong>in</strong>vasive assessment of oestrouscycles <strong>in</strong> the common wombat (Vombatus urs<strong>in</strong>us) and thesouthern hairy-nosed wombat (Lasiorh<strong>in</strong>us latifrons). AnimReprod Sci 72, 245–257.Pelican KM, Wildt DE, Pukazhenthi B, Howard J, 2006:Ovarian control for assisted reproduction <strong>in</strong> the domesticcat and wild felids. Theriogenology 66, 37–48.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


82 M Dehnhard, S Naidenko, A Frank, B Braun, F Go¨ ritz and K JewgenowPenfold LM, Monfort SL, Wolfe BA, Cit<strong>in</strong>o SB, Wildt DE,2005: Reproductive physiology and artificial <strong>in</strong>sem<strong>in</strong>ationstudies <strong>in</strong> wild and captive gerenuk (Litocranius walleriwalleri). Reprod Fertil Dev 17, 707–714.Pereira RJG, Polegato BF, De Souza S, Negrao JA, DuarteJMB, 2006: Monitor<strong>in</strong>g ovarian cycles and pregnancy <strong>in</strong>brown brocket deer (Mazama gouazoubira) by measurementof fecal progesterone metabolites. Theriogenology 65, 387–399.Pukazhenthi B, Laroe D, Crosier A, Bush LM, Sp<strong>in</strong>dler R,Pelican KM, Bush M, Howard JG, Wildt DE, 2006:Challenges <strong>in</strong> cryopreservation of clouded leopard (Neofelisnebulosa) spermatozoa. Theriogenology 66, 1790–1796.Ryder O, 1993: The Przewalski’s horse: prospects for re<strong>in</strong>troduction<strong>in</strong>to the wild. Conserv Biol 7, 13–15.Scheibe KM, Dehnhard M, Meyer HHD, Scheibe A, 1999:Non<strong>in</strong>vasive monitor<strong>in</strong>g of reproductive function by determ<strong>in</strong>ationof faecal progestagens and sexual behaviour <strong>in</strong> aherd of Przewalski mares <strong>in</strong> a semireserve. Acta Theriol 44,451–463.Schwarzenberger F, 2007: The many uses of non-<strong>in</strong>vasivefaecal steroid monitor<strong>in</strong>g <strong>in</strong> zoo and wildlife species. Int ZooYB 41, 52–74.Stewart DR, Stabenfeldt GH, 1985: Relax<strong>in</strong> activity <strong>in</strong> thepregnant cat. Biol Reprod 32, 848–854.Stoops MA, Bond JB, Bateman HL, Campbell MK, LevensGP, Bowsher TR, Ferrell ST, Swanson WF, 2007: Comparisonof different sperm cryopreservation procedures on postthawquality and heterologous <strong>in</strong> vitro fertilisation success <strong>in</strong>the ocelot (Leopardus pardalis). Reprod Fertil Dev 19, 685–694.Swanson WF, 2006: Application of assisted reproduction forpopulation management <strong>in</strong> felids: the potential and realityfor conservation of small cats. Theriogenology 66, 49–58.Swanson WF, Magarey GM, Herrick JR, 2007: Spermcryopreservation <strong>in</strong> endangered felids: develop<strong>in</strong>g l<strong>in</strong>kageof <strong>in</strong> situ-ex situ populations. Soc Reprod Fertil Suppl 65,417–432.Wasser SK, Hunt KE, Brown JL, Cooper K, Crockett CM,Bechert U, Millspaugh JJ, Larson S, Monfort SL, 2000: Ageneralized fecal glucocorticoid assay for use <strong>in</strong> a diversearray of nondomestic mammalian and avian species. GenComp Endocr<strong>in</strong>ol 120, 260–275.Author’s address (for correspondence): M Dehnhard, Leibniz-Institutefor Zoo Biology and Wildlife Research, PF 601103, D-10252 Berl<strong>in</strong>,Germany. E-mail: dehnhard@izw-berl<strong>in</strong>.deConflict of <strong>in</strong>terest: All authors declare no conflict of <strong>in</strong>terests.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 83–88 (2008); doi: 10.1111/j.1439-0531.2008.01146.xISSN 0936-6768Reproductive Biotechnology and Gene Mapp<strong>in</strong>g: Tools for Conserv<strong>in</strong>g Rare Breedsof LivestockJA LongAnimal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Center, US Department of Agriculture, Beltsville, MD, USAContentsToday’s livestock diversity orig<strong>in</strong>ated from the wild ancestorspecies and was subsequently shaped through the processes ofmutation, genetic drift, and natural and human selection. Onlya subset of the diversity present <strong>in</strong> the ancestral speciessurvives <strong>in</strong> the domestic counterparts. A 2007 report releasedby UN Food and Agriculture Organization ‘The State of theWorld’s Animal Genetic Resources’, compiled from surveysconducted <strong>in</strong> 169 countries, found that nearly 70% of theworld’s rema<strong>in</strong><strong>in</strong>g livestock breeds live <strong>in</strong> develop<strong>in</strong>g countries.The UN report was presented to more than 300 policy makers,scientists, breeders, and livestock keepers at the First InternationalTechnical Conference on Animal Genetic Resources,held <strong>in</strong> September 2007 <strong>in</strong> Interlaken, Switzerland. Theconference aims were to adopt a global plan of action forconserv<strong>in</strong>g animal genetic resources as its ma<strong>in</strong> outcome. Inthis paper, the current and potential contributions of reproductiveand molecular biotechnology are considered as tools ofconserv<strong>in</strong>g rare breeds of livestock.IntroductionOf the 50 000 known mammalian and avian species, arelatively small proportion has been domesticated.Approximately 40 livestock species, shaped by a longhistory of domestication and development, contribute totoday’s agriculture and food production. Livestockbreed development has been and cont<strong>in</strong>ues to be adynamic process of genetic change driven by selectionpressures, <strong>in</strong>clud<strong>in</strong>g environmental factors and human<strong>in</strong>tervention through controlled breed<strong>in</strong>g and husbandry,which has resulted <strong>in</strong> a great variety ofgenetically dist<strong>in</strong>ct breeds. The livestock breeds developedover thousands of years has, until recently, causeda net <strong>in</strong>crease <strong>in</strong> genetic diversity over time. Dur<strong>in</strong>g thepast 100 years, however, there has been a net loss ofdiversity because of an <strong>in</strong>creased rate of ext<strong>in</strong>ction oflivestock breeds and varieties [UN Food and AgricultureOrganization (FAO) (2007)]. The number of breedslost over the past 8 years is rapidly approach<strong>in</strong>g the rateof ext<strong>in</strong>ction that occurred from 1900 to 1999 (Table 1).Losses have been accelerated by the rapid <strong>in</strong>tensificationof livestock production, a failure to evaluate localbreeds, and <strong>in</strong>appropriate breed replacement or crossbreed<strong>in</strong>gfacilitated by the availability of high perform<strong>in</strong>gbreeds (FAO 2007). As an example of <strong>in</strong>appropriatebreed replacement, Uganda’s <strong>in</strong>digenous, droughthardyAnkole cattle could face ext<strong>in</strong>ction with<strong>in</strong>20 years because they are be<strong>in</strong>g rapidly supplanted byHolste<strong>in</strong>-Friesians, a breed which produces much moremilk. Dur<strong>in</strong>g a recent drought, however, farmers whohad Ankole cattle were able to walk them long distancesto reach water sources while those who had traded theAnkole for imported breeds lost entire herds. Thephysiology and graz<strong>in</strong>g behaviour of the importedbreeds typically are not adapted to the natural pasturesand climate of Africa, especially when drought strikes(Kay 1997). In another example, cross-breed<strong>in</strong>g hasalmost decimated purebred populations of the EastAfrican Red Maasai sheep, which is renowned for itsdisease resistance to gastro<strong>in</strong>test<strong>in</strong>al parasites and highproductivity under extremely challeng<strong>in</strong>g environments.In the mid-1970s, as a result of a subsidized dissem<strong>in</strong>ationprogram, many farmers <strong>in</strong> Kenya cross-bred theirRed Maasai flocks with the less-hardy Dorpers sheep,which subsequently proved unsuitable <strong>in</strong> many productionareas. In 1992, the International LivestockResearch Institute (ILRI) undertook an extensive search<strong>in</strong> Kenya and northern parts of the United Republic ofTanzania, and was only able to locate a very smallnumber of purebred animals, which later showed somelevels of genetic contam<strong>in</strong>ation (Gibson and Candiff2000).The ext<strong>in</strong>ction of a breed or population means theloss of its unique adaptive attributes, which are underthe control of many <strong>in</strong>teract<strong>in</strong>g genes and are the resultsof complex <strong>in</strong>teractions between the genotype and theenvironment. Figure 1 illustrates the current status oflivestock breeds (FAO 2007). The regions with thehighest proportion of their breeds classified as at risk areEurope (28% of mammalian breeds; 49% of avianbreeds) and North America (20% of mammalian breeds;79% of avian breeds). Europe and North America arethe regions that have the most highly specializedlivestock <strong>in</strong>dustries, <strong>in</strong> which production is dom<strong>in</strong>atedby a small number of breeds. In recent years, many ofthe world’s small farmers have abandoned their traditionalanimals <strong>in</strong> favour of higher yield<strong>in</strong>g stockimported from Europe and the USA. For example, <strong>in</strong>1994 local breeds comprised 72% of the sow population<strong>in</strong> northern Vietnam; with<strong>in</strong> 8 years, however, this figurehad dropped to just 26%. Of the country’s 14 local pigbreeds, five are now listed as vulnerable, two areconsidered critical state and three are fac<strong>in</strong>g ext<strong>in</strong>ction.The 2007 report, ‘The State of the World’s AnimalGenetic Resources’, compiled by the FAO, with contributionsby the ILRI and other research groups,surveyed farm animals <strong>in</strong> 169 countries. Nearly 70%of the entire world’s rema<strong>in</strong><strong>in</strong>g unique livestock breedsare found <strong>in</strong> develop<strong>in</strong>g countries, which as describedabove, are at risk from the importation and farm<strong>in</strong>g ofexogenous livestock breeds. Renowned organizationssuch as the ILRI and FAO have spearheaded the firstimportant step for conservation by conduct<strong>in</strong>g livestockÓ 2008 No claim to orig<strong>in</strong>al government works


84 JA LongTable 1. Livestock breed ext<strong>in</strong>ction rates over timeYear span Number of breeds Percent of breedBefore 1900 15 21900–1999 111 16After 1999 62 9Unspecified a 502 73Total 690 100a Unspecified = no year of ext<strong>in</strong>ction <strong>in</strong>dicated. Adapted from FAO (2007).breed surveys. These surveys are time-consum<strong>in</strong>g andlogistically complex; however, ILRI scientists havedeveloped and <strong>in</strong>tegrated the <strong>Domestic</strong> Animal GeneticResources Information System with the FAO’s <strong>Domestic</strong>Animal Diversity Information System to streaml<strong>in</strong>ethe process. These web-based <strong>in</strong>formation systemsprovide a means for curation and dissem<strong>in</strong>ation ofvaluable <strong>in</strong>formation that will support development ofconservation priorities, as well as provide a database foruse of reproductive and molecular biotechnologies toma<strong>in</strong>ta<strong>in</strong> valuable genetic resources.Impact of Reproductive Biotechnology onLivestock ConservationModern reproductive biotechnologies, such as artificial<strong>in</strong>sem<strong>in</strong>ation, embryo transfer, <strong>in</strong> vitro fertilization,gamete ⁄ embryo micromanipulation, semen sex<strong>in</strong>g, genomeresource bank<strong>in</strong>g and somatic cell nuclear transfer(clon<strong>in</strong>g) have enormous potential for conserv<strong>in</strong>g rarebreeds of livestock. The advent of artificial <strong>in</strong>sem<strong>in</strong>ation<strong>in</strong> the 1940s with cattle and semen cryopreservation <strong>in</strong>the 1950s with poultry was <strong>in</strong>strumental <strong>in</strong> the successfultransfer of genetic material between and among livestockpopulations and breeds. There are several anecdotaland published reports of apply<strong>in</strong>g thesetechnologies to conserv<strong>in</strong>g rare livestock breeds. TheRare Breeds Program <strong>in</strong> Colonial Williamsburg, VA(USA) makes use of semen cryopreservation for livestockbreeds dat<strong>in</strong>g from the 17th and 18th centuries,<strong>in</strong>clud<strong>in</strong>g Devon cattle, Leicester sheep, Ossabaw pigsand American Cream horses, although actual numbersof offspr<strong>in</strong>g from artificial <strong>in</strong>sem<strong>in</strong>ation with frozen⁄ thawed semen have not been documented to date.The Hamilton Rare Breeds Foundation <strong>in</strong> Hartland, VT(USA) has pioneered <strong>in</strong> the use of frozen semen <strong>in</strong> thePoitou Donkey, an ancient breed dat<strong>in</strong>g back over2000 years, and is the first group to have produced foalsfrom the rarest breed of donkey <strong>in</strong> existence today.Embryo transfer, pioneered <strong>in</strong> agricultural species <strong>in</strong>the 1930s, also has been reported <strong>in</strong> use with heritagelivestock breeds. In collaboration with the Swiss VillageFarm (SVF) Foundation <strong>in</strong> Newport, RI (USA),frozen ⁄ thawed embryos from the Tennessee Myotonicor fa<strong>in</strong>t<strong>in</strong>g goat breed were surgically transferred <strong>in</strong>totwo surrogate Nubian does (a common domestic breed)and resulted <strong>in</strong> the birth of one healthy buck (Matsaset al. 2005). In 2006, the SVF Foundation reported asecond birth from <strong>in</strong>terspecies embryo transfer: anendangered Gulf Coast lamb born to a Santa Cruzewe. The Gulf Coast sheep provides a good example forthe importance of preserv<strong>in</strong>g the unique genetic attributesof heritage livestock breeds, as the Gulf Coast issheep is extremely resistant to parasites and nearlyimpervious to the foot rot that plagues many other ov<strong>in</strong>ebreeds.One of the most promis<strong>in</strong>g areas of reproductivebiotechnology is the creation of genetic resource banksas a conservation tool for rare livestock breeds. Theconcept of bank<strong>in</strong>g gametes, embryos and DNA materialfor conservation purposes is not new, as the idea hasRisk status of the world's mammalian breeds <strong>in</strong> January 2006: absolute (table) and percentage (chart) figures by region100%80%60%40%20%0%Africa Asia Europe Lat<strong>in</strong> americaNear & middleEastNorth americaSouthwestpacificInternationalTransboundrybreedsUnknown 384 469 459 304107 79 8058 1940Critical 13 23182 9012 97 255Critical-ma<strong>in</strong>ta<strong>in</strong>ed 0 451 4 000059Endangered 26 50249 21622 11 22 407Endangered-ma<strong>in</strong>ta<strong>in</strong>ed 4 3142 9011 0160Ext<strong>in</strong>ct 35 45 481 21 549 6 1643Not-at-risk 187 776 664 81 85 13 17 312 2135WorldFig. 1. Proportion of the world’s breeds by risk status category. Adapted from FAO (2007)Ó 2008 No claim to orig<strong>in</strong>al government works


Biotechnology Methods for Preserv<strong>in</strong>g Rare Livestock 85Table 2. Rare and historic breed livestock genetic resource banksNameAvian Resource CenterCenter for Genetic ResourcesNational Bureau of Animal Genetic ResourcesINRA’s National CryobankInternational Livestock Research InstituteLivestock Research InstituteNordic Genebank for Farm <strong>Animals</strong>Rare Breeds Gene BankSwiss Village Farm FoundationUSDA’s National Animal Germplasm ProgramCountryCanada ⁄ British ColumbiaThe NetherlandsIndiaFranceKenyaTaiwanNorwayNew ZealandUSAUSAbeen widely discussed for use <strong>in</strong> preserv<strong>in</strong>g endangeredwildlife populations (Wildt 1992, 2000; Long et al. 1996;Wildt et al. 1997; Holt and Pickard 1999; Andrabi andMaxwell 2007). Table 2 lists the pre-dom<strong>in</strong>ant germplasm⁄ genetic repositories for rare livestock breedsaround the world. One notable success story thatillustrates the potential benefits of genetic resourcebanks <strong>in</strong>volves the Dutch Friesian cattle breed. In1879, the cattle population <strong>in</strong> the prov<strong>in</strong>ce of Frieslandconsisted ma<strong>in</strong>ly of Red Pied cattle registered as a redand white phenotype <strong>in</strong> the Friesian Cattle herd book.Black and white cattle progressively became morepopular than the orig<strong>in</strong>al red and white; by 1970, therewere only 50 farmers registered as own<strong>in</strong>g a total of2500 Red and White Friesian cattle. The susta<strong>in</strong>edimport of Holste<strong>in</strong>-Friesians from the United States andCanada further eroded the population, to where only 21Red and White cattle (4 males and 17 females) rema<strong>in</strong>ed<strong>in</strong> 1993. A group of owners started the Foundation forNative Red and White Friesian Cattle and, <strong>in</strong> collaborationwith the Nordic Genebank for <strong>Animals</strong>, developeda breed<strong>in</strong>g program. Frozen ⁄ thawed semen thatwas preserved the 1970s and 1980s and subsequentlystored <strong>in</strong> the Genebank was used to breed females undera contract system. Result<strong>in</strong>g male progeny were raisedby breeders, and semen from these males was collected,frozen and later used under new contracts. The breed<strong>in</strong>creased <strong>in</strong> number, reach<strong>in</strong>g 256 registered liv<strong>in</strong>gfemales and 12 liv<strong>in</strong>g males <strong>in</strong> 2004. Currently, a total of11 780 semen doses from 43 bulls are stored <strong>in</strong> theGenebank and kept available for artificial <strong>in</strong>sem<strong>in</strong>ation(FAO 2007). Another example of successful usage ofsemen from storage repositories <strong>in</strong>volves the endangeredGauloise dore´e chicken, the oldest patrimonial poultrybreed <strong>in</strong> France. Us<strong>in</strong>g frozen ⁄ thawed semen and an<strong>in</strong>tensive breed<strong>in</strong>g program, current stocks have proventhe restoration of more than 96% of the <strong>in</strong>itial genome(Blesbois et al. 2007). This po<strong>in</strong>t is particularly importantfor avian breeds, as neither the female gamete norembryo has not been successfully cryopreserved and,unlike mammals where the male gamete determ<strong>in</strong>esgender, birds have a ZZ male ⁄ ZW female sex-determ<strong>in</strong><strong>in</strong>gsystem.More recent reproductive biotechnologies such assomatic cell nuclear clon<strong>in</strong>g also have enormouspotential for conserv<strong>in</strong>g rare breeds of livestock.Another rare breed success story <strong>in</strong>volves the clon<strong>in</strong>gof the Enderby Island Cow, the last survivor of theworld’s rarest cattle breed. In 1992, members of NewZealand’s Rare Breeds Conservation Society found freshhoof-pr<strong>in</strong>ts of two cattle on Enderby Island. Fivemonths later, the world’s only surviv<strong>in</strong>g Enderby Islandcow and her heifer calf were captured. Unfortunately,the calf subsequently died of unknown causes, leav<strong>in</strong>gthe cow as the only survivor of her breed <strong>in</strong> the world. Atotal of 35 embryo transfers were conducted, andresulted <strong>in</strong> the birth of a s<strong>in</strong>gle male calf. In a lasteffort to save the breed, somatic cell nuclear transfer wasused to produce heifer clones from the cow (Wells et al.1998). To date, two of the three surviv<strong>in</strong>g clones haves<strong>in</strong>ce given natural birth to two heifer calves.Somatic cell clon<strong>in</strong>g also was used to produce liveoffspr<strong>in</strong>g from the rare European mouflon sheep, abreed found on Sard<strong>in</strong>ia, Corsica and Cyprus wherethere is thought to be fewer than 1000 mature <strong>in</strong>dividuals<strong>in</strong> the wild. Loi et al. (2001) <strong>in</strong>jected enucleatedsheep oocytes from a closely-related domestic breed withsomatic granulosa cells recovered from the ovaries oftwo adult female mouflons found dead <strong>in</strong> the pasture.Blastocyst-stage cloned embryos were transferred <strong>in</strong>tosheep foster mothers and two pregnancies were established,one of which produced an apparently normalmouflon lamb. What is remarkable about this example isthat although the nuclear donor cells were recoveredfrom dead animals and considered non-viable, thesepost-mortem cells were able to generate normal embryosand offspr<strong>in</strong>g. This example supports the use of clon<strong>in</strong>gfor the expansion of critically endangered populations,both with<strong>in</strong> a concerted conservation program and <strong>in</strong>extreme situations <strong>in</strong>volv<strong>in</strong>g sudden death (Loi et al.2001). Despite these reports of the positive impact ofreproductive biotechnology on the conservation of rarelivestock breeds, there are too few examples of artificial<strong>in</strong>sem<strong>in</strong>ation, germplasm cryopreservation, or embryotransfer be<strong>in</strong>g used <strong>in</strong> conjunction with rare livestock.Application of Molecular Biotechnology Toolsfor Livestock ConservationAt the molecular level, the genetic diversity presentwith<strong>in</strong> a livestock species is a reflection of differences <strong>in</strong>DNA sequences, or allelic diversity, across the functionalDNA regions, or genes affect<strong>in</strong>g animal developmentand performance. The complete and partialsequenc<strong>in</strong>g of major livestock genomes (chicken, 2004;bov<strong>in</strong>e, 2005; rabbit, 2006; pig, 2007) provides a wealthof <strong>in</strong>formation useful for many aspects of livestockbreed conservation from identify<strong>in</strong>g ancestral breeds tounderstand<strong>in</strong>g disease resistance.Gene mapp<strong>in</strong>g has been used to as a tool tounderstand livestock orig<strong>in</strong> and diversity <strong>in</strong> severallivestock species. For example, 5 dist<strong>in</strong>ct maternalmitochondrial major l<strong>in</strong>eages have been identified <strong>in</strong>domestic goats (Luikart et al. 2001; Sultana et al. 2003;Joshi et al. 2004); while the Asian mouflon is purportedto be the only progenitor of domestic sheep (Hiendlederet al. 1998). The ancestor of the domestic pig is the wildboar (Sus scrofa), with at least 16 dist<strong>in</strong>ct subspecies ofwild boar have been described <strong>in</strong> Eurasia and NorthAfrica. A recent survey of mitochondrial DNA diversityamong Eurasian domestic pigs and wild boar revealed acomplex picture of pig domestication, with at least fiveÓ 2008 No claim to orig<strong>in</strong>al government works


86 JA Longor six dist<strong>in</strong>ct centres across the geographical range ofthe wild species (Larson et al. 2005).<strong>Domestic</strong>ation of cattle has been particularly welldocumented through gene mapp<strong>in</strong>g, with clear evidenceof three dist<strong>in</strong>ct <strong>in</strong>itial domestication events for threedist<strong>in</strong>ct aurochs (Bos primigenius) subspecies. Bos primigeniusprimigenius and B. p. opisthonomous, are theancestors of the humpless B. taurus cattle of the NearEast and Africa, respectively, with domestication occurr<strong>in</strong>gapproximately 9000 years ago (Wendorf and Schild1994). Humped Zebu cattle (B. <strong>in</strong>dicus) are believed tohave been domesticated at a later date, approximately7000–8000 years ago (Loftus et al. 1994; Bradley et al.1996; Bradley and Magee 2006). F<strong>in</strong>ally, the domesticchicken (Gallus domesticus) is descended from the wildred jungle fowl (G. gallus). While previous molecularstudies suggested a s<strong>in</strong>gle domestic orig<strong>in</strong> <strong>in</strong> SoutheastAsia (Fumihito et al. 1994, 1996), at least six dist<strong>in</strong>ctmaternal genetic l<strong>in</strong>eages have now been identified (Liuet al. 2006).In genetic diversity studies, the most frequently usedmarkers are microsatellites and these are the mostpopular markers <strong>in</strong> livestock genetic characterizationstudies (Sunnucks 2001). Their high mutation rate andco-dom<strong>in</strong>ant nature permit the estimation of with<strong>in</strong>- andbetween-breed genetic diversity, and genetic admixtureamong closely related breeds. There are a few examplesof large-scale analyses of the genetic diversity oflivestock species. For example, chicken and pig diversitythroughout Europe have been reported (Hillel et al.2003; SanCristobal et al. 2006). Sheep diversity wasassessed at a large regional scale <strong>in</strong> northern Europeancountries (Tapio et al. 2005); while Can˜ on et al. (2006)studied goat diversity <strong>in</strong> Europe and the Middle East.Probably the most comprehensive study of this type <strong>in</strong>livestock is a cont<strong>in</strong>ent-wide study of African cattle(Hanotte et al. 2002), which revealed the genetic signaturesof the orig<strong>in</strong>s, secondary movements and differentiationof African cattle. For most livestock breeds,however, a comprehensive review is still lack<strong>in</strong>g.S<strong>in</strong>gle nucleotide polymorphisms (SNPs) are used asan alternative to microsatellites <strong>in</strong> genetic diversitystudies (Marsjan and Oldenbroek 2007). S<strong>in</strong>gle nucleotidepolymorphisms are variations at s<strong>in</strong>gle nucleotideswhich do not change the overall length of theDNA sequence <strong>in</strong> the region and occur throughoutthe genome. With this perspective, large-scale projectsare ongo<strong>in</strong>g <strong>in</strong> several livestock species to identifymillions (Wong et al. 2004) and validate severalthousands of SNPs, and identify haplotype blocks <strong>in</strong>the genome.Mitochondrial DNA (mtDNA) polymorphisms havebeen extensively used <strong>in</strong> phylogenetic and geneticdiversity analyses. The haploid mtDNA, carried by themitochondria <strong>in</strong> the cell cytoplasm, has a maternal modeof <strong>in</strong>heritance (<strong>in</strong>dividuals <strong>in</strong>herit the mtDNA fromtheir dams and not from their sires) and a high mutationrate; it does not recomb<strong>in</strong>e. These characteristics enablebiologists to reconstruct evolutionary relationshipsbetween and with<strong>in</strong> species by assess<strong>in</strong>g the patterns ofmutations <strong>in</strong> mtDNA. MtDNA markers may alsoprovide a rapid way of detect<strong>in</strong>g hybridization betweenlivestock species or subspecies (Nijman et al. 2003).An alternative approach to the identification ofgenome regions carry<strong>in</strong>g relevant genes has recentlybeen proposed. It consists of the detection of ‘selectionsignatures’ via a ‘population genomics’ approach (Blacket al. 2001; Luikart et al. 2003). Population genomicsutilizes phenotypic data at the breed level (or subpopulationswith<strong>in</strong> a breed), rather than at the <strong>in</strong>dividuallevel. The population genomics approach also canidentify genes subjected to strong selection pressureand eventually fixed with<strong>in</strong> breeds and, <strong>in</strong> particular,genes <strong>in</strong>volved <strong>in</strong> adaptation to extreme environmentsor disease resistance. Population genomics relies on thepr<strong>in</strong>ciple that loci across the genome are <strong>in</strong>fluenced bygenome-wide evolutionary forces (e.g. genetic drift, geneflow), whereas locus-specific forces, such as selection,impr<strong>in</strong>t a particular pattern of variability on l<strong>in</strong>ked locionly (Luikart et al. 2003). By compar<strong>in</strong>g the geneticdiversity of many loci across the genome, it is thenpossible to reveal loci display<strong>in</strong>g an atypical variationpattern, which are likely to be l<strong>in</strong>ked to those genomicregions affected by selection (Black et al. 2001). Therefore,<strong>in</strong> contrast to candidate-gene-based methods,strategies mak<strong>in</strong>g use of population genomics do notfocus on a few loci only, but rather depict the effect ofselection over the whole genome (Storz 2005).Another new frontier emerg<strong>in</strong>g from the concept ofpopulation genomics is landscape genomics. Livestockby def<strong>in</strong>ition are adapted to the landscape (e.g. temperature,altitude, ra<strong>in</strong>fall, disease challenge, nutritionalchallenge and human selection). The aim of landscapegenomics is to learn from the co-evolution of livestockand production systems and use the knowledge ga<strong>in</strong>edto better match different breeds with production circumstances.A novel approach for evaluat<strong>in</strong>g populationgenomics is based on a spatial analysis methoddesigned to detect signatures of natural selection with<strong>in</strong>the genome of domestic and wild animals (Joost et al.2007). Spatial analysis method goes a step furthercompared to classical approaches, as it is designed toidentify environmental parameters associated withselected markers (FAO 2007). By overlay<strong>in</strong>g populationgenomic analyses (e.g. ‘signatures of selection’) withother sets of <strong>in</strong>formation such as agro-ecological mapsor other environmental ⁄ production <strong>in</strong>formation, it canbe determ<strong>in</strong>ed what genetic materials are candidates foruse <strong>in</strong> which parts of the globe. The concept oflandscape genomics is promis<strong>in</strong>g, as this comb<strong>in</strong>es georeferenc<strong>in</strong>gof breed distributions, spatial ⁄ global geneticdiversity, climatic, ecological, epidemiological and productionsystem <strong>in</strong>formation which will facilitate anddirect priority decisions for breed conservation.Future Challenges and OpportunitiesLack of <strong>in</strong>formation on the world’s livestock resources,such as what livestock breeds ⁄ populations exist, theirgeographical location and their genetic characteristics, isa major impediment to their susta<strong>in</strong>able use. The currentdocumented numbers of breeds is likely an underestimation,as a large proportion of <strong>in</strong>digenous livestockpopulations are <strong>in</strong> the develop<strong>in</strong>g world and have yetto be described at phenotypic and genotypic levels(Hanotte and Jianl<strong>in</strong> 2005). Additionally, the geneticÓ 2008 No claim to orig<strong>in</strong>al government works


Biotechnology Methods for Preserv<strong>in</strong>g Rare Livestock 87characterization of all rema<strong>in</strong><strong>in</strong>g wild ancestral populationsand closely related species is critical as these are theonly rema<strong>in</strong><strong>in</strong>g sources of putative alleles of economicvalues that might have been lost dur<strong>in</strong>g domesticationevents. Moreover, the development and use of reproductivebiotechnology, particularly genetic resourcebanks, is critical for the preservation and managementof the rema<strong>in</strong><strong>in</strong>g agricultural resources. There is a largegap between developed and develop<strong>in</strong>g countries <strong>in</strong> theability to use reproductive and molecular biotechnologyfor sett<strong>in</strong>g and ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g conservation priorities. Therecent International Technical Conference on AnimalGenetic Resources was a timely event that presentedmany areas for global concern and provided leadershipfor sett<strong>in</strong>g conservation priorities. It is particularlyimportant to conserve the current livestock geneticresources because the ancestors of most of our exist<strong>in</strong>glivestock species no longer exist. Genetically diverselivestock populations provide a greater range of optionsfor meet<strong>in</strong>g future challenges, whether associated withenvironmental change, emerg<strong>in</strong>g disease threats, newknowledge of human nutritional requirements, fluctuat<strong>in</strong>gmarket conditions or chang<strong>in</strong>g societal needs.ReferencesAndrabi S, Maxwell W, 2007: A review on reproductivebiotechnologies for conservation of endangered mammalianspecies. Anim Reprod Sci 99, 223–243.Black WC, Baer CF, Antol<strong>in</strong> MF, DuTeau NM, 2001:Population genomics: genome-wide sampl<strong>in</strong>g of <strong>in</strong>sectpopulations. Annu Rev Entomol 46, 441–469.Blesbois E, Seigneur<strong>in</strong> F, Grasseau I, Limouz<strong>in</strong> C, Besnard J,Gourichon D, Coquerelle G, Rault P, Tixier-Boichard M,2007: Semen cryopreservation for ex situ management ofgenetic diversity <strong>in</strong> chicken: creation of the French aviancryobank. Poultry Sci 86, 555–564.Bradley DG, Magee D, 2006: Genetics and the orig<strong>in</strong>s ofdomestic cattle. In: Zeder MA, Emshwiller E, Smith BD,Bradley DG (eds), Document<strong>in</strong>g <strong>Domestic</strong>ation: NewGenetics and Archaeological Paradigm. University of CaliforniaPress, California, U S A, pp. 317–328.Bradley DG, MacHugh DE, Cunn<strong>in</strong>gham P, Loftus RT, 1996:Mitochondrial DNA diversity and the orig<strong>in</strong>s of African andEuropean cattle. Proc Natl Acad Sci U S A 93, 5131–5135.Can˜ on J, Garcıa D, Garcıa-Atance MA, Obexer-Ruff G,Lenstra JA, Ajmone-Marsan P, Dunner S, 2006: TheECONOGENE Consortium. Geographical partition<strong>in</strong>g ofgoat diversity <strong>in</strong> Europe and the Middle East. Anim Genet37, 327–334.FAO, 2007: Multiple chapters. In: Rischkowsky B, Pill<strong>in</strong>g D(eds), The State of the World’s Animal Genetic Resourcesfor Food and Agriculture. FAO, Rome.Fumihito A, Miyake T, Sumi S, Takada M, Ohno S, KondoN, 1994: One subspecies of the red junglefowl (Gallus gallusgallus) suffices as the matriarchic ancestor of all domesticbreeds. Proc Natl Acad Sci U S A 91, 12505–12509.Fumihito A, Miyake T, Takada M, Sh<strong>in</strong>gu R, Endo T,Gojobori T, Kondo N, Ohno S, 1996: Monophyletic orig<strong>in</strong>and unique dispersal patterns of domestic fowls. Proc NatlAcad Sci U S A 93, 6792–6795.Gibson JP, Candiff LV 2000: Develop<strong>in</strong>g straight-breed<strong>in</strong>gand cross-breed<strong>in</strong>g structures for extensive graz<strong>in</strong>g systemswhich utilize exotic animal genetic resources. ICAR TechnicalSeries No. 3. In: Galal S, Boyazoglu J, Hammond K(eds), Proceed<strong>in</strong>gs of Workshop on Develop<strong>in</strong>g Breed<strong>in</strong>gStrategies for Low Input Animal Production Environments.ICAR, Villa del Ragno, Via Nomentana, Rome Italy, pp.207–241.Hanotte O, Bradley DG, Ochieng JW, Verjee Y, Hill EW,2002: African pastoralism: genetic impr<strong>in</strong>ts of orig<strong>in</strong>s andmigrations. Science 296, 336–339.Hiendleder S, Ma<strong>in</strong>z K, Plante Y, Lewalski H, 1998: Analysisof mitochondrial DNA <strong>in</strong>dicates that the domestic sheep arederived from two different ancestral maternal sources: noevidences for the contribution from Urial and Argali sheep.J Heredity 89, 113–120.Hillel J, Groenen MA, Tixier-Boichard M, Korol AB, DavidL, Kirzhner VM, Burke T, Barre-Dirie A, Crooijmans RP,Elo K, Feldman MW, Freidl<strong>in</strong> PJ, 2003: Biodiversity of 52chicken populations assessed by microsatellite typ<strong>in</strong>g ofDNA pools. Genet Sel Evol 35, 533–557.Holt WV, Pickard AR, 1999: Role of reproductive technologies<strong>in</strong> genetic resource banks <strong>in</strong> animal conservation. RevReprod 4, 143–150.Joost S, Bon<strong>in</strong> A, Bruford MW, Despres L, Conord C,Erhardt G, Taberlet P, 2007: A spatial analysis method(SAM) to detect candidate loci for selection: towards alandscape genomics approach to adaptation. Mol Ecol 16,3955–3969.Joshi MB, Rout PK, Mandal AK, Tyler-Smith C, S<strong>in</strong>gh L,Thangaray K, 2004: Phylogeography and orig<strong>in</strong>s of Indiandomestic goats. Mol Biol Evol 21, 454–462.Kay RNB, 1997: Responses of African livestock and wildherbivores to drought. J Arid Environ 37, 683–694.Larson G, Dobney K, Albarella U, Fang M, Matisoo-Smith E,Rob<strong>in</strong>s J, Lowden S, F<strong>in</strong>layson H, Brand T, Willerslev E,Rowley-Conwy P, Andersson L, Cooper A, 2005: Worldwidephylogeography of wild boar reveals multiple centersof pig domestication. Sci 307, 1618–1621.Liu YP, Wu GS, Yao YG, Miao YW, Luikart G, Baig M,Beja-Pereira A, D<strong>in</strong>g ZL, Palanichamy MG, Zhang YP,2006: Multiple maternal orig<strong>in</strong>s of chickens: out of theAsian jungles. Mol Phylogene Evol 38, 12–19.Loftus RT, MacHugh DE, Bradley DG, Sharp PM, Cunn<strong>in</strong>ghamP, 1994: Evidence for two <strong>in</strong>dependent domesticationof cattle. Proc Natl Acad Sci U S A 91, 2757–2761.Loi P, Ptak G, Barboni B, Fulka J, Cappai P, Cl<strong>in</strong>ton M, 2001:Genetic rescue of an endangered mammal by cross-speciesnuclear transfer us<strong>in</strong>g post-mortem somatic cells. NatureBiotechnol 19, 962–964.Long JA, Larson SE, Wasser SK, 1996: Safeguard<strong>in</strong>g diversity:challenges <strong>in</strong> develop<strong>in</strong>g a genome resource bank forCalifornia sea otters. Endangered Species Update 13, 57–60.Luikart GL, Gielly L, Excoffier L, Vigne JD, Bouvet J,Taberlet P, 2001: Multiple maternal orig<strong>in</strong>s and weakphylogeographic structure <strong>in</strong> domestic goats. Proc NatlAcad Sci U S A 98, 5927–5930.Marsjan PA, Oldenbroek JK2007: Molecular markers, a toolfor explor<strong>in</strong>g genetic diversity. In: Rischkowsky B, Pill<strong>in</strong>g D(eds), The State of the World’s Animal Genetic Resourcesfor Food and Agriculture, Section C, Part 4. FAO, Rome,pp. 359–379.Matsas D, Huntress V, Lev<strong>in</strong>e H, Ayres S, Am<strong>in</strong>i J, Duby R,Borden P, Saperste<strong>in</strong> G, Overstrom E2005: Preservation ofheritage livestock breeds: <strong>in</strong>tegrated program to cryopreservegermplasm from Tennessee Myotonic goats. ReprodFertil Dev 17, 195 (Abstract).Nijman IJ, Otsen M, Verkaar EL, de Ruijter C, Hanekamp E,2003: Hybridization of banteng (Bos javanicus) and zebu(Bos <strong>in</strong>dicus) revealed by mitochondrial DNA, satelliteDNA, AFLP and microsatellites. Heredity 90, 10–16.SanCristobal M, Chevalet C, Haley CS, Joosten R, Ratt<strong>in</strong>kAP, Harlizius B, Groenen MA, Amigues Y, Boscher MY,Ó 2008 No claim to orig<strong>in</strong>al government works


88 JA LongRussell G, Law A, Davoli R, Russo V, Desautes C,Alderson L, Fimland E, Bagga M, Delgado JV, Vega-PlaJL, Mart<strong>in</strong>ez AM, Ramos M, Glodek P, Meyer JN, Gand<strong>in</strong>iGC, Matass<strong>in</strong>o D, Plastow GS, Siggens KW, Laval G,Archibald AL, Milan D, Hammond K, Cardell<strong>in</strong>o R,2006: Genetic diversity with<strong>in</strong> and between Europeanpig breeds us<strong>in</strong>g microsatellite markers. Anim Genet 37,189–198.Storz JF, 2005: Us<strong>in</strong>g genome scans of DNA polymorphismto <strong>in</strong>fer adaptive population divergence. Mol Ecol 14,671–688.Sultana S, Mannen H, Tsuji S, 2003: Mitochondrial DNAdiversity of Pakistani goats. Anim Genet 34, 417–421.Sunnucks P, 2001: Efficient genetic markers for populationbiology. Tree 15, 199–203.Tapio M, Tapio I, Grislis Z, Holm LE, Jeppsson S, KantanenJ, Miceikiene I, Olsaker I, Vi<strong>in</strong>alass H, Eythorsdottir E,2005: Native breeds demonstrate high contributions to themolecular variation <strong>in</strong> northern European sheep. Mol Ecol14, 3951–3963.Wells DN, Misica PM, Tervit HR, Vivanco WH, 1998: Adultsomatic cell nuclear transfer is used to preserve the lastsurviv<strong>in</strong>g cow of the Enderby Island cattle breed. ReprodFertil Dev 10, 369–378.Wendorf F, Schild R, 1994: Are the early Holecene cattle <strong>in</strong> theEastern Sahara domestic or wild? Evol Anthropol 3, 118–128.Wildt DE, 1992: Genetic resource bank<strong>in</strong>g for conserv<strong>in</strong>gwildlife species: Justification, examples and becom<strong>in</strong>gorganized on a global basis. Anim Reprod Sci 28, 247–257.Wildt DE, 2000: Genome resource bank<strong>in</strong>g for wildliferesearch, management, and conservation. ILAR J 41, 228–234.Wildt DE, Rall WF, Crister JK, Monfort SL, Seal US, 1997:Genome resource banks: ‘liv<strong>in</strong>g collections’ for biodiversityconservation. Bioscience 47, 689–698.Wong GK, Liu B, Wang J, Zhang Y, Yang X, Zhang Z, MengQ, Zhou J, Li D, Zhang J, Ni P, Li S, Ran L, Li H, Zhang J,Li R, Li S, Zheng H, L<strong>in</strong> W, Li G, Wang XZhao W, Li J, YeC, Dai M, Ruan J, Zhou Y, Li Y, He X, Zhang Y, Wang J,Huang X, Tong W, Chen J, Ye J, Chen C, Wei N, Li G,Dong L, Lan F, Sun Y, Zhang Z, Yang Z, Yu Y, Huang Y,He D, Xi Y, Wei D, Qi Q, Li W, Shi J, Wang M, Xie F,Wang J, Zhang X, Wang P, Zhao Y, Li N, Yang N, DongW, Hu S, Zeng C, Zheng W, Hao B, Hillier LW, Yang SP,Warren WC, Wilson RKBrandstro¨m M, Ellegren H, CrooijmansRP, van der Poel JJ, Bovenhuis H, Groenen MA,Ovcharenko I, Gordon L, Stubbs L, Lucas S, Glav<strong>in</strong>a T,Aerts A, Kaiser P, Rothwell L, Young JR, Rogers S, WalkerBA, van Hateren A, Kaufman J, Bumstead N, Lamont SJ,Zhou H, Hock<strong>in</strong>g PM, Morrice D, de Kon<strong>in</strong>g DJ, Law A,Bartley N, Burt DW, Hunt H, Cheng HH, Gunnarsson U,Wahlberg P, Andersson L, K<strong>in</strong>dlund E, Tammi MT,Andersson B, Webber C, Pont<strong>in</strong>g CP, Overton IM, BoardmanPE, Tang H, Hubbard SJ, Wilson SA, Yu J, Wang J,Yang H, International Chicken Polymorphism Map Consortium,2004: A genetic variation map for chicken with 2.8million s<strong>in</strong>gle-nucleotide polymorphisms. Nature 432, 717–722.Author’s address (for correspondence): JA Long, Animal Biosciencesand Biotechnology Laboratory, Beltsville Agricultural Research Center,US Department of Agriculture, Beltsville, MD 20705, USA. E-mail: jlong@anri.barc.usda.govConflict of <strong>in</strong>terest: The author declares no conflict of <strong>in</strong>terest.Ó 2008 No claim to orig<strong>in</strong>al government works


Reprod Dom Anim 43 (Suppl. 2), 89–95 (2008); doi: 10.1111/j.1439-0531.2008.01147.xISSN 0936-6768Genetic Improvement of Dairy Cow Reproductive PerformanceB BerglundDepartment of Animal Breed<strong>in</strong>g and Genetics, Swedish University of Agricultural Sciences and Centre for Reproductive Biology <strong>in</strong> Uppsala, Uppsala,SwedenContentsThe welfare of cow along with profitability <strong>in</strong> production areimportant issues <strong>in</strong> susta<strong>in</strong>able animal breed<strong>in</strong>g programmes.Along with an <strong>in</strong>tense ⁄ <strong>in</strong>tensive selection for <strong>in</strong>creased milkyield, reproductive performance has decl<strong>in</strong>ed <strong>in</strong> many countries,<strong>in</strong> part due to an unfavourable genetic relationship. Thelargely unchanged genetic trend <strong>in</strong> female fertility and calv<strong>in</strong>gtraits for Scand<strong>in</strong>avian Red breeds shows that it is possible toavoid deterioration <strong>in</strong> these traits if they are properly considered<strong>in</strong> the breed<strong>in</strong>g programme. Today’s breed<strong>in</strong>g is <strong>in</strong>ternationalwith a global selection and extensive use of the best bulls.The Nordic countries have traditionally recorded and performedgenetic evaluation for a broad range of functional traits<strong>in</strong>clud<strong>in</strong>g reproduction. In recent years many other countrieshave also implemented genetic evaluation for these traits. Thus,the relative emphasis of dairy cattle breed<strong>in</strong>g objectives hasgradually shifted from production to functional traits such asreproduction. Improved ways of record<strong>in</strong>g traits, e.g. physiologicalmeasures, early <strong>in</strong>dicator traits, assisted reproductivetechniques and <strong>in</strong>creased knowledge of genes and their regulationmay improve the genetic selection strategies and havelarge impact on present and future genetic evaluation programmes.Extensive data bases with phenotypic record<strong>in</strong>gs oftraits for <strong>in</strong>dividuals and their pedigree are a prerequisite.Quantitative trait loci have been associated to the reproductivecomplex. Most important traits, <strong>in</strong>clud<strong>in</strong>g reproduction traitsare regulated by a multitude of genes and environmental factors<strong>in</strong> a complex relationship, however. Genomic selection mighttherefore be important <strong>in</strong> future breed<strong>in</strong>g programmes. Informationon s<strong>in</strong>gle nucleotide polymorphism has already been<strong>in</strong>troduced <strong>in</strong> the selection programmes of some countries.IntroductionThere are strong motives for <strong>in</strong>clud<strong>in</strong>g reproduction <strong>in</strong>genetic selection programmes. A good reproductiveperformance is crucial for economic as well as ethicalreasons. Without reproduction there will be no animalproduction. The unfavourable genetic correlation withmilk production has led to a decl<strong>in</strong>e <strong>in</strong> reproduction <strong>in</strong>dairy cattle, at least <strong>in</strong> part due to an <strong>in</strong>sufficientconsideration of this trait when select<strong>in</strong>g for a highermilk production. There are several reports that the<strong>in</strong>creas<strong>in</strong>g use of Holste<strong>in</strong> genetics has caused decl<strong>in</strong><strong>in</strong>gfertility as well as calv<strong>in</strong>g performance dur<strong>in</strong>g the latestdecades (e.g. Berglund and Philipsson 1992; Royal et al.2000; Lucy 2001; Hansen et al. 2004). This has causeddeterioration <strong>in</strong> fertility <strong>in</strong> countries that heavily used thisbreed even if they have had reproductive performanceboth <strong>in</strong> the breed<strong>in</strong>g goal and <strong>in</strong> the selection criteria. For<strong>in</strong>stance, <strong>in</strong> Swedish Holste<strong>in</strong>s (SH), daughter fertilityhas fallen by approximately 1.2 <strong>in</strong>dex units per year overthe last fifteen years until now (L<strong>in</strong>dhe´ B, 2007: SvenskAvel, Skara, Sweden, personal communication), whereasthe Swedish Red breed (SRB), with similar milk yieldlevels and genetic trend, has largely ma<strong>in</strong>ta<strong>in</strong>ed fertility.A national breed<strong>in</strong>g programme consider<strong>in</strong>g reproductiontraits for more than three decades may expla<strong>in</strong> this.However for Holste<strong>in</strong>s the <strong>in</strong>clusion of fertility <strong>in</strong> theSwedish breed<strong>in</strong>g goal has not been enough to withstandthe large imports of genetic material from countries thathave low, or no weight<strong>in</strong>g on fertility <strong>in</strong> their breed<strong>in</strong>gobjective.In 2006 the World Holste<strong>in</strong> Friesian Federation<strong>in</strong>itiated a survey of the status on fertility <strong>in</strong> the Holste<strong>in</strong>population around the world. The conclusion from thissurvey was that fertility is a problem and actions need tobe taken both <strong>in</strong>ternationally and with<strong>in</strong> each country(Sørensen et al. 2007). Many countries have implementedgenetic evaluation for fertility traits <strong>in</strong> recentyears. More traits are gradually be<strong>in</strong>g evaluated andmore sophisticated evaluation methods are be<strong>in</strong>g implemented.Maybe the decl<strong>in</strong>e <strong>in</strong> fertility now has levelledout and reached a plateau, as was concluded from an<strong>in</strong>ternational conference on fertility <strong>in</strong> dairy cows held <strong>in</strong>Liverpool, 2007 (Crowe 2007, personal communication).Still the low level of reproductive performance is aproblem. <strong>Reproduction</strong> problems are among the mostcommon reason for cull<strong>in</strong>g <strong>in</strong> dairy production. This isthe major cause for <strong>in</strong>voluntary cull<strong>in</strong>g, e.g. <strong>in</strong> Swedishdairy cattle (Swedish Dairy Association 2007).Today’s breed<strong>in</strong>g is <strong>in</strong>ternational, <strong>in</strong>tensive and usesmodern reproductive and molecular genetic techniques.The welfare of cows along with profitability <strong>in</strong> productionare important issues <strong>in</strong> susta<strong>in</strong>able animal breed<strong>in</strong>gprogrammes. Nielsen et al. (2006) suggested that whendef<strong>in</strong><strong>in</strong>g breed<strong>in</strong>g goals for susta<strong>in</strong>able production,breed<strong>in</strong>g organizations should predict the selectionresponse based on market economic value and addnon-market value for traits with unacceptable selectionresponses. New ways of measur<strong>in</strong>g, record<strong>in</strong>g andanalys<strong>in</strong>g traits, new reproductive techniques and<strong>in</strong>creased knowledge of genes and their regulation mayimprove the genetic selection strategies and have a largeimpact on genetic evaluation programmes. This paperwill focus on the genetic improvement of the femalefertility and calv<strong>in</strong>g traits.Difficulties <strong>in</strong> Selection for <strong>Reproduction</strong>Many reproduction traits are difficult to handle <strong>in</strong>parameter estimation and genetic evaluation. In generalheritabilities are low, usually less than 5%, ma<strong>in</strong>ly due toa large <strong>in</strong>fluence of management and environmentaleffects. Most traits are not normally distributed and havecensored records, which complicates the analysis of thetraits. In addition to the other features of the reproductiveÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


90 B Berglundtraits calv<strong>in</strong>g performance traits are <strong>in</strong>fluenced by aneffect of the mother and a direct effect of the young(Ste<strong>in</strong>bock et al. 2003; Hansen et al. 2004) and it may bedifficult to correctly estimate these (co)variances. To getselection response <strong>in</strong> reproduction traits a well developedrecord<strong>in</strong>g scheme is needed and a breed<strong>in</strong>g structureallow<strong>in</strong>g large daughter group sizes.Several sub-traits<strong>Reproduction</strong>, especially female fertility, is a complextrait composed of several sub-traits. Thus it is importantto <strong>in</strong>clude all important aspects of fertility to achieve agood and expected selection response. A challenge <strong>in</strong>do<strong>in</strong>g this is the data collection and the quality of data.Traditionally, most fertility traits are based on calv<strong>in</strong>gand <strong>in</strong>sem<strong>in</strong>ation data and each trait has its strengthsand weaknesses. For female fertility there are measuresreflect<strong>in</strong>g the ability to resume oestrous cycles aftercalv<strong>in</strong>g and the ability to conceive, or measurescomb<strong>in</strong><strong>in</strong>g these abilities like calv<strong>in</strong>g to last <strong>in</strong>sem<strong>in</strong>ation(CLI), often also called days open (DO). Informationabout treatments and cull<strong>in</strong>g for reproductivedisorders are recorded and used ma<strong>in</strong>ly <strong>in</strong> the Nordiccountries. In Sweden scores for heat symptoms arerecorded as well. Heat detection plays a considerablerole for the economy. A visible manifestation of oestrusand a high oestrus detection rate is important especiallywhen us<strong>in</strong>g AI and <strong>in</strong> countries where hormones foroestrus synchronization are not generally used. Dobsonet al. (2007) reported from the UK that the number ofoestrous animals stand<strong>in</strong>g-to-be-mounted has decl<strong>in</strong>edfrom 80% to 50% over the past 30–50 years.Antagonistic genetic correlations to other traitsThe antagonistic relationship between fertility and milkproduction is well-known and was shown already byJanson and Andre´ asson (1981). These correlations wereconfirmed <strong>in</strong> studies by Roxstro¨ m et al. (2001b), whereunfavourable genetic correlation for, e.g. number of<strong>in</strong>sem<strong>in</strong>ations per service period, <strong>in</strong>terval from calv<strong>in</strong>gto first AI and treatments for reproductive disorders tomilk production was shown. The correlations rangedfrom 0.2 to 0.4, <strong>in</strong>creas<strong>in</strong>g with lactation number,possibly as a consequence of a higher energy demandwith <strong>in</strong>creas<strong>in</strong>g production level and steeper lactationcurves. Due to the antagonistic genetic correlation tomilk production, undesirable trends are expected forthe reproductive traits if they are not <strong>in</strong>cluded <strong>in</strong> thebreed<strong>in</strong>g goal. Even if <strong>in</strong>cluded <strong>in</strong> the breed<strong>in</strong>g objectivethere is a risk for deterioration of this trait due to thelow heritability if too small a breed<strong>in</strong>g goal weight ordaughter groups are used. Ethic values (cow welfare,consumer preference, etc.) should be put <strong>in</strong>to thebreed<strong>in</strong>g goal weight and those are not easily accessible.Moreover, there are unfavourable correlations amongthe reproduction traits that are important to consider.Negative energy balance and reproductionA top produc<strong>in</strong>g cow might not cope with the energyrequirements for both milk production and ma<strong>in</strong>ta<strong>in</strong>edreproduction and health even though the cows have acapacity to mobilize from their energy reserves. In ourstudies of Swedish Red (SRB) and Swedish Holste<strong>in</strong>s(SH) we have seen that SH has a th<strong>in</strong>ner layer ofsubcutaneous fat than SRB which may be important <strong>in</strong>this aspect (Hjertén 2006). A negative energy balance isassociated with <strong>in</strong>ferior embryo quality. A review paperon metabolic changes and embryo quality was given byChagas et al. (2007). Hayhurst et al. (2007a) found asignificant genetic variation <strong>in</strong> embryo quality andestimated a heritability of 0.13 for this trait, imply<strong>in</strong>ga possibility of genetically select<strong>in</strong>g cattle with <strong>in</strong>herentquality to produce high quality embryos.Body condition score (BCS) is an <strong>in</strong>ternationallyaccepted, rapid, <strong>in</strong>expensive and non-<strong>in</strong>vasive method ofestimat<strong>in</strong>g body energy reserves <strong>in</strong> dairy cattle (Berryet al. 2007). Even though ma<strong>in</strong>ly used for managementpurposes, BCS may also add to the <strong>in</strong>formation <strong>in</strong>genetic evaluations, especially when data on more directtraits are lack<strong>in</strong>g. BCS is moderately heritable (0.09–0.45) and favourably correlated to fertility and survival.BCS is used as a predictor trait for genetic merit forfertility <strong>in</strong> some countries, e.g. the Netherlands (De Jong2005) and <strong>in</strong> Irish and UK cattle (Berry et al. 2007).Current Status <strong>in</strong> Breed<strong>in</strong>g ProgrammesThe general breed<strong>in</strong>g goal for the reproduction of cows israther similar <strong>in</strong> different countries and may be formulatedas follows: cows that return to normal cyclicity earlyafter calv<strong>in</strong>g show strong and regular heats, and whichconceive when <strong>in</strong>sem<strong>in</strong>ated at a correct time. Furthermore,the cow should carry her pregnancy to term, have agood calv<strong>in</strong>g ability and give birth to viable calves.Female fertilityEarly studies by Janson (1980) showed that even thoughheritability for fertility traits was low, the additivegenetic variation was shown to be substantial. Furthermore,the importance of <strong>in</strong>clud<strong>in</strong>g both <strong>in</strong>terval andpregnancy measures at different ages <strong>in</strong> the geneticevaluation for fertility was underl<strong>in</strong>ed. These resultswere later confirmed <strong>in</strong> large field studies by Roxstro¨ met al. (2001a,b). The Nordic countries have traditionallyrecorded and performed genetic evaluation for a broadrange of functional traits <strong>in</strong>clud<strong>in</strong>g reproduction, but <strong>in</strong>recent years many other countries have also implementedgenetic evaluation for these traits. Thus, therelative emphasis of dairy cattle breed<strong>in</strong>g objectives hasgradually shifted from production to functional traitssuch as reproduction dur<strong>in</strong>g the past couple of decades(Miglior et al. 2005).Breed<strong>in</strong>g values for daughter fertility were <strong>in</strong>troduced<strong>in</strong> Sweden as early as 1972, and have s<strong>in</strong>ce been used <strong>in</strong>selection. In the Nordic countries, the <strong>in</strong>tegration of cowdata bases (pedigree, milk record<strong>in</strong>g, AI and diseasedata) has facilitated selection for reproductive traits.Three of the Nordic (Denmark, F<strong>in</strong>land and Sweden)breed<strong>in</strong>g organizations have had a jo<strong>in</strong>t genetic evaluationand breed<strong>in</strong>g programme s<strong>in</strong>ce 2005 (for more<strong>in</strong>formation see http://www.nordicebv.<strong>in</strong>fo). The Nordicfertility <strong>in</strong>dex <strong>in</strong>cludes the traits number of AI per serviceÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Genetic Improvement of Dairy Cow Reproductive Performance 91period for heifers (h) and cows (c), <strong>in</strong>terval from calv<strong>in</strong>gto first AI (c), <strong>in</strong>terval from first to last AI (h, c) andreproductive treatments (c). S<strong>in</strong>ce the mid 1990s geneticevaluation for fertility has gradually been <strong>in</strong>troduced <strong>in</strong>several countries, e.g. the Netherlands (Hoekstra et al.1994), UK (Wall et al. 2003) and US (VanRaden et al.2004). An <strong>in</strong>ternational genetic evaluation for fertilitytraits was <strong>in</strong>troduced for Holste<strong>in</strong> populations <strong>in</strong>February 2007 by Interbull and an evaluation of fertility<strong>in</strong> the other ma<strong>in</strong> breeds of the Interbull membercountries is under development (Jorjani 2007) (for more<strong>in</strong>formation see http://www.<strong>in</strong>terbull.org).Genetic trend for fertilityL<strong>in</strong>dhe´ and Philipsson (2001) found a clear unfavourablegenetic trend <strong>in</strong> female fertility for SwedishHolste<strong>in</strong>s and a slightly favourable genetic trend forthe Swedish Red breed. In the Norwegian Red breed notrend or a slightly favourable genetic trend for fertilitytraits was found (Chang et al. 2006). Traditional breed<strong>in</strong>gstrategies have been very successful <strong>in</strong> select<strong>in</strong>g highyield<strong>in</strong>g dairy cows (Fig. 1a,b). While functional traitssuch as reproductive performance have decl<strong>in</strong>ed <strong>in</strong> manycountries, especially for Holste<strong>in</strong>s, the largelyunchanged genetic trend <strong>in</strong> female fertility and calv<strong>in</strong>gtraits for Danish, F<strong>in</strong>nish and Swedish Red (Ayrshire(a) 115DFS/SWE scale DFS/SWE scale1059585(b) 115751985 1990 1995 2000Years1059585751985 1990 1995 2000YearsMIFAPRSCLOCECFDOCMFig. 1. (a) Genetic trends for functional traits and milk productiontraits for Red (Ayrshire type) breeds. DFS ⁄ SWE, Danish, F<strong>in</strong>nish andSwedish Red breeds on a Swedish scale for breed<strong>in</strong>g values (Jorjani2007, personal communication); MI, milk yield; FA, fat yield; PR,prote<strong>in</strong> yield; SC, somatic cell scores; LO, longevity; CE, direct calv<strong>in</strong>gease; CF, calv<strong>in</strong>g to first <strong>in</strong>sem<strong>in</strong>ation; DO, days open; CM, cl<strong>in</strong>icalmastitis. (b) Genetic trends for functional traits and milk productiontraits for Holste<strong>in</strong>sMIFAPRSCLOCECFDOCMtype) breeds (Fig. 1a,b) shows that it is possible to avoida deterioration <strong>in</strong> these traits if they are properlyconsidered <strong>in</strong> the breed<strong>in</strong>g programme. While forDanish, Swedish and F<strong>in</strong>nish Holste<strong>in</strong>s the breed<strong>in</strong>gprogramme could not fully compensate for the use offoreign bull father genetic material for which functionaltraits such as reproduction were not known.Calv<strong>in</strong>g performance traitsFor first-parity Holste<strong>in</strong> cows, calf mortality is a greatproblem (Berglund and Philipsson 1992) and nowadaysreports are many of their high stillbirth rates, 10–13% atfirst calv<strong>in</strong>g. Stillbirths are approximately twice as high forHolste<strong>in</strong> first-calvers as for Swedish Red (Fig. 2). The<strong>in</strong>creas<strong>in</strong>g trend for stillbirth <strong>in</strong> Holste<strong>in</strong>s has not beenaccompanied by an <strong>in</strong>crease <strong>in</strong> calv<strong>in</strong>g difficulty and thedivergent phenotypic trends may be an illustration of avitality problem. For second-calvers hardly any differences<strong>in</strong> calv<strong>in</strong>g difficulty and stillbirth exist between the breeds.In a post-mortem exam<strong>in</strong>ation of 76 calves from firstcalv<strong>in</strong>gHolste<strong>in</strong>s, one-third of the calves were bornwithout any visible defects or <strong>in</strong>juries (Berglund et al.2003) and only half of the calves were born with signs of adifficult calv<strong>in</strong>g, <strong>in</strong>dicat<strong>in</strong>g a vitality problem. The geneticcorrelations for stillbirth at first and second calv<strong>in</strong>g were0.45–0.48 for Swedish Holste<strong>in</strong>s (Ste<strong>in</strong>bock et al. 2003),but 0.83–0.85 for the Swedish Red breed (Ste<strong>in</strong>bock et al.2006). Thus there seem to be a genetic difference <strong>in</strong>vitality of calves between these breeds at first calv<strong>in</strong>g.There are probably multifactorial reasons beh<strong>in</strong>d<strong>in</strong>creas<strong>in</strong>g stillbirth rates. Adamec et al. (2005) recentlyshowed a consistently unfavourable effect of <strong>in</strong>breed<strong>in</strong>gon calv<strong>in</strong>g difficulty and stillbirth <strong>in</strong> US Holste<strong>in</strong>s, withlargest effects for first parity births. McParland et al.(2007) found that <strong>in</strong>breed<strong>in</strong>g had a deleterious effectupon most of the traits studied such as dystocia, stillbirthand calv<strong>in</strong>g <strong>in</strong>terval <strong>in</strong> Irish Holste<strong>in</strong>-Friesians. A maximizedgenetic ga<strong>in</strong> should be balanced aga<strong>in</strong>st a m<strong>in</strong>imized<strong>in</strong>breed<strong>in</strong>g. There are now programmes to be built<strong>in</strong>to the genetic evaluation programmes that maximizegenetic ga<strong>in</strong> while m<strong>in</strong>imiz<strong>in</strong>g <strong>in</strong>breed<strong>in</strong>g.Because calv<strong>in</strong>g difficulty and stillbirth ma<strong>in</strong>ly is aproblem for first parity cows, this category should be thema<strong>in</strong> source of <strong>in</strong>formation for genetic evaluation. Forcerta<strong>in</strong> breeds such as the Swedish Red breed the reliability<strong>in</strong> breed<strong>in</strong>g values for calv<strong>in</strong>g ability could be <strong>in</strong>creasedby <strong>in</strong>clud<strong>in</strong>g higher calv<strong>in</strong>g numbers (Ste<strong>in</strong>bock et al.2006). Genetic evaluations should consider both calv<strong>in</strong>gdifficulty and stillbirth as calf and maternal traits.Calv<strong>in</strong>g performance has been recorded <strong>in</strong> Swedens<strong>in</strong>ce the 1960s. The number of countries record<strong>in</strong>gcalv<strong>in</strong>g traits is <strong>in</strong>creas<strong>in</strong>g (Mark et al. 2005), and an<strong>in</strong>ternational genetic evaluation was <strong>in</strong>troduced <strong>in</strong> 2005(Jacobsen and Fikse 2005). Many of the member countriesof Interbull genetically evaluate calv<strong>in</strong>g difficulty andmost of them now also evaluate bulls for stillbirth, e.g. arout<strong>in</strong>e evaluation for stillbirth <strong>in</strong> Holste<strong>in</strong>s was implemented<strong>in</strong> the US <strong>in</strong> 2006 (Cole et al. 2007). Difficultcalv<strong>in</strong>g and stillbirth reduce reproductive performance(Bicalho et al. 2007), and genes associated with difficultbirth also reduce reproductive success (Lo´ pez de Maturanaet al. 2007).Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


92 B Berglund12108Percent64201986 1989 1992 1995 1998 2001 2004YearsStillbirths SH Calv<strong>in</strong>g diff. SH Stillbirths SRB Calv<strong>in</strong>g diff. SRBFig. 2. Phenotypic trends <strong>in</strong> stillbirth rate and calv<strong>in</strong>g difficulty for first calvers of SLB ⁄ Swedish Holste<strong>in</strong>s (SH) and Swedish Red (SRB) (Swedishmilk record<strong>in</strong>g statistics)Genetic trend for calv<strong>in</strong>g traitsMark et al. (2005) reported a slightly negative genetictrend from the Interbull genetic evaluations for maternalstillbirth <strong>in</strong> the Holste<strong>in</strong> breed s<strong>in</strong>ce the early 1990swhile there was no clear trend <strong>in</strong> the other calv<strong>in</strong>g traits.For the Swedish and Norwegian Red breeds, no orslightly positive genetic trends for calv<strong>in</strong>g performancehave been observed (Her<strong>in</strong>gstad et al. 2007; L<strong>in</strong>dhé,2007, personal communication). The genetic trends fordirect calv<strong>in</strong>g ease for Danish, F<strong>in</strong>nish and Swedish Redbreed and Holste<strong>in</strong>s can be seen as one of the functionaltraits <strong>in</strong> Fig. 1a,b.Genetic defects caus<strong>in</strong>g reproduction lossMutations <strong>in</strong> s<strong>in</strong>gle genes occur regularly and may causecongenital genetic defects. These are commonly recessively<strong>in</strong>herited (http://www.omia.angis.org.au). Onerecent example is the complex vertebral malformation(CVM) <strong>in</strong> the Holste<strong>in</strong> breed, first described by Agerholmet al. (2001). Complex vertebral malformation has had amajor impact on the reproductive performance <strong>in</strong>Holste<strong>in</strong>s (Agerholm 2007). In a study of SwedishHolste<strong>in</strong>s, carriers of the CVM-gene had <strong>in</strong>ferior NRrate compared with non-carrier bulls reflect<strong>in</strong>g a higher<strong>in</strong>tra-uter<strong>in</strong>e mortality (Berglund et al. 2004). It isimportant to report all k<strong>in</strong>ds of malformations and tohave national control programmes for congenital defects<strong>in</strong> order to avoid multiplication of deleterious genescaus<strong>in</strong>g reproductive losses and animal welfare problems.Crossbreed<strong>in</strong>g as a tool to enhance fertilityDecreas<strong>in</strong>g fertility and <strong>in</strong>creas<strong>in</strong>g calf mortality <strong>in</strong>Holste<strong>in</strong>s have become a large problem. In some countries,e.g. <strong>in</strong> the USA, Scand<strong>in</strong>avian Red and other fertilebreeds are now crossed <strong>in</strong>to Holste<strong>in</strong>s to improve theHolste<strong>in</strong> reproduction. Crossbreed<strong>in</strong>g may help elevatethe level of traits comb<strong>in</strong><strong>in</strong>g breeds with favourable traitsand by explor<strong>in</strong>g heterosis effects, but the cont<strong>in</strong>uousgenetic improvement has to be done <strong>in</strong> the pure breeds.He<strong>in</strong>s et al. (2006a) reported that calves from Holste<strong>in</strong>first-calf heifers sired by Scand<strong>in</strong>avian Red (NorwegianRed and Swedish Red) bulls had significantly less calv<strong>in</strong>gdifficulty (5.5%) and lower stillbirth rate (7.7%) thancalves sired by Holste<strong>in</strong> bulls (16.4% difficult calv<strong>in</strong>g and15.1% stillbirth). Moreover, DO were significantlyshorter for all crossbred groups studied and were129 days for Scand<strong>in</strong>avian Red ⁄ Holste<strong>in</strong> crossbredsand 150 days for pure Holste<strong>in</strong>s (He<strong>in</strong>s et al. 2006b).New Tools for Genetic Improvement of<strong>Reproduction</strong>Improved ways of record<strong>in</strong>g traits, e.g. direct measur<strong>in</strong>gof physiological measures may offer valuable opportunitiesto improve the genetic evaluation of fertility.Indicator traits are valuable because fertility has a lowheritability and is expressed late <strong>in</strong> life. More advancedand expensive record<strong>in</strong>g technologies may be used <strong>in</strong>nucleus herds and the genetic progress may be enhancedby us<strong>in</strong>g modern reproductive techniques such asMOET. Reproductive techniques like sex<strong>in</strong>g, clon<strong>in</strong>gand transfer of genetic material will impact present andfuture selection strategies <strong>in</strong> breed<strong>in</strong>g programmes.The amount of <strong>in</strong>formation on the molecular level israpidly <strong>in</strong>creas<strong>in</strong>g. The sequenc<strong>in</strong>g of the bov<strong>in</strong>e genomewas completed <strong>in</strong> 2003 (http://www.hgsc.bcm.tmc.edu/projects/bov<strong>in</strong>e). Markers are connected to the phenotypicexpression of several traits <strong>in</strong> the reproductioncomplex. Extensive data bases with phenotypic record<strong>in</strong>gsof traits for <strong>in</strong>dividuals and their pedigree are aprerequisite. Gene expression profiles may <strong>in</strong>crease ourunderstand<strong>in</strong>g of the mechanisms beh<strong>in</strong>d reproductivefunctions and their phenotypic expression. As genes fortraits are identified, genetic selection strategies can beimproved. Most important traits, <strong>in</strong>clud<strong>in</strong>g reproductiontraits are regulated by a multitude of genes and environmentalfactors <strong>in</strong> a complex relationship, however.Progesterone-based measures of female fertilityEarly measures of the reproductive function of cow canbe obta<strong>in</strong>ed from progesterone profiles. Progesterone-Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Genetic Improvement of Dairy Cow Reproductive Performance 93based measures of fertility have higher heritability thanthe traditional measures of fertility (Royal et al. 2002;Petersson et al. 2007). Veerkamp et al. (1998) suggestedthat selection on days to first luteal activity (CLA) basedon monthly analysis of progesterone <strong>in</strong> milk may addfurther accuracy to the genetic evaluation for fertility.Van der Lende et al. (2004) suggested to select siresbased on a measure when 50% of the daughters of a sirehad an active corpus luteum (CLA 50%) based on 3- to6-week <strong>in</strong>tervals of progesterone sampl<strong>in</strong>g. Peterssonet al. (2007) found that direct selection on progesteronebased measures of fertility may <strong>in</strong>crease the accuracy <strong>in</strong>the genetic evaluation for an early start of cyclic ovarianactivity after calv<strong>in</strong>g compared to the commonly usedmeasure CFI, even with an <strong>in</strong>frequent sampl<strong>in</strong>g such as<strong>in</strong> the regular milk record<strong>in</strong>g system. Progesteroneanalysis <strong>in</strong> the first monthly collected milk samplescould also be used as a management tool. Peterssonet al. (2008) showed that four out of five cows withdelayed cyclicity could be predicted with<strong>in</strong> 60 days aftercalv<strong>in</strong>g enabl<strong>in</strong>g an earlier treatment. The cost forprogesterone analysis of milk samples could thereforebenefit from management returns as well as fromimprovements <strong>in</strong> genetic ga<strong>in</strong> for fertility.Automization of record<strong>in</strong>gsAutomatic milk<strong>in</strong>g systems, e.g. robotic milk<strong>in</strong>g systemsmay allow automization of record<strong>in</strong>gs. Commercialsystems for on-l<strong>in</strong>e record<strong>in</strong>gs allow<strong>in</strong>g herd-profiles offertility (e.g. based on progesterone analysis) and healthparameters are underway (Friggens and Løvendahl2007). These may offer record<strong>in</strong>gs with a high accuracythat could be built <strong>in</strong>to selection programmes. Løvendahland Chagunda (2006) used activity meters forautomatic heat controls and estimated a heritability of0.17 for this trait.Juvenile predictorsThe genes controll<strong>in</strong>g fertility are present and potentiallyexpressed early <strong>in</strong> life, but it takes at least four yearsbefore a bull has milk<strong>in</strong>g daughters and can be progenytested for fertility. Thus early fertility predictors would bevery valuable. These could be reproductive hormones ormetabolic traits. Hayhurst et al. (2007b) suggested apossibility of us<strong>in</strong>g the correlation between the prepubertalresponse to gonadotroph<strong>in</strong> releas<strong>in</strong>g hormone <strong>in</strong>bull calves and the fertility of their daughters as a possibleselection tool. In another study by Hayhurst et al.(2007c), it was suggested that selection for bull calveswith lower concentrations of glucose and FFA couldresult <strong>in</strong> female offspr<strong>in</strong>g with genetically better fertility.Gene mapp<strong>in</strong>g studiesThe identification of quantitative trait loci (QTL) is afirst step towards novel selection methods based on bothphenotypic and molecular <strong>in</strong>formation. Us<strong>in</strong>g QTL <strong>in</strong>selection is most beneficial for low heritability traits, sexlimited traits and traits expressed late <strong>in</strong> life such asdaughter fertility. Holmberg and Andersson-Eklund(2006) found regions with several QTLs on chromosome9 and 11 for both reproduction and health traits. A QTLfor non-return rate was f<strong>in</strong>e mapped to an <strong>in</strong>terval ofless than 3 cM on chromosome 9 (Holmberg et al.2007). Marker assisted selection (MAS) can be used forpre-selection among full-sibs before progeny test<strong>in</strong>g byaccount<strong>in</strong>g for the Mendelian sampl<strong>in</strong>g and also toavoid genetic defects for which there are availablemarkers. For quantitative traits the benefit from MAS islimited by the proportion of the genetic varianceexpla<strong>in</strong>ed by known QTL. The marker density <strong>in</strong> theQTL region can be <strong>in</strong>creased by use of s<strong>in</strong>gle nucleotidepolymorphism (SNP) markers. S<strong>in</strong>gle nucleotide polymorphismsare s<strong>in</strong>gle base-pair differences between<strong>in</strong>dividuals with<strong>in</strong> a species and where the differentvariants (most often only two allelic forms) are relativelycommon <strong>in</strong> a population.Expression profiles of genes regulat<strong>in</strong>g reproductionGene expression profil<strong>in</strong>g is a relatively recent toolcontribut<strong>in</strong>g to our knowledge about the various processesunderly<strong>in</strong>g reproduction such as the function ofgenes and their products determ<strong>in</strong><strong>in</strong>g the phenotype forreproduction. Beerda and Veerkamp (2006) summarizedgene expression studies related to reproduction <strong>in</strong> cattle,sheep and sw<strong>in</strong>e <strong>in</strong> a paper at the World Congress onGenetics Applied to Livestock Production <strong>in</strong> Brazil.They concluded that a vast amount of <strong>in</strong>formation hasalready been achieved <strong>in</strong> this area and as <strong>in</strong>formationwill grow exponentially over the next few years theyunderl<strong>in</strong>ed the importance of a major effort <strong>in</strong> br<strong>in</strong>g<strong>in</strong>gall <strong>in</strong>formation together <strong>in</strong> a ‘broadly accessible ontology’.The need for compil<strong>in</strong>g and analys<strong>in</strong>g largeamounts of molecular data has created a new field ofscience referred to as bio<strong>in</strong>formatics. An EU <strong>in</strong>tegratedproject SABRE was started <strong>in</strong> 2006, which aims toprovide fundamental knowledge on the genomics andepigenetics on, e.g. reproduction traits <strong>in</strong> dairy cattle tobe used <strong>in</strong> selection for improved reproduction efficiency.An update on studies on expression profiles ofgenes regulat<strong>in</strong>g dairy cow fertility was given at theInternational conference on fertility <strong>in</strong> dairy cows <strong>in</strong>Liverpool, 2007 (Beerda and Veerkamp 2007).Genomic selection and <strong>in</strong>tegration of molecular data <strong>in</strong>togenetic evaluation programmesGenomics is the study of variation of base pairs <strong>in</strong> thenucleic acids and genomic selection means us<strong>in</strong>g this<strong>in</strong>formation <strong>in</strong> genetic selection programmes. The availabilityof large arrays of SNPs is chang<strong>in</strong>g the approachof predict<strong>in</strong>g breed<strong>in</strong>g values from molecular <strong>in</strong>formation.Genomic selection (GMAS) uses all markers, orrather haplotypes consist<strong>in</strong>g of a pair of contiguousSNPs, spann<strong>in</strong>g the genome for prediction of breed<strong>in</strong>gvalues. Thus, by summ<strong>in</strong>g the effects of all markerhaplotypes <strong>in</strong> the genome of a bull calf, a breed<strong>in</strong>g valueis obta<strong>in</strong>ed directly at birth whereby the generation<strong>in</strong>terval can be considerably shortened. This <strong>in</strong>formationcan be used directly, hence theoretically no progenytest<strong>in</strong>g is needed. Muir (2007) showed that by us<strong>in</strong>gGMAS for traits of high (0.5) or low heritability (0.1) theaccuracy of selection <strong>in</strong>creased between 10% and 30%.Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


94 B BerglundVan Raden (2007) calculated measures of relationshipand <strong>in</strong>breed<strong>in</strong>g by use of genomic measures and demonstratedga<strong>in</strong>s <strong>in</strong> reliability as compared to us<strong>in</strong>g traditionalmethods, and comparatively more for lowheritability traits than high heritability traits. A disadvantageis that the <strong>in</strong>formation may only be used forsome generations, maybe 7–8 generations, because theestimated effects of SNP haplotypes will change overtime. Furthermore, without progeny test<strong>in</strong>g <strong>in</strong> eachgeneration the risk for late detection of undesired sideeffectsof selection might <strong>in</strong>crease.Along with <strong>in</strong>creas<strong>in</strong>g knowledge at the genomic level,effort is put on how to <strong>in</strong>tegrate these data <strong>in</strong> the geneticevaluation systems. Gengler and Verkenne (2007) statedthat it will be a challenge to <strong>in</strong>tegrate molecular andphenotypic data and that SNPs and genomic selectionwill not change this situation.Genomic selection based on 3000 SNP markers toselect young bulls was <strong>in</strong>troduced <strong>in</strong> the Netherlands <strong>in</strong>October 2006 (Van der Beek 2007). Denmark andNorway have also recently <strong>in</strong>troduced SNP <strong>in</strong>formation<strong>in</strong> their genetic selection programmes (MS Lund andS Lien, personal communication). Large scale projectsare ongo<strong>in</strong>g <strong>in</strong> several livestock species to identify andvalidate several thousands of SNPs <strong>in</strong> haplotype blocks<strong>in</strong> the genome. SNP chips with 60 000 SNPs will soonbe available.Conclud<strong>in</strong>g RemarksThe genetic trend for functional traits <strong>in</strong> the Red(Ayrshire) breeds shows that it is possible to ma<strong>in</strong>ta<strong>in</strong>a good dairy cow fertility and calv<strong>in</strong>g performancealong with selection for <strong>in</strong>creased levels of milk production,if the traits are properly considered. It ispossible to make improvements <strong>in</strong> breed<strong>in</strong>g, as well as <strong>in</strong>nutrition and management to keep a good reproductiveability of the cow and a good health and well-be<strong>in</strong>g <strong>in</strong>susta<strong>in</strong>able breed<strong>in</strong>g programmes. Along with <strong>in</strong>creas<strong>in</strong>gknowledge at a molecular level, large changes maybe expected <strong>in</strong> genetic evaluation programmes.AcknowledgementsI would like to acknowledge Professor Erl<strong>in</strong>g Strandberg at theDepartment of Animal Breed<strong>in</strong>g and Genetics, SLU, for valuablecomments on the manuscript. Associate Professor Hosse<strong>in</strong> Jorjani atthe Interbull office (Jorjani 2007, communication), SLU, for k<strong>in</strong>dlyprovid<strong>in</strong>g Interbull genetic trends and Associate Professor BengtL<strong>in</strong>dhé at Svensk Avel (L<strong>in</strong>dhé 2007, communication) for k<strong>in</strong>dlyprovid<strong>in</strong>g national genetic trends.ReferencesAdamec V, Cassell BG, Smith EP, Pearson RE, 2005: Effectsof <strong>in</strong>breed<strong>in</strong>g <strong>in</strong> the dam on dystocia and stillbirths <strong>in</strong> USHolste<strong>in</strong>s. J Dairy Sci 89, 307–314.Agerholm JS, 2007: Complex vertebral malformation <strong>in</strong>Holste<strong>in</strong> cattle: the story so far. Per<strong>in</strong>atal death <strong>in</strong> domesticanimals. Proc 20th NKVet Symp, Reykjavik, Iceland, p. 88.Agerholm JS, Bendixen C, Andersen O, Arnbjerg J, 2001:Complex vertebral malformation <strong>in</strong> Holste<strong>in</strong> calves. J VetDiagn Invest 13, 283–289.Beerda B, Veerkamp RF, 2006: Functional genomics of femalereproduction. 8th World Congress on Genetics Applied toLivestock Production, Belo Horizonte, Brazil. Proc Commun11–10 (http://www.wcgalp8.org.br).Beerda B, Veerkamp RF, 2007: Expression profiles of genesregulat<strong>in</strong>g dairy cow fertility: recent f<strong>in</strong>d<strong>in</strong>gs, ongo<strong>in</strong>gactivities and future possibilities. Proc Fertility <strong>in</strong> DairyCows – bridg<strong>in</strong>g the gaps, 30–31 August 2007, LiverpoolHope University, Liverpool, UK, p. 33.Berglund B, Philipsson J, 1992: Increas<strong>in</strong>g stillbirth rates <strong>in</strong> theSwedish Friesian population. Paper <strong>in</strong> 43rd Annual Meet<strong>in</strong>gof the European Association on Animal Production,Madrid, 14–17 September 1992.Berglund B, Ste<strong>in</strong>bock L, Elvander M, 2003: Causes ofstillbirth and time of death <strong>in</strong> Swedish Holste<strong>in</strong> calvespost-mortem exam<strong>in</strong>ed. Acta Vet Scand 44, 111–120.Berglund B, Persson A, Stålhammar H, 2004: Effects ofcomplex vertebral malformation on fertility <strong>in</strong> SwedishHolste<strong>in</strong> cattle. Acta Vet Scand 45, 161–165.Berry D, Roche J, Coffey M, 2007: Body condition score andfertility – more than just a feel<strong>in</strong>g. Proc Fertility <strong>in</strong> DairyCows – bridg<strong>in</strong>g the gaps, 30–31 August 2007, LiverpoolHope University, Liverpool, UK, p. 28.Bicalho RC, Galvao KN, Cheong SH, Gilbert RO, WarnikLD, Guard CL, 2007: Effect of stillbirths on dam survivaland reproduction performance <strong>in</strong> Holste<strong>in</strong> dairy cows.J Dairy Sci 90, 2797–2803.Chagas LM, Bass JJ, Blache D, Burke CR, Kay JK, L<strong>in</strong>dsayDR, Lucy MC, Mart<strong>in</strong> GB, Meier S, Rhodes FM, RocheJR, Thatcher WW, Webb R, 2007: Invited review: Newperspectives on the roles of nutrition and metabolic priorities<strong>in</strong> the subfertility of high-produc<strong>in</strong>g dairy cows. J DairySci 90, 4022–4032.Chang YM, Andersen-Ranberg IM, Her<strong>in</strong>gstad B, Gianola D,Klemetsdal G, 2006: Bivariate analysis of number of servicesto conception and days open <strong>in</strong> Norwegian Red us<strong>in</strong>g acensored threshold-l<strong>in</strong>ear model. J Dairy Sci 89, 772–778.Cole JB, Wiggans GR, VanRaden PM, Miller RH, 2007:Stillbirth (co)variance components for a sire-maternalgrandsire threshold model and development of a calv<strong>in</strong>gability <strong>in</strong>dex for sire selection. J Dairy Sci 90, 2489–2496.De Jong G, 2005: Usage of predictors for fertility <strong>in</strong> the geneticevaluation application <strong>in</strong> the Netherlands. Interbull Bull 33,69–73.Dobson H, Walker S, Morris M, Routly J, Smith R, 2007:Why is it gett<strong>in</strong>g more difficult to successfully artificially<strong>in</strong>sem<strong>in</strong>ate cows? Proc Fertility <strong>in</strong> Dairy Cows – Bridg<strong>in</strong>gthe Gaps, 30–31 August 2007, Liverpool Hope University,Liverpool, UK, p. 4.Friggens N, Løvendahl P, 2007: Where did she come from?Where is she go<strong>in</strong>g? The potential of on-farm fertility profiles.Proc. Fertility <strong>in</strong> Dairy Cows – Bridg<strong>in</strong>g the Gaps, 30–31August 2007, LiverpoolHope University, Liverpool,UK, p.23.Gengler N, Verkenne C, 2007: Bayesian approach to <strong>in</strong>tegratemolecular data <strong>in</strong>to genetic evaluations. Interbull Bull 37,37–41.Hansen M, Lund MS, Pedersen J, Christensen LG, 2004:Genetic parameters for stillbirth <strong>in</strong> Danish Holste<strong>in</strong> cowsus<strong>in</strong>g a Bayesian threshold model. Livest Prod Sci 91, 23–33.Hayhurst C, Firth M, Christie MF, Royal MD, 2007a:Estimation of genetic variation <strong>in</strong> embryo quality: Is therepotential to genetically select cattle with the <strong>in</strong>herent abilityto produce high quality embryos? Proc Fertility <strong>in</strong> DairyCows – Bridg<strong>in</strong>g the Gaps, 30–31 August 2007, LiverpoolHope University, Liverpool, UK, p. 13.Hayhurst C, Fl<strong>in</strong>t APF, Lovendahl P, Wolliams JA, RoyalMD, 2007b: Prepubertal predictors for fertility <strong>in</strong> dairycattle: potential use of metabolic hormones. Proc BSASAnnual Conference 2007: Southport, UK, 58.Hayhurst C, Sorensen MK, Royal MD, Løvendahl P, 2007c:Metabolic regulation <strong>in</strong> Danish bull calves and the relation-Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Genetic Improvement of Dairy Cow Reproductive Performance 95ship to the fertility of their female offspr<strong>in</strong>g. J Dairy Sci 90,3909–3916.He<strong>in</strong>s BJ, Hansen LB, Seykora AJ, 2006a: Fertility andsurvival of pure Holste<strong>in</strong>s versus crossbreds of Holste<strong>in</strong> withNormande, Montbeliarde, and Scand<strong>in</strong>avian Red. J DairySci 89, 4944–4951.He<strong>in</strong>s BJ, Hansen LB, Seykora AJ, 2006b: Calv<strong>in</strong>g difficultyand stillbirths of pure Holste<strong>in</strong>s versus crossbreds ofHolste<strong>in</strong> with Normande, Montbeliarde, and Scand<strong>in</strong>avianRed. J Dairy Sci 89, 2805–2810.Her<strong>in</strong>gstad B, Chang YM, Svendsen M, Gianola D, 2007:Genetic analysis of calv<strong>in</strong>g difficulty and stillbirth <strong>in</strong>Norwegian Red cows. J Dairy Sci 90, 3500–3507.Hjerte´ n J, 2006: Relationships between body condition score,subcutaneous fat, live weight and reproduction <strong>in</strong> SwedishHolste<strong>in</strong> and Swedish Red and White Cattle. In Swedish.Summary <strong>in</strong> English. MSc Thesis 281, SLU, Department ofAnimal Breed<strong>in</strong>g and Genetics, Uppsala, Sweden.Hoekstra J, van der Lugt AW, van der Werf JHJ, OuweltjesW, 1994: Genetic and phenotypic parameters for milkproduction and fertility traits <strong>in</strong> upgraded dairy cattle.Livest Prod Sci 40, 225–232.Holmberg M, Andersson-Eklund L, 2006: Quantitative traitloci affect<strong>in</strong>g fertility and calv<strong>in</strong>g traits <strong>in</strong> Swedish dairycattle. J Dairy Sci 89, 3664–3671.Holmberg M, Sahana G, Andersson-Eklund L, 2007: F<strong>in</strong>emapp<strong>in</strong>g of a quantitative trait locus on chromosome 9affect<strong>in</strong>g non-return rate <strong>in</strong> Swedish dairy cattle. J AnimBreed 124, 257–263.Jacobsen JH, Fikse F, 2005: Feasibility of MACE for calv<strong>in</strong>gtraits for non-Holste<strong>in</strong> breeds. Interbull Bull 33, 28–31.Janson L, 1980: Studies on fertility traits <strong>in</strong> Swedish dairycattle. II. Genetic parameters. Acta Agr Scand 30, 427–436.Janson L, Andre´ asson B, 1981: Studies on fertility traits <strong>in</strong>Swedish dairy cattle. IV. Genetic and phenotypic correlationbetween milk yield and fertility. Acta Agr Scand 31, 313–322.Jorjani J, 2007: International genetic evaluation of femalefertility traits <strong>in</strong> five major breeds. Interbull Bull 37, 144–147.L<strong>in</strong>dhe´ B, Philipsson J, 2001: Genetic trends <strong>in</strong> the twoSwedish dairy cattle breeds SRB and SLB <strong>in</strong> 1985–1999.SLU, Department of Animal Breed<strong>in</strong>g and Genetics, Publ.138, Uppsala, Sweden.Lo´ pez de Maturana E, Legarra A, Varona L, Ugarte E, 2007:Analysis of fertility and dystocia <strong>in</strong> Holste<strong>in</strong>s us<strong>in</strong>g recursivemodels to handle censored and categorical data. J Dairy Sci90, 2012–2024.Løvendahl P, Chagunda MGG, 2006: Assessment of fertility <strong>in</strong>dairy cows based on electronic monitor<strong>in</strong>g of their physicalactivity. 8th World Congress on Genetics Applied toLivestock Production, Belo Horizonte, Brazil. Proc Commun18-04 (http://www.wcgalp8.org.br).Lucy MC, 2001: Reproductive loss <strong>in</strong> high-produc<strong>in</strong>g dairycattle: where will it end? J Dairy Sci 84, 1277–1293.Mark T, Jakobsen JH, Jorjani H, Fikse WF, Philipsson J,2005: International trends <strong>in</strong> record<strong>in</strong>g and genetic evaluationof functional traits <strong>in</strong> dairy cattle. 56th Annual Meet ofthe European Association for Animal Production, Uppsala,Sweden, Abstract no. 536.McParland S, Kearney JF, Rath M, Berry DP, 2007:Inbreed<strong>in</strong>g effects on milk production, calv<strong>in</strong>g performance,fertility and conformation <strong>in</strong> Irish Holste<strong>in</strong>-Friesians.J Dairy Sci 90, 4411–4419.Miglior F, Muir BL, Van Doormaal BJ, 2005: Selection <strong>in</strong>dices<strong>in</strong> Holste<strong>in</strong> cattle of various countries. J Dairy Sci 88, 1255–1263.Muir WM, 2007: Genomic selection: a break through forapplication of marker assisted selection to traits of lowheritability, promise and concerns. Paper at the 58th AnnualMeet of the European Association for Animal Production,Dubl<strong>in</strong>, Ireland.Nielsen HM, Christensen LG, Ødega˚ rd J, 2006: A method todef<strong>in</strong>e breed<strong>in</strong>g goals for susta<strong>in</strong>able dairy cattle production.J Dairy Sci 89, 3615–3625.Petersson K-J, Berglund B, Strandberg E, Gustafsson H, Fl<strong>in</strong>tAPF, Wolliams JA, Royal MD, 2007: Genetic analysis ofpostpartum measures of luteal activity <strong>in</strong> dairy cows.J Dairy Sci 90, 427–434.Petersson K-J, Strandberg E, Gustafsson H, Royal MD,Berglund B, 2008: Detection of delayed cyclicity <strong>in</strong> dairycows based on progesterone content <strong>in</strong> monthly milksamples. Prev Vet Med (<strong>in</strong> press).Roxstro¨ m A, Strandberg E, Berglund B, Emanuelson U,Philipsson J, 2001a: Genetic and environmental correlationsamong female fertility traits and milk production <strong>in</strong> differentparities of Swedish Red and White dairy cattle. Acta AgricScand A Anim Sci 51, 7–14.Roxstro¨ m A, Strandberg E, Berglund B, Emanuelson U,Philipsson J, 2001b: Genetic and environmental correlationsamong female fertility traits and the ability to show oestrus,and milk production. Acta Agric Scand A Anim Sci 51, 192–199.Royal MD, Mann GE, Fl<strong>in</strong>t APF, 2000: Strategies forrevers<strong>in</strong>g the trend towards subfertility <strong>in</strong> dairy cattle. VetJ 160, 53–60.Royal MD, Fl<strong>in</strong>t APF, Wolliams JA, 2002: Genetic andphenotypic relationships among endocr<strong>in</strong>e and traditionalfertility traits and production traits <strong>in</strong> Holste<strong>in</strong>-Friesiandairy cows. J Dairy Sci 85, 958–967.Sørensen AC, Lawlor T, Ruiz F, 2007: A survey on fertility <strong>in</strong>the Holste<strong>in</strong> populations of the world. Proc Fertility <strong>in</strong>Dairy Cows – Bridg<strong>in</strong>g the Gaps, 30–31 August 2007,Liverpool Hope University, Liverpool, UK, p. 17.Ste<strong>in</strong>bock L, Näsholm A, Berglund B, Johansson K, PhilipssonJ, 2003: Genetic effects on stillbirth and calv<strong>in</strong>g difficulty<strong>in</strong> Swedish Holste<strong>in</strong>s at first and second calv<strong>in</strong>g. J Dairy Sci86, 2228–2235.Ste<strong>in</strong>bock L, Johansson K, Näsholm A, Berglund B, PhilipssonJ, 2006: Genetic effects on stillbirth and calv<strong>in</strong>g difficulty<strong>in</strong> Swedish Red dairy cattle at first and second calv<strong>in</strong>g. ActaAgric Scand A, 56, 65–72.Van der Beek S, 2007: Effect of genomic selection on nationaland <strong>in</strong>ternational genetic evaluations. Interbull Bull 37, 115–118.Van der Lende T, Kaal LMTE, Roelofs RMG, Veerkamp RF,Schrooten C, Bovenhuis H, 2004: Infrequent milk progesteronemeasurements <strong>in</strong> daughters enable bull selection forcow fertility. J Dairy Sci 87, 3953–3957.VanRaden PM, 2007: Genomic measures of relationship and<strong>in</strong>breed<strong>in</strong>g. Interbull Bull 37, 33–36.VanRaden PM, Sanders AH, Tooker ME, Miller RH,Norman HD, Kuhn MT, Wiggans GR, 2004: Developmentof a national genetic evaluation for cow fertility. J Dairy Sci87, 2285–2292.Veerkamp RF, Oldenbroek JK, van der Lende T, 1998: The useof milk progesterone measurements for genetic improvementof fertility traits <strong>in</strong> dairy cattle. Interbull Bull 18, 62–67.Wall E, Brotherstone S, Wolliams JA, Banos G, Coffey MP,2003: Genetic evaluation of fertility us<strong>in</strong>g direct andcorrelated traits. J Dairy Sci 86, 4093–4102.Author’s address (for correspondence): B Berglund, Department ofAnimal Breed<strong>in</strong>g and Genetics, Swedish University of AgriculturalSciences, Box 7023, SE-750 07, Uppsala, Sweden. E-mail: britt.berglund@hgen.slu.seConflict of <strong>in</strong>terest: The author declares no conflict of <strong>in</strong>terest.Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 96–103 (2008); doi: 10.1111/j.1439-0531.2008.01148.xISSN 0936-6768Nutrient Prioritization <strong>in</strong> Dairy Cows Early Postpartum: Mismatch BetweenMetabolism and Fertility?JLMR Leroy 1 , T Vanholder 1 , ATM Van Knegsel 2 , I Garcia-Ispierto 3 and PEJ Bols 11 Veter<strong>in</strong>ary Centre Someren, Someren; 2 Adaptation Physiology Group and Animal Nutrition Group, Wagen<strong>in</strong>gen Institute of Animal Sciences,Wagen<strong>in</strong>gen University, Wagen<strong>in</strong>gen, The Netherlands; 3 Department of Animal Health and Anatomy, Autonomous University of Barcelona,Barcelona, Spa<strong>in</strong>ContentsFor several decades, researchers worldwide report a decrease<strong>in</strong> fertility <strong>in</strong> high-yield<strong>in</strong>g dairy cows, most probably based onconflict<strong>in</strong>g metabolic and reproductive needs. The dairy herdmanager’s success at improv<strong>in</strong>g milk production has beenaccompanied by a negative trend for the most visible reproductiveparameters such as calv<strong>in</strong>g <strong>in</strong>tervals, number of daysopen and number of <strong>in</strong>sem<strong>in</strong>ations needed per pregnancy. Inparallel, many research groups studied the metabolic andendocr<strong>in</strong>e factors that <strong>in</strong>fluence follicular growth and thedevelopmental competence of oocytes and embryos. In thepast, herd managers and reproductive biologists each tried totackle the same problems with limited consultation. Morerecently, the situation has improved significantly and theriogenologists,nutritionists and veter<strong>in</strong>arians now conductresearch <strong>in</strong> multidiscipl<strong>in</strong>ary teams. This review paper starts<strong>in</strong> a general way by discuss<strong>in</strong>g nutrient prioritization towardsthe udder to guarantee milk production and by describ<strong>in</strong>g<strong>in</strong>teractions between the somatotropic and gonadotropic axis.It then focuses on the consequences of the negative energybalance on follicular growth and environment, oocyte andembryo quality, not only by summariz<strong>in</strong>g the currentlyaccepted hypotheses but also based on clear scientific evidenceat the follicular level. All this, with one question <strong>in</strong> m<strong>in</strong>d: isthere a mismatch between metabolism and fertility and whatcan the dairy manager learn from research to tackle theproblem of reduced fertility?IntroductionDisappo<strong>in</strong>t<strong>in</strong>g reproductive performance <strong>in</strong> high-produc<strong>in</strong>gdairy herds is a global problem, characterized asmultifactorial and urged a multidiscipl<strong>in</strong>ary approach <strong>in</strong>which animal scientists, veter<strong>in</strong>arians and molecularbiologists were required to unravel the complex pathogenesisof this ‘subfertility syndrome’. After all, produc<strong>in</strong>ga calf at regular <strong>in</strong>tervals is considered a prerequisitefor profitable lactational performance (Royal et al.2000; Huirne et al. 2002). After giv<strong>in</strong>g birth, the processof becom<strong>in</strong>g pregnant aga<strong>in</strong> <strong>in</strong> dairy cows starts withclearance and <strong>in</strong>volution of the uterus followed byresumption of ovarian activity. This should result <strong>in</strong> thecompletion of the growth of a healthy follicle, enclos<strong>in</strong>ga competent oocyte, and ultimately <strong>in</strong> oestrus, ovulation,fertilization and uter<strong>in</strong>e attachment by a viableembryo. Any upset of these balanced and f<strong>in</strong>e-tunedbiological and mechanical events leads to fail<strong>in</strong>g reproduction– and this is exactly where the shoe p<strong>in</strong>ches <strong>in</strong>our modern dairy herds.The subfertility syndrome can be divided <strong>in</strong>to twomajor sub-problems. First of all, up to 50% of moderndairy cows display abnormal oestrus cycles postpartumlead<strong>in</strong>g to prolonged calv<strong>in</strong>g to first <strong>in</strong>sem<strong>in</strong>ation<strong>in</strong>tervals (Opsomer et al. 1998). In this context, especially<strong>in</strong>stability with<strong>in</strong> the hypothalamo–pituitary–ovarian–uter<strong>in</strong>e axis has been studied thoroughly (Lucy2001; Butler 2003). The concomitant reduced oestrusexpression or even anoestrus, cyst formation anddelayed first ovulation have been extensively documented(Beam and Butler 1997; de Vries and Veerkamp2000; Lopez et al. 2004; Vanholder et al. 2006a).Secondly, attention was focussed on disappo<strong>in</strong>t<strong>in</strong>gconception rates (Bousquet et al. 2004) and the <strong>in</strong>creas<strong>in</strong>glyhigh <strong>in</strong>cidence of early embryonic mortality(Dunne et al. 1999; Mann and Lamm<strong>in</strong>g 2001; Bilodeau-Goeseelsand Kastelic 2003). Fertilization ofoocytes from high-genetic merit cows resulted <strong>in</strong> significantlylower blastocyst yields <strong>in</strong> vitro, irrespective ofmilk production as such (Snijders et al. 2000). Embryoquality was also reduced <strong>in</strong> high-produc<strong>in</strong>g dairy cowscompared with non-lactat<strong>in</strong>g counterparts (Wiltbanket al. 2001; Leroy et al. 2005a). A high proportion ofnon-viable embryos were found <strong>in</strong> lactat<strong>in</strong>g cowscompared with non-lactat<strong>in</strong>g cows (Sartori et al.2002). Approximately 70–80% of the total embryonicand foetal losses typically occur dur<strong>in</strong>g the earlyembryonic, pre-attachment period (Santos et al. 2004a)(for review, see Leroy et al. 2007).Modern dairy cows, albeit sub-fertile, produce vastamounts of milk ma<strong>in</strong>ly because of significant geneticimprovements, comb<strong>in</strong>ed with nutritional managementoptimized towards lactation. Based on almostunchanged heifer fertility, we can conclude that thereproductive processes of modern dairy cattle areessentially normal when lactation demands are notimposed (Lucy 2007). Why do modern dairy cowsprioritize milk production at the expense of susta<strong>in</strong>edreproductive efficiency? In this review, we aim to answerthis question. Are high milk yields and good fertilityoutcomes conflict<strong>in</strong>g <strong>in</strong>terests metabolically speak<strong>in</strong>g?From Phylogenetically Driven to GeneticallyEnforced Nutrient Prioritization: TheConsequences on MetabolismFrom a biological po<strong>in</strong>t of view, it makes sense formammals <strong>in</strong> early lactation to favour milk productionover fertility: this we can refer to as nutrient prioritization(Lucy 2003). As nutrition becomes scarce, thelactat<strong>in</strong>g dam will preferentially <strong>in</strong>vest the limitedresources <strong>in</strong> the survival of liv<strong>in</strong>g offspr<strong>in</strong>g rather thangambl<strong>in</strong>g on the oocyte that is yet to be ovulated,Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Nutrient Prioritization and Fertility <strong>in</strong> Dairy Cows 97fertilized and cared for dur<strong>in</strong>g an entire gestation. Thismaternal catabolic mechanism, also genetically programmed,should maximize the chance of survival of thenewborn offspr<strong>in</strong>g (Silvia 2003). Over the past 40 years,the focus of dairy <strong>in</strong>dustry has been on maximiz<strong>in</strong>g milkyield, thereby creat<strong>in</strong>g a ‘nutrient highway’ from thedaily ration and body reserves (estimated on 74% bodyfat and 6% body prote<strong>in</strong>: Tamm<strong>in</strong>ga et al. 1997) directlyto the udder to susta<strong>in</strong> milk production.Nutrient requirements of the gravid uterus late <strong>in</strong>gestation impose a catabolic status on the dairy cow.Follow<strong>in</strong>g parturition, an additional demand for glucose,fatty acids and prote<strong>in</strong> is established as milkproduction starts. Dur<strong>in</strong>g this transition period, cowsare unable to compensate for such <strong>in</strong>creased energydemands by <strong>in</strong>creas<strong>in</strong>g feed <strong>in</strong>take, and this results <strong>in</strong>negative energy balance (NEB). Drastically reduced<strong>in</strong>sul<strong>in</strong> concentrations br<strong>in</strong>g approximate energy mobilizationand partition<strong>in</strong>g of energy to the udder.Hypo<strong>in</strong>sul<strong>in</strong>aemia promotes gluconeogenesis <strong>in</strong> the liver(up to 4 kg glucose each day) and acts as a massivelipolytic trigger. The mobilized non-esterified fatty acids(NEFAs) serve as an alternative energy source for othertissues to preserve glucose, which is preferentially usedby the mammary gland to form lactose (Vernon 2002).NEFAs are predom<strong>in</strong>antly transported to the liverwhere they are oxidized to provide energy or transformed<strong>in</strong>to ketone bodies, aga<strong>in</strong> an alternative energysource elsewhere <strong>in</strong> the body. An aberrant over-load ofthe liver by NEFAs can <strong>in</strong>duce steatosis and disturbedliver function (Herdt 2000). Hormone-sensitive lipases<strong>in</strong> adipose tissue of high-yield<strong>in</strong>g dairy cows have an<strong>in</strong>creased sensitivity to lipolytic stimuli (such as low<strong>in</strong>sul<strong>in</strong>, and high catecholam<strong>in</strong>es or glucocorticoidsconcentrations). In other words, high-yield<strong>in</strong>g dairycows have been genetically selected to partition evenmore energy reserves <strong>in</strong>to milk production (Coffey et al.2004). A higher dietary energy <strong>in</strong>take will thereforeresult <strong>in</strong> greater milk production, but a similar energyimbalance rema<strong>in</strong>s, with no beneficial effects on bodycondition score (BCS) at all (Patton et al. 2006).A series of biological mechanisms br<strong>in</strong>g an approximateprioritization for milk production at the cost ofbody reserves <strong>in</strong> early postpartum dairy cows. First ofall, the udder benefits because it does not need <strong>in</strong>sul<strong>in</strong> tofacilitate glucose uptake <strong>in</strong>to cells by the glucosetransportmolecules, GLUT 1 and 3, while most othertissues predom<strong>in</strong>antly express <strong>in</strong>sul<strong>in</strong>-dependent GLUT4 (Zhao et al. 1996). Secondly, us<strong>in</strong>g repeatedly <strong>in</strong>travenousglucose-tolerance tests, we recently found atemporary suppression of pancreatic function <strong>in</strong> earlypostpartum high-yield<strong>in</strong>g dairy cows and this wascorrelated with elevated NEFA concentrations (Bossaertet al. 2007). In vitro, high NEFA levels have toxiceffects on pancreatic cells (Cnop et al. 2001; Maedleret al. 2001). Thirdly, <strong>in</strong> the early postpartum period, low<strong>in</strong>sul<strong>in</strong> concentrations uncouple the growth hormone(GH)–<strong>in</strong>sul<strong>in</strong> like growth factor 1 (IGF-I) axis <strong>in</strong> theliver because of down-regulation of GH 1A receptorsand this can be restored by <strong>in</strong>creas<strong>in</strong>g <strong>in</strong>sul<strong>in</strong> (Butleret al. 2003). As IGF-I production <strong>in</strong> the liver issuppressed, the negative feedback of IGF-I is removedat the level of the hypothalamus ⁄ pituitary, and GHconcentrations <strong>in</strong>crease. High GH concentrations notonly stimulate milk production but also provoke livergluconeogenesis and lipolysis <strong>in</strong> adipocytes. The result<strong>in</strong>ghigh blood NEFA and GH concentrations antagonize<strong>in</strong>sul<strong>in</strong> action and create a further state ofperipheral <strong>in</strong>sul<strong>in</strong> resistance (Lucy 2007; Pires et al.2007). In this way even more glucose is conserved to beavailable for lactose synthesis.Fatter cows tend to mobilize more body fat because ofreduced appetite (Garnsworthy and Topps 1982). It isbroadly accepted that genetic selection for milk productionresults <strong>in</strong> greater BCS loss, further suggest<strong>in</strong>g thatenergy is partitioned towards the udder (Roche et al.2006). An excessive BCS loss dur<strong>in</strong>g the transitionperiod is a major risk factor for health and fertilitydisorders (Roche et al. 2007), which stresses the importanceof BCS monitor<strong>in</strong>g early postpartum as a managementtool (Chagas et al. 2007).Interactions Between the Somatotropic and theGonadotropic AxisExtensive scientific research has shown that mechanismsthat regulate energy and nutrient distribution <strong>in</strong> thesomatotropic system may affect the reproductive systemat different levels of the hypothalamo–pituitary–ovarianaxis (Roche 2006; Chagas et al. 2007). With<strong>in</strong> thehypothalamus, <strong>in</strong>teractions between the gonadotropicand somatotropic systems may occur <strong>in</strong> the pre-opticarea (Blache et al. 2006, 2007). This region produces thereleas<strong>in</strong>g hormones that control the secretion of bothgonadotrop<strong>in</strong>s and somatotrop<strong>in</strong> (Kacsoh 2000). Inaddition, it plays a crucial role <strong>in</strong> <strong>in</strong>tegrat<strong>in</strong>g appetite(Wynne et al. 2005), oestrus behaviour (Pfaff 2005) andsens<strong>in</strong>g of the nutritional status (Wade and Jones 2004).Consequently, metabolic <strong>in</strong>puts <strong>in</strong> the hypothalamusmay have divergent effects on the gonadotropic andsomatotropic axis, i.e. stimulation of GH productionmay be accompanied by <strong>in</strong>hibition of GnRH secretion(Zieba et al. 2005). The hormones ⁄ metabolites that aremost likely to exert a signall<strong>in</strong>g function are glucose and<strong>in</strong>sul<strong>in</strong>. Low postpartum <strong>in</strong>sul<strong>in</strong> and glucose concentrationssuppress hypothalamic GnRH secretion andsubsequent pituitary LH release (Disk<strong>in</strong> et al. 2003;Ohkura et al. 2004). By activation of specific neurons <strong>in</strong>the forebra<strong>in</strong>, peptides such as neuropeptide Y andcatecholam<strong>in</strong>es are released, which suppress the hypothalamicGnRH pulse generator (Ichimaru et al. 2001;Disk<strong>in</strong> et al. 2003; Wade and Jones 2004). Othermetabolic signals may <strong>in</strong>volve lept<strong>in</strong> and NEFA,although their role currently rema<strong>in</strong>s unclear (Lieferset al. 2003; Wade and Jones 2004; Amstalden et al.2005).At ovarian level, follicular growth and developmentseems to be directly <strong>in</strong>fluenced by altered <strong>in</strong>sul<strong>in</strong>, IGF-I,lept<strong>in</strong> and NEFA levels. Because <strong>in</strong>sul<strong>in</strong> locally stimulatesfollicular growth, maturation and steroidogenesis,reduced postpartum concentrations are l<strong>in</strong>ked to ovariandysfunction (Gutierrez-Aguilar 1997; Landau et al.2000; Armstrong et al. 2002a; Butler et al. 2004; Vanholderet al. 2005a; Kawashima et al. 2007). Long-termtreatment with exogenous bov<strong>in</strong>e somatotrop<strong>in</strong> clearly<strong>in</strong>creased the number of small follicles (Bols et al. 1998).Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


98 JLMR Leroy, T Vanholder, ATM Van Knegsel, I Garcia-Ispierto and PEJ BolsGong (2002) showed that the beneficial effects of <strong>in</strong>sul<strong>in</strong>on ovarian functions are <strong>in</strong>dependent of changes <strong>in</strong>GnRH ⁄ LH release. In ovarian cells, <strong>in</strong>sul<strong>in</strong>-<strong>in</strong>dependentGLUT-1 and GLUT-3 are the major glucose transporters,while the <strong>in</strong>sul<strong>in</strong>-dependent GLUT-4 only plays asupportive role (Nishimoto et al. 2006). Hence, <strong>in</strong>sul<strong>in</strong>may exert its effects through mechanisms other thanmediat<strong>in</strong>g glucose uptake.Together with <strong>in</strong>sul<strong>in</strong>, the IGF system plays animportant role <strong>in</strong> follicle growth and development byact<strong>in</strong>g directly on ovarian cells (Spicer and Echternkamp1995; Gutierrez-Aguilar 1997; Beam and Butler 1999;Webb et al. 1999; Gong 2002). Consequently, lowcirculat<strong>in</strong>g IGF-1 concentrations negatively <strong>in</strong>fluencethe onset of postpartum ovarian activity and seem<strong>in</strong>volved <strong>in</strong> the development of cystic ovarian follicles(Beam and Butler 1997; Kawashima et al. 2007; Ortegaet al. 2007). The effects of lept<strong>in</strong> on steroidogenesis andcell proliferation are, yet, dependent on the circulat<strong>in</strong>gconcentrations of IGF-1, LH and <strong>in</strong>sul<strong>in</strong> (Spicer andFrancisco 1997, 1998; Spicer et al. 2000).At ovarian level, NEFAs may affect follicular growthand development by act<strong>in</strong>g directly on follicle cells.Add<strong>in</strong>g NEFAs <strong>in</strong> vitro, at concentrations measured <strong>in</strong>follicular fluid (FF) dur<strong>in</strong>g NEB has detrimental effectson follicle cell viability and function (Leroy et al. 2005b;Vanholder et al. 2005b, 2006b).In conclusion, metabolic changes <strong>in</strong>duced by thesomatotropic system to susta<strong>in</strong> a high milk yield alsoaffect the reproductive system. By act<strong>in</strong>g at differentlevels of the hypothalamo–pituitary–ovarian axis,altered hormone and metabolite levels exert a negativeeffect on follicle growth, development and probablyovulation.Consequences for Oocyte QualityResearchers assume the existence of a carry-over effectof the adverse metabolic conditions dur<strong>in</strong>g primaryfollicle growth early postpartum on the health of thepre-ovulatory follicle 2–3 months later (Britt 1992).Such follicles may be less capable of produc<strong>in</strong>g adequateamounts of oestrogens and progesterone (follow<strong>in</strong>govulation) and might be doomed to conta<strong>in</strong> an oocyteof <strong>in</strong>ferior quality (Britt 1992; Roth et al. 2001a). Thedevelopmental capacity of the oocyte is <strong>in</strong>tr<strong>in</strong>sicallyl<strong>in</strong>ked to the growth phase and health of the develop<strong>in</strong>gfollicle (Bilodeau-Goeseels and Panich 2002; Suttonet al. 2003; Lequarre et al. 2005). Last but not least, dietcomposition can also alter the endocr<strong>in</strong>e and metabolicmicro-environment of the develop<strong>in</strong>g oocyte (Bolandet al. 2001; McEvoy et al. 2001; Kenny et al. 2002).A deviant endocr<strong>in</strong>e environment, because of or as aconsequence of a NEB can alter oocyte quality throughvarious mechanisms such as sp<strong>in</strong>dle formation, prolongedfollicular growth and resumption of meioticprogression, all of which has been extensively reviewedearlier (Leroy et al. 2007). Only a few studies haveexam<strong>in</strong>ed possible effects of NEB-associated low glucose,elevated b-hydroxybutyrate (b-OHB) or NEFAconcentrations on oocyte quality. Apart from <strong>in</strong>directeffects of hypoglycaemia <strong>in</strong> early postpartum dairy cows(through an effect on LH secretion or ovarian responsivenessto gonadotroph<strong>in</strong>s), hypoglycaemic conditions(e.g. cl<strong>in</strong>ical ketosis) are reflected <strong>in</strong> the microenvironmentof the pre-ovulatory oocyte, and can compromisethe oocyte’s developmental capacity, because glucose isan <strong>in</strong>dispensable molecule for proper oocyte maturation(Bilodeau-Goeseels 2006; for review see Sutton et al.2003; Leroy et al. 2004, 2006). Kruip and Kemp (1999)suggested possible direct toxic effects of high NEFAconcentrations at the ovarian level of the ovary. Indeed,<strong>in</strong> an <strong>in</strong> vitro maturation model, saturated long-cha<strong>in</strong>fatty acids, reduced rates of maturation, fertilization,cleavage and blastocyst formation. Apoptosis, and evencumulus cell necrosis, dur<strong>in</strong>g maturation could expla<strong>in</strong>these observations (Leroy et al. 2005b). F<strong>in</strong>ally, elevatedammonia and urea concentrations <strong>in</strong> the FF, because ofan unbalanced diet and prote<strong>in</strong> catabolism were toxicfor the oocyte (S<strong>in</strong>clair et al. 2000; De Wit et al. 2001;Leroy et al. 2004).Early Pregnancy <strong>in</strong> High-produc<strong>in</strong>g DairyCows: Embryo QualityEarly embryonic death is a major cause of reproductivefailure <strong>in</strong> dairy cows account<strong>in</strong>g for up to a total 80%pregnancy losses (Santos et al. 2004a). There are fourmajor factors imp<strong>in</strong>g<strong>in</strong>g on embryo quality <strong>in</strong> thespecific case of high-produc<strong>in</strong>g dairy cows: gametequality, corpus luteum quality comb<strong>in</strong>ed with thecirculat<strong>in</strong>g progesterone concentration, uter<strong>in</strong>e <strong>in</strong>volutionand nutrition. Yet, only those that are related toNEB will be discussed.Adverse pre-ovulatory conditions, such as NEB, mayhave carry-over effects on embryo metabolism andviability result<strong>in</strong>g <strong>in</strong> early embryonic mortality (Yaakubet al. 1999; Lozano et al. 2003; Rhoads et al. 2006).Dist<strong>in</strong>guish<strong>in</strong>g oocyte effects on embryo quality frompost-fertilization <strong>in</strong>fluences is extremely difficult. Only atleast a 6-day-old embryo can be transferred to arecipient to assess the impact of uter<strong>in</strong>e environmenton embryo quality. Lucy (2007) supports the conceptthat fertility could be improved <strong>in</strong> dairy cows by us<strong>in</strong>gembryo transfer and thus circumvent<strong>in</strong>g the period ofoocyte and early embryonic development.Well-timed and balanced post-mat<strong>in</strong>g progesteroneconcentrations are vital for zygote viability as progesteronemodulates the endometrial secretions, and thusoptimal uter<strong>in</strong>e receptivity (McEvoy et al. 1995). It hasbeen suggested that disappo<strong>in</strong>t<strong>in</strong>g pregnancy results <strong>in</strong>modern dairy<strong>in</strong>g are partially caused by the retardedonset of the progesterone rise and suboptimal progesteroneconcentrations dur<strong>in</strong>g the luteal phase (Mannand Lamm<strong>in</strong>g 2001). Furthermore, the typical NEBobserved early postpartum can reduce the number ofovulatory oestrous cycles preced<strong>in</strong>g AI which mayhamper adequate uterus preparation (Butler 2003).Villa-Godoy et al. (1988) showed that cows <strong>in</strong> NEBpostpartum had lower progesterone concentrationsdur<strong>in</strong>g the first three ovarian cycles follow<strong>in</strong>g calv<strong>in</strong>g.Despite larger volumes of luteal tissue, compared withnon-lactat<strong>in</strong>g heifers, maximal progesterone concentrations<strong>in</strong> lactat<strong>in</strong>g cows are lower, possibly because of ahigher rate of degradation <strong>in</strong> the liver (Sangsritavonget al. 2002; Sartori et al. 2004; Wiltbank et al. 2006).Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Nutrient Prioritization and Fertility <strong>in</strong> Dairy Cows 99Good postpartum uter<strong>in</strong>e <strong>in</strong>volution, compris<strong>in</strong>gendometrium repair and evacuation of bacteriallycontam<strong>in</strong>ated contents, is of critical importance forreproductive performance (for review, see Roche et al.2006). Because of a reduced immune response,negative energy status can dramatically delay thisprocess, jeopardiz<strong>in</strong>g future fertility (Wathes et al.2007).Apart from <strong>in</strong>terest <strong>in</strong> the consequences of NEB andassociated endocr<strong>in</strong>e and metabolic imbalances, there isa grow<strong>in</strong>g focus towards the effect of milk yieldpromot<strong>in</strong>gdiets that are rich <strong>in</strong> energy and prote<strong>in</strong>(for review, see Leroy et al. 2007). From these studies wecan learn that optimum nutritional conditions forfollicle growth and ovulation are not compatible withembryo survival and ma<strong>in</strong>tenance of pregnancy(O’Callaghan and Boland 1999).F<strong>in</strong>ally, heat stress can cause a reduction <strong>in</strong> drymatter <strong>in</strong>take which prolongs the period of NEB,decreas<strong>in</strong>g plasma concentrations of <strong>in</strong>sul<strong>in</strong>, glucoseand IGF-I, while <strong>in</strong>creas<strong>in</strong>g GH and NEFA (Drew1999; Butler 2001). In addition, there is a direct effect ofheat stress on FSH (<strong>in</strong>creased) and oestradiol(decreased) plasma concentrations (Wolfenson et al.1997; Wilson et al. 1998). This causes not only poorexpression of oestrus, but also delayed follicle selectionand thus has potentially adverse effects on oocytequality (Roth et al. 2001a,b). Heat stress may furthermore<strong>in</strong>crease uter<strong>in</strong>e temperature by decreas<strong>in</strong>g bloodflow to the uterus. These changes <strong>in</strong>hibit embryonicdevelopment (Rivera and Hansen 2001), <strong>in</strong>crease earlyembryonic loss and reduce the proportion of successful<strong>in</strong>sem<strong>in</strong>ations (Garcia-Ispierto et al. 2007). The impactof this heat effect decreases as the embryo develops(Paula-Lopes et al. 2003).All the factors described above, potentially affect<strong>in</strong>gfertility, are diagrammatically presented <strong>in</strong> Fig. 1.Some Clues to Modify the Somatotropic Axis <strong>in</strong>Order to Generate Acceptable Fertility ResultsTackl<strong>in</strong>g the multifactorial problem of subfertility <strong>in</strong>dairy cows is a real challenge as the ‘skewed somatotropicaxis’ described above is not the only reason forthe decl<strong>in</strong>e <strong>in</strong> fertility. Evolv<strong>in</strong>g farm systems, grow<strong>in</strong>gherd sizes, <strong>in</strong>creased managerial demands comb<strong>in</strong>edwith a reduced labour <strong>in</strong>put per animal and <strong>in</strong>creasedsusceptibility to diseases, all <strong>in</strong>terfere with the ability ofthe dairy cow to be successfully bred (Mallard et al.1998). All these factors should be carefully addressedwhen formulat<strong>in</strong>g management advice to the dairy farmmanager, but this is beyond the scope of the presentpaper.Metabolic disorders (hypocalcaemia, ketosis andacidosis) and <strong>in</strong>fectious diseases dur<strong>in</strong>g the puerperiumare all key risk factors for efficient reproductive performance(Grohn and Rajala-Schultz 2000; Santos et al.2004b). Balanced and sophisticated birth managementcomb<strong>in</strong>ed with strict follow-up of cow health statusearly postpartum is vital to prevent a drop <strong>in</strong> theanimal’s appetite. Accurate and repeated assessment ofBCS to estimate changes <strong>in</strong> body reserves is critical.M<strong>in</strong>imiz<strong>in</strong>g BCS changes and thus the ‘exhaustion ofthe energy reserves’ early postpartum requires anoptimal dietary strategy by reduc<strong>in</strong>g energy <strong>in</strong>takedur<strong>in</strong>g the first weeks of the dry period then an<strong>in</strong>creased energy supply (carbohydrates) shortly prepartum(for review, see Overton and Waldron 2004).Yet, the beneficial effect of extra pre-partum energy onpostpartum energy balance is a matter of debate(Grummer 2007). Furthermore, adequate dietary modulationsdur<strong>in</strong>g the demand<strong>in</strong>g early postpartum periodare a promis<strong>in</strong>g approach although difficult to achieve(Grummer 2007). Therefore, Van Knegsel et al. (2007a)fed on an isocaloric and isonitrogenous basis, a ma<strong>in</strong>lyglucogenic (by-pass starch) or a ma<strong>in</strong>ly lipogenic diet(beetpulp, MEGALAC and palm oil) and showed thatthe glucogenic (or ‘<strong>in</strong>sul<strong>in</strong>ogenic’) diet, stimulates energypartition<strong>in</strong>g towards body reserves <strong>in</strong> early lactation.Cows fed the glucogenic diet had lower NEB andreduced body fat mobilization, which led to milk fatdepression and less energy partitioned to milk (VanKnegsel et al. 2007a). In a follow-up study (Van Knegselet al. 2007b), multiparous cows fed with glucogenic dietalso had higher plasma <strong>in</strong>sul<strong>in</strong> concentrations andtended to resume ovarian activity earlier(20.4 ± 2.1 days) compared to cows fed with morelipogenic diet (26.1 ± 2.1 days).Fig. 1. Interaction between geneticselection for milk production andfertility. The imposed metabolicand endocr<strong>in</strong>e changes susta<strong>in</strong><strong>in</strong>gmilk production <strong>in</strong> comb<strong>in</strong>ationwith the <strong>in</strong>creased susceptibility toheat stress, diseases and suboptimalmanagement conditions allnegatively affect reproductive performanceof the high-produc<strong>in</strong>gdairy cowBreed<strong>in</strong>g priorityfor milkproduction traitsBreed<strong>in</strong>g valuefor fertility+Increased susceptibility to:Metabolic/<strong>in</strong>fectious diseasesHeat stressSuboptimal management:DietHous<strong>in</strong>g conditionsHerd sizeLabour per animalHeat detection+NUTRIENT PRIORITIZATION :Genetically drivendepletion of body reservesMETABOLIC ADAPTATIONS TOCATABOLIC STATUSNEB, BCS loss, reduced appetiteLow <strong>in</strong>sul<strong>in</strong>, low IGF-I, high GHLow lept<strong>in</strong>High NEFA, high ketones, high urea,low glucosePeriphere <strong>in</strong>sul<strong>in</strong> resistanceFERTILITYHypothalamo-pituitary-ovarian-axisoocyte/embryo qualitycorpus luteum qualityfollicular, oviductal, uter<strong>in</strong>e environmentDirect or carry over effects– –Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


100 JLMR Leroy, T Vanholder, ATM Van Knegsel, I Garcia-Ispierto and PEJ BolsIt is speculated that changes <strong>in</strong> management are morelikely to have a positive effect on EB. Shorten<strong>in</strong>g or evenskipp<strong>in</strong>g the dry period improves dry matter <strong>in</strong>takeperipartum, reduces milk production <strong>in</strong> early lactation,improves energy balance and reduces the number ofdays postpartum till resumption of ovarian activity(Gumen et al. 2005; Rastani et al. 2005).Besides this, grow<strong>in</strong>g attention is be<strong>in</strong>g paid to dietaryfatty acid content and composition provided by supplementedby-pass fats dur<strong>in</strong>g the early postpartum period.Not the effect on energy balance as such but improvedsteroid secretion and alteration of the fatty acid profile(more x-3 poly-unsaturated fatty acids), result<strong>in</strong>g <strong>in</strong>modified prostagland<strong>in</strong> metabolism (Thatcher et al.2006). Suppression of milk fat synthesis by supplementationof rumen-protected conjugated l<strong>in</strong>oleic acids(trans-10, cis-12) has been suggested to restrict energyloss through milk (Castaneda-Gutierrez et al. 2005).Yet, <strong>in</strong> spite of several recent <strong>in</strong>terest<strong>in</strong>g papers, theoutcome on energy balance and fertility are equivocal.An extensive description and clear overview of nutritionalstrategies support<strong>in</strong>g the metabolic demandsdur<strong>in</strong>g the transition period are given by Overton andWaldron (2004) and are beyond the scope of the presentpaper.F<strong>in</strong>ally, genetic selection programmes <strong>in</strong> the dairy<strong>in</strong>dustry have emphasized milk production traits byun<strong>in</strong>tended mobilization of cow body reserves. This loss<strong>in</strong> BCS is not only dependent on the available mass ofadipose tissue but also on a genetically determ<strong>in</strong>ed setpo<strong>in</strong>tfor BCS. This set-po<strong>in</strong>t is correlated with reproductiveoutcome (Lucy 2007). Therefore, not onlyfertility traits as such (Royal et al. 2000), but alsovariables compris<strong>in</strong>g changes <strong>in</strong> BCS early postpartumshould be <strong>in</strong>cluded <strong>in</strong> genetic selection criteria.ConclusionsIntense selection for milk production has resulted <strong>in</strong> animmense priority for the high-produc<strong>in</strong>g dairy cow topartition energy to milk, at the cost of body reserves.This has resulted <strong>in</strong> excessive NEB and poor reproductiveperformance. Thus, milk production and reproductiveperformance have conflict<strong>in</strong>g <strong>in</strong>terests <strong>in</strong>high-produc<strong>in</strong>g dairy cows. Metabolites and metabolichormones associated with energy prioritiz<strong>in</strong>g for milkproduction (NEFA, <strong>in</strong>sul<strong>in</strong>, glucose, IGF-1, b-OH)<strong>in</strong>fluence fertility, <strong>in</strong>directly by modulat<strong>in</strong>g the somatotropic⁄ gonadotropic axis, as well as directly at theovary, follicle or uter<strong>in</strong>e environment. Strict follow-upperipartum to monitor health and BCS loss and directtreatment of (<strong>in</strong>fectious or metabolic) disorders <strong>in</strong> earlylactation will limit fertility disorders postpartum. Furthermore,a series of promis<strong>in</strong>g management, geneticselection and nutritional strategies have been proposed,which have the potential to shift the somatotropic axisprioritiz<strong>in</strong>g energy partition<strong>in</strong>g of milk to a somatotropicaxis with an <strong>in</strong>creased priority for body reserves toimprove fertility. Yet, research <strong>in</strong> this area is limited.Explor<strong>in</strong>g such strategies, compar<strong>in</strong>g their benefit oreven comb<strong>in</strong><strong>in</strong>g two or more strategies is an extremely<strong>in</strong>terest<strong>in</strong>g area of research, and essential to improvehealth and welfare of the modern dairy cow.ReferencesAmstalden M, Harms PG, Welsh TH Jr, Randel RD, WilliamsGL, 2005: Effects of lept<strong>in</strong> on gonadotrop<strong>in</strong>-releas<strong>in</strong>ghormone release from hypothalamic-<strong>in</strong>fundibular explantsand gonadotrop<strong>in</strong> release from adenohypophyseal primarycell cultures: further evidence that fully nourished cattle areresistant to lept<strong>in</strong>. Anim Reprod Sci 85, 41–52.Armstrong DG, Baxter G, Hogg CO, Woad KJ, 2002a:Insul<strong>in</strong>-like growth factor (IGF) system <strong>in</strong> the oocyte andsomatic cells of bov<strong>in</strong>e preantral follicles. <strong>Reproduction</strong> 123,789–797.Beam SW, Butler WR, 1997: Energy balance and ovarianfollicle development prior to first ovulation postpartum <strong>in</strong>dairy cows receiv<strong>in</strong>g three levels of dietary fat. Biol Reprod56, 133–142.Beam SW, Butler WR, 1999: Effects of energy balance onfollicular development and first ovulation <strong>in</strong> postpartumdairy cows. J Reprod Fertil Suppl 54, 411–424.Bilodeau-Goeseels S, 2006: Effect of culture media and energysources on the <strong>in</strong>hibition of nuclear maturation <strong>in</strong> bov<strong>in</strong>eoocytes. Theriogenology 66, 297–306.Bilodeau-Goeseels S, Kastelic JP, 2003: Factors affect<strong>in</strong>gembryo survival and strategies to reduce embryonic mortality<strong>in</strong> cattle. Can J Anim Sci 83, 659–671.Bilodeau-Goeseels S, Panich P, 2002: Effects of quality ondevelopment and transcriptional activity <strong>in</strong> early bov<strong>in</strong>eembryos. Anim Reprod Sci 71, 143–155.Blache D, Zhang S, Mart<strong>in</strong> GB, 2006: Dynamic and <strong>in</strong>tegrativeaspects of the regulation of reproduction by metabolicstatus <strong>in</strong> male sheep. Reprod Nutr Dev 46, 379–390.Blache D, Chagas LM, Mart<strong>in</strong> GB, 2007: Nutritional <strong>in</strong>puts<strong>in</strong>to the reproductive neuroendocr<strong>in</strong>e control system – amultidimensional perspective. <strong>Reproduction</strong>, Supplement64, 124–139.Boland MP, Lonergan P, O’Callaghan D, 2001: Effect ofnutrition on endocr<strong>in</strong>e parameters, ovarian physiology, andoocyte and embryo development. Theriogenology 55, 1323–1340.Bols PEJ, Ysebaert MT, Le<strong>in</strong> A, Coryn M, Van Soom A, deKruif A, 1998: Effects of long-term treatment with bov<strong>in</strong>esomatotrop<strong>in</strong> on follicular dynamics and subsequent oocyteand blastocyst yield <strong>in</strong> an OPU-IVF program. Theriogenology49, 983–995.Bossaert P, Leroy JLMR, Cools S, Opsomer G, 2007: Impactof metabolic and endocr<strong>in</strong>e parameters on ovulation <strong>in</strong> highyield<strong>in</strong>gdairy cows (abstract). Reprod Domest Anim 42,118.Bousquet D, Bouchard E, Du Tremblay D, 2004: Decreas<strong>in</strong>gfertility <strong>in</strong> dairy cows: myth or reality? Le Médec<strong>in</strong>Véte´r<strong>in</strong>aire 34, 59–61.Britt JH, 1992: Impacts of early postpartum metabolism onfollicular development and fertility. Proc Annu ConventionAm Assoc Bov<strong>in</strong>e Pract 24, 39–43.Butler WR, 2001: Nutritional effects on resumption of ovariancyclicity and conception rate <strong>in</strong> postpartum dairy cows.Anim Sci Occas Publ 26, 133–145.Butler WR, 2003: Energy balance relationships with folliculardevelopment, ovulation and fertility <strong>in</strong> pp dairy cows.Livestock Prod Sci 83, 211–218.Butler ST, Marr AL, Pelton SH, Radcliff RP, Lucy MC, ButlerWR, 2003: Insul<strong>in</strong> restores GH responsiveness dur<strong>in</strong>glactation-<strong>in</strong>duced negative energy balance <strong>in</strong> dairy cattle:effects on expression of IGF-I and GH receptor 1A. JEndocr<strong>in</strong>ol 176, 205–217.Butler ST, Pelton SH, Butler WR, 2004: Insul<strong>in</strong> <strong>in</strong>creases 17bestradiolproduction by the dom<strong>in</strong>ant follicle of the firstpostpartum follicle wave <strong>in</strong> dairy cows. <strong>Reproduction</strong> 127,537–545.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Nutrient Prioritization and Fertility <strong>in</strong> Dairy Cows 101Castaneda-Gutierrez E, Overton TR, Butler WR, BaumanDE, 2005: Dietary supplements of two doses of calcium saltsof conjugated l<strong>in</strong>oleic acid dur<strong>in</strong>g the transition period andearly lactation. J Dairy Sci 88, 1078–1089.Chagas LM, Bass JJ, Blache D, Burke CR, Kay JK,L<strong>in</strong>dsay DR, Lucy MC, Mart<strong>in</strong> GB, Meier S, RhodesFM, Roche JR, Thatcher WW, Webb R, 2007: Newperspectives on the roles of nutrition and metabolicpriorities <strong>in</strong> the subfertility of high-produc<strong>in</strong>g dairy cows.J Dairy Sci 90, 4022–4032.Cnop M, Hannaert JC, Hoorens A, Eizirik DL, Pipeleers DG,2001: Inverse relationship between cytotoxicity of free fattyacids <strong>in</strong> pancreatic islet cells and cellular triglycerideaccumulation. Diabetes 50, 1771–1777.Coffey MP, Simm G, Oldham JD, Hill WG, Brotherstone S,2004: Genotype and diet effects on energy balance <strong>in</strong> the firstthree lactations of dairy cows. J Dairy Sci 87, 4318–4326.De Wit AAC, Cesar MLF, Kruip TAM, 2001: Effect of ureadur<strong>in</strong>g <strong>in</strong> vitro maturation on nuclear maturation andembryo development of bov<strong>in</strong>e cumulus-oocyte-complexes.J Dairy Sci 84, 1800–1804.Disk<strong>in</strong> MG, Mackey DR, Roche JF, Sreenan JM, 2003:Effects of nutrition and metabolic status on circulat<strong>in</strong>ghormones and ovarian follicle development <strong>in</strong> cattle. AnimReprod Sci 78, 345–370.Drew B, 1999: Practical nutrition and management of heifersand high yield<strong>in</strong>g dairy cows for optimal fertility. CattlePract 7, 243–248.Dunne LD, Disk<strong>in</strong> MG, Boland MP, O’Farrell KJ, SreenanJM, 1999: The effect of pre- and post-<strong>in</strong>sem<strong>in</strong>ation plane ofnutrition on embryo survival <strong>in</strong> beef heifers. Anim Sci 69,411–417.Garcia-Ispierto I, Lopez-Gatius F, Bech-Sabat G, SantolariaP, Yaniz JL, Nogareda C, De Rensis F, Lopez-Bejar M,2007: Climate factors affect<strong>in</strong>g conception rate of highproduc<strong>in</strong>g dairy cows <strong>in</strong> north-eastern Spa<strong>in</strong>. Theriogenology67, 1379–1385.Garnsworthy PC, Topps JH, 1982: The effect of bodycondition score at calv<strong>in</strong>g on their food <strong>in</strong>take and performancewhen given complete diets. Anim Prod 35, 113–119.Gong JG, 2002: Influence of metabolic hormones and nutritionon ovarian follicle development <strong>in</strong> cattle: practicalimplications. Domest Anim Endocr<strong>in</strong>ol 23, 229–241.Grohn YT, Rajala-Schultz PJ, 2000: Epidemiology of reproductiveperformance <strong>in</strong> dairy cows. Anim Reprod Sci 60,605–614.Grummer RR, 2007: Strategies to improve fertility of highyield<strong>in</strong>g dairy farms: management of the dry period.Theriogenology 68, S281–S288.Gumen A, Rastani RR, Grummer RR, Wiltbank MC, 2005:Reduced dry periods and vary<strong>in</strong>g prepartum diets alterpostpartum ovulation and reproductive measures. J DairySci 88, 2401–2411.Gutierrez-Aguilar CG, 1997: The effect of nutrition andmetabolic hormones on follicular development <strong>in</strong> cattle.PhD Dissertation, University of Ed<strong>in</strong>burgh, pp. 146–166.Herdt TH, 2000: Rum<strong>in</strong>ant adaptation to negative energybalance. Vet Cl<strong>in</strong> North Am Food Anim Pract 16, 215–230.Huirne RBM, Saatkamp HW, Bergevoet RHM, 2002: Economicanalysis of common health problems <strong>in</strong> dairy cattle.In: Kaske M, Scholz H, Ho¨ ltersh<strong>in</strong>ken M(eds), TwelfthWorld Buiatrics Congress, Hannover, Germany, 18–23August 2002, pp. 420–431.Ichimaru T, Mori Y, Okamura H, 2001: A possible role ofneuropeptide Y as a mediator of undernutrition to thehypothalamic gonadotrop<strong>in</strong>-releas<strong>in</strong>g hormone pulse generator<strong>in</strong> goats. Endocr<strong>in</strong>ology 142, 2389–2498.Kacsoh B, 2000: Endocr<strong>in</strong>e Physiology. McGraw Hill BookCo., New York, NY.Kawashima C, Fukihara S, Maeda M, Kaneko E, MontoyaCA, Matsui M, Shimizu T, Matsunaga N, Kida K, MiyakeY, Schams D, Miyamoto A, 2007: Relationship betweenmetabolic hormones and ovulation of dom<strong>in</strong>ant follicledur<strong>in</strong>g the first follicular wave post-partum <strong>in</strong> high-produc<strong>in</strong>gdairy cows. <strong>Reproduction</strong> 133, 155–163.Kenny DA, Humpherson PG, Leese HJ, Morris DG, TomosAD, Disk<strong>in</strong> MG, Sreenan JM, 2002: Effect of elevatedsystemic concentrations of ammonia and urea on themetabolite and ionic composition of oviductal fluid <strong>in</strong>cattle. Biol Reprod 66, 1797–1804.Kruip TAM, Kemp B, 1999: Voed<strong>in</strong>g en vruchtbaarheid bijlandbouwhuisdieren. Tijdschr Diergeneeskd 124, 462–467.Landau S, Braw-Tal R, Kaim M, Bor A, Bruckental I, 2000:Preovulatory follicular status and diet affect the <strong>in</strong>sul<strong>in</strong> andglucose content of follicles <strong>in</strong> high-yield<strong>in</strong>g dairy cows.Anim Reprod Sci 64, 181–197.Lequarre AS, Vigneron C, Ribaucour F, Holm P, Donnay I,Dalbies-Tran R, Callesen H, Mermillod P, 2005: Influenceof antral follicle size on oocyte characteristics and embryodevelopment <strong>in</strong> the bov<strong>in</strong>e. Theriogenology 63, 841–859.Leroy JLMR, Vanholder T, Delange JR, Opsomer G, VanSoom A, Bols PEJ, Dewulf J, de Kruif A, 2004: Metabolicchanges <strong>in</strong> follicular fluid of the dom<strong>in</strong>ant follicle <strong>in</strong> highyield<strong>in</strong>gdairy cows early post partum. Theriogenology 62,1131–1143.Leroy JLMR, Opsomer G, De Vliegher S, Vanholder T,Goossens L, Geldhof A, Bols PEJ, de Kruif A, Van SoomA, 2005a: Comparison of embryo quality <strong>in</strong> high-yield<strong>in</strong>gdairy cows, <strong>in</strong> dairy heifers and <strong>in</strong> beef cows. Theriogenology64, 2022–2036.Leroy JLMR, Vanholder T, Mateusen B, Christophe A,Opsomer G, de Kruif A, Genicot G, Van Soom A, 2005b:Non-esterified fatty acids <strong>in</strong> follicular fluid of dairy cowsand their effect on developmental capacity of bov<strong>in</strong>e oocytes<strong>in</strong> vitro. <strong>Reproduction</strong> 130, 485–495.Leroy JLMR, Vanholder T, Opsomer G, Van Soom A, deKruif A, 2006: The <strong>in</strong> vitro development of bov<strong>in</strong>e oocytesafter maturation <strong>in</strong> glucose and beta-hydroxybutyrate concentrationsassociated with negative energy balance <strong>in</strong> dairycows. Reprod Domest Anim 41, 119–123.Leroy JLMR, Opsomer G, Van Soom A, Goovaerts IGF, BolsPEJ, 2007: Reduced fertility <strong>in</strong> high yield<strong>in</strong>g dairy cows: arethe oocyte and embryo <strong>in</strong> danger? Part I: The importance ofnegative energy balance and altered corpus luteum functionto the reduction of oocyte and embryo quality <strong>in</strong> highyield<strong>in</strong>g dairy cows (review). Reprod Domest Anim, <strong>in</strong>press. DOI:10.1111/j.1439–0531.2007.00960.Liefers SC, Veerkamp RF, te Pas MFW, Delavaud C,Chilliard Y, van der Lende T, 2003: Lept<strong>in</strong> concentrations<strong>in</strong> relation to energy balance, milk yield, <strong>in</strong>take, live weightand estrus <strong>in</strong> dairy cows. J Dairy Sci 86, 799–807.Lopez H, Satter LD, Wiltbank MC, 2004: Relationshipbetween level of milk production and oestrus behavior oflactat<strong>in</strong>g dairy cows. Anim Reprod Sci 81, 209–223.Lozano JM, Lonergan P, Boland MP, O’Callaghan D, 2003:Influence of nutrition on the effectiveness of superovulationprogrammes <strong>in</strong> ewes: effect on oocyte quality andpost-fertilization development. <strong>Reproduction</strong> 125, 543–553.Lucy MC, 2001: Reproductive loss <strong>in</strong> high-produc<strong>in</strong>g dairycattle: where will it end? J Dairy Sci 84, 1277–1293.Lucy MC, 2003: Mechanisms l<strong>in</strong>k<strong>in</strong>g nutrition and reproduction<strong>in</strong> postpartum cows. Reprod Suppl. 61, 415–427.Lucy MC, 2007: Fertility <strong>in</strong> high-produc<strong>in</strong>g dairy cows:reasons for decl<strong>in</strong>e and corrective strategies for susta<strong>in</strong>ableimprovement. Reprod Suppl. 64, 237–254.Maedler K, Sp<strong>in</strong>as GA, Dyntar D, Moritz W, Kaiser N,Donath MY, 2001: Dist<strong>in</strong>ct effects of saturated and mono-Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


102 JLMR Leroy, T Vanholder, ATM Van Knegsel, I Garcia-Ispierto and PEJ Bolsunsaturated fatty acids on beta-cell turnover and function.Diabetes 50, 69–76.Mallard BA, Dekkers JC, Ireland MJ, Leslie KE, Sharif S,Vankampen CL, Wagter L, Wilkie BN, 1998: Alteration <strong>in</strong>immune responsiveness dur<strong>in</strong>g the peripartum period and itsramification on dairy cow and calf health. J Dairy Sci 81,585–595.Mann GE, Lamm<strong>in</strong>g GE, 2001: Relationship between maternalendocr<strong>in</strong>e environment, early embryo development and<strong>in</strong>hibition of the luteolytic mechanism <strong>in</strong> cows. <strong>Reproduction</strong>121, 175–180.McEvoy TG, Rob<strong>in</strong>son JJ, Aitken RP, F<strong>in</strong>dlay PA, PalmerRM, Robertson IS, 1995: Dietary-<strong>in</strong>duced suppression ofpre-ovulatory progesterone concentrations <strong>in</strong> superovulatedewes impairs the subsequent <strong>in</strong> vivo and <strong>in</strong> vitro developmentof their ova. Anim Reprod Sci 39, 89–107.McEvoy TG, Rob<strong>in</strong>son JJ, Ashworth CJ, Rooke JA, S<strong>in</strong>clairKD, 2001: Feed and forage toxicants affect<strong>in</strong>g embryosurvival and fetal development. Theriogenology 55, 113–129.Nishimoto H, Matsutani R, Yamamoto S, Takahashi T,Hayashi KG, Miyamoto A, Hamano S, Tetsuka M, 2006:Gene expression of glucose transporter (GLUT) 1, 3 and 4<strong>in</strong> bov<strong>in</strong>e follicle and corpus luteum. J Endocr<strong>in</strong>ol 188, 111–119.O’Callaghan D, Boland MP, 1999: Nutritional effects onovulation, embryo development and the establishment ofpregnancy <strong>in</strong> rum<strong>in</strong>ants. Anim Sci 68, 299–314.Ohkura S, Ichimaru T, Itoh F, Matsuyama S, Okamura H,2004: Further evidence for the role of glucose as a metabolicregulator of hypothalamic gonadotrop<strong>in</strong>-releas<strong>in</strong>g hormonepulse generator activity <strong>in</strong> goats. Endocr<strong>in</strong>ology 145, 3239–3246.Opsomer G, Coryn M, Deluyker H, de Kruif A, 1998: Ananalysis of ovarian dysfunction <strong>in</strong> high yield<strong>in</strong>g dairy cowsafter calv<strong>in</strong>g based on progesterone profiles. Reprod DomestAnim 33, 193–204.Ortega HH, Palomar MM, Acosta JC, Salvetti NR, DallardBE, Lorente JA, Barbeito CG, Gimeno EJ, 2008: Insul<strong>in</strong>likegrowth factor I <strong>in</strong> sera, ovarian follicles and follicularfluid of cows with spontaneous or <strong>in</strong>duced cystic ovariandisease. Res Vet Sci 84, 419–427.Overton TR, Waldron MR, 2004: Nutrtional management oftransition dairy cows: strategies to optimize metabolichealth. J Dairy Sci 87, E105–E119.Patton J, Kenny DA, Mee JF, O’Mara FP, Wathes DC, CookM, Murphy JJ, 2006: Effect of milk<strong>in</strong>g frequency and diet onmilk production, energy balance and reproduction <strong>in</strong> dairycows. J Dairy Sci 89, 1478–1487.Paula-Lopes FF, Al-Katanani YM, Majewski AC, McDowellLR, Hansen PJ, 2003: Manipulation of antioxidant statusfails to improve fertility of lactat<strong>in</strong>g cows or survival of heatshockedembryos. J Dairy Sci 86, 2343–2351.Pfaff D, 2005: Hormone-driven mechanisms <strong>in</strong> the centralnervous system facilitate the analysis of mammalian behaviours.J Endocr<strong>in</strong>ol 184, 447–453.Pires JAA, Souza AH, Grummer RR, 2007: Induction ofhyperlipidemia by <strong>in</strong>travenous <strong>in</strong>fusion of Tallow emulsioncauses <strong>in</strong>sul<strong>in</strong> resistance <strong>in</strong> Holste<strong>in</strong> cows. J Dairy Sci 90,2735–2744.Rastani RR, Grummer RR, Bertics SJ, Gumen A, WiltbankMC, Mashek DG, Schwab MC, 2005: Reduc<strong>in</strong>g dry periodlength to simplify feed<strong>in</strong>g transition cows: milk production,energy balance, and metabolic profiles. J Dairy Sci 88, 1004–1014.Rhoads ML, Rhoads RP, Gilbert RO, Toole R, Butler WR,2006: Detrimental effects of high plasma urea nitrogen levelson viability of embryos from lactat<strong>in</strong>g dairy cows. AnimReprod Sci 91, 1–10.Rivera RM, Hansen PJ, 2001: Development of cultured bov<strong>in</strong>eembryos after exposure to high temperatures <strong>in</strong> the physiologicalrange. <strong>Reproduction</strong> 121, 107–115.Roche JF, 2006: The effect of nutritional management of thedairy cow on reproductive efficiency. Anim Reprod Sci 96,282–296.Roche JR, Berry DP, Kolver ES, 2006: Holste<strong>in</strong>-Friesianstra<strong>in</strong>s and feed effects on milk production, bodyweight andbody condition score profiles <strong>in</strong> graz<strong>in</strong>g dairy cows. J DairySci 89, 3532–3543.Roche JR, Macdonald KA, Burke CR, Lee JM, Berry DP,2007: Associations among body condition score, bodyweight and reproductive performance <strong>in</strong> seasonal-calv<strong>in</strong>gdairy cattle. J Dairy Sci 90, 376–391.Roth Z, Meweidan R, Shaham-Albalancy A, Braw-Tal R,Wolfenson D, 2001a: Delayed effect of heat stress on steroidproduction <strong>in</strong> medium-size and preovulatory bov<strong>in</strong>e follicles.<strong>Reproduction</strong> 121, 745–751.Roth Z, Arav A, Bor A, Zeron Y, Braw-Tal R, Wolfenson D,2001b: Improvement of quality of oocytes collected <strong>in</strong> theautumn by enhanced removal of impaired follicles frompreviously heat-stressed cows. <strong>Reproduction</strong> 122, 737–744.Royal M, Mann GE, Fl<strong>in</strong>t APF, 2000: Strategies for revers<strong>in</strong>gthe trend towards subfertility <strong>in</strong> dairy cattle. Vet J 160, 53–60.Sangsritavong S, Combs DK, Sartori R, Armentano LE,Wiltbank MC, 2002: High feed <strong>in</strong>take <strong>in</strong>crease liver bloodflow and metabolism of progesterone and estradiol-17b <strong>in</strong>dairy cattle. J Dairy Sci 85, 2831–2842.Santos JEP, Thatcher WW, Chebel RC, Cerri RLA, GalvaoKN, 2004a: The effect of embryonic death rates <strong>in</strong> cattle onthe efficacity of estrus synchronization programs. AnimReprod Sci 82 ⁄ 83, 513–535.Santos JE, Cerri RL, Ballou MA, Higg<strong>in</strong>botham GE, Kirk JH,2004b: Effect of tim<strong>in</strong>g of first cl<strong>in</strong>ical mastitis occurrence onlactational and reproductive performance of Holste<strong>in</strong> dairycows. Anim Reprod Sci 80, 31–45.Sartori R, Sartor-Bergfelt R, Mertens SA, Guenther JN,Parrish JJ, Wiltbank MC, 2002: Fertilization and earlyembryonic development <strong>in</strong> heifers and lactat<strong>in</strong>g cows <strong>in</strong>summer and lactat<strong>in</strong>g and dry cows <strong>in</strong> w<strong>in</strong>ter. J Dairy Sci 85,2803–2812.Sartori R, Haughian JM, Shaver RD, Rosa GJM, WiltbankMC, 2004: Comparison of ovarian function and circulat<strong>in</strong>gsteroids <strong>in</strong> estrous cycles of Holste<strong>in</strong> heifers and lactat<strong>in</strong>gcows. J Dairy Sci 87, 905–920.Silvia WJ, 2003: Address<strong>in</strong>g the decl<strong>in</strong>e <strong>in</strong> reproductiveperformance of lactat<strong>in</strong>g dairy cows: a researcher’s perspective.Vet Sci Tomorrow 3, 1–5.S<strong>in</strong>clair KD, Kuran M, Gebbie FE, Webb R, McEvoy TG,2000: Nitrogen metabolism and fertility <strong>in</strong> cattle: II.Development of oocytes recovered from heifers offered dietsdiffer<strong>in</strong>g <strong>in</strong> their rate of nitrogen release <strong>in</strong> the rumen. JAnim Sci 78, 2670–2680.Snijders SE, Dillon P, O’Callaghan D, Boland MP, 2000:Effect of genetic merit, milk yield, body condition andlactation number on <strong>in</strong> vitro oocyte development <strong>in</strong> dairycows. Theriogenology 53, 981–989.Spicer LJ, Echternkamp SE, 1995: The ovarian <strong>in</strong>sul<strong>in</strong> and<strong>in</strong>sul<strong>in</strong>-like growth factor system with an emphasis ondomestic animals. Domest Anim Endocr<strong>in</strong>ol 12, 223–245.Spicer LJ, Francisco CC, 1997: The adipose obese geneproduct, lept<strong>in</strong>: evidence of a direct <strong>in</strong>hibitory role <strong>in</strong>ovarian function. Endocr<strong>in</strong>ology 138, 3374–3379.Spicer LJ, Francisco CC, 1998: Adipose obese gene product,lept<strong>in</strong>, <strong>in</strong>hibits bov<strong>in</strong>e ovarian thecal cell steroidogenesis.Biol Reprod 58, 207–212.Spicer LJ, Chamberla<strong>in</strong> CS, Francisco CC, 2000: Ovarianaction of lept<strong>in</strong>: effects on <strong>in</strong>sul<strong>in</strong>-like growth factor-I-Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Nutrient Prioritization and Fertility <strong>in</strong> Dairy Cows 103stimulated function of granulosa and thecal cells. Endocr 12,53–59.Sutton ML, Gilchrist RB, Thompson JG, 2003: Effects of <strong>in</strong>vivo and <strong>in</strong> vitro environments on the metabolism of thecumulus-oocyte complex and its <strong>in</strong>fluence on oocyte developmentalcapacity. Hum Reprod Update 9, 35–48.Tamm<strong>in</strong>ga S, Luteijn PA, Meijer RGM, 1997: Changes <strong>in</strong>composition and energy content of liveweight loss <strong>in</strong> dairycows with time after parturition. Livestock Prod Sci 52, 129–155.Thatcher WW, Bilby TR, Bartolome JA, Silvestre F, StaplesCR, Santos JEP, 2006: Strategies for improv<strong>in</strong>g fertility <strong>in</strong>the modern dairy cow. Theriogenology 65, 30–44.Van Knegsel ATM, Van den Brand H, Dijkstra J, van StraalenWM, Heetkamp MJW, Tamm<strong>in</strong>ga S, Kemp B, 2007a:Dietary energy source <strong>in</strong> dairy cows <strong>in</strong> early lactation:energy partition<strong>in</strong>g and milk composition. J Dairy Sci 90,1467–1476.Van Knegsel ATM, Van den Brand H, Dijkstra J, van StraalenWM, Jorritsma R, Tamm<strong>in</strong>ga S, Kemp B, 2007b: Effect ofglucogenic vs. lipogenic diets on energy balance, bloodmetabolites, and reproduction <strong>in</strong> primiparous and multiparousdairy cows <strong>in</strong> early lactation. J Dairy Sci 90, 3397–3409.Vanholder T, Leroy JLMR, Dewulf J, Duchateau L, CorynM, de Kruif A, 2005a: Hormonal and metabolic profiles ofhigh-yield<strong>in</strong>g dairy cows prior to ovarian cyst formation orfirst ovulation post partum. Reprod Domest Anim 40, 460–469.Vanholder T, Leroy JLMR, Van Soom A, Opsomer G, MaesD, Coryn M, de Kruif A, 2005b: Effect of non-esterifiedfatty acids on bov<strong>in</strong>e granulosa cell steroidogenesis andproliferation <strong>in</strong> vitro. Anim Reprod Sci 87, 33–44.Vanholder T, Opsomer G, de Kruif A, 2006a: Aetiology andpathogenesis of cystic ovarian follicles <strong>in</strong> dairy cattle: areview. Reprod Nutr Dev 46, 105–119.Vanholder T, Leroy JLMR, Van Soom A, Maes D, Coryn M,Fiers T, de Kruif A, Opsomer G, 2006b: Effect of nonesterifiedfatty acids on bov<strong>in</strong>e theca cell steroidogenesis andproliferation <strong>in</strong> vitro. Anim Reprod Sci 92, 51–63.Vernon RG, 2002: Nutrient partition<strong>in</strong>g, lipid metabolism andrelevant imbalances. Proceed<strong>in</strong>gs of the 12th World BuiatricsCongress, Hannover, Germany, 18–23 August, 2002.Villa-Godoy A, Hughes TL, Emery RS, Chap<strong>in</strong> LT, FogwellRL, 1988: Association between energy balance and lutealfunction <strong>in</strong> lactat<strong>in</strong>g dairy cows. J Dairy Sci 71, 1063–1072.de Vries MJ, Veerkamp RF, 2000: Energy balance of dairycattle <strong>in</strong> relation to milk production variables and fertility.J Dairy Sci 83, 62–69.Wade GN, Jones JE, 2004: Neuroendocr<strong>in</strong>ology of nutritional<strong>in</strong>fertility. Am J Physiol. Regul, Integrative Comp Physiol287, 1277–1296.Wathes DC, Fenwick M, Cheng Z, Bourne N, Llewellyn S,Morris DG, Kenny D, Murphy J, Fitzpatrick R, 2007:Influence of negative energy balance on cyclicity and fertility<strong>in</strong> the high produc<strong>in</strong>g dairy cow. Theriogenology 68, S232–S241.Webb R, Garnsworthy PC, Gong JG, Rob<strong>in</strong>son RS, WathesDC, 1999: Consequences for reproductive function ofmetabolic adaptation to load. Anim Sci Occasion Publ 24,99–112.Wilson SJ, Marion RS, Spa<strong>in</strong> JN, Spiers DE, Keisler DH,Lucy MC, 1998: Effect of controlled heat stress on ovarianfunction <strong>in</strong> dairy cattle: I. Lactat<strong>in</strong>g cows. J Dairy Sci 1,2124–2131.Wiltbank MC, Sartori R, Sangsritavong S, Lopez H, HaughianJM, Fricke PM, Gumen A, 2001: Novel effects ofnutrition on reproduction <strong>in</strong> lactat<strong>in</strong>g dairy cows. J DairySci Suppl 84, 32 (abstract).Wiltbank M, Lopez H, Sartori R, Sangsritavong S, Gu¨ men A,2006: Changes <strong>in</strong> reproductive physiology of lactat<strong>in</strong>g dairycows due to elevated steroid metabolism. Theriogenology65, 17–29.Wolfenson D, Lew BJ, Thatcher WW, Graber Y, Meidan R,1997: Seasonal and acute heat stress effects on steroidproduction by dom<strong>in</strong>ant follicles <strong>in</strong> cow. Anim Reprod Sci47, 9–19.Wynne K, Stanley S, McGowan B, Bloom S, 2005: Appetitecontrol. J Endocr<strong>in</strong>ol 184, 291–318.Yaakub H, O’Callaghan D, Boland MP, 1999: Effect ofroughage type and concentrate supplementation on folliclenumbers and <strong>in</strong> vitro fertilisation on follicle numbers and <strong>in</strong>vitro fertilisation and development of oocytes recoveredfrom beef heifers. Anim Reprod Sci 44, 1–12.Zhao F, Dixon WT, Kennelly JJ, 1996: Localization and geneexpression of glucose transporters <strong>in</strong> bov<strong>in</strong>e mammarygland. Comprehens Biochem Physiol 115, 127–134.Zieba DA, Amstalden M, Williams GL, 2005: Regulatoryroles of lept<strong>in</strong> <strong>in</strong> reproduction and metabolism: a comparativereview. Domest Anim Endocr<strong>in</strong>ol 29, 166–185.Author’s address (for correspondence): J Leroy, Laboratory forVeter<strong>in</strong>ary Physiology, Department of Veter<strong>in</strong>ary Sciences, Facultyof Pharmaceutical, Biomedical and Veter<strong>in</strong>ary Sciences, University ofAntwerp, Universiteitsple<strong>in</strong> 1, Gebouw U, B-2610 Wilrijk, Belgium.E-mail: jo.leroy@ua.ac.beConflict of <strong>in</strong>terests: All authors declare no conflict of <strong>in</strong>terests.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 104–112 (2008); doi: 10.1111/j.1439-0531.2008.01149.xISSN 0936-6768Corpus Luteum–Endometrium–Embryo Interactions <strong>in</strong> the Dairy Cow: Underly<strong>in</strong>gMechanisms and Cl<strong>in</strong>ical RelevanceRS Rob<strong>in</strong>son 1 , AJ Hammond 2 , DC Wathes 3 , MG Hunter 2 and GE Mann 21 School of Veter<strong>in</strong>ary Medic<strong>in</strong>e and Science; 2 School of Biosciences, University of Nott<strong>in</strong>gham, Sutton Bon<strong>in</strong>gton Campus, Loughborough, Leics;3 Department of Veter<strong>in</strong>ary Basic Sciences, Royal Veter<strong>in</strong>ary College, Hawkshead Lane, North Mymms, Hatfield, Hertfordshire, UKContentsConception rates of dairy cows are currently decl<strong>in</strong><strong>in</strong>g at anestimated 1% every year. Approximately, 35% of embryos failto prevent luteolysis dur<strong>in</strong>g the first three weeks of gestation.Interactions between the corpus luteum, endometrium andembryo are critical to the successful establishment of pregnancyand <strong>in</strong>adequacies will result <strong>in</strong> the mortality of theembryo. For example, as little as a one day delay <strong>in</strong> the postovulatoryrise of progesterone has serious consequences forembryo development and survival. Recently, we found thatLH support, degree of vascularization and luteal cell steroidogeniccapacity were not the major factors responsible forthis luteal <strong>in</strong>adequacy, but are nevertheless essential for lutealdevelopment and function. Progesterone act<strong>in</strong>g on its receptor<strong>in</strong> the endometrium stimulates the production of endometrialsecretions on which the free-liv<strong>in</strong>g embryo is dependent.However, their exact composition and effects of <strong>in</strong>adequateprogesterone rema<strong>in</strong>s to be determ<strong>in</strong>ed. The embryo isrecognized through its secretion of <strong>in</strong>terferon tau (IFNT),which suppresses luteolytic pulses of prostagland<strong>in</strong> F 2a . In thecow, it is most likely that IFNT <strong>in</strong>hibits oxytoc<strong>in</strong> receptor upregulationdirectly and does not require the prior <strong>in</strong>hibition ofoestrogen receptor a (ESR1). Unravell<strong>in</strong>g the precise lutealendometriumand embryo <strong>in</strong>teractions is essential for us tounderstand pregnancy establishment and development ofstrategies to reverse the decl<strong>in</strong><strong>in</strong>g fertility of dairy cows.IntroductionOver the past 20 years, <strong>in</strong> the UK, calv<strong>in</strong>g rate to firstservice has fallen from approximately 60% to 40%(Royal et al. 2000), a decl<strong>in</strong>e of 1% every year. Similardecl<strong>in</strong>es have been seen <strong>in</strong> the USA (Lucy 2001).However, while conception rates <strong>in</strong> lactat<strong>in</strong>g cows havefallen, conception rates <strong>in</strong> maiden heifers have not.While the decl<strong>in</strong>e <strong>in</strong> fertility <strong>in</strong> lactat<strong>in</strong>g cows is welldocumented, the precise causes are still the subject ofconsiderable debate, although <strong>in</strong>volvement of <strong>in</strong>creasedproductivity is irrefutable. The tim<strong>in</strong>g and extent ofpregnancy losses have been reviewed extensively bySreenan and Disk<strong>in</strong> (1986) and Peters (1996). Yet, whileit is clear that <strong>in</strong> a typical dairy herd


CL–Endometrium–Embryo Interactions 105mechanisms beh<strong>in</strong>d <strong>in</strong>adequate production of thesehormones.Pre-ovulatory Events and Subsequent EmbryoDevelopmentThe post-ovulatory embryo–endometrium–ovary <strong>in</strong>teractionsbeg<strong>in</strong> with the <strong>in</strong>timate <strong>in</strong>ter-play between thefollicle and oocyte. Moreover, follicular and oocyte‘quality’ are undoubtedly l<strong>in</strong>ked (Wathes et al. 2003;Hunter et al. 2005). The mammalian oocyte undergoessignificant changes while enclosed with<strong>in</strong> the follicle,particularly when it becomes dom<strong>in</strong>ant and ovulationapproaches. Oocytes recovered from large antral folliclesdevelop to blastocysts at significantly higher ratesthan those from smaller follicles (Lonergan et al. 1994),suggest<strong>in</strong>g that the stage of follicle development atwhich the oocyte is removed is important <strong>in</strong> determ<strong>in</strong><strong>in</strong>gits development. Various studies have shown thatnot only a higher proportion of <strong>in</strong> vivo-matured oocytesreach the blastocyst stage compared with their <strong>in</strong> vitromaturedcounterparts (Greve et al. 1987; Rizos et al.2002), but also that it is the late pre-ovulatory stagethat is critical for oocytes to obta<strong>in</strong> developmentalcapacity (Humblot et al. 2005). This suggests that the<strong>in</strong> vitro environment is <strong>in</strong>adequate to support oocytematuration and stimulate the correct pattern of geneexpression, as it occurs <strong>in</strong> vivo. Precisely, what is lack<strong>in</strong>g<strong>in</strong> the <strong>in</strong> vitro environment is currently unknown, but isthe focus of much research (Lonergan 2007). Inaddition to affect<strong>in</strong>g oocyte competence, abnormalities<strong>in</strong> pre-ovulatory follicle function (such as that whichoccurs, for example, dur<strong>in</strong>g resumption of ovulation <strong>in</strong>post-partum dairy cows) may also detrimentally affectoestradiol production and the number and differentiatedstate of granulosa and theca cells; this maysubsequently impact on the growth and developmentof the corpus luteum (CL) and its ability to produceprogesterone (Wathes et al. 2003; Rob<strong>in</strong>son et al.2005).Post-ovulatory Period: The Initiation ofMaternal–Embryonic Interactions (Day 1–5)There are three critical events dur<strong>in</strong>g the post-ovulatoryperiod. First, the cow must create an appropriateoviductal environment and secretions to nurture thedevelopment of the zygote to a morula. The oviductprovides support for the free-liv<strong>in</strong>g embryo not only <strong>in</strong>the form of nutrients (e.g. ions, am<strong>in</strong>o acids andglucose) but also local growth factors such as <strong>in</strong>sul<strong>in</strong>likegrowth factors (IGF) 1 and 2. The IGFs may besecreted <strong>in</strong>to the oviductal lumen, where they could actdirectly on the develop<strong>in</strong>g embryo. Alternatively, theymay stimulate oviductal secretions by act<strong>in</strong>g on theIGF type 1 receptor (IGF1R), which is located <strong>in</strong> theglandular epithelial cells (Pushpakumara et al. 2002).These hypotheses are supported by the observationsthat IGF1 was able to stimulate the formation of<strong>in</strong> vitro blastocysts (Moreira et al. 2002). Furthermore,the oviductal expression patterns of IGFBP2 andIGFBP6 are altered for cows <strong>in</strong> negative energy balance(Fenwick et al. 2008). As both these two b<strong>in</strong>d<strong>in</strong>gprote<strong>in</strong>s have higher aff<strong>in</strong>ity for IGF2 than IGF1, thislikely alters the IGF signall<strong>in</strong>g pathways with<strong>in</strong> theoviductal environment. The ovary is the pr<strong>in</strong>cipalconductor for these changes to occur <strong>in</strong> a timely andsite-specific manner. For example, oestradiol up-regulatesIGF1 mRNA expression <strong>in</strong> the oviduct (Pushpakumaraet al. 2002) thereby creat<strong>in</strong>g an ‘IGF-rich’environment for the imm<strong>in</strong>ent arrival of the embryo. Inturn, the embryo regulates the bioavailability of theseIGFs by secret<strong>in</strong>g IGF-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s (IGFBP),thereby creat<strong>in</strong>g the appropriate environment for itsdevelopment (Watson et al. 1999). Secondly, embryonictransport through the oviduct requires the coord<strong>in</strong>atedaction of cilia and oviductal smooth muscle contractionsto enable the timely entrance of the morula <strong>in</strong>tothe uterus.Thirdly, the formation of the CL and the rise <strong>in</strong> postovulatoryprogesterone are of critical importance. Thisfollicular-luteal transition is a dynamic process <strong>in</strong>volv<strong>in</strong>ga series of biochemical and morphological changes<strong>in</strong> the pre-ovulatory follicle <strong>in</strong>duced by the LH surge(Reynolds and Redmer 1999; Niswender et al. 2000;Wathes et al. 2003). These <strong>in</strong>clude basement membranebreakdown, theca and granulosa differentiation <strong>in</strong>tosmall and large luteal cells, respectively, tissue remodell<strong>in</strong>gand growth. Concomitantly, there is <strong>in</strong>creasedsteroidogenesis (through the up-regulation of 3b-HSDand p450scc) and a switch <strong>in</strong> steroidogenesis toprogesterone synthesis (through the down-regulationof aromatase). Progesterone production is also <strong>in</strong>creasedthrough the up-regulation of key regulatoryprote<strong>in</strong>s, such as steroid acute regulatory prote<strong>in</strong> andperipheral-type benzodiazep<strong>in</strong>e receptors, which controlthe rate-limit<strong>in</strong>g transport of cholesterol from thecytoplasm to the <strong>in</strong>ner mitochondrial membrane. Progesteroneproduction and its <strong>in</strong>tricate control have beenextensively <strong>in</strong>vestigated and expertly reviewed (e.g.Niswender et al. 2000). Consequently, peripheral progesteroneconcentrations start to <strong>in</strong>crease approximatelyby day 4 and by day 8–10 reach maximalconcentrations. It is this rapid decl<strong>in</strong>e <strong>in</strong> oestradiolconcentrations and subsequent rise <strong>in</strong> the progesteronethat provides the timely control of both oviductal andendometrial functions. Indeed, Green et al. (2005)showed that as early as day 5, the embryos from cowswith greater progesterone concentrations showed moreadvanced development.Formation of the Blastocyst and Progesterone(Day 5–12)Upon entry <strong>in</strong>to the uterus, the blastocyst developswith the formation of the blastocoele surrounded bythe trophectoderm and it is <strong>in</strong> this w<strong>in</strong>dow thatprogesterone is likely to have its greatest <strong>in</strong>fluence onthe development of the embryo (Mann et al. 1999).This is supported by the observation that progesteronereceptor (PGR) levels are maximal <strong>in</strong> both endometrialglands and sup-epithelial stroma from day 4 to day 10(Boos et al. 1996; Kimm<strong>in</strong>s and MacLaren 2001;Rob<strong>in</strong>son et al. 2001). Thus, it is likely that oestradiol<strong>in</strong>itiates PGR up-regulation dur<strong>in</strong>g the follicularphase. PGR levels further <strong>in</strong>crease <strong>in</strong> the early lutealÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


106 RS Rob<strong>in</strong>son, AJ Hammond, DC Wathes, MG Hunter and GE Mannphase while progesterone concentrations are still lowand ⁄ or are further stimulated by the rise <strong>in</strong> plasmaoestradiol from the first dom<strong>in</strong>ant follicle on approximatelyday 4–5 of the oestrous cycle. Then, PGRconcentrations decl<strong>in</strong>e after a period of progesteroneexposure. Similar observations have been reported <strong>in</strong>sheep (Wathes and Hamon 1993; Spencer and Bazer1995).The endometrial glands synthesize, secrete and ⁄ ortransport a complex mixture of am<strong>in</strong>o acids, ions,glucose, transport prote<strong>in</strong>s and growth factor calledhistotroph (Bazer 1975). These secretions are essentialfor the development of the blastocyst, which is freeliv<strong>in</strong>gdur<strong>in</strong>g the pre-attachment period. The requirementof these secretions has been demonstrated us<strong>in</strong>g anov<strong>in</strong>e uter<strong>in</strong>e gland knock out (UGKO) model (Grayet al. 2001). In this model, uter<strong>in</strong>e gland development issuppressed by progesterone adm<strong>in</strong>istration to the neonatalewe lamb. In UGKO sheep, embryos develop<strong>in</strong>itially but are arrested at the blastocyst stage, <strong>in</strong>dicat<strong>in</strong>gthat glandular secretions are essential for posthatch<strong>in</strong>gdevelopment. It is progesterone that controlsthese endometrial secretions act<strong>in</strong>g through glandularPGR. This is supported by observations that rapidblastocyst development correlates with ris<strong>in</strong>g progesteroneconcentrations (Sreenan and Disk<strong>in</strong> 1986; Mannand Lamm<strong>in</strong>g 2001). Recent transcriptomic analyseshave helped <strong>in</strong> the identification of the molecularpathways that are <strong>in</strong>volved <strong>in</strong> regulat<strong>in</strong>g endometrialfunctions dur<strong>in</strong>g the bov<strong>in</strong>e oestrous cycle (Bauersachset al. 2005; Mitko et al. 2008). These studies havehighlighted the genes associated with proliferation,cytoskeleton, extra-cellular matrix structure, remodell<strong>in</strong>gand cell motility which were up-regulated dur<strong>in</strong>goestrus. While, genes up-regulated dur<strong>in</strong>g the lutealphase were <strong>in</strong>volved <strong>in</strong> various cellular processes<strong>in</strong>clud<strong>in</strong>g the immune response, prostagland<strong>in</strong> metabolism,cell adhesion, angiogenesis, tricarboxylic acid cycleenzymes, tight junctions and transport prote<strong>in</strong>s. Thelatter group <strong>in</strong>cluded those <strong>in</strong>volved <strong>in</strong> glutamate, metalion, selenium and thyroid hormone transport. These areparticularly <strong>in</strong>terest<strong>in</strong>g as they are likely to control thecomposition of the histotroph. These observationssupport the hypothesis that luteal <strong>in</strong>adequacy adverselyaffects embryonic development through the deficient upregulationof these transporters. As well as hav<strong>in</strong>gfunctional effects, progesterone also exerts profoundstructural changes <strong>in</strong> the endometrium dur<strong>in</strong>g thisperiod. For example, progesterone <strong>in</strong>duces the reduction<strong>in</strong> endometrial area and <strong>in</strong>creases the glandular ductdensity (Wang et al. 2007). This observation raises theexcit<strong>in</strong>g hypothesis that poor embryo development is theresult of reduced glandular formation. F<strong>in</strong>ally, an<strong>in</strong>terest<strong>in</strong>g observation from our studies <strong>in</strong> sheep andcows is the presence of immunosta<strong>in</strong><strong>in</strong>g of non-nuclearprogesterone receptor along the apical surface of thelum<strong>in</strong>al epithelium (Wathes and Hamon 1993; Rob<strong>in</strong>sonet al. 2001). Furthermore, the appearance of thissta<strong>in</strong><strong>in</strong>g pattern was regulated through the oestrouscycle with the highest <strong>in</strong>tensity be<strong>in</strong>g observed on day 12(Rob<strong>in</strong>son et al. 2001). The function and regulation ofthis membrane-located progesterone receptor rema<strong>in</strong>s tobe elucidated.Blastocyst Elongation, Pregnancy Recognitionand Interferon tau (Day 12–20)Inhibition of the luteolytic mechanismThe embryo establishes pregnancy by block<strong>in</strong>g theluteolytic pulses of prostagland<strong>in</strong> (PG) F 2a . The key<strong>in</strong>itiation step <strong>in</strong> luteolysis is the up-regulation of theoxytoc<strong>in</strong> receptor (OXTR) <strong>in</strong> the endometrial lum<strong>in</strong>alepithelium (Wathes and Lamm<strong>in</strong>g 1995; Mann et al.1999). The exact mechanism by which OXTR are upregulatedand the role that oestrogen receptor a (ESR1)plays has been widely debated. However, it is acceptedthat progesterone <strong>in</strong>itially suppresses OXTR and ESR1expression, but after a period of progesterone exposure(10 days) this block is lost (Lamm<strong>in</strong>g and Mann 1995;Spencer and Bazer 1995; Wathes et al. 1996). Also,dur<strong>in</strong>g the follicular phase, ris<strong>in</strong>g oestradiol concentrations<strong>in</strong>duce further up-regulation of OXTR and ESR1(Spencer and Bazer 1995; Rob<strong>in</strong>son et al. 2001). Oestradiolis unlikely to have a direct effect on OXTR, as noclassical oestrogen response element (ERE) has beenlocalized <strong>in</strong> either the bov<strong>in</strong>e (Telgmann et al. 2003) orov<strong>in</strong>e (Flem<strong>in</strong>g et al. 2006) OXTR promoter region.However, there are three ERE half-sites and oestradiolactivation of the OXTR is likely to require either steroidreceptor co-factors, such as SRC1e (Telgmann et al.2003) or the transcription factor SP-1 (Flem<strong>in</strong>g et al.2006). It is always important to note that OXTR are notonly temporally, but also spatially regulated with<strong>in</strong> theendometrium. However, the role of the ESR1 <strong>in</strong> the<strong>in</strong>itiation of OXTR up-regulation rema<strong>in</strong>s equivocal.Spencer et al. (1998) have <strong>in</strong>dicated that ESR1 appearsbefore OXTR <strong>in</strong> sheep, while others have shown thatOXTR appears <strong>in</strong> the lum<strong>in</strong>al epithelium before ESR1<strong>in</strong> both sheep (Wathes and Hamon 1993) and dairy cows(Rob<strong>in</strong>son et al. 1999, 2001). The latter is supported byLeung and Wathes (2000) who showed that OXTR wereup-regulated spontaneously <strong>in</strong> lum<strong>in</strong>al epithelial cells<strong>in</strong> vitro, <strong>in</strong> the absence of oestradiol, but oestradiol didspeed-up the process. Interpretation of the relativetim<strong>in</strong>g of the up-regulation of OXTR and ESR1 genesis further complicated by the variation of tim<strong>in</strong>g ofOXTR up-regulation (rang<strong>in</strong>g from day 14 to day 18)and partially expla<strong>in</strong>s the variation <strong>in</strong> oestrous cyclelength. The possible mechanisms <strong>in</strong>volved are thevariation <strong>in</strong> the tim<strong>in</strong>g of PGR down-regulation or thatOXTR up-regulation is <strong>in</strong>fluenced by the number offollicular waves (either two or three). The latter would<strong>in</strong>dicate that OXTR expression is <strong>in</strong>fluenced by theovary through a mechanism other than progesterone oroestradiol.Dur<strong>in</strong>g early pregnancy, the blastocyst undergoesrapid elongation <strong>in</strong>creas<strong>in</strong>g from 10 cm by day 16, pr<strong>in</strong>cipally because of the rapidtrophoblast growth (Rob<strong>in</strong>son et al. 2006a; Spenceret al. 2007). The elongation of the blastocyst <strong>in</strong>itiatesIFNT production, which is detectable <strong>in</strong> uter<strong>in</strong>e flushesfrom day 12 to day 25 (Roberts et al. 2003). Consequently,IFNT concentrations <strong>in</strong> uter<strong>in</strong>e lum<strong>in</strong>al fluiddramatically <strong>in</strong>crease from day 14 to day 18 (Mannet al. 1999). This is because of the rapid growth of thetrophoblast dur<strong>in</strong>g embryo expansion rather thanchanges <strong>in</strong> IFNT mRNA expression (Rob<strong>in</strong>son et al.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


CL–Endometrium–Embryo Interactions 1072006a). IFNT is widely recognized as the maternalrecognition of pregnancy signal <strong>in</strong> rum<strong>in</strong>ants (Robertset al. 2003) and establishes pregnancy by suppress<strong>in</strong>gthe up-regulation of OXTR and ESR1 (Rob<strong>in</strong>son et al.1999, 2001). Furthermore, recomb<strong>in</strong>ant IFNT can<strong>in</strong>hibit OXTR expression both <strong>in</strong> vivo (Spencer andBazer 1995, Spencer et al. 1998) and <strong>in</strong> vitro (Telgmannet al. 2003). However, what rema<strong>in</strong>s to be elucidated iswhether IFNT acts directly on the OXTR gene (Mannet al. 1999) or through suppression of the ESR1 gene(Spencer et al. 1996). The former hypothesis is supportedby the observations that the bov<strong>in</strong>e OXTRpromoter region conta<strong>in</strong>s an <strong>in</strong>terferon response element(IRE) and that <strong>in</strong>terferon-regulatory factors(IRF)-1 and -2 b<strong>in</strong>d to this site (Telgmann et al. 2003).Moreover, ESR1 was present <strong>in</strong> the lum<strong>in</strong>al epitheliumof pregnant cows from day 12 to day 14 (Rob<strong>in</strong>son et al.2001). Although Flem<strong>in</strong>g et al. (2006) reported thatIRFs had no effect on the ov<strong>in</strong>e OXTR promoter andmay represent a species difference.Interferon tau-stimulated genesInterferon tau also stimulates a plethora of endometrialgenes, which are likely to be important mediators forconceptus development and attachment to the endometrium.For example, IFNT suppresses the immunesystem by silenc<strong>in</strong>g genes, such as b2-microglobul<strong>in</strong>and major histocompatibility complex class 1 polypeptide-relatedsequence, thereby prevent<strong>in</strong>g the rejection ofthe conceptus allograft. IFNT also stimulates genes<strong>in</strong>volved <strong>in</strong> cell proliferation (e.g. Wnt7a), uter<strong>in</strong>ereceptivity and conceptus attachment (e.g. Galect<strong>in</strong> 15,muc<strong>in</strong>s, catheps<strong>in</strong> L). For extensive reviews on theeffects of IFNT on endometrial functions, see Demmerset al. (2001) and Spencer et al. (2007, 2008).Endometrial–embryonic <strong>in</strong>teractionsThe endometrium and embryo secrete a number ofcytok<strong>in</strong>es and growth factors (Martal et al. 1997). These<strong>in</strong>clude the IGF system, fibroblast growth factor (FGF)-1 and -2, transform<strong>in</strong>g growth factor (TGF) a and b andepidermal growth factor (EGF). The precise role ofthese factors is currently unclear, but they are likely toplay an important role <strong>in</strong> the regulation of pre-attachmentblastocyst development, uter<strong>in</strong>e receptivity andstimulation of IFNT secretion (Roberts et al. 2003;Imakawa et al. 2004). There is a strong l<strong>in</strong>k betweenmetabolic status and reproductive function (Watheset al. 2003). Thus, the IGF system is particularly<strong>in</strong>terest<strong>in</strong>g as IGF1 is an important <strong>in</strong>termediary ofmetabolic status. For example, systemic lower IGF1concentrations at two weeks post-partum were associatedwith a longer time to conception as a consequenceof impaired ovarian and uter<strong>in</strong>e function (Wathes et al.2003, 2007; Taylor et al. 2004). IGF1 and its b<strong>in</strong>d<strong>in</strong>gprote<strong>in</strong>s (IGFBPs) are expressed <strong>in</strong> the endometriumand are regulated dur<strong>in</strong>g the oestrous cycle. IGF1mRNA is present <strong>in</strong> the endometrium through theoestrous cycle (Geisert et al. 1991; Rob<strong>in</strong>son et al. 2000)with up-regulation dur<strong>in</strong>g oestrus and dur<strong>in</strong>g the midlutealphase (Rob<strong>in</strong>son et al. 2000). Moreover, IGF1(Kirby et al. 1996; Rob<strong>in</strong>son et al. 2000) and IGF2(Geisert et al. 1991; Bilby et al. 2006) mRNA expressionis <strong>in</strong>creased dur<strong>in</strong>g early pregnancy. This raises thepossibility that either the embryo up-regulates thesegenes or that embryo development was enhanced <strong>in</strong>animals with <strong>in</strong>creased levels of IGF1 and ⁄ or IGF2.Moreover, IGF1 <strong>in</strong> concert with IGF2 can stimulate theproduction of IFNT (Ko et al. 1991). These observationscomb<strong>in</strong>ed with the fact that systemic IGF1concentrations are attenuated <strong>in</strong> cattle with poor fertility(Wathes et al. 2003), <strong>in</strong>dicate that IGFs are likely toplay a key role <strong>in</strong> regulat<strong>in</strong>g endometrial function andsubsequent embryonic development. The endometrial–embryonic <strong>in</strong>teractions through the IGF system aresummarized <strong>in</strong> Fig. 1.The bioavailability of IGFs is <strong>in</strong>fluenced by theIGFBPs, which are generally considered to sequesterIGFs, but can also promote IGF action. Interest<strong>in</strong>gly,IGFBP levels are differentially regulated dur<strong>in</strong>g pregnancywith IGFBP2 mRNA and IGFBP3 mRNA be<strong>in</strong>gdown-regulated (Geisert et al. 1991; Rob<strong>in</strong>son et al.2000), while IGFBP1 mRNA was up-regulated <strong>in</strong> thepresence of an embryo (Rob<strong>in</strong>son et al. 2000). Theexpression pattern of IGFBP1 mRNA was particularly<strong>in</strong>terest<strong>in</strong>g as it was expressed <strong>in</strong> the lum<strong>in</strong>al epitheliumIGFBP1IGFBP2IGF2MyometriumHistotrophIGF1RIGF1R (proliferation)IGF1IGF2(IGF1)IGFBP1 IGFBP2IGFBP3 IGFBP5IGF1R(proliferation)IGF1Systemic IGF1IGFBP5IGFBP3E 2ConceptusIFNTIGFBP1PGRCaruncleBlood vesselFig. 1. A work<strong>in</strong>g hypothesis of the <strong>in</strong>ter-relationships between thecomponents of the <strong>in</strong>sul<strong>in</strong>-like growth factor (IGF) system dur<strong>in</strong>gearly pregnancy <strong>in</strong> the cow. IGF-1 (endometrial-derived) and IGF2(embryonic-derived) stimulate conceptus growth and secretion of<strong>in</strong>terferon tau (IFNT). These effects are likely to be modulated byIGF-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> (IGFBP) 1 (from lum<strong>in</strong>al epithelium; dark-greybox) and IGFBP2 (from sub-epithelial stroma, light-grey box). Based onits expression patterns, IGFBP1 expression is stimulated by IFNT butsuppressed by progesterone. IGF1 act<strong>in</strong>g through IGF type 1 receptors(IGF1R) also stimulates the secretion of required histotroph from theendometrial glands. IGF2 action, modulated by IGFBP3 and IGFBP5 islikely to stimulate caruncular development. IGFBP3 may also play arole <strong>in</strong> regulat<strong>in</strong>g the availability of systemic IGF1, while IGFBP5was localized to the myometrium. The dotted l<strong>in</strong>e <strong>in</strong>dicates proposedregulatory mechanisms; the solid dot represents up-regulation_Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


108 RS Rob<strong>in</strong>son, AJ Hammond, DC Wathes, MG Hunter and GE Mannand its up-regulation co<strong>in</strong>cided with that of blastocystelongation. It is feasible that IGFBP1 <strong>in</strong>creases IGFavailability for the embryo by regulat<strong>in</strong>g the transportof endometrial IGF1 to the uter<strong>in</strong>e lumen. Moreover,the embryo may enhance this process by stimulat<strong>in</strong>gIGFBP1 expression even more. In human implantation,there is considerable evidence for a specific role ofIGFBP1 <strong>in</strong> enhanc<strong>in</strong>g trophoblast <strong>in</strong>vasiveness (Hamiltonet al. 1998). Also IGFBP1 mRNA and endometrialPGR were <strong>in</strong>versely correlated suggest<strong>in</strong>g that progesteronesuppresses IGFBP1 mRNA expression and <strong>in</strong>dicat<strong>in</strong>gthat IGFBP1 transcription is switched on oncethe progesterone block has been lost. This has implications<strong>in</strong> cows with a delayed post-ovulatory rise <strong>in</strong>progesterone <strong>in</strong> which the tim<strong>in</strong>g of this IGFBP1 upregulationwould be shifted effect<strong>in</strong>g IGF bio-availability.The embryo may further <strong>in</strong>crease the availability ofIGF1 and ⁄ or IGF2 by suppress<strong>in</strong>g the expression of the<strong>in</strong>hibitory IGFBP2 and IGFBP3.Luteal Inadequacy: Consequences and CausesThe tim<strong>in</strong>g of the post-ovulatory progesterone rise iscritical to ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g the appropriate synchronybetween the ovary-endometrium and embryo and anyluteal <strong>in</strong>adequacy has dramatic effects. For example,Mann and Lamm<strong>in</strong>g (2001) demonstrated that as littleas one day delay <strong>in</strong> the rise <strong>in</strong> post-ovulatory progesteronesignificantly reduced the subsequent developmentof the embryo. Similarly, Garrett et al. (1988) showedthat supplementation with progesterone on day 1 to day4 <strong>in</strong>creased embryo development. More recently, Mannet al. (2006) demonstrated that early progesteronesupplementation from day 5 to day 9, but not latersupplementation from day 12 to day 16, <strong>in</strong>creased bothtrophoblastic length fivefold and uter<strong>in</strong>e IFNT concentrations.Pre-ovulatory follicleWhile the consequences of luteal <strong>in</strong>adequacy on embryonicdevelopment are well-characterized, the underly<strong>in</strong>gcause of this aberrant luteal function is poorly understood.A major limitation to <strong>in</strong>vestigat<strong>in</strong>g the aetiologyhas been the lack of a robust, physiological model <strong>in</strong>which, from the follicular phase, it can be predictedwhether the progesterone rise would be delayed ornormal. We have recently developed such a model bymanipulat<strong>in</strong>g the dynamics of the follicular phase by<strong>in</strong>duc<strong>in</strong>g luteolysis <strong>in</strong> the presence of either a large(>10 mm) or a small (30 h afteroestrus) also results <strong>in</strong> post-ovulatory luteal <strong>in</strong>adequacy(Mann and Lamm<strong>in</strong>g 2001; Kaim et al. 2003). Us<strong>in</strong>gour model, with detailed endocr<strong>in</strong>ology and ultrasonographicmonitor<strong>in</strong>g of ovarian function, we found noevidence that delayed ovulation per se (LH surge toovulation time) resulted <strong>in</strong> a delayed rise <strong>in</strong> plasma postovulatoryprogesterone (Rob<strong>in</strong>son et al. 2005).Luteal development, luteotrophic support andsteroidogenic capacityThe rates of luteal growth and angiogenesis are such thatthey are only equalled by the fastest grow<strong>in</strong>g tumours.This enables the CL to grow from 0.5 g immediatelypost-ovulation to >5 g with<strong>in</strong> 10 days (Reynolds andRedmer 1999). Our group <strong>in</strong> two <strong>in</strong>dependent studies hasshown that the weight of CL is highly correlated withplasma progesterone concentrations on day 5 postoestrus(Rob<strong>in</strong>son et al. 2006b; Green et al. 2007).However, this relationship is reduced by day 8 andcompletely lost by day 16 (Rob<strong>in</strong>son et al. 2006b). Thus,it would appear that the rate of luteal development (e.g.compactness, luteal hypertrophy) <strong>in</strong>fluences luteal functionrather than size per se. Other potential causes ofluteal <strong>in</strong>adequacy were <strong>in</strong>sufficient luteotrophic support(e.g. number of LH pulses, basal LH and LH pulseamplitude) and aberrant steroidogenic capacity (numberof small ⁄ large luteal cells, <strong>in</strong> vitro progesterone productionunder basal or LH-stimulated conditions). Wefound that none of these factors are associated withluteal <strong>in</strong>adequacy (Rob<strong>in</strong>son et al. 2006b).AngiogenesisIn order to meet these demands, the growth of bloodvessels and establishment of a blood supply (angiogen-Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


CL–Endometrium–Embryo Interactions 109esis) is essential (Reynolds and Redmer 1999; Schamsand Berisha 2004). Indeed, the mature CL is so vascularthat the majority of luteal cells are adjacent to one ormore capillaries (Reynolds and Redmer 1999; Rob<strong>in</strong>sonet al. 2006b) and the CL has one of the greatest bloodflow rates per unit of tissue. Moreover, luteal blood flowis closely associated with the <strong>in</strong>creased plasma progesteroneconcentrations (Acosta et al. 2003). There isevidence that promot<strong>in</strong>g angiogenesis <strong>in</strong> vivo improvesovarian function <strong>in</strong> gilts (Shimizu et al. 2003) andconversely that <strong>in</strong>hibit<strong>in</strong>g angiogenesis suppresses thepost-ovulatory rise <strong>in</strong> progesterone (Yamashita et al.2008). Thus, angiogenesis is essential for the developmentand function of the CL. Us<strong>in</strong>g our luteal <strong>in</strong>adequacymodel, the degree of vascularization (vonWillebrandt factor; endothelial cell marker), pericytecoverage (a-smooth muscle act<strong>in</strong>) and proliferation<strong>in</strong>dex (Ki67) were determ<strong>in</strong>ed. While we found differencesbetween day 5 and day 8 (<strong>in</strong>creased endothelialcell area and decreased proliferation <strong>in</strong>dex), no differencesbetween the high- and low-progesterone groupswere detected (Rob<strong>in</strong>son et al. 2006b). However, the<strong>in</strong>itiation of tissue growth occurs on approximately day1–2, thus days 5 and 8 might be too late to detectFig. 2. The relationship between area of SMA (pericyte marker)sta<strong>in</strong><strong>in</strong>g and plasma progesterone on day 8 of the oestrous cycle. Thearea of SMA and progesterone were positively correlated (r 2 = 0.33;p < 0.05), suggest<strong>in</strong>g that the degree of blood vessel maturation<strong>in</strong>fluences luteal production of progesteronedifferences. Interest<strong>in</strong>gly, the area of pericyte sta<strong>in</strong><strong>in</strong>gwas positively correlated with progesterone concentrationson day 8, <strong>in</strong>dicat<strong>in</strong>g that degree of vasculaturematuration might <strong>in</strong>fluence progesterone production(Fig. 2). This adds to evidence that pericytes might bekey drivers of angiogenesis and vascular function.Vascular endothelial growth factor A (VEGFA) andfibroblast growth factor 2 (FGF2) are potent stimulatorsof endothelial cell proliferation and migration(Carmeliet 2003; Ferrara et al. 2003) and VEGFA isgenerally considered to be the pr<strong>in</strong>cipal growth factorcontroll<strong>in</strong>g angiogenesis. Contrary to expectation, weshowed that FGF2 production, rather than VEGFA, isdynamic dur<strong>in</strong>g the follicle-luteal transition and earlyCL development <strong>in</strong> the cow (Rob<strong>in</strong>son et al. 2006b,2007). Follicular fluid concentrations of FGF2 <strong>in</strong>creasedsixfold after the LH surge and were ma<strong>in</strong>ta<strong>in</strong>edat high concentrations <strong>in</strong> the collapsed follicle. Thissuggested that the somewhat overlooked angiogenicfactor FGF2 plays a more important role <strong>in</strong> the<strong>in</strong>itiation of angiogenesis post-ovulation, as suggestedby Gospodarowicz et al. (1985), while VEGFA plays amore constitutive role <strong>in</strong> the ma<strong>in</strong>tenance of thedevelop<strong>in</strong>g capillaries ⁄ blood vessels. This hypothesisand others are currently be<strong>in</strong>g tested us<strong>in</strong>g a physiologicallyrelevant luteal angiogenesis culture systemrecently developed <strong>in</strong> our laboratory (Fig. 3; Rob<strong>in</strong>sonet al. 2008).This novel physiological system <strong>in</strong>volves culture of amixed population of luteal cells (<strong>in</strong>clud<strong>in</strong>g small, largeluteal, endothelial and fibroblast cells) <strong>in</strong> a specializedendothelial cell medium on fibronect<strong>in</strong>-coated wells. Theendothelial cells develop tubule-like structures and formhighly organized <strong>in</strong>tricate networks, which superficiallyresemble a capillary bed (Fig. 3; Rob<strong>in</strong>son et al. 2008).Furthermore, we found that this endothelial networkformation was <strong>in</strong>creased by both FGF2 and VEGFA<strong>in</strong>dependently, but required the addition of both factorsfor maximal development (Rob<strong>in</strong>son et al. 2008; Fig. 3-a,b). The strength of this system is that it will enable usto elucidate the role of these other cell types (e.g.pericytes) <strong>in</strong> promot<strong>in</strong>g and direct<strong>in</strong>g the formation ofthese endothelial tubule-like structures with a long-termaim of manipulat<strong>in</strong>g angiogenesis and hence progesteroneproduction <strong>in</strong> vivo.(a)(b)Fig. 3. A novel culture system to<strong>in</strong>vestigate luteal angiogenesis.Endothelial cell networks (vonWillebrandt factor, green) oftubule-like structures developedafter 9 days <strong>in</strong> (a) the absence and(b) the presence of 1 ng ⁄ ml fibroblastgrowth factor (FGF) 2 +1ng⁄ ml vascular endothelialgrowth factor A (VEGFA). Theaddition of FGF2 and VEGFA<strong>in</strong>creased endothelial cell areaby 10-fold. The cells werecountersta<strong>in</strong>ed with DAPI(4¢,6-diamid<strong>in</strong>o-2-phenyl<strong>in</strong>dole)(blue)Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


110 RS Rob<strong>in</strong>son, AJ Hammond, DC Wathes, MG Hunter and GE MannConclusionsThe establishment of pregnancy <strong>in</strong> cattle <strong>in</strong>volves aseries of highly coord<strong>in</strong>ated <strong>in</strong>teractions between theovary, endometrium and embryo. First, the appropriatetim<strong>in</strong>g of these <strong>in</strong>teractions is critical. In particular, thetim<strong>in</strong>g of the post-ovulatory rise <strong>in</strong> progesterone determ<strong>in</strong>esboth the transport of the embryo and thestimulation of appropriate endometrial secretions requiredfor the rapid development of the embryo. Any<strong>in</strong>adequacy <strong>in</strong> the post-ovulatory progesterone leads todelayed embryo development and embryo mortality.The ‘status’ of the pre-ovulatory follicle and rate ofsubsequent luteal development have profound effects onluteal function. Secondly, the precise <strong>in</strong>terplay betweenthe embryo and the endometrium on which it isdependent are also crucial. The embryo must blockluteolysis to establish pregnancy. It does this <strong>in</strong> the cowby secret<strong>in</strong>g IFNT, which <strong>in</strong> turn directly suppresses theup-regulation of the OXTR <strong>in</strong> the lum<strong>in</strong>al epithelium.Embryonic development is <strong>in</strong>fluenced by a number ofendometrial growth factors, such as the IGFs. Thesefactors are likely mediators of the cow’s metabolic statusand are thus excit<strong>in</strong>g targets for development ofstrategies to alleviate the decl<strong>in</strong><strong>in</strong>g fertility.AcknowledgementsThis work has been funded by M<strong>in</strong>istry of Agriculture, Fisheries andFood, DEFRA, BBSRC, the Wellcome Trust and the Milk DevelopmentCouncil. We greatly appreciate the technical assistance of staff atthe University of Nott<strong>in</strong>gham and Royal Veter<strong>in</strong>ary College withoutwhich this work would not have been possible.ReferencesAcosta TJ, Hayahsi KG, Ohtani M, Miyamoto A, 2003: Localchanges <strong>in</strong> blood flow with<strong>in</strong> the pre-ovulatory follicle walland early corpus luteum <strong>in</strong> cows. <strong>Reproduction</strong> 125, 759–767.Bauersachs S, Ulbrich SE, Gross K, Schmidt SE, Meyer HH,E<strong>in</strong>spanier R, Wenigerk<strong>in</strong>d H, Vermehren M, Blum H,S<strong>in</strong>owatz F, Wolf E, 2005: Gene expression profil<strong>in</strong>g ofbov<strong>in</strong>e endometrium dur<strong>in</strong>g the oestrous cycle: detection ofmolecular pathways <strong>in</strong>volved <strong>in</strong> functional changes. J MolEndocr<strong>in</strong>ol 34, 889–908.Bazer FW, 1975: Uter<strong>in</strong>e prote<strong>in</strong> secretions: relationshipto development of the conceptus. J Anim Sci 41, 1376–1382.Bilby TR, Sozzi A, Lopez MM, Silvestre FT, Ealy AD, StaplesCR, Thatcher WW, 2006: Pregnancy, bov<strong>in</strong>e somatotrop<strong>in</strong>,and dietary n-3 fatty acids <strong>in</strong> lactat<strong>in</strong>g dairy cows: I.Ovarian, conceptus, and growth hormone-<strong>in</strong>sul<strong>in</strong>-likegrowth factor system responses. J Dairy Sci 89, 3360–3374.Boos A, Meyer W, Schwarz R, Grunert E, 1996: Immunohistochemicalassessment of oestrogen receptor and progesteronereceptor distribution <strong>in</strong> biopsy samples of the bov<strong>in</strong>eendometrium collected throughout the oestrous cycle. AnimReprod Sci 44, 11–21.Carmeliet P, 2003: Angiogenesis <strong>in</strong> health and disease. NatMed 9, 653–660.Demmers KJ, Derecka K, Fl<strong>in</strong>t AP, 2001: Trophoblast<strong>in</strong>terferon and pregnancy. <strong>Reproduction</strong> 121, 41–49.Fenwick MA, Llewellyn S, Fitzpatrick R, Kenny DA, MurphyJJ, Patton J, Wathes DC, 2008: Negative energy balance <strong>in</strong>dairy cows is associated with specific changes <strong>in</strong> IGFb<strong>in</strong>d<strong>in</strong>gprote<strong>in</strong> expression <strong>in</strong> the oviduct. <strong>Reproduction</strong>135, 63–75.Ferrara N, Gerber HP, LeCouter J, 2003: The biology ofVEGF and its receptors. Nat Med 9, 669–676.Flem<strong>in</strong>g JG, Spencer TE, Safe SH, Bazer FW, 2006: Estrogenregulates transcription of the ov<strong>in</strong>e oxytoc<strong>in</strong> receptor genethrough GC-rich SP1 promoter elements. Endocr<strong>in</strong>ology147, 899–911.Garrett JE, Geisert RD, Zavy MT, Morgan GL, 1988:Evidence for maternal regulation of early conceptus growthand development <strong>in</strong> beef cattle. J Reprod Fert 84, 437–446.Geisert RD, Lee CY, Simmen FA, Zavy MT, Fliss AE, BazerFW, Simmen RC, 1991: Expression of messenger RNAsencod<strong>in</strong>g <strong>in</strong>sul<strong>in</strong>-like growth factor-I, -II, and <strong>in</strong>sul<strong>in</strong>-likegrowth factor b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>-2 <strong>in</strong> bov<strong>in</strong>e endometriumdur<strong>in</strong>g the estrous cycle and early pregnancy. Biol Reprod45, 975–983.Gospodarowicz D, Cheng J, Lui GM, Baird A, Esch F, BohlenP, 1985: Corpus luteum angiogenic factor is related tofibroblast growth factor. Endocr<strong>in</strong>ology 117, 2383–2391.Gray CA, Taylor KM, Ramsey WS, Hill JR, Bazer FW, BartolFF, Spencer TE, 2001: Endometrial glands are required forpreimplantation conceptus elongation and survival. BiolReprod 2001 64, 1608–1613.Green MP, Hunter MG, Mann GE, 2005: Relationshipsbetween maternal hormone secretion and embryo developmenton day 5 of pregnancy <strong>in</strong> dairy cows. Anim Reprod Sci88, 179–189.Green MP, Mann GE, Hunter MG, 2007: Luteal characteristicsand progesterone production on day 5 of the bov<strong>in</strong>eoestrous cycle. Reprod Domest Anim 42, 643–647.Greve T, Xu KP, Callesen H, Hyttel P, 1987: In vivodeveloment of <strong>in</strong> vitro fertilized bov<strong>in</strong>e oocytes matured <strong>in</strong>vivo versus <strong>in</strong> vitro. J IVF Embryo Transfer 4, 281–285.Hamilton GS, Lysiak JJ, Han VK, Lala PK, 1998: Autocr<strong>in</strong>eparacr<strong>in</strong>eregulation of human trophoblast <strong>in</strong>vasiveness by<strong>in</strong>sul<strong>in</strong>-like growth factor (IGF)-II and IGF-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>(IGFBP)-1. Exp Cell Res 244, 147–156.Humblot P, Holm P, Lonergan P, Wrenzycki C, Lequarre´ AS,Joly CG, Herrmann D, Lopes A, Rizos D, Niemann H,Callesen H, 2005: Effect of stage of follicle growth dur<strong>in</strong>gsuperovulation on developmental competence of bov<strong>in</strong>eoocytes. Theriogenology 63, 1149–1166.Hunter MG, Rob<strong>in</strong>son RS, Mann GE, Webb R, 2005:Endocr<strong>in</strong>e and paracr<strong>in</strong>e control of follicular developmentand ovulation rate <strong>in</strong> farm species. Anim Reprod Sci 82 ⁄ 83,461–477.Imakawa K, Chang KT, Christenson RK, 2004: Pre-implantationconceptus and maternal uter<strong>in</strong>e communications:molecular events lead<strong>in</strong>g to successful implantation. JReprod Dev 50, 155–169.Kaim M, Bloch A, Wolfenson D, Braw-Tal R, Rosenberg M,Voet H, Folman Y, 2003: Effects of GnRH adm<strong>in</strong>istered tocows at the onset of estrus on tim<strong>in</strong>g of ovulation, endocr<strong>in</strong>eresponses, and conception. J Dairy Sci 86, 2012–2021.Kimm<strong>in</strong>s S, MacLaren LA, 2001: Oestrous cycle and pregnancyeffects on the distribution of oestrogen and progesteronereceptors <strong>in</strong> bov<strong>in</strong>e endometrium. Placenta 22, 742–748.Kirby CJ, Thatcher WW, Collier RJ, Simmen FA, Lucy MC,1996: Effects of growth hormone and pregnancy on expressionof growth hormone receptor, <strong>in</strong>sul<strong>in</strong>-like growth factor-I and <strong>in</strong>sul<strong>in</strong>-like growth factor b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>-2 and -3genes <strong>in</strong> bov<strong>in</strong>e uterus, ovary and oviduct. Biol Reprod 55,996–1002.Ko Y, Lee CJ, Ott TL, Davis MA, Simmen RCM, Bazer FW,Simmen FA, 1991: Insul<strong>in</strong>-like growth factors <strong>in</strong> sheeputer<strong>in</strong>e fluids: concentrations and relationship to ov<strong>in</strong>etrophoblastic prote<strong>in</strong>-1 production dur<strong>in</strong>g early pregnancy.Biol Reprod45, 135–142.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


CL–Endometrium–Embryo Interactions 111Koos RD, 1989: Potential relevance of angiogenic factors toovarian physiology. Sem<strong>in</strong> Reprod Endocr<strong>in</strong>ol 7, 29–40.Lamm<strong>in</strong>g GE, Mann GE, 1995: Control of endometrialoxytoc<strong>in</strong> receptors and prostagland<strong>in</strong> F2 alpha production<strong>in</strong> cows by progesterone and oestradiol. J Reprod Fertil 103,69–73.Leung ST, Wathes DC, 2000: Oestradiol regulation of oxytoc<strong>in</strong>receptor expression <strong>in</strong> cyclic bov<strong>in</strong>e endometrium. JReprod Fertil 119, 287–292.Lonergan P, 2007: State-of-the-art embryo technologies <strong>in</strong>cattle. Soc Reprod Fert Suppl 64, 315–325.Lonergan P, Monaghan P, Rizos D, Boland MP, Gordon I,1994: Effect of follicle size on bov<strong>in</strong>e oocyte quality anddevelopmental competence follow<strong>in</strong>g maturation, fertilizationand culture <strong>in</strong> vitro. Mol Reprod Dev 31, 63–67.Lucy MC, 2001: Reproductive loss <strong>in</strong> high-produc<strong>in</strong>g dairycattle: where will it end? J Dairy Sci 84, 1277–1293.Mann GE, Lamm<strong>in</strong>g GE, 1999: The <strong>in</strong>fluence of progesteronedur<strong>in</strong>g early pregnancy <strong>in</strong> cattle. Reprod Domest Anim 34,269–274.Mann GE, Lamm<strong>in</strong>g GE, 2001: Relationship between maternalendocr<strong>in</strong>e environment, early embryo development and<strong>in</strong>hibition of the luteolytic mechanism <strong>in</strong> cows. <strong>Reproduction</strong>121, 175–180.Mann GE, Lamm<strong>in</strong>g GE, Rob<strong>in</strong>son RS, Wathes DC, 1999:The regulation of <strong>in</strong>terferon-tau production and uter<strong>in</strong>ehormone receptors dur<strong>in</strong>g early pregnancy. J Reprod FertSuppl 54, 317–328.Mann GE, Fray MD, Lamm<strong>in</strong>g GE, 2006: Effects of time ofprogesterone supplementation on embryo development and<strong>in</strong>terferon-s production <strong>in</strong> the cow. Vet J 171, 500–503.Martal J, Chene N, Camous S, Huynh L, Lantier F, HermierP, L’Haridon R, Charpigny G, Charlier M, Chaouat G,1997: Recent developments and potentialities for reduc<strong>in</strong>gembryo mortality <strong>in</strong> rum<strong>in</strong>ants: the role of IFNs and othercytok<strong>in</strong>e <strong>in</strong> early pregnancy. Reprod Fert Dev 9, 355–380.Mitko K, Ulbrich SE, Wenigerk<strong>in</strong>d H, S<strong>in</strong>owatz F, Blum H,2008: Dynamic changes <strong>in</strong> messenger RNA profiles ofbov<strong>in</strong>e endometrium dur<strong>in</strong>g the oestrous cycle. <strong>Reproduction</strong>135, 225–240.Moreira F, Paula-Lopes FF, Hansen PJ, Bad<strong>in</strong>ga L, ThatcherWW, 2002: Effects of growth hormone and <strong>in</strong>sul<strong>in</strong>-likegrowth factor-I on development of <strong>in</strong> vitro derived bov<strong>in</strong>eembryos. Theriogenology 57, 895–907.Niswender GD, Juengel JL, Silva PJ, Rollyson MK, McIntushEW, 2000: Mechanisms controll<strong>in</strong>g the function and lifespan of the corpus luteum. Physiol Rev 80, 1–29.Peters AR, 1996: Embryo mortality <strong>in</strong> the cow. Anim BreedAbstr 64, 587–598.Pushpakumara PG, Rob<strong>in</strong>son RS, Demmers KJ, Mann GE,S<strong>in</strong>clair KD, Webb R, Wathes DC, 2002: Expression ofthe <strong>in</strong>sul<strong>in</strong>-like growth factor (IGF) system <strong>in</strong> the bov<strong>in</strong>eoviduct at oestrus and dur<strong>in</strong>g early pregnancy. <strong>Reproduction</strong>123, 859–868.Reynolds LP, Redmer DA, 1999: Growth and development ofthe corpus luteum. J Reprod Fertil Suppl 54, 181–191.Rizos D, Lonergan P, Ward F, Duffy P, Boland MP, 2002:Consequences of bov<strong>in</strong>e oocyte maturation, fertilization orearly embryo development <strong>in</strong> vitro versus <strong>in</strong> vivo: implicationsfor blastocyst yield and blastocyst quality. MolReprod Dev 61, 234–248.Roberts RM, Ezashi T, Rosenfeld CS, Ealy AD, Kubisch HM,2003: Evolution of the <strong>in</strong>terferon tau genes and theirpromoters, and maternal-trophoblast <strong>in</strong>teractions <strong>in</strong> controlof their expression. Reprod Suppl 61, 239–251.Rob<strong>in</strong>son RS, Mann GE, Lamm<strong>in</strong>g GE, Wathes DC, 1999:The effect of pregnancy on the expression of uter<strong>in</strong>eoxytoc<strong>in</strong>, oestrogen and progesterone receptors dur<strong>in</strong>g earlypregnancy <strong>in</strong> the cow. J Endocr<strong>in</strong>ol 160, 21–33.Rob<strong>in</strong>son RS, Mann GE, Gadd TS, Lamm<strong>in</strong>g GE, WathesDC, 2000: The expression of the IGF system <strong>in</strong> the bov<strong>in</strong>euterus throughout the oestrous cycle and early pregnancy.J Endocr<strong>in</strong>ol 165, 231–243.Rob<strong>in</strong>son RS, Mann GE, Lamm<strong>in</strong>g GE, Wathes DC, 2001:Expression of oxytoc<strong>in</strong>, oestrogen and progesterone receptors<strong>in</strong> uter<strong>in</strong>e biopsy samples throughout the oestrouscycle and early pregnancy <strong>in</strong> cows. <strong>Reproduction</strong> 122, 965–979.Rob<strong>in</strong>son RS, Hammond AJ, Hunter MG, Mann GE, 2005:The <strong>in</strong>duction of a delayed post-ovulatory progesterone rise<strong>in</strong> dairy cows: a novel model. Domest Anim Endocr<strong>in</strong>ol 28,285–295.Rob<strong>in</strong>son RS, Fray MD, Wathes DC, Lamm<strong>in</strong>g GE, MannGE, 2006a: In vivo expression of <strong>in</strong>terferon tau mRNA bythe embryonic trophoblast and uter<strong>in</strong>e concentrations of<strong>in</strong>terferon tau prote<strong>in</strong> dur<strong>in</strong>g early pregnancy <strong>in</strong> the cow.Mol Reprod Dev 73, 470–474.Rob<strong>in</strong>son RS, Hammond AJ, Nickl<strong>in</strong> LT, Mann GE, HunterMG, 2006b: Endocr<strong>in</strong>e and cellular characteristics ofcorpora lutea from cows with a delayed post-ovulatoryprogesterone rise. Domest Anim Endocr<strong>in</strong>ol 31, 154–172.Rob<strong>in</strong>son RS, Nickl<strong>in</strong> LT, Hammond AJ, Schams D, HunterMG, Mann GE, 2007: FGF2 is more dynamic than VEGFAdur<strong>in</strong>g the follicle-luteal transition <strong>in</strong> the cow. Biol Reprod77, 28–36.Rob<strong>in</strong>son RS, Hammond AJ, Mann GE, Hunter MG, 2008: Anovel physiological culture system that mimics lutealangiogenesis. <strong>Reproduction</strong> 135, 405–413.Royal MD, Darwash AO, Fl<strong>in</strong>t AP, Webb R, Woolliams JA,Lamm<strong>in</strong>g GE, 2000: Decl<strong>in</strong><strong>in</strong>g fertility <strong>in</strong> dairy cattle:changes <strong>in</strong> traditional and endocr<strong>in</strong>e parameters of fertility.Anim Sci 70, 487–501.Schams D, Berisha B, 2004: Regulation of corpus luteumfunction <strong>in</strong> cattle: an overview. Reprod Domest Anim 39,241–251.Shimizu T, Jiang JY, Iijima K, Miyabayashi K, Ogawa Y,Sasada H, Sato E, 2003: Induction of follicular developmentby direct s<strong>in</strong>gle <strong>in</strong>jection of vascular endothelial growthfactor gene fragments <strong>in</strong>to the ovary of m<strong>in</strong>iature gilts. BiolReprod 69, 1388–1393.Spencer TE, Bazer FW, 1995: Temporal and spatial alterations<strong>in</strong> uter<strong>in</strong>e estrogen receptor and progesterone receptor geneexpression dur<strong>in</strong>g the estrous cycle and early pregnancy <strong>in</strong>the ewe. Biol Reprod 53, 1527–1543.Spencer TE, Mirando MA, Mayes JS, Watson GH, Ott TL,Bazer FW, 1996: Effects of <strong>in</strong>terferon-tau and progesteroneon oestrogen-stimulated expression of receptors for oestrogen,progesterone and oxytoc<strong>in</strong> <strong>in</strong> the endometrium ofovariectomized ewes. Reprod Fertil Dev 8, 843–853.Spencer TE, Ott TL, Bazer FW, 1998: Expression of <strong>in</strong>terferonregulatory factors one and two <strong>in</strong> the ov<strong>in</strong>e endometrium:effects of pregnancy and ov<strong>in</strong>e <strong>in</strong>terferon tau. Biol Reprod58, 1154–1162.Spencer TE, Johnson GA, Bazer FW, Burghardt RC, Palmar<strong>in</strong>iM, 2007: Pregnancy recognition and conceptus implantation<strong>in</strong> domestic rum<strong>in</strong>ants: roles of progesterone,<strong>in</strong>terferons and endogenous retroviruses. Reprod FertilDev 19, 65–78.Spencer TE, Sandra O, Wolf E, 2008: Genes <strong>in</strong>volved <strong>in</strong>conceptus-endometrial <strong>in</strong>teractions <strong>in</strong> rum<strong>in</strong>ants: <strong>in</strong>sightsfrom reductionism and thoughts on holistic approaches.<strong>Reproduction</strong> 135, 165–179.Sreenan JM, Disk<strong>in</strong> MG, 1986: The extent and tim<strong>in</strong>g ofembryonic mortality <strong>in</strong> the cow. In: Sreenan JM, Disk<strong>in</strong>MG (eds), Embryo Mortality <strong>in</strong> Farm <strong>Animals</strong>. Mart<strong>in</strong>usNijhoff, Dordrecht, pp. 1–11.Taylor VJ, Cheng Z, Pushpakumara PG, Beever DE, WathesDC, 2004: Relationships between the plasma concentrationsÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


112 RS Rob<strong>in</strong>son, AJ Hammond, DC Wathes, MG Hunter and GE Mannof <strong>in</strong>sul<strong>in</strong>-like growth factor-I <strong>in</strong> dairy cows and theirfertility and milk yield. Vet Rec 155, 583–588.Telgmann R, Bathgate RA, Jaeger S, Tillmann G, Ivell R,2003: Transcriptional regulation of the bov<strong>in</strong>e oxytoc<strong>in</strong>receptor gene. Biol Reprod 68, 1015–1026.Wang CK, Rob<strong>in</strong>son RS, Fl<strong>in</strong>t AP, Mann GE, 2007:Quantitative analysis of changes <strong>in</strong> endometrial glandmorphology dur<strong>in</strong>g the bov<strong>in</strong>e oestrous cycle and theirassociation with progesterone levels. <strong>Reproduction</strong> 134,365–371.Wathes DC, Hamon M, 1993: Localization of oestradiol,progesterone and oxytoc<strong>in</strong> receptors <strong>in</strong> the uterus dur<strong>in</strong>g theoestrous cycle and early pregnancy of the ewe. J Endocr<strong>in</strong>ol138, 479–492.Wathes DC, Lamm<strong>in</strong>g GE, 1995: The oxytoc<strong>in</strong> receptor,luteolysis and the ma<strong>in</strong>tenance of pregnancy. J Reprod FertSuppl 49, 53–67.Wathes DC, Mann GE, Payne JH, Riley PR, Stevenson KR,Lamm<strong>in</strong>g GE, 1996: Regulation of oxytoc<strong>in</strong>, oestradiol andprogesterone receptor concentrations <strong>in</strong> different uter<strong>in</strong>eregions by oestradiol, progesterone and oxytoc<strong>in</strong> <strong>in</strong> ovariectomizedewes. J Endocr<strong>in</strong>ol 151, 375–393.Wathes DC, Taylor VJ, Cheng Z, Mann GE, 2003: Folliclegrowth, corpus luteum function and their effects on embryodevelopment <strong>in</strong> postpartum dairy cows. Reprod Suppl 61,219–237.Wathes DC, Fenwick M, Cheng Z, Bourne N, Llewellyn S,Morris DG, Kenny D, Murphy J, Fitzpatrick R, 2007:Influence of negative energy balance on cyclicity and fertility<strong>in</strong> the high produc<strong>in</strong>g dairy cow. Theriogenology 68S, S232–S241.Watson AJ, Westhus<strong>in</strong> ME, W<strong>in</strong>ger QA, 1999: IGF paracr<strong>in</strong>eand autocr<strong>in</strong>e <strong>in</strong>teractions between conceptus and oviduct.J Reprod Fert Suppl 54, 303–315.Yamashita H, Kamada D, Shirasuna K, Matsui M, Shimizu T,Kida K, Berisha B, Schams D, Miyamoto A, 2008: Effect oflocal neutralization of basic fibroblast growth factor orvascular endothelial growth factor by a specific antibody onthe development of the corpus luteum <strong>in</strong> the cow. MolReprod Dev, <strong>in</strong> press.Zelenik AJ, Schuler HM, Reichert JR, 1981: Gonadotroph<strong>in</strong>b<strong>in</strong>d<strong>in</strong>gsites <strong>in</strong> the rhesus monkey ovary: role of vasculature<strong>in</strong> the selective distribution of human chorionic gonadotroph<strong>in</strong>to the preovulatory follicle. Endocr<strong>in</strong>ology 109, 356–362.Author’s address (for correspondence): R Rob<strong>in</strong>son, School ofVeter<strong>in</strong>ary Medic<strong>in</strong>e and Science, University of Nott<strong>in</strong>gham, SuttonBon<strong>in</strong>gton Campus, Loughborough, Leics LE12 5RD, UK. E-mail:bob.rob<strong>in</strong>son@nott<strong>in</strong>gham.ac.ukConflict of <strong>in</strong>terest: All authors declare no conflict of <strong>in</strong>terests.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 113–121 (2008); doi: 10.1111/j.1439-0531.2008.01150.xISSN 0936-6768Improved Detection of Reproductive Status <strong>in</strong> Dairy Cows Us<strong>in</strong>g Milk ProgesteroneMeasurementsNC Friggens 1 , M Bjerr<strong>in</strong>g 1 , C Ridder 1,2 , S Højsgaard 1 and T Larsen 11 Faculty of Agricultural Sciences, University of Aarhus, Research Centre Foulum, Tjele; 2 Lattec I ⁄ S, Hillerød, DenmarkContentsThis study tested a model for predict<strong>in</strong>g reproductive statusfrom <strong>in</strong>-l<strong>in</strong>e milk progesterone ‘measurements. The model isthat of Friggens and Chagunda [Theriogenology 64 (2005)155]. Milk progesterone measurements (n = 55 036) represent<strong>in</strong>g578 lactations from 380 cows were used to test themodel. Two types of known oestrus were identified: (1)confirmed oestrus (at which <strong>in</strong>sem<strong>in</strong>ation resulted <strong>in</strong> aconfirmed pregnancy, n = 121) and (2) ratified oestrus (wherethe shape of the progesterone profile matched that of theaverage progesterone profile of a confirmed oestrus, n = 679).The model detected 99.2% of the confirmed oestruses. This<strong>in</strong>cluded a number of cases (n = 16) where the smoothedprogesterone did not decrease below 4 ng ⁄ ml. These cows hadsignificantly greater concentrations of progesterone, bothm<strong>in</strong>imum and average, suggest<strong>in</strong>g that between cow variationexists <strong>in</strong> the absolute level of the progesterone profile. Us<strong>in</strong>gratified oestruses, model sensitivity was 93.3% and specificitywas 93.7% for detection of oestrus. Exam<strong>in</strong>ation of falsepositives showed that they were largely associated with lowconcentrations of progesterone, fluctuat<strong>in</strong>g around the4ng⁄ ml threshold. The distribution of time from <strong>in</strong>sem<strong>in</strong>ationuntil the model detected pregnancy failure had a median of22 days post-<strong>in</strong>sem<strong>in</strong>ation. In this test, the model was runus<strong>in</strong>g limited <strong>in</strong>puts, the potential benefits of <strong>in</strong>clud<strong>in</strong>gadditional non-progesterone <strong>in</strong>formation were not evaluated.Despite this, the model performed at least as well as otheroestrus detection systems.IntroductionProgesterone measurements have been used for sometime now, and are accepted as a valid <strong>in</strong>dicator, forassess<strong>in</strong>g the reproductive status of dairy cows (Bulmanand Lamm<strong>in</strong>g 1978; Royal et al. 2000). Implicit <strong>in</strong> thisprocess is a set of biological rules that are used toconvert the progesterone data <strong>in</strong>to different reproductivestatuses (e.g. postpartum anoestrus, luteal andfollicular phases of the oestrous cycle and pregnancy).Traditionally, these rules were expert op<strong>in</strong>ions usuallyapplied manually to progesterone data (e.g. Lamm<strong>in</strong>gand Darwash 1998). As measur<strong>in</strong>g technology hasadvanced from the orig<strong>in</strong>al radioimmunoassays,through laboratory based ELISA methods (Waldmann1993), to on-farm manual tests (Simersky et al. 2007),biosensors (Delwiche et al. 2001), and ultimatelytowards automated <strong>in</strong>-l<strong>in</strong>e systems for measur<strong>in</strong>g milkprogesterone (Lattec 2007), these rules become bothexplicit and fixed. This is because automated systemsnecessarily <strong>in</strong>corporate a model to process and condensethe raw data, provid<strong>in</strong>g the end-user with the relevantbiological <strong>in</strong>terpretation <strong>in</strong> a palatable form.The fact that these biological rules are codified allowsthem to be tested. Test<strong>in</strong>g the ability of these rules tocorrectly categorize reproductive status becomes <strong>in</strong>creas<strong>in</strong>glyimportant as automated <strong>in</strong>-l<strong>in</strong>e progesterone monitor<strong>in</strong>gsystems are implemented on commercial farms.Clearly, the value of a progesterone monitor<strong>in</strong>g systemdepends to a large extent upon how good the biologicalmodel is. The objective of this study was to test the abilityof a model based on <strong>in</strong>-l<strong>in</strong>e milk progesterone measurementsto predict reproductive status. The model tested isthat of Friggens and Chagunda (2005).Time-series models exist for detect<strong>in</strong>g oestrus frommilk traits other than progesterone (de Mol et al. 1999;Firk et al. 2003), and decision strategies for <strong>in</strong>terpret<strong>in</strong>gprogesterone data have been suggested (Lamm<strong>in</strong>g andDarwash 1998; Opsomer et al. 1998; Prandi et al. 1999;Delwiche et al. 2001). However, to our knowledge, theonly published full biological model to predict reproductivestatus based on a time-series of milk progesteronemeasurements is that of Friggens and Chagunda(2005).The normal procedure for test<strong>in</strong>g a new predictor of agiven state is to compare it with some ‘gold standard’reference measure. However, <strong>in</strong> the present case themodel is based on the measure, progesterone, which isthe accepted gold standard for assess<strong>in</strong>g the reproductivestatus of dairy cows (Peters and Ball 1995; Cavalieriet al. 2003). Any conventional test of progesteroneaga<strong>in</strong>st another <strong>in</strong>dicator (e.g. visually determ<strong>in</strong>edoestrus or pedometers) rapidly reverses polarity, becom<strong>in</strong>g<strong>in</strong>stead a test of the other <strong>in</strong>dicator aga<strong>in</strong>stprogesterone. Thus, test<strong>in</strong>g of such a model requires anon-conventional approach, which to a large extentbecomes an exploration of the shapes of the progesteroneprofiles. Consequently, this study also characterizeskey aspects of progesterone profiles accord<strong>in</strong>g toreproductive status.Materials and MethodsTest data collectionMilk progesterone measurements were made on proportionalwhole milk samples collected from all milk<strong>in</strong>gcows <strong>in</strong> one research herd (Danish Cattle ResearchCentre) dur<strong>in</strong>g the period 12 September 2002 to 30September 2006. Cows were milked <strong>in</strong> a robotic milk<strong>in</strong>gsystem with free traffic (mean no. visits ⁄ day = 2.4). Oneprogesterone measure was to be made daily dur<strong>in</strong>g thefirst 120 days from calv<strong>in</strong>g. For the rema<strong>in</strong>der oflactation, progesterone measurements were to be madeevery second day. Actual average <strong>in</strong>tervals betweenprogesterone samples before and after 120 days fromcalv<strong>in</strong>g were: 1.4 and 2.4 days, respectively. ProgesteroneÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


114 NC Friggens, M Bjerr<strong>in</strong>g, C Ridder, S Højsgaard and T Larsenwas analysed us<strong>in</strong>g the Ridgeway ELISA-kit (RidgewayScience Ltd, Gloucestershire, UK). Milk samples werepipetted, diluted and distributed us<strong>in</strong>g a Biomek 2000 Ó(Laboratory Automation Workstation, Beckman Coulter,Fullerton, CA, USA). Milk samples (25 ll, diluted1 + 2 with water) were handled accord<strong>in</strong>g to themanufacturer’s <strong>in</strong>structions, however <strong>in</strong>cubation R4was <strong>in</strong>creased to 1 h 30 m<strong>in</strong>. Plates were read us<strong>in</strong>g aspectrophotometer ⁄ fluorometer, Fluostar Ó (BMG Labtechnologies,Offenburg, Germany) (575 nm). Analyseswere performed <strong>in</strong> 96-well plates; two sets of sevenstandards (0–30 ng ⁄ ml), locally made us<strong>in</strong>g milk froman ovariectomized cow and ethanolic progesteronesolutions, and two sets of two control samples wereused for every analysis and plate. For the low and highcontrols, the <strong>in</strong>tra assay precision (CV%) was 14.9 and1.4, respectively; the <strong>in</strong>ter-assay precision (CV%) was32.7 and 20.1, respectively; and the average <strong>in</strong>accuracy(bias) was +0.82 and )0.60 ng ⁄ ml, respectively.The time series of milk progesterone measurementswas exam<strong>in</strong>ed for gaps longer than 14 days with noprogesterone values. All data follow<strong>in</strong>g such gaps wereexcluded. Further, the entire cow-lactation was excludedif there was <strong>in</strong>sufficient data to identify the end of thepostpartum anoestrus period (i.e. the time series did notstart at calv<strong>in</strong>g and there were less than five measurementsbefore the cow was detected to have elevatedprogesterone and resumed oestrous cycles). The result<strong>in</strong>gdata represented <strong>in</strong>formation dur<strong>in</strong>g 578 lactationsfrom 380 cows. The total number of milk progesteronemeasurements was 55 036.Tim<strong>in</strong>g of <strong>in</strong>sem<strong>in</strong>ations was based on visualdetection of oestrus backed up by activity meter<strong>in</strong>dications (DeLaval, Tumba, Sweden). Farm staffhad no access to the progesterone data. Visualobservations for oestrus signs were made daily at06:00, 12:00 and 16:00, each period lasted 20 m<strong>in</strong>. Cowsshow<strong>in</strong>g oestrus earlier than 35 days from calv<strong>in</strong>g werenot <strong>in</strong>sem<strong>in</strong>ated to that oestrus. All cows were<strong>in</strong>sem<strong>in</strong>ated to oestrus regardless of other managementfactors such as age, milk yield, and whether the cow wasdesignated for subsequent cull<strong>in</strong>g. Insem<strong>in</strong>ations werecarried out the same day for oestruses detected at themorn<strong>in</strong>g observation, and the next morn<strong>in</strong>g foroestruses detected <strong>in</strong> the afternoon and even<strong>in</strong>g.The model be<strong>in</strong>g testedThe model be<strong>in</strong>g tested here has been described <strong>in</strong> detailby Friggens and Chagunda (2005). Briefly, the model isbased on the cow always be<strong>in</strong>g <strong>in</strong> one of threereproductive states (Status), these are: postpartumanoestrus (Status 0), oestrus cycl<strong>in</strong>g (Status 1), andpotentially pregnant (Status 2). In the model thesestatuses are mutually exclusive. The Status the cow wasfound to be <strong>in</strong> by the previous run of the model is alwaysthe Status at the start of the current run. By default, thecow is assumed to be <strong>in</strong> Status 0 at calv<strong>in</strong>g. The model isdynamic and determ<strong>in</strong>istic, designed to run each time anew trigger <strong>in</strong>put occurs us<strong>in</strong>g both the current andprevious values. The ma<strong>in</strong> model <strong>in</strong>puts are a smoothed(extended Kalman filter) progesterone concentrationand slope. Insem<strong>in</strong>ations and pregnancy determ<strong>in</strong>ationsare also <strong>in</strong>cluded as <strong>in</strong>puts. To make use of other knowneffectors of reproductive performance that are notnecessarily reflected <strong>in</strong> progesterone concentrations,the model is designed to <strong>in</strong>corporate a number ofadditional <strong>in</strong>puts. However, the model is designed tofunction <strong>in</strong> the absence of these additional <strong>in</strong>puts(although with some loss of accuracy <strong>in</strong> some outputs).The model outputs are all reproductive Status specificwith the exception of days to next sample (DNS) whichis calculated <strong>in</strong> each model run regardless of reproductiveStatus. Days to next sample is an important outputwith respect to the progesterone measurements be<strong>in</strong>gautomated. It is designed to feedback to the sampl<strong>in</strong>gsystem so that the frequency of milk sampl<strong>in</strong>g (i.e.progesterone measurement) can be varied accord<strong>in</strong>g tothe predicted likelihood of a future reproductive eventsuch as onset of oestrus cycles. The other model outputsare: risk of prolonged postpartum anoestrus, risk andtype of ovarian cyst (i.e. prolonged luteal or follicularphase), onset of oestrus, likelihood of a potential<strong>in</strong>sem<strong>in</strong>ation succeed<strong>in</strong>g and likelihood of be<strong>in</strong>g pregnant(follow<strong>in</strong>g oestrus).The shift from Status 0 (postpartum anoestrus) toStatus 1 (oestrus cycl<strong>in</strong>g) is caused by two consecutivesmoothed progesterone measurements above the thresholdvalue (LThresh) which is 4 ng ⁄ ml. (If EOD <strong>in</strong>putsare available these can also provoke this shift). Asdescribed by Friggens and Chagunda (2005), once a cowis <strong>in</strong> Status 1 the shift to Status 2 is caused by thesmoothed progesterone value decreas<strong>in</strong>g belowLThresh. This shift generates a model-detected <strong>in</strong>dicationof oestrus. If no <strong>in</strong>sem<strong>in</strong>ation is recorded <strong>in</strong> thefollow<strong>in</strong>g 5 days, the cow automatically reverts to Status1. If there is a timely <strong>in</strong>sem<strong>in</strong>ation and subsequentprogesterone concentrations <strong>in</strong>crease, then the cowrema<strong>in</strong>s <strong>in</strong> status 2 (potentially pregnant) as long asthe model predicted likelihood of pregnancy rema<strong>in</strong>sabove a pregnancy threshold. Typically, if progesteroneconcentrations decrease, then the model shifts backfrom Status 2 to Status 1.The model tested <strong>in</strong> the present study conta<strong>in</strong>ed thefollow<strong>in</strong>g major modification relative to the modeldescribed by Friggens and Chagunda (2005). Visual<strong>in</strong>spection of the earliest progesterone profiles collectedshowed a number of oestrus-like decreases <strong>in</strong> progesteronethat did not decrease below LThresh. To dealwith this an additional threshold, HThresh was <strong>in</strong>troduced(6 ng ⁄ ml). This allowed identification of these‘high-progesterone oestruses’ conditional on the maximumprogesterone concentration <strong>in</strong> the preced<strong>in</strong>g cyclebe<strong>in</strong>g greater than 15 ng ⁄ ml, and the present decreasebe<strong>in</strong>g >10 days s<strong>in</strong>ce a preced<strong>in</strong>g oestrus.The model tested <strong>in</strong> the present study also conta<strong>in</strong>edthe follow<strong>in</strong>g m<strong>in</strong>or modifications relative to the modeldescribed by Friggens and Chagunda (2005). The slopeof the progesterone profile is used, for example <strong>in</strong> thefunction calculat<strong>in</strong>g the predicted likelihood of AIsuccess. The slope can have extreme values particularlywith short <strong>in</strong>tervals between measurements, thus anadjusted slope (a sigmoid transformation) was used<strong>in</strong>stead. Over the normal range this makes virtually nodifference but it caps the extremes. In Status 2, when thecalculated likelihood of pregnancy (LikePreg) decreasesÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reproductive Status Assessed by Milk Progesterone 115below a pregnancy threshold, the cow is deemed nolonger pregnant and reverts to Status 1. In the currentimplementation of the model, a modification was madesuch that once the cow was more than 30 days after<strong>in</strong>sem<strong>in</strong>ation, 2 consecutive low LikePreg (i.e. lowprogesterone) values are required to cause the changeback to Status 1. F<strong>in</strong>ally, the DNS functions <strong>in</strong> theluteal phase and dur<strong>in</strong>g pregnancy were modified tobetter optimize the sampl<strong>in</strong>g frequency relative to timeof expected next oestrus (equations <strong>in</strong> Appendix).Because sampl<strong>in</strong>g frequency <strong>in</strong> the present study wasnot based on DNS, this change had no bear<strong>in</strong>g on theresults presented here, except for those relat<strong>in</strong>g directlyto chang<strong>in</strong>g the sampl<strong>in</strong>g frequency.Model test<strong>in</strong>gThe model has many features that warrant test<strong>in</strong>g but <strong>in</strong>the present study we have chosen to focus on thedetection of oestrus, end of postpartum anoestrus, andonset of pregnancy, because these are the major outputs,and those by which the usefulness of the model <strong>in</strong>commercial application will be primarily judged. Forthis study, the model was run <strong>in</strong> its most basic form withno additional <strong>in</strong>puts other than <strong>in</strong>sem<strong>in</strong>ations.Oestrus detectionTest<strong>in</strong>g the ability of the model to identify oestrusimplies that there are a series of known oestruses with<strong>in</strong>the data aga<strong>in</strong>st which to compare the progesteronebasedmodel detected oestruses. Both visual observationsof oestrus and activity measurement-based oestrusalarms were available on the test farm. However, it iswell known from other studies (Ross<strong>in</strong>g and Spahr1992), and was patently clear from <strong>in</strong>spection of thedata, that these external oestrus detection methods werenot sufficiently reliable for the purpose of test<strong>in</strong>g themodel. Accord<strong>in</strong>gly, an alternative approach to def<strong>in</strong><strong>in</strong>ga reference set of known oestruses was made. Two typesof known oestrus were identified: confirmed and ratifiedoestruses. A confirmed oestrus was def<strong>in</strong>ed as an oestrusat which <strong>in</strong>sem<strong>in</strong>ation resulted <strong>in</strong> a confirmed pregnancy.A ratified oestrus is one <strong>in</strong> which the shape of theprogesterone profile matches that of the average progesteroneprofile of the confirmed oestruses.Confirmed oestrusesInsem<strong>in</strong>ations that occurred with<strong>in</strong> the time w<strong>in</strong>dow±10 days around the time-po<strong>in</strong>t 284 days before asubsequent calv<strong>in</strong>g or 40 days before a positive pregnancydeterm<strong>in</strong>ation (rectal palpation 6 weeks post<strong>in</strong>sem<strong>in</strong>ation)were assumed to be associated with a true,confirmed oestrus (because pregnancy resulted). Thestart of these confirmed oestruses was def<strong>in</strong>ed as thetime of the first smoothed milk progesterone measurement


116 NC Friggens, M Bjerr<strong>in</strong>g, C Ridder, S Højsgaard and T Larsenthresholds were applied. The probabilities had to begreater than 0.95 and the weighted number of observationshad to be greater than 3. The weighted number ofobservations was calculated as:X(absðt tmidpo<strong>in</strong>t Þþ1Þ 1 ;where t is time from the midpo<strong>in</strong>t of the segment for anygiven observation. For observations 7.5, 5, 2.5 and0 days from the midpo<strong>in</strong>t the <strong>in</strong>dividual weights were;0.12, 0.17, 0.29 and 1, respectively. The threshold ofthree for the sum of the <strong>in</strong>dividual weights correspondsto an observation frequency of two progesterone measurementsper 3 days for evenly spread observations, ora hole of )3 to +3 days from the midpo<strong>in</strong>t <strong>in</strong> anotherwise complete set of observations.In order to avoid the same oestrus be<strong>in</strong>g repeatedlyidentified as the roll<strong>in</strong>g 15-day w<strong>in</strong>dow moves throughthe progesterone time series, the follow<strong>in</strong>g filter<strong>in</strong>g ruleswere applied. In addition to p be<strong>in</strong>g >0.95, p for thepreced<strong>in</strong>g observation had to be >0.90. Once anobservation was ratified, then subsequent observationsmeet<strong>in</strong>g the above criteria were not accepted as ratifiedunless the probability had decreased to 8 ng ⁄ ml(Mahalanobis distance is scale <strong>in</strong>variant, i.e. it assessesthe shape of the segment and not the absolute level ofthe segment). The total number of ratified oestruses was679. The shape of the progesterone profile shown <strong>in</strong>Fig. 1 is that which is associated with oestruses thatoccur after a prior period of luteal activity. Oestrusesthat occur <strong>in</strong> conjunction with the end of the postpartumanoestrus period do not match this profile andhave thus been excluded from the test of the models’ability to detect oestrus.Pregnancy determ<strong>in</strong>ationThe ability of the model to identify cows as pregnantfollow<strong>in</strong>g <strong>in</strong>sem<strong>in</strong>ation was evaluated <strong>in</strong> two ways. Bydef<strong>in</strong>ition, the progesterone profile follow<strong>in</strong>g confirmedoestruses should always be identified by the model as thecow be<strong>in</strong>g <strong>in</strong> Status 2 (pregnant). The proportion ofcases <strong>in</strong> which this was so was calculated, and anyexceptions were exam<strong>in</strong>ed. For those <strong>in</strong>sem<strong>in</strong>ations thatdid not result <strong>in</strong> a confirmed pregnancy (n = 375), thedistribution of time <strong>in</strong>tervals between <strong>in</strong>sem<strong>in</strong>ation andmodel detected pregnancy failure (return from Status 2to 1) was exam<strong>in</strong>ed.Sampl<strong>in</strong>g frequencyThe effects of reduc<strong>in</strong>g the progesterone measurementfrequency on model performance were evaluated bygenerat<strong>in</strong>g new data sets from the orig<strong>in</strong>al, full, progesteronedata <strong>in</strong> which a proportion of the orig<strong>in</strong>alprogesterone measures were removed. This was done byimpos<strong>in</strong>g a m<strong>in</strong>imum <strong>in</strong>terval between consecutiveprogesterone measurements (with<strong>in</strong> each cow-lactationtime-series). For m<strong>in</strong>imum <strong>in</strong>tervals of 1, 2, 3, 4 and5 days, the result<strong>in</strong>g data sets conta<strong>in</strong>ed: 42 459, 26 455,16 756, 8713 and 1535 progesterone measurementsrespectively. With each of these reduced data sets, themodel was run and the proportion of ratified oestrusesthat was detected by the model us<strong>in</strong>g the orig<strong>in</strong>al dataset was calculated. In addition, the model was run us<strong>in</strong>gonly those progesterone measurements that it wouldhave requested if it had been runn<strong>in</strong>g <strong>in</strong> real-timeaccord<strong>in</strong>g to the model output DNS function. This is avariable sampl<strong>in</strong>g frequency as described previously(and <strong>in</strong> Appendix). The total number of progesteronesamples used <strong>in</strong> this case was 11 957.Results and DiscussionConfirmed oestrusesOut of 121 confirmed oestruses, 104 were associatedwith a model-detected oestrus based on a decrease <strong>in</strong>smoothed progesterone below 4 ng ⁄ ml. An additional16 of the confirmed oestruses were model-detected onthe basis of the supplementary oestrus detection rulethat smoothed progesterone decreased below 6 ng ⁄ mlhav<strong>in</strong>g previously been >15 ng ⁄ ml. Thus, the model asimplemented detected 99.2% of the confirmed oestruses,whereas the simpler model (4 ng ⁄ ml threshold) resulted<strong>in</strong> 85.6% of confirmed oestruses be<strong>in</strong>g detected.The majority of studies <strong>in</strong> the literature have used3ng⁄ ml as a threshold for detect<strong>in</strong>g oestrus (Lamm<strong>in</strong>gand Bulman 1976). Thus, a discrepancy exists betweenthis def<strong>in</strong>ition and the fact that <strong>in</strong>sem<strong>in</strong>ation of cowshav<strong>in</strong>g these ‘high progesterone oestruses’ still resulted<strong>in</strong> conception. A possible explanation for this discrepancycould be related to the smooth<strong>in</strong>g of the progesteroneprofiles. The very nature of smooth<strong>in</strong>g is that it isresistant to extreme values. Indeed, this was one reasonfor us<strong>in</strong>g a threshold of 4 rather than 3 ng ⁄ ml <strong>in</strong> themodel. So, if <strong>in</strong>sufficient low progesterone values areavailable, the smoothed profile may not decrease below4ng⁄ ml even when <strong>in</strong>dividual observations are


Reproductive Status Assessed by Milk Progesterone 1174 ng ⁄ ml (31 of 59 of false negatives). Of these, <strong>in</strong> 24cases smoothed progesterone did not decrease below6ng⁄ ml. This high-progesterone oestrus <strong>in</strong>creas<strong>in</strong>glyseems to be a real biological phenomenon (Fig. 2). Ofthe rema<strong>in</strong><strong>in</strong>g 28 false negatives, enlarg<strong>in</strong>g the w<strong>in</strong>dow()3 to + 5 dfo) around oestrus for align<strong>in</strong>g modeldetected and ratified oestruses by ±3 days of the oestruscaptured 21 cases. Accept<strong>in</strong>g these 21 cases as a matchreduced the number of false negatives to 38 result<strong>in</strong>g <strong>in</strong> amodel sensitivity of 93.3%. This sensitivity comparedvery favourably with the sensitivities reported for othermethods of oestrus detection (Firk et al. 2002), evenwhen these methods are used <strong>in</strong> very favourablecircumstances such as high <strong>in</strong>tensity visual oestrusdetection (85.7% Van Eerdenburg 2006) or after oestrussynchronisation (91.3% for tail pa<strong>in</strong>t, 81.4% forpedometers Cavalieri et al. 2003). Only the comb<strong>in</strong>edactivity, milk yield, milk temperature and conductivitymodel of de Mol et al. (1999) had a sensitivity (94%)that matched that for the ratified oestruses. Althoughthis sensitivity was less than the 99% found us<strong>in</strong>g theconfirmed oestruses.In addition to enlarg<strong>in</strong>g the number of oestrusesaccepted as ‘true’, the profile match<strong>in</strong>g procedure allowsexam<strong>in</strong>ation of those cases <strong>in</strong> which the model <strong>in</strong>dicatedoestrus <strong>in</strong> the absence of a ratified oestrus, and thusallows calculation of model specificity. There were 171model-detected oestruses (exclud<strong>in</strong>g first <strong>in</strong>creases <strong>in</strong>progesterone >4 ng ⁄ ml) that did not match a ratifiedoestrus (i.e. false positives). Thus, the model had apositive predictive value (proportion of model detectedoestruses that are true oestruses) of 72.2%[445 ⁄ (445 + 171)]. The total number of progesteronemeasures <strong>in</strong> the period of oestrus cycl<strong>in</strong>g (Status = 1and profile match<strong>in</strong>g weight


118 NC Friggens, M Bjerr<strong>in</strong>g, C Ridder, S Højsgaard and T Larsenbasic specificity of 98.7%. However, this assumes thatfor any given progesterone observation oestrus could be<strong>in</strong>dicated. This was not the case, because <strong>in</strong> the model,once oestrus was <strong>in</strong>dicated, a wait<strong>in</strong>g period of 5 dayswas mandatory before a new oestrus can be <strong>in</strong>dicated(Friggens and Chagunda 2005). Allow<strong>in</strong>g for thiswait<strong>in</strong>g period, the total number of progesterone measurementsfor which oestrus could be determ<strong>in</strong>ed was2 723 result<strong>in</strong>g <strong>in</strong> a specificity of 93.7%. This specificityis similar to that found by de Mol et al. (1999).Exam<strong>in</strong>ation of the false positives showed that theywere largely associated with low progesterone concentrationsfluctuat<strong>in</strong>g around the 4 ng ⁄ ml threshold(Fig. 3). Nearly 53% (90 ⁄ 171) of the false positivescame at the end of ‘oestrus’ cycles <strong>in</strong> which themaximum progesterone concentration did not exceed10 ng ⁄ ml. Only <strong>in</strong> 56 out of the 171 cases had themaximum progesterone concentration <strong>in</strong> the preceed<strong>in</strong>gcycle exceeded 15 ng ⁄ ml. Clearly, every time such afluctuat<strong>in</strong>g profile decreased below 4 ng ⁄ ml, the model<strong>in</strong>dicated oestrus. This is the major drawback of asimple threshold-based rule, which cannot be overcomeby a simple smooth<strong>in</strong>g of the progesterone profiles. Thenumber of false positives could be reduced by simplyus<strong>in</strong>g a much greater threshold, thus <strong>in</strong>creas<strong>in</strong>g modelspecificity. Unfortunately, a greater threshold has asignificant unwanted cost <strong>in</strong> terms of reduc<strong>in</strong>g modelsensitivity (de Mol et al. 2001). Instead, each timeoestrus is detected the model calculates a likelihood of apotential <strong>in</strong>sem<strong>in</strong>ation succeed<strong>in</strong>g. Because this calculation<strong>in</strong>cludes <strong>in</strong>formation about the height and length ofthe preced<strong>in</strong>g oestrus cycle, these false positive oestrusesare associated with a very low likelihood of <strong>in</strong>sem<strong>in</strong>ationsucceed<strong>in</strong>g. Average values of the predicted likelihoodof a potential <strong>in</strong>sem<strong>in</strong>ation succeed<strong>in</strong>g when the maximumprogesterone concentration <strong>in</strong> the preced<strong>in</strong>g cycledid not exceed 5, 10 or 15 ng ⁄ ml were: 0.01, 0.17 and0.45 respectively (on a scale 0–0.9). Thus, false positivesProgesterone (ng/ml)30241812600 30 60 90 120 150 180Days from calv<strong>in</strong>gFig. 3. An example of a progesterone profile show<strong>in</strong>g a fluctuat<strong>in</strong>g lowprogesterone after calv<strong>in</strong>g. Because the smoothed progesterone profile(solid l<strong>in</strong>e) repeatedly decreases below 4 ng/ml dur<strong>in</strong>g this period, themodel repeatedly detected false positive oestruses. Raw, unsmoothedprogesterone values are shown by the open circles. Pluses <strong>in</strong>dicatemodel-determ<strong>in</strong>ed Status 0 (postpartum an oestrus), solid triangles<strong>in</strong>dicate Status 1 (oestrus cycl<strong>in</strong>g), and solid squares <strong>in</strong>dicate Status 2(potentially pregnant). The downward po<strong>in</strong>t<strong>in</strong>g open triangles <strong>in</strong>dicate<strong>in</strong>sem<strong>in</strong>ations, the upwards po<strong>in</strong>t<strong>in</strong>g open triangles <strong>in</strong>dicate pregnancydeterm<strong>in</strong>ations (1 ‘‘ng/ml’’ = not pregnant, 9 ‘‘ng/ml’’ = pregnant)are obvious to the end user of the system. The predictedlikelihood of a potential <strong>in</strong>sem<strong>in</strong>ation succeed<strong>in</strong>g alsoprovides the end user with helpful <strong>in</strong>formation formak<strong>in</strong>g a decision about whether or not to <strong>in</strong>sem<strong>in</strong>atethe cow. This has been explored further by Friggens andLøvendahl (2008).In the model, the first <strong>in</strong>crease <strong>in</strong> the smoothedprogesterone profile above 4 ng ⁄ ml <strong>in</strong>dicates the end ofthe postpartum anoestrus and co<strong>in</strong>cidentally the firstoestrus. For those first oestruses that were ratified, theaverage difference between the time of model detectedfirst oestrus and ratified oestrus was 1.5 days(SD = 1.6). The distribution of the differences was leftskewed by those cases <strong>in</strong> which a period of fluctuationoccurred around the 4-ng ⁄ ml threshold (Fig. 3). Themedian difference was 2.2 days. Thus, the model is <strong>in</strong>good agreement with the ratified oestruses. However,because the ‘Mahalanobis template’ was based onoestruses that occurred after a prior period of lutealactivity, it did not identify all first <strong>in</strong>creases <strong>in</strong> progesteroneabove the threshold. The distribution of thesemodel-detected durations of postpartum anoestrus wassimilar to that reported by Royal et al. (2002).Pregnancy determ<strong>in</strong>ationOf the 121 confirmed pregnancies, 108 rema<strong>in</strong>ed <strong>in</strong>Status 2 (model-detected pregnant) from conceptionuntil at least positive pregnancy determ<strong>in</strong>ation (by rectalpalpation), the sensitivity of the model for detect<strong>in</strong>gpregnancy was thus 89.3%. Of the 11 cases <strong>in</strong> which themodel falsely detected the cow as no longer pregnant, 2were due to gaps <strong>in</strong> the progesterone data, 5 failedbecause the model judged that the <strong>in</strong>sem<strong>in</strong>ation wasmistimed, and 6 were caused by decreases <strong>in</strong> progesteroneconcentration. These decreases were <strong>in</strong> some cases<strong>in</strong>dist<strong>in</strong>guishable from those one would detect if the cowwas return<strong>in</strong>g to oestrus follow<strong>in</strong>g early embryo loss(e.g. a sudden decrease to < 10 ng ⁄ ml) but these cowsrema<strong>in</strong>ed pregnant and subsequently calved. Giventhese types of cases, it is hard to envisage be<strong>in</strong>g ableto substantially improve pregnancy detection withoutsimultaneously <strong>in</strong>creas<strong>in</strong>g the number of false negatives.However, of greater important to the end user isdetect<strong>in</strong>g true pregnancy failure.The distribution of model detected pregnancy lengths,for those pregnancies that did not result <strong>in</strong> a positivepregnancy determ<strong>in</strong>ation or a calf, provides useful<strong>in</strong>formation (Fig. 4). The distribution of time from<strong>in</strong>sem<strong>in</strong>ation to model-detected pregnancy failure isbimodal reflect<strong>in</strong>g the two reasons for the modelassess<strong>in</strong>g the cow to not be pregnant: (1) <strong>in</strong>appropriatetim<strong>in</strong>g of <strong>in</strong>sem<strong>in</strong>ation and (2) progesterone concentrationstoo low (first and second peaks, respectively). Themedian of the distribution was 22 days, reflect<strong>in</strong>g thesecond peak. By 27 days 60% of pregnancy failures hadoccurred, and by 34 days 70% had occurred. Thesefigures reflect a typical time pattern of early embryonicloss (Sreenan et al. 2001). Reliable detection of pregnancyus<strong>in</strong>g progesterone is not possible much earlierthan 18–23 days post-<strong>in</strong>sem<strong>in</strong>ation because the progesteroneprofile dur<strong>in</strong>g the luteal phase is not affected bypresence of an embryo (Bulman and Lamm<strong>in</strong>g 1978).Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reproductive Status Assessed by Milk Progesterone 119Proportion of cases0.160.140.120.100.080.060.040.020.0010 20 30 40 50 60 70 80 90 100110120130140150160170Days from calv<strong>in</strong>gFig. 4. Distribution of pregnancy lengths for cows <strong>in</strong> which pregnancyfailed (model detected reproductive status reverted from pregnancy tooestrus cycl<strong>in</strong>g)The present results <strong>in</strong>dicate that after 23 days the modelreliably detected pregnancy failure. This is substantiallyearlier than the tim<strong>in</strong>g of reliable pregnancy determ<strong>in</strong>ationby rectal palpation at 6 weeks post-<strong>in</strong>sem<strong>in</strong>ation(Peters and Ball 1995).Sampl<strong>in</strong>g frequencyThe above results were generated from a data set <strong>in</strong>which average time between collection of samples was1.4 days (for the period 0–120 days from calv<strong>in</strong>g).Because these results are expected to be <strong>in</strong>fluenced bysampl<strong>in</strong>g frequency, we exam<strong>in</strong>ed the effect of reduc<strong>in</strong>gsampl<strong>in</strong>g frequency. As shown <strong>in</strong> Fig. 5, the relativeproportion of ratified oestruses detected by the modeldecreased with decreas<strong>in</strong>g sampl<strong>in</strong>g frequency. Theseresults were achieved by simulat<strong>in</strong>g an evenly spreadreduction <strong>in</strong> samples and thus sampl<strong>in</strong>g frequency.However, this contrasts with the way the model is set upProportion of oestruses detected1.00.80.60.40.20.01.0 1.5 2.0 2.5 3.0 3.5 4.0 4.5 5.0Interval between progesterone measurementsFig. 5. The effect of reduc<strong>in</strong>g progesterone sampl<strong>in</strong>g frequency(<strong>in</strong>creas<strong>in</strong>g the average <strong>in</strong>terval between progesterone measurements[units = days]) on the proportion of ratified oestruses detected by themodel. The proportion is expressed relative to the case with the fullprogesterone data set. The solid l<strong>in</strong>e <strong>in</strong>dicates the effect of reducedsampl<strong>in</strong>g frequency us<strong>in</strong>g fixed sampl<strong>in</strong>g schedules. The solid circle<strong>in</strong>dicates the proportion of oestruses detected when us<strong>in</strong>g the modeloutput days to next sample function that results <strong>in</strong> a variable sampl<strong>in</strong>gfrequencyto request samples. In a situation <strong>in</strong> which the model ispart of an automated <strong>in</strong>-l<strong>in</strong>e progesterone measur<strong>in</strong>gsystem, it controls sampl<strong>in</strong>g frequency through the DNSfunction. The DNS function implements a variablesampl<strong>in</strong>g frequency that is low immediately afteroestrus, and then <strong>in</strong>creases as a function of bothduration of luteal phase and any evidence of a decrease<strong>in</strong> progesterone (Friggens and Chagunda 2005; see alsoAppendix). Us<strong>in</strong>g this variable function to reduce thesampl<strong>in</strong>g frequency <strong>in</strong> the test data resulted <strong>in</strong> anaverage sampl<strong>in</strong>g frequency of 3.3 days, but the proportionalreduction <strong>in</strong> oestrus detection was only 0.73.This is notably smaller than the 0.45 reduction for theequivalent equally distributed sampl<strong>in</strong>g frequency(Fig. 5). Even <strong>in</strong> this conservative test of sampl<strong>in</strong>gfrequency effects there is a clear benefit of hav<strong>in</strong>g avariable sampl<strong>in</strong>g frequency.General considerationsThis paper provides a test of one particular model for<strong>in</strong>terpret<strong>in</strong>g progesterone time-series data. In this testthe model was run us<strong>in</strong>g limited <strong>in</strong>puts, for simplicitythe potential benefits of <strong>in</strong>clud<strong>in</strong>g additional non-progesterone<strong>in</strong>formation such as <strong>in</strong>put from other externaloestrus-detection methods (e.g. pedometers), bodyenergy status, and urea concentrations were not evaluated.Despite this, the model performed at least as wellas other oestrus-detection systems (Firk et al. 2002;Cavalieri et al. 2003; Van Eerdenburg 2006). In thiscontext, it would have been relevant to compare thismodel with other models for <strong>in</strong>terpret<strong>in</strong>g progesteroneprofiles. At the time of writ<strong>in</strong>g, we could f<strong>in</strong>d no otherpublished progesterone models, thus comparison islimited to specific aspects such as oestrus detectionmeasured by <strong>in</strong>dicators other than progesterone (virtuallyall tests use progesterone as the reference measure).Although the present model performed substantiallybetter <strong>in</strong> terms of sensitivity than the majority ofmethods <strong>in</strong> the literature (Firk et al. 2002; Cavalieri etal. 2003; Van Eerdenburg 2006), this is not <strong>in</strong> itself adef<strong>in</strong>itive assessment of the models quality. As demonstratedby numerous authors, sensitivity and specificitycan be altered by vary<strong>in</strong>g the thresholds used, andgenerally have an <strong>in</strong>verse relation to each other (de Molet al. 2001; Faust<strong>in</strong>i et al. 2007). In the context of timeseriesmeasurements such as progesterone profiles,sensitivity and specificity measures are particularlyunsatisfactory because they are calculated for fixedtime-po<strong>in</strong>ts or w<strong>in</strong>dows (Friggens et al. 2007). Forexample, Faust<strong>in</strong>i et al. (2007) reported a sensitivity forprogesterone determ<strong>in</strong>ation of pregnancy of 98.2%,substantially greater than the 89.3% reported <strong>in</strong> thepresent study, whereas Romano et al. (2006) reported asensitivity of 74.5%. The difference was <strong>in</strong> large partbecause of the time (days post-<strong>in</strong>sem<strong>in</strong>ation) at whichthe sensitivity was calculated. Indeed, Romano et al.(2006) reported <strong>in</strong>creas<strong>in</strong>g sensitivity with <strong>in</strong>creas<strong>in</strong>gdays post-<strong>in</strong>sem<strong>in</strong>ation. Clearly, factors such as thechosen time-po<strong>in</strong>t and sampl<strong>in</strong>g frequency make directcomparison of methods difficult.It should also be noted that <strong>in</strong> the present study,progesterone measurements were made <strong>in</strong> a laboratoryÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


120 NC Friggens, M Bjerr<strong>in</strong>g, C Ridder, S Højsgaard and T Larsenby a classical ELISA method. It may be expected thatsuch a method is more precise than the types ofmeasurement result<strong>in</strong>g from <strong>in</strong> situ biosensors. Conversely,milk samples used <strong>in</strong> the present study werecollected from cows milked robotically, result<strong>in</strong>g <strong>in</strong>variable <strong>in</strong>ter milk<strong>in</strong>g <strong>in</strong>tervals, and thus variable milkfat content (Friggens and Rasmussen 2001). This<strong>in</strong>troduces a greater variability <strong>in</strong> the milk progesteronemeasurements (Waldmann et al. 1999) than wouldbe expected from milk samples collected more conventionally.On balance, the results presented <strong>in</strong> thisstudy probably reflect what can be expected undercommercial conditions, but this rema<strong>in</strong>s to be quantified.ConclusionsThe type of model needed to condense and <strong>in</strong>terpretprogesterone profile data <strong>in</strong> real-time on-farm has beentested and found to perform substantially better <strong>in</strong> termsof sensitivity of oestrus detection than other exist<strong>in</strong>gdetection systems. The model also was shown to providevaluable <strong>in</strong>formation about other aspects of reproductivestatus such as pregnancy determ<strong>in</strong>ation and commencementof luteal activity.AcknowledgementsWe gratefully acknowledge the valuable contribution of the farm staffat KFC, Jens Clausen, Carsten Berthelsen, Peter Løvendahl, JesNielsen, and Connie Middelhede for their efforts <strong>in</strong> secur<strong>in</strong>g thismassive number of samples. This study, which was part of the Biosensproject, was funded by the Danish M<strong>in</strong>istry of Food, Agriculture andFisheries and the Danish Cattle Association.AppendixDays to next sampl<strong>in</strong>g functions for the luteal phase and dur<strong>in</strong>gpregnancyThe orig<strong>in</strong>al days to next sampl<strong>in</strong>g (DNS) function <strong>in</strong> the lutealphase (Friggens and Chagunda 2005) decreased sampl<strong>in</strong>g frequencyas duration of oestrus cycle (cyclen) <strong>in</strong>creased towards 21 but thenstayed low <strong>in</strong> prolonged luteal phases. The function was changed <strong>in</strong>two ways. The default luteal DNS function (DNS1LdefR) waschanged from a decl<strong>in</strong><strong>in</strong>g sigmoid to a decl<strong>in</strong><strong>in</strong>g sigmoid plus asubsequent climb<strong>in</strong>g sigmoid (i.e. it rises aga<strong>in</strong> after expectedcyclen). The slope modifier was adjusted to use the absolutedifference between the maximum progesterone concentration s<strong>in</strong>celast oestrus (CycMax) and current progesterone concentration ratherthan the slope:DayFromNextOest = abs((Date - DayOest) - Cyclen)MaxStep ¼ Cyclen DNSLPropDNS1Ldef ¼ MaxStep expð expðDNSLRatðDayFromNextOest DNSLlagÞÞÞSlopeMod ¼ expðexpðSModRat*((CycMax - Level)(CycMax/SModT))))DNS ¼ DNS1Ldef SlopeModwhere date is the current day, DayOest is the day of the preced<strong>in</strong>goestrus, and level is the smoothed progesterone concentration (ng ⁄ ml).DNSLProp, DNSLRat, DNSLlag, SModRat, and SModT are constantswith the follow<strong>in</strong>g values: 0.25, )0.4, 5, 0.75 and 4, respectively.The DNS function dur<strong>in</strong>g pregnancy was modified <strong>in</strong> a similar way toconcentrate sampl<strong>in</strong>g frequency around the time approximately22 days post-<strong>in</strong>sem<strong>in</strong>ation and reduce it thereafter:PregStepT = Cyclen + PregStepLagMaxStepPreg ¼ðTopPregStep BotPregStepÞ expð expðPregStepRat ððRunTime AITimeÞPregStepTÞÞÞ þ BotPregStepDNS2def ¼ MaxStepPreg expð expðDNSLRatðDayFromNextOest DNSLlagÞÞÞTimeFromAIFun ¼ expðexpðTfAIRat*((RunTimeAITime) - TfAIT)))offsetLevTime ¼ expð expðPLevRat ðLevel ðPLevTþ DNS2LevToffsetÞTimeFromAIFunRÞÞÞDNS2LevTime ¼ð1 ð1 TimeFromAIFunÞÞ offsetLevTime þð1 TimeFromAIFunÞDNSR ¼ DNS2def DNS2LevTimewhere RunTime is the current time and AITime is the time of<strong>in</strong>sem<strong>in</strong>ation. PregStepLag, TopPregStep, BotPregStep, PregStepRat,TfAIRat, TfAIT, PLevRat, PLevT and DNS2LevToffset are constantswith the follow<strong>in</strong>g values: 6, 10, 5, )0.4, )0.3, 12, )0.5, 12 and 6,respectively.ReferencesBulman DC, Lamm<strong>in</strong>g GE, 1978: Milk progesterone levels<strong>in</strong> relation to conception, repeat breed<strong>in</strong>g and factors<strong>in</strong>fluenc<strong>in</strong>g acyclicity <strong>in</strong> dairy cows. J Reprod Fertil 54,447–458.Cavalieri J, Eagles VE, Ryan M, Macmillan KL, 2003:Comparison of four methods for detection of oestrus <strong>in</strong>dairy cows with resynchronised oestrous cycles. Aust Vet J81, 422–425.de Mol RM, Keen A, Kroeze GH, Achten JMFH, 1999:Description of a detection model for oestrus and diseases <strong>in</strong>dairy cattle based on time series analysis comb<strong>in</strong>ed with aKalman filter. Comput Electron Agr 22, 171–185.de Mol RM, Ouweltjes W, Kroeze GH, Hendriks MMWB,2001: Detection of estrus and mastitis: field performance ofa model. Appl Eng Agr 17, 399–407.Delwiche MJ, Tang X, BonDurant RH, Munro CJ, 2001:Estrus detection with a progesterone biosensor. TransASAE 44, 2003–2008.Faust<strong>in</strong>i M, Battocchio M, Vigo D, Prandi A, Veronesi MC,Com<strong>in</strong> A, Cairoli F, 2007: Pregnancy diagnosis <strong>in</strong> dairycows by whey progesterone analysis: an ROC approach.Theriogenology 67, 1386–1392.Firk R, Stamer E, Junge W, Krieter J, 2002: Automation ofoestrus detection <strong>in</strong> dairy cows: a review. Livest Prod Sci 75,219–232.Firk R, Stamer E, Junge W, Krieter J, 2003: Improv<strong>in</strong>g oestrusdetection by comb<strong>in</strong>ation of activity measurements with<strong>in</strong>formation about previous oestrus cases. Livest Prod Sci82, 97–103.Friggens NC, Chagunda MGG, 2005: Prediction of thereproductive status of cattle on the basis of milk progesteronemeasures: model description. Theriogenology 64,155–190.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reproductive Status Assessed by Milk Progesterone 121Friggens NC, Chagunda MGG, Bjerr<strong>in</strong>g M, Ridder C,Højsgaard S, Larsen T, 2007: Estimat<strong>in</strong>g degree of mastitisfrom time-series measurements <strong>in</strong> milk: a test of a modelbased on lactate dehydrogenase measurements. J Dairy Sci90, 5415–5427.Friggens NC, Løvendahl P, 2008: The potential of on-farmfertility profiles: <strong>in</strong>-l<strong>in</strong>e progesterone and activitymeasurements. In: Friggens NC, Royal MD, Smith RF(eds.), Fertility <strong>in</strong> Dairy Cows: Bridg<strong>in</strong>g the Gaps, BSASOccasional Publication. British Society of Animal Science,Ed<strong>in</strong>burgh, UK (<strong>in</strong> press).Friggens NC, Rasmussen MD, 2001: Milk quality assessment<strong>in</strong> automatic milk<strong>in</strong>g systems: account<strong>in</strong>g for the effects ofvariable <strong>in</strong>tervals between milk<strong>in</strong>gs on milk composition.Livest Prod Sci 73, 45–54.Lamm<strong>in</strong>g GE, Bulman DC, 1976: Use of milk progesteroneradioimmunoassay <strong>in</strong> diagnosis and treatment of subfertility<strong>in</strong> dairy cows. Br Vet J 132, 507–517.Lamm<strong>in</strong>g GE, Darwash AO, 1998: The use of milk progesteroneprofiles to characterise components of subfertility <strong>in</strong>milked dairy cows. Anim Reprod Sci 52, 175–190.Lattec, 2007: http://www.herdnavigator.com/pages/id33.asp.Mahalanobis PC, 1936: On the generalised distance <strong>in</strong> statistics.Proc Natl Inst Sci India 12, 49–55.Opsomer G, Coryn M, Deluyker H, De Kruif A, 1998: Ananalysis of ovarian dysfunction <strong>in</strong> high yield<strong>in</strong>g dairy cowsafter calv<strong>in</strong>g based on progesterone profiles. ReprodDomest Anim 33, 193–204.Peters AR, Ball PJH, 1995: <strong>Reproduction</strong> <strong>in</strong> Cattle, 2nd edn.Blackwell Science, Oxford, UK.Prandi A, Mess<strong>in</strong>a M, Tondoio A, Motta M, 1999:Correlation between reproductive efficiency, as determ<strong>in</strong>edby new mathematical <strong>in</strong>dexes, and the body condition score<strong>in</strong> dairy cows. Theriogenology 52, 1251–1265.Romano JE, Thompson JA, Forrest DW, Westhus<strong>in</strong> ME,Tomaszweski MA, Kraemer DC, 2006: Early pregnancydiagnosis by transrectal ultrasonography <strong>in</strong> dairy cattle.Theriogenology 66, 1034–1041.Ross<strong>in</strong>g W, Spahr SL, 1992: Automation <strong>in</strong> mach<strong>in</strong>e milk<strong>in</strong>g.In: Bramley AJ, Dodd FH, Me<strong>in</strong> GA (eds.), Mach<strong>in</strong>eMilk<strong>in</strong>g and Lactation. Queen City Pr<strong>in</strong>ters Inc., Burl<strong>in</strong>gton,pp. 311–342.Royal MD, Darwash AO, Fl<strong>in</strong>t APF, Webb R, Woolliams JA,Lamm<strong>in</strong>g GE, 2000: Decl<strong>in</strong><strong>in</strong>g fertility <strong>in</strong> dairy cattle:changes <strong>in</strong> traditional and endocr<strong>in</strong>e parameters of fertility.Anim Sci 70, 487–501.Royal MD, Pryce JE, Woolliams JA, Fl<strong>in</strong>t APF, 2002: Thegenetic relationship between commencement of luteal activityand calv<strong>in</strong>g <strong>in</strong>terval, body condition score, production,and l<strong>in</strong>ear type traits <strong>in</strong> Holste<strong>in</strong>-Friesian dairy cattle. JDairy Sci 85, 3071–3080.Simersky R, Swaczynova J, Morris DA, Franek M, Strnad M,2007: Development of an ELISA-based kit for the on-farmdeterm<strong>in</strong>ation of progesterone <strong>in</strong> milk. Vet Med 52, 19–28.Sreenan JM, Disk<strong>in</strong> MG, Morris DG, 2001: Embryo survivalrate <strong>in</strong> cattle: a major limitation to the achievement of highfertility. In: Disk<strong>in</strong> MG (ed), British Society of AnimalScience Occasional Publication, 26 (1). British Society ofAnimal Science, Ed<strong>in</strong>burgh, pp. 93–104.Van Eerdenburg FJCM, 2006: Estrus detection <strong>in</strong> dairy cattle:how to beat the bull. Vlaams Diergen Tijds 75, 61–69.Waldmann A, 1993: Enzyme immunoassay (EIA) for milkprogesterone us<strong>in</strong>g a monoclonal antibody. Anim ReprodSci 34, 19–30.Waldmann A, Ropstad E, Landsverk K, Sørensen K, SølverødL, Dahl E, 1999: Level and distribution of progesterone <strong>in</strong>bov<strong>in</strong>e milk <strong>in</strong> relation to storage <strong>in</strong> the mammary gland.Anim Reprod Sci 56, 79–91.Author’s address (for correspondence): NC Friggens, Faculty ofAgricultural Sciences, University of Aarhus, Research Centre Foulum,PO Box 50, 8830 Tjele, Denmark. E-mail: n.friggens@agrsci.dkConflict of <strong>in</strong>terest: NC Friggens has received a research grant for thiswork, part-funded by Lattec I ⁄ S; C Ridder is employed by Lattec I ⁄ S;all rema<strong>in</strong><strong>in</strong>g authors declare no conflict of <strong>in</strong>terests.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Supp. 2), 122–128 (2008); doi: 10.1111/j.1439-0531.2008.01151.xISSN 0936-6768Genetic Aspects of <strong>Reproduction</strong> <strong>in</strong> SheepDR NotterDepartment of Animal and Poultry Sciences, Virg<strong>in</strong>ia Polytechnic Institute and State University, Blacksburg, USAContentsMa<strong>in</strong>tenance of high levels of realized fertility (def<strong>in</strong>ed as thepercentage of ewes that lamb) and appropriate levels offecundity are critical for efficient sheep production. Theoptimal level of fecundity <strong>in</strong> most situations is well below themaximum atta<strong>in</strong>able level and can be targeted by comb<strong>in</strong><strong>in</strong>gselection among and with<strong>in</strong> breeds with use of an expand<strong>in</strong>garray of s<strong>in</strong>gle-gene mutations affect<strong>in</strong>g ovulation rate andlitter size. The heritability of litter size is approximately 0.10,allow<strong>in</strong>g changes of up to 2% ⁄ year from simple mass selection.Mutations <strong>in</strong> several genes associated with the transform<strong>in</strong>ggrowth factor b superfamily (BMPRIB, GDF9 and sex-l<strong>in</strong>kedBMP15) can <strong>in</strong>crease ovulation rates by 0.7–1.5 ova <strong>in</strong>heterozygous ewes. However, ewes that are homozygous forBMP15 or GDF9 mutations are sterile, so use of thesemutations requires carefully structured breed<strong>in</strong>g programmes.Improvements <strong>in</strong> fertility may be critical for autumn lamb<strong>in</strong>gor programmes that aspire to lamb throughout the year.Selection to improve fertility <strong>in</strong> spr<strong>in</strong>g mat<strong>in</strong>gs has beensuccessful; selected adult ewes have lamb<strong>in</strong>g rates of 80–85%<strong>in</strong> October and early November. The selected ewes have adramatically reduced seasonal anestrus, and many ewescont<strong>in</strong>ue to cycle dur<strong>in</strong>g spr<strong>in</strong>g and summer. Major genesaffect<strong>in</strong>g seasonal breed<strong>in</strong>g have not been identified <strong>in</strong> sheep.Polymorphisms <strong>in</strong> the melaton<strong>in</strong> receptor 1a gene appear to beassociated with seasonal breed<strong>in</strong>g <strong>in</strong> some, but not all breeds.However, functional genomic studies of genes associated withcircadian and circannual rhythms have potential to revealadditional candidate genes <strong>in</strong>volved <strong>in</strong> seasonal breed<strong>in</strong>g.IntroductionImplementation of effective programmes of reproductivemanagement <strong>in</strong> commercial sheep production<strong>in</strong>volves synchronization of genetic potentials forreproductive ability with the production environment.The production objective <strong>in</strong> such situations is generallymaximization of farm profit. If meat is the ma<strong>in</strong> output,atta<strong>in</strong>ment of this objective commonly <strong>in</strong>volvesmaximiz<strong>in</strong>g the annual number and ⁄ or total weight oflambs marketed. Wool may make an additionalcontribution to <strong>in</strong>come, but unless wool quality is veryhigh, <strong>in</strong>creases <strong>in</strong> wool production cannot compensatefor even m<strong>in</strong>or losses <strong>in</strong> meat production. However,reproductive goals <strong>in</strong> sheep dairy<strong>in</strong>g, and particularlythe economic benefit from <strong>in</strong>creas<strong>in</strong>g litter size, may besecondary to the primary goal of <strong>in</strong>creas<strong>in</strong>g milkproduction.Opportunities to <strong>in</strong>crease ovulation rates <strong>in</strong> sheep arevast, <strong>in</strong>volv<strong>in</strong>g both well-established polygenic breeddifferences (Fahmy 1996) and access to a number ofs<strong>in</strong>gle-gene mutations with major effects on ovulationrate and litter size. Mutant alleles may be <strong>in</strong>trogressed<strong>in</strong>to different genetic backgrounds with relative ease,allow<strong>in</strong>g large <strong>in</strong>creases <strong>in</strong> ovulation rate without othermajor changes <strong>in</strong> genetic background or environmentaladaptation. Genetic management of ovulation rate andlitter size thus <strong>in</strong>volves def<strong>in</strong>ition of the optimum littersize (which is commonly well below the potentiallyatta<strong>in</strong>able maximum) and the development of breed<strong>in</strong>gschemes to establish and ma<strong>in</strong>ta<strong>in</strong> the desired level offecundity.Seasonal breed<strong>in</strong>g limits both diversification and<strong>in</strong>tensification of production. Under extensiveconditions, the breed<strong>in</strong>g season of established, locallyadapted breeds generally has evolved to compliment theenvironmental conditions and desired production calendarand serves to ensure that lamb<strong>in</strong>g occurs at appropriatetimes. However, <strong>in</strong>tensification of animalproduction to meet <strong>in</strong>creased consumer demands formeat and other animal products, both locally and <strong>in</strong>expand<strong>in</strong>g global markets, provides both motivation andopportunity to modify the traditional lamb<strong>in</strong>g schedules.In some cases, these modifications require only a shift <strong>in</strong>the breed<strong>in</strong>g season, but there is also an opportunity to<strong>in</strong>crease the frequency of lamb<strong>in</strong>g, with 7- to 9-monthlamb<strong>in</strong>g <strong>in</strong>tervals as an apparent practical m<strong>in</strong>imum. Amassive literature exists catalogu<strong>in</strong>g attempts to useexogenous hormone treatments or photoperiodic manipulationto <strong>in</strong>duce fertile mat<strong>in</strong>gs outside the normalbreed<strong>in</strong>g season. Correspond<strong>in</strong>g <strong>in</strong>formation now existsto show that useful levels of additive genetic variation <strong>in</strong>duration of the breed<strong>in</strong>g season are present among andwith<strong>in</strong> breeds, and the development of sheep with adramatically reduced breed<strong>in</strong>g season will be discussed.The understand<strong>in</strong>g of the genetic control of circadianand circannual timekeep<strong>in</strong>g is likewise expand<strong>in</strong>g rapidly,but access to quantitative trait loci (QTL) andfunctional mutations associated with seasonal breed<strong>in</strong>g<strong>in</strong> sheep rema<strong>in</strong>s limited.This review will thus focus on genetic mechanismscontroll<strong>in</strong>g ovulation rate and seasonal breed<strong>in</strong>g <strong>in</strong>sheep, and on strategies to synchronize geneticpotentials for reproductive traits with the productionenvironment.Genetic Control of Ovulation Rate and LitterSizePolygenic associationsA summary of heritability estimates for several reproductivetraits is shown <strong>in</strong> Table 1 (Safari et al. 2005).Estimates of heritability for litter size are generallyconsistent <strong>in</strong> the literature, averag<strong>in</strong>g 0.11 (n = 102). Asdiscussed by Bradford (1985), these parameters wouldsuggest a maximum response to s<strong>in</strong>gle-trait mass selectionfor litter size of approximately 2% per year. Thus, agenetic ga<strong>in</strong> of 0.20 lambs born per ewe lamb<strong>in</strong>g froms<strong>in</strong>gle-trait mass selection would require approximatelyÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Genetic Aspects of <strong>Reproduction</strong> <strong>in</strong> Sheep 12310 years (or three sheep generations). These heritabilityestimates would generally be considered low, but evaluationof the potential for genetic change must alsoconsider the variation present <strong>in</strong> the population, quantifiedby the phenotypic coefficient of variation (CV) ofapproximately 35% for litter size. The predictedresponse to mass selection is proportional to the productof (1) the selection <strong>in</strong>tensity (i), (2) the heritability of thetrait (h 2 ), (3) the phenotypic SD and (4) the <strong>in</strong>verse ofthe generation <strong>in</strong>terval (t). For fixed i and t, the responseto mass selection as a percentage of mean performanceis thus directly proportional to the product (h 2 · CV).A comparison of values for h 2 and CV for litter size <strong>in</strong>Table 1 to values for other traits shows that the modestselection responses predicted for traits such as litter sizedo not arise directly from a shortage of heritablevariation. They <strong>in</strong>stead reflect delays <strong>in</strong> age at measurementwhich limit annual selection response by extend<strong>in</strong>gthe generation <strong>in</strong>terval or, alternatively, necessitate thatselection be based on the litter size of the dam (therebyreduc<strong>in</strong>g accuracy of the evaluation). If we compare theanticipated selection response <strong>in</strong> litter size with that forpost-wean<strong>in</strong>g body weight, we f<strong>in</strong>d that the heritabilityof body weight is much higher (0.28 vs 0.11), but thatthe phenotypic CV is considerably lower (12.5 vs 35%;Safari et al. 2005) lead<strong>in</strong>g to a slightly higher value forh 2 · CV for litter size (3.85) than for body weight (3.50).A particular powerful demonstration of the powerof selection to change litter size comes from anexperiment to study the response to selection fortw<strong>in</strong>n<strong>in</strong>g <strong>in</strong> cattle (Echternkamp et al. 2002). Thisexperiment was <strong>in</strong>itiated <strong>in</strong> 1982 with an <strong>in</strong>itialTable 1. Estimates of heritability (h 2 ), phenotypic coefficient ofvariation (CV), and relative selection response as a percentage of themean (h 2 .CV) for reproductive traits <strong>in</strong> sheep a Phenotypich 2 CV (%) h 2 CV bEwe fertility c 0.07 49 3.4Ovulation rate 0.19 28 5.3Embryo survival 0.01 27 0.0Litter size 0.11 35 3.9Lamb survival 0.04 ⁄ 0.05 47 1.9Ram scrotal circumference 0.23 10 2.3a Safari et al. (2005).b See text for <strong>in</strong>terpretation of h 2 . CV values.c Percentage of ewes that lamb.<strong>in</strong>cidence of tw<strong>in</strong> births of less than 3%. By 1998,the <strong>in</strong>cidence of tw<strong>in</strong>n<strong>in</strong>g had <strong>in</strong>creased to over 48%.Factors that contributed to success <strong>in</strong> this project<strong>in</strong>cluded <strong>in</strong>itial <strong>in</strong>tensive screen<strong>in</strong>g of the US cattlepopulation to identify cows with a history of tw<strong>in</strong>births, use of repeated laparoscopic measures ofovulation rate at consecutive cycles to <strong>in</strong>crease theaccuracy of assessment without correspond<strong>in</strong>glylengthen<strong>in</strong>g generation <strong>in</strong>terval, and use of progenytest<strong>in</strong>g and artificial <strong>in</strong>sem<strong>in</strong>ation to maximize use ofsuperior sires. These genetic changes were achievedthrough the traditional quantitative strategies, but thepopulation has now also been used to detect QTLsthat affect tw<strong>in</strong>n<strong>in</strong>g <strong>in</strong> cattle. However, the currentexperiment has reached a position of dim<strong>in</strong>ish<strong>in</strong>greturns because of <strong>in</strong>creases <strong>in</strong> triplet pregnancies. Incontrast to the situation <strong>in</strong> sheep, the presence ofmore than one foetus <strong>in</strong> a s<strong>in</strong>gle uter<strong>in</strong>e hornnormally results <strong>in</strong> term<strong>in</strong>ation of pregnancy <strong>in</strong> cattle.Mutations affect<strong>in</strong>g ovulation rate and litter sizeTable 2, derived from the review by Davis (2005), listsmutations that result <strong>in</strong> <strong>in</strong>creased ovulation rates. Thesemutations all <strong>in</strong>volve gene products and receptorsassociated with the transform<strong>in</strong>g growth factor bsuperfamily (McNatty et al. 2005). Thus, the FecB Bmutation first reported <strong>in</strong> Booroola Mer<strong>in</strong>o is nowknown to result from a po<strong>in</strong>t mutation <strong>in</strong> the bonemorphogenic prote<strong>in</strong> receptor IB (BMPRIB) gene(Wilson et al. 2001), which is also sometimes referredto as activ<strong>in</strong>-like k<strong>in</strong>ase 6 (ALK6). This mutation iswidespread <strong>in</strong> prolific Asian sheep breeds <strong>in</strong>clud<strong>in</strong>g theIndian Garole, Javanese Th<strong>in</strong>-tail, and Ch<strong>in</strong>ese Hu andHan as well as the Booroola Mer<strong>in</strong>o, but appears to beabsent <strong>in</strong> prolific European breeds (Davis et al. 2002,2006). The Garole has long been postulated to be thesource of the FecB B mutation <strong>in</strong> the Booroola Mer<strong>in</strong>o(Turner 1982), but the presence of mutation <strong>in</strong> Ch<strong>in</strong>eseHu and Han sheep suggests a central Asian orig<strong>in</strong>. TheHu and Han are relatively closely related and derivedfrom Mongolian sheep breeds (Lu et al. 2005). Lu et al.(2005) likewise cite historical studies by Xie (1985)assert<strong>in</strong>g that establishment of the Hu breed predatedemergence of the Han breed.Mutations <strong>in</strong> the X-l<strong>in</strong>ked bone morphogenic prote<strong>in</strong>15 (BMP15) gene were first reported <strong>in</strong> New ZealandRomney, but mutations <strong>in</strong> this gene also exist <strong>in</strong> theTable 2. S<strong>in</strong>gle-gene mutationsaffect<strong>in</strong>g ovulation rate (OR) <strong>in</strong>sheep a Gene a Breed(s) Name ChromosomeOR effect1 copy 2 copiesBMPRIB Mer<strong>in</strong>o, Javenese Th<strong>in</strong>-tail,Booroola (FecB B ) 6 +1.5 3.0Garole, Hu, HanBMP15 Romney Inverdale ⁄ Hanna (FecX I ⁄ FecX H ) b X +1.0 – cBelclaire Belclaire (FecX B ) X +1.0 – cBelclaire, Cambridge Galway (FecX G ) X +0.7 – cLaucaune Laucaune (FecX L ) X +1.5 – cGDF9 Belclaire, Cambridge High fertility (FecG H ) 5 +1.4 – ca Derived from Davis (2005) and Bod<strong>in</strong> et al. (2007).b FecX I and FecX H represent two separate mutations with similar effects.c Ewes that are homozygous for these mutations are sterile.Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


124 DR NotterBelclaire and Cambridge breeds <strong>in</strong> Ireland and the UnitedK<strong>in</strong>gdom (Davis 2005) and <strong>in</strong> the French Laucaune(Bod<strong>in</strong> et al. 2007). Thus, five separate mutations <strong>in</strong>BMP15 are associated with <strong>in</strong>creases <strong>in</strong> ovulation rate.The unique characteristic of BMP15 mutations is that an<strong>in</strong>crease <strong>in</strong> ovulation rate relative to the wild type isobserved only <strong>in</strong> <strong>in</strong>dividuals that are heterozygous for themutation. Homozygous <strong>in</strong>dividuals have abnormal ovariandevelopment and rudimentary ‘streak’ ovaries, andare sterile. A po<strong>in</strong>t mutation <strong>in</strong> the growth differentiationfactor 9 (GDF9) gene on chromosome 5 <strong>in</strong> Belclaire andCambridge sheep produces phenotypic effects that aresimilar to those of the X-l<strong>in</strong>ked BMP15 mutations, withan effect on ovulation rate of approximately +1.4 ova <strong>in</strong>heterozygotes, but abnormal ovarian development andsterility <strong>in</strong> homozygotes.Programmes to use s<strong>in</strong>gle-gene mutations to <strong>in</strong>creasefecundity usually must be carefully structured to avoidproduction of homozygous females. Under pastoralconditions, the relatively high <strong>in</strong>cidence of quadrupletand higher births generally limits the desirability ofhomozygous FecB B ⁄ FecB B <strong>in</strong>dividuals. However, mat<strong>in</strong>gsof homozygous sires to ewes of breeds that do notcarry the mutation allows production of heterozygousdaughters. The average effect of such a substitution is to<strong>in</strong>crease the realized litter size by approximately 1.0 lambs ⁄litter, which may be useful <strong>in</strong> breeds that producepredom<strong>in</strong>antly s<strong>in</strong>gle lambs, provided the environmentand management system can be adjusted to accommodatetw<strong>in</strong> and occasional triplet births. The occurrence ofFecB B <strong>in</strong> phenotypically diverse breeds also providesopportunity to source FecB B genes for <strong>in</strong>trogression frombreeds with similar levels of adaptation to the targetbreed. Utilization of BMP15 or GDF9 mutations requiresuse of DNA tests to identify males that are homozygousfor the mutation and can therefore be used to reliablyproduce heterozygous daughters. Ma<strong>in</strong>tenance of a flockthat is homozygous for these mutations is precluded bythe sterility of the homozygous females.Screen<strong>in</strong>g of ewes to identify exceptional <strong>in</strong>dividualspermits identification of animals that may carry mutantalleles with<strong>in</strong> exist<strong>in</strong>g, adapted populations. However, itis likewise essential that the production system beconfigured <strong>in</strong> such a way that allows producers tobenefit from <strong>in</strong>creases <strong>in</strong> litter size, and identification ofan appropriate target litter size distribution is a necessaryfirst step <strong>in</strong> develop<strong>in</strong>g more prolific l<strong>in</strong>es. This taskis not as easy as it sounds, because effects of ewe age, theewe age distribution, effects of lamb<strong>in</strong>g season, and, <strong>in</strong>the case of s<strong>in</strong>gle-gene mutants, genetic backgroundmust all be considered. In many pastoral environments,tw<strong>in</strong> births are considered ideal, and there is oftenconsiderable discrim<strong>in</strong>ation aga<strong>in</strong>st triplet births. Borget al. (2007) demonstrated that atta<strong>in</strong>ment of more than60–65% tw<strong>in</strong> births on a whole-flock basis generallyrequires a correspond<strong>in</strong>g significant <strong>in</strong>crease <strong>in</strong> thefrequency of triplets.Genetic Control of Seasonal Breed<strong>in</strong>gPolygenic controlHeritability estimates for realized fertility (i.e. thepercentage of ewes that lamb) are very low (Table 1),almost always below 0.10 and often below 0.05. Thisresult is not particularly surpris<strong>in</strong>g. Natural selection forfertility is necessarily <strong>in</strong>tense, and lowly fertile ewes leavefew progeny. In well-adapted populations ma<strong>in</strong>ta<strong>in</strong>edunder long-established breed<strong>in</strong>g conditions, the percentageof ewes that lamb is commonly high to very high(e.g. 85–95%) and potential for genetic improvement <strong>in</strong>fertility is limited. However, this will not necessarily bethe case when animals are moved to different productionconditions or different lamb<strong>in</strong>g seasons.The potential for genetic improvement <strong>in</strong> fertility isconstra<strong>in</strong>ed by the current level of fertility. As discussedearlier, the potential rate of genetic change is proportionalto the CV of the trait. If fertility is expressed as ab<strong>in</strong>ary trait separat<strong>in</strong>g ewes that lamb from those thatdo not, the associated phenotypic variance is given asp(1 – p), where p is the proportion of ewes that lamb.For values of p between 0 and 1, p(1 – p) is maximizedat p = 0.5, and the anticipated selection responsedecl<strong>in</strong>es as p <strong>in</strong>creases towards the maximum valuep = 1.0. The phenotypic CV of 49% for fertility <strong>in</strong>Table 1 corresponds to a value of p = 0.87, and wouldbe expected to double at p = 0.5. Thus, the selection toimprove the fertility will become progressively lesseffective as mean fertility <strong>in</strong>creases with little potentialfor further improvement <strong>in</strong> highly fertile breeds.Conversely, opportunities for genetic improvement <strong>in</strong>fertility may be anticipated when animals are matedoutside their normal breed<strong>in</strong>g season or managed <strong>in</strong>various systems of accelerated lamb<strong>in</strong>g <strong>in</strong> order to allowmore than one lamb<strong>in</strong>g <strong>in</strong> a 12-month period (Notter2002). There have been several attempts to develop l<strong>in</strong>esof sheep with reduced seasonality of breed<strong>in</strong>g, but mostwere term<strong>in</strong>ated before yield<strong>in</strong>g measurable changes <strong>in</strong>duration or tim<strong>in</strong>g of the seasonal anestrus. However, apopulation developed us<strong>in</strong>g classical selection proceduresat Virg<strong>in</strong>ia Tech (Al-Shorepy and Notter 1997;Notter et al. 1998; Notter and Cockett 2005) experienceda dramatic reduction <strong>in</strong> duration of the seasonalanestrus. This flock was derived from crosses among theDorset (50%), Rambouillet (25%) and F<strong>in</strong>nish Landrace(25%) breeds, all of which have some capacity tomate and conceive <strong>in</strong> spr<strong>in</strong>g and summer. S<strong>in</strong>gle-traitselection was imposed from 1988 to 1998 and was basedon the ability of the ewes to mate and conceive <strong>in</strong>natural, s<strong>in</strong>gle-sire mat<strong>in</strong>gs over an 8-week periodbeg<strong>in</strong>n<strong>in</strong>g May 1.In phase 1 of the study (1988–1993; Al-Shorepy andNotter 1997), ewes were exposed to vasectomized ramsfor 2 weeks before the start of mat<strong>in</strong>g to attempt tostimulate ovulation, and selection of replacement ewesand rams was based on the mean fertility of the dam.The average fertility of adult ewes <strong>in</strong> the base populationwas approximately 60%, which would nearly maximizethe phenotypic SD and, therefore, anticipated selectionresponse, but the estimated heritability of spr<strong>in</strong>g fertilitywas only 0.08, <strong>in</strong>dicat<strong>in</strong>g that the heritability of ewefertility <strong>in</strong> spr<strong>in</strong>g mat<strong>in</strong>gs was not higher than theaverage estimate of 0.07 reported by Safari et al. (2005).The estimated cumulative and annual selectionresponses <strong>in</strong> fertility <strong>in</strong> phase 1 were 6.5% and1.4% ⁄ year, respectively. Ewe age <strong>in</strong>fluenced both meanfertility and selection response. Fertility of 7-month-oldÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Genetic Aspects of <strong>Reproduction</strong> <strong>in</strong> Sheep 125autumn-born ewe lambs placed with rams <strong>in</strong> spr<strong>in</strong>gaveraged only 10% (vs approximately 60% for unselectedadult ewes) and did not respond to selection.In phase 2 of the study (1994–1998; Notter et al.1998), ewes were no longer exposed to vasectomizedrams before breed<strong>in</strong>g. The distribution of lamb<strong>in</strong>gdur<strong>in</strong>g phase 2 was characterized by an immediate startat 140–145 days after the start of breed<strong>in</strong>g, and mostewes (other than ewe lambs) that lambed did so with<strong>in</strong>3–4 weeks after the start of lamb<strong>in</strong>g. This pattern<strong>in</strong>dicated that ewes were cycl<strong>in</strong>g when rams were<strong>in</strong>troduced on May 1 and were not <strong>in</strong>duced to cycle bythe ram effect (Mart<strong>in</strong> et al. 1986). A system of breed<strong>in</strong>gvalue prediction was also implemented to maximizeaccuracy of selection. Trends <strong>in</strong> estimated breed<strong>in</strong>gvalues (EBV) <strong>in</strong> selected and control l<strong>in</strong>es are shown <strong>in</strong>Fig. 1, with a f<strong>in</strong>al difference <strong>in</strong> spr<strong>in</strong>g fertility <strong>in</strong> adultewes of 17% (87 vs 70%). Fertility of ewe lambs,however, rema<strong>in</strong>ed unresponsive to selection and wasstill below 15% <strong>in</strong> selected ewes at the end of the study.The selection experiment ended <strong>in</strong> 1998, with subsequentstudies devoted to characteriz<strong>in</strong>g the selectedanimals. An area of concern was to determ<strong>in</strong>e whetherthe selection had truly lengthened the breed<strong>in</strong>g season orhad simply shifted the period of anestrus to avoid thespr<strong>in</strong>g breed<strong>in</strong>g season. To that end, V<strong>in</strong>cent et al.(2000) monitored mat<strong>in</strong>g behaviour of high and low BVewes that were cont<strong>in</strong>uously ma<strong>in</strong>ta<strong>in</strong>ed with the samevasectomized rams from mid-January through late Julyof 1992, 1993 and 1995. Ewes were checked twice weeklyfor mat<strong>in</strong>g marks. They were identified as anestrouswhenever consecutive estruses were separated by morethan 21 days, and the numbers of days of anestrus weresummed over the period. High BV ewes (average EBVfor fertility of 12.6%) were anestrous for an average ofonly 28 days whereas ewes with average BV (mean EBVof 0.3%) were anestrous for an average of 70 days(p < 0.001). Each 1-day <strong>in</strong>crease <strong>in</strong> EBV was associatedwith a decrease <strong>in</strong> days of anestrus of 2.15 ± 0.72 days(p < 0.01). Results of this study confirm that selectionfor ability to lamb <strong>in</strong> autumn resulted <strong>in</strong> a reduction <strong>in</strong>the duration of the seasonal anestrus.A second study (J. M. Smith and D. R. Notter,unpublished) evaluated 68 selected ewes ma<strong>in</strong>ta<strong>in</strong>ed<strong>in</strong>doors with vasectomized rams from mid-January untilearly July. One half of the ewes were placed underconditions that mimicked ambient photoperiods andFall fertility EBV (%)20151050–5Selection l<strong>in</strong>eControl l<strong>in</strong>e1988 1990 1992 1994 1996 1998Year of birthFig. 1. Changes <strong>in</strong> estimated breed<strong>in</strong>g values (EBV) for fall fertility,def<strong>in</strong>ed as the percentage of ewes that lamb <strong>in</strong> fall, over time <strong>in</strong> selectedand control l<strong>in</strong>esone half were ma<strong>in</strong>ta<strong>in</strong>ed under constant 16-h days.Each group was ma<strong>in</strong>ta<strong>in</strong>ed with three vasectomizedrams and the same rams rema<strong>in</strong>ed with each groupthroughout the study. The number of days of anestruswas not affected by the photoperiod; results for 4-yearoldand older ewes were similar to those of V<strong>in</strong>cent et al.(2000) with an average of 33 days of anestrus, but theaverage duration of anestrus was longer for 3-year-old(56 days) and 2-year-old ewes (71 days). At the end ofthe study on July 6, 10 elite ewes that had missed at mostone cycle were identified. These ewes came from bothlight treatments and were comb<strong>in</strong>ed, placed with fourvasectomized rams (two from each of the orig<strong>in</strong>al lighttreatments), and ma<strong>in</strong>ta<strong>in</strong>ed under 16-h days for anadditional 10 weeks (until approximately the fall equ<strong>in</strong>ox)to <strong>in</strong>vestigate their ability to cont<strong>in</strong>ue cycl<strong>in</strong>g undersusta<strong>in</strong>ed long days. Blood samples were collected twiceweekly and assayed for circulat<strong>in</strong>g progesterone toconfirm ovarian status. All ewes were found to becycl<strong>in</strong>g at the start of this period, but all but one of theewes subsequently missed at least one cycle whenma<strong>in</strong>ta<strong>in</strong>ed under cont<strong>in</strong>uous long days and the relativelyhigh temperatures of mid-August <strong>in</strong> Blacksburg,Virg<strong>in</strong>ia. However, all but two of the 10 ewes re<strong>in</strong>itiatedcycles by September 7, when ewes were removed fromthe study and jo<strong>in</strong>ed with <strong>in</strong>tact rams outdoors forbreed<strong>in</strong>g. These results suggest that even these elite ewesmay, under extended long days, have experienced aremnant anestrous period of 1–2 cycles <strong>in</strong> late summer.A study to assess late-summer ovarian activity <strong>in</strong> greaterdetail is currently underway.Selection to improve fertility <strong>in</strong> accelerated lamb<strong>in</strong>gsystems is particularly challeng<strong>in</strong>g because of thecomplexity of the systems (Notter 2002). The varioussystems used <strong>in</strong> practice often <strong>in</strong>volve 3–5 mat<strong>in</strong>gseasons per year. Open ewes are commonly jo<strong>in</strong>edwith rams at consecutive mat<strong>in</strong>g seasons until conceptionoccurs, and ewes re-enter the system shortlyafter wean<strong>in</strong>g of their lambs. Ewes <strong>in</strong> these systemstend to become stratified, with high- and low-fertilityewes mov<strong>in</strong>g through the system <strong>in</strong> separate groups,but with periodic <strong>in</strong>fusions of ewes with newly weanedlambs. Differences <strong>in</strong> fertility among seasons are oftenlarge (Lewis et al. 1996) and must be properlyconsidered <strong>in</strong> statistical models to ensure separationof genetic and seasonal environmental effects. Thechoice of the trait(s) that best characterizes geneticdifferences <strong>in</strong> performance <strong>in</strong> accelerated systems islikewise not clear.Lewis et al. (1996) and Banos et al. (2002) studiedgenetic control of reproductive performance <strong>in</strong> an<strong>in</strong>tensive accelerated-lamb<strong>in</strong>g system developed at CornellUniversity and <strong>in</strong>volv<strong>in</strong>g five breed<strong>in</strong>g seasons peryear. Over 6 years, fertility varied from 15% <strong>in</strong> June to69% <strong>in</strong> October. Heritability estimates for the numberof exposures (mat<strong>in</strong>g seasons) per conception were0.09 ± 0.06 at first lamb<strong>in</strong>g, 0.15 ± 0.09 at secondlamb<strong>in</strong>g and 0.05 ± 0.06 at the third and subsequentlamb<strong>in</strong>gs. Heritability estimates for age at first lamb<strong>in</strong>gand first lamb<strong>in</strong>g <strong>in</strong>terval were 0.09 ± 0.06 and0.25 ± 0.11, respectively, but the heritability estimatefor the second and subsequent lamb<strong>in</strong>g <strong>in</strong>tervals wasonly 0.03 ± 0.04. These results suggest some potentialÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


126 DR Notterto use early performance, and particularly the firstlamb<strong>in</strong>g <strong>in</strong>terval, as a selection criterion, but recordsoccurr<strong>in</strong>g after the second lamb<strong>in</strong>g contributed littleadditional <strong>in</strong>formation.In a related study, Vanimisetti (2006) estimatedgenetic parameters for realized fertility <strong>in</strong> a commercialPolypay flock with three annual lamb<strong>in</strong>g seasons.Heritability estimates were near zero for all lamb<strong>in</strong>g<strong>in</strong>tervals, but significant additive genetic effects wereobserved for ewe lamb fertility at first mat<strong>in</strong>g and forage at the first, second and third lamb<strong>in</strong>g, with heritabilityestimates of 0.14, 0.39, 0.28 and 0.36, respectively(all p < 0.01). These measures were strongly <strong>in</strong>tercorrelated,with absolute values of genetic correlationsabove 0.85, suggest<strong>in</strong>g that the ability of the ewes tomate and conceive at the first opportunity and thereforelamb at young ages was the best <strong>in</strong>dicator of geneticmerit and could be relatively easily evaluated bymonitor<strong>in</strong>g ewe ages at lamb<strong>in</strong>g.Possible major genes affect<strong>in</strong>g seasonalityLoss-of-function mutations comparable to those thatdisrupt regulation of ovulation rate have not yet beenidentified for seasonal breed<strong>in</strong>g. The most promis<strong>in</strong>ggenetic marker has been a restriction fragment lengthpolymorphism (RFLP) polymorphism <strong>in</strong> the gene thatencodes melaton<strong>in</strong> receptor 1a (MTNR1A). This, andother, polymorphisms <strong>in</strong> MTNR1A were described byBarrett et al. (1997), Messer et al. (1997) and Pelletieret al. (2000), and their frequencies and effects werereviewed by Notter and Cockett (2005). Briefly,significant associations between seasonal breed<strong>in</strong>gperformance and MTNR1A genotype were reported<strong>in</strong> both the moderately seasonal Mer<strong>in</strong>o d’Arles breed(Pelletier et al. 2000) and the Virg<strong>in</strong>ia Tech out-ofseason-breed<strong>in</strong>gl<strong>in</strong>e (Notter et al. 2003). In theVirg<strong>in</strong>ia Tech population, adult ewes with at leastone copy of the favourable RFLP allele had11.2 ± 5.1% higher spr<strong>in</strong>g fertility than ewes thatwere homozygous for the alternative allele (p = 0.03),and this marker accounted for 23.8% of the additivevariance <strong>in</strong> spr<strong>in</strong>g fertility.A review of allelic frequencies <strong>in</strong> various breeds foundthat the putative favourable MTNR1A allele was oftenpresent at relatively high frequencies <strong>in</strong> breedscommonly considered to be quite seasonal (Notter andCockett 2005). Hernandez et al. (2005) likewise reportedthat MTNR1A genotype had no association with lengthof the breed<strong>in</strong>g season <strong>in</strong> the relatively seasonal Ile-de-France breed. These results suggest that effects of thepolymorphism may depend on genetic background andare not necessarily detectable <strong>in</strong> highly seasonal breeds,thereby limit<strong>in</strong>g its usefulness to that of a breed-specificgenetic marker.Knowledge of functional genomic control of circadianand circannual rhythms is expand<strong>in</strong>g <strong>in</strong> both laboratoryspecies and sheep. Several clock genes have beencatalogued and shown to both express circadianrhythms <strong>in</strong> gene expression and be <strong>in</strong>ducible by changes<strong>in</strong> photoperiod. Studies of clock gene expression <strong>in</strong> thesuprachiasmatic nucleus and pars tuberalis (PT) ofsheep have shown that expression of the cryptochrome 1gene (cry1) is transiently stimulated by exposure toelevated melaton<strong>in</strong> levels at the onset of the dark phasewhereas expression of the period 1 gene (per1) co<strong>in</strong>cideswith onset of the light phase (L<strong>in</strong>coln et al. 2003a;Hazlerigg et al. 2004; Wagner et al. 2008). Dimerizationof the prote<strong>in</strong> products of cry1 and per1 <strong>in</strong> thecytoplasm is required to allow their translocation <strong>in</strong>tothe nucleus, where this prote<strong>in</strong> complex is known toeffect expression of other clock genes (Reppert andWeaver 2001). L<strong>in</strong>coln et al. (2003b) hypothesized thatthe photoperiodically mediated time <strong>in</strong>terval betweenpeak expression of cry1 and per1 <strong>in</strong> the PT can act as an<strong>in</strong>dicator of day length, allow<strong>in</strong>g these cells to functionas ‘calendar cells’. Both cry1 and per1 are immediatelyresponsive to changes <strong>in</strong> photoperiod but expression ofa third clock gene <strong>in</strong> the ov<strong>in</strong>e PT, RevErba, has beenshown to vary with photoperiodic history (Hazlerigget al. 2004), provid<strong>in</strong>g an as-yet hypothetical mechanismto expla<strong>in</strong> the spontaneous changes <strong>in</strong> reproductive stateobserved <strong>in</strong> animals ma<strong>in</strong>ta<strong>in</strong>ed under constant stimulatoryor <strong>in</strong>hibitory photoperiods.Endocr<strong>in</strong>e regulation of seasonal breed<strong>in</strong>g <strong>in</strong>volveschanges <strong>in</strong> sensitivity of the bra<strong>in</strong> to steroid(predom<strong>in</strong>antly oestradiol) negative feedback. Amechanism for <strong>in</strong>teraction of circulat<strong>in</strong>g steroids withmelaton<strong>in</strong> from the p<strong>in</strong>eal gland is therefore required.Recent studies of effects of kisspept<strong>in</strong> and its associatedGPR54 receptor <strong>in</strong> the hypothalamus may provide thismiss<strong>in</strong>g l<strong>in</strong>k. Kisspept<strong>in</strong>, the product of the KiSS1 gene,stimulates GnRH release from the hypothalamus <strong>in</strong> anumber of species and nearly all GnRH neurons appearto co-express GPR54 (Smith 2008). Smith et al. (2007)demonstrated the presence of steroid receptors <strong>in</strong> KiSS1neurons <strong>in</strong> the arcuate nucleus of the ewe and found thatexpression of KiSS1 <strong>in</strong> the arcuate nucleus <strong>in</strong>creasesdur<strong>in</strong>g transition from anestrus to the breed<strong>in</strong>g season.Revel et al. (2006) reported that exogenous melaton<strong>in</strong>could override effects of photoperiod on KiSS1 expression<strong>in</strong> Syrian hamster, <strong>in</strong>dicat<strong>in</strong>g that melaton<strong>in</strong>mediates photoperiodic effects on kisspept<strong>in</strong> secretion.The sensitivity of KiSS1 neurons to both melaton<strong>in</strong> andsex steroids may provide the miss<strong>in</strong>g l<strong>in</strong>k enabl<strong>in</strong>gseasonal changes <strong>in</strong> steroid sensitivity <strong>in</strong> the hypothalamus.These functional genomic discoveries have identifiedan array of candidate genes with potential effects onseasonality. Screen<strong>in</strong>g of large populations for ewes withunusual seasonal breed<strong>in</strong>g patterns and screen<strong>in</strong>g ofthese candidate gene regions for functional sequencevariants may uncover useful mutations similar to thosefound for ovulation rate and litter size.ConclusionsThe establishment of appropriate levels of prolificacyand patterns of seasonal fertility is critical to efficientsheep production. The potential of classical selectionmethods to create change <strong>in</strong> lowly heritable reproductivetraits is often discounted, but has been shown to besubstantial. A number of s<strong>in</strong>gle-gene mutations can alsobe used to enhance ovulation rates and litter size, butcomparable mutations affect<strong>in</strong>g seasonal breed<strong>in</strong>g havenot yet been detected.Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Genetic Aspects of <strong>Reproduction</strong> <strong>in</strong> Sheep 127ReferencesAl-Shorepy SA, Notter DR, 1997: Response to selection forfertility <strong>in</strong> a fall-lamb<strong>in</strong>g sheep flock. J Anim Sci 75, 2033–2040.Banos G, Lewis RM, Notter DR, Hogue DE, 2002: Geneticprofile of fertility and prolificacy of maiden and mature ewesmanaged <strong>in</strong> a frequent lamb<strong>in</strong>g system. In: Proceed<strong>in</strong>gs ofthe 7th World Congress on Genetics Applied to LivestockProduction, CD-ROM communication No. 08-21, 4 pp.,Cedex, France.Barrett P, Conway S, Jockers R, Strosberg AD, Guardiola-Lemaitre B, Delagrange P, Morgan PJ, 1997: Clon<strong>in</strong>g andfunctional analysis of a polymorphic variant of the ov<strong>in</strong>eMel 1a melaton<strong>in</strong> receptor. Biochem Biophys Acta 1356,299–307.Bod<strong>in</strong> L, Di Pasquale E, Fabre S, Bontoux M, Monget P,Persane L, Mulsant P, 2007: A novel mutation <strong>in</strong> bonemorphogenic prote<strong>in</strong> 15 gene caus<strong>in</strong>g defective prote<strong>in</strong>secretion is associated with both <strong>in</strong>creased ovulation rateand sterility <strong>in</strong> Lacaune sheep. Endocr<strong>in</strong>ology 148,393–400.Borg RC, Notter DR, Kuehn LA, Kott RW, 2007: Breed<strong>in</strong>gobjectives for Targhee sheep. J Anim Sci 85, 2815–2829.Bradford GE, 1985: Selection for litter size. In: Land RB,Rob<strong>in</strong>son DW (eds), Genetics of <strong>Reproduction</strong> <strong>in</strong> Sheep.Butterworths, London, pp. 3–18.Davis GH, 2005: Major genes affect<strong>in</strong>g ovulation rate <strong>in</strong> sheep.Genet Sel Evol 37(Suppl. 1), 11–23.Davis GH, Galloway SM, Ross IK, Gregan SM, Ward J,Nimbkar BV, Ghalsasi PM, Nimbkar B, Gray GD,Subandriyo, Inounu I, Tiesnamurti B, Martyniuk E, EythorsdottirE, Mulsant P, Lecerf F, Hanrahan JP, BradfordGE, Wilson T, 2002: DNA tests <strong>in</strong> prolific sheep from eightcountries provide new evidence on orig<strong>in</strong> of the Booroola(FecB) mutation. Biol Reprod 66, 1869–1874.Davis GH, Balakrishnan L, Ross IK, Wilson T, Galloway SM,Lumsden BM, Hanrahan JP, Mullen M, Mao XZ, WangGL, Zhao ZS, Zeng YQ, Rob<strong>in</strong>son JJ, Mavrogenis AP,Papachristoforou C, Peter C, Baumung R, Cardyn P,Boujenane I, Cockett NE, Eythorsdottir E, Arranz JJ,Notter D, 2006: Investigation of the Booroola (FecB) andInverdale (FecX I ) mutations <strong>in</strong> 21 prolific breeds and stra<strong>in</strong>sof sheep sampled <strong>in</strong> 13 countries. Anim Reprod Sci 92,87–96.Echternkamp SE, Thallman RM, Kappes SM, Gregory KE,2002: Increased tw<strong>in</strong>n<strong>in</strong>g and cow productivity <strong>in</strong> beef cattleselected for ovulation and tw<strong>in</strong>n<strong>in</strong>g rate. In: Proceed<strong>in</strong>gs ofthe 7th World Congress on Genetics Applied to LivestockProduction. CD-ROM communication no. 08-12, 4, pp.,Cedex, France.Fahmy MH (ed.), 1996: Prolific Sheep. CAB International,Wall<strong>in</strong>gford, UK.Hazlerigg DG, Andersson H, Johnston JE, L<strong>in</strong>coln G,2004: Molecular characterization of the long-day response<strong>in</strong> the Soay sheep, a seasonal mammal. Curr Biol 14,334–339.Hernandez X, Bod<strong>in</strong> L, Chesneau D, Guillaume D, Alla<strong>in</strong> D,Chem<strong>in</strong>eau P, Malpaux B, Migaud M, 2005: Relationshipbetween MT1 melaton<strong>in</strong> receptor gene polymorphism andseasonal physiological responses <strong>in</strong> Ile-de-France ewes.Reprod Nutr Dev 45, 151–162.Lewis RM, Notter DR, Hogue DE, Magee BH, 1996: Ewefertility <strong>in</strong> the STAR accelerated lamb<strong>in</strong>g system. J Anim Sci74, 1511–1522.L<strong>in</strong>coln G, Andersson H, Hazlerigg D, 2003a: Clock genes andthe long-term regulation of prolact<strong>in</strong> secretion: evidence fora photoperiod ⁄ circannual timer <strong>in</strong> the pars tuberalis. JNeuroendocr<strong>in</strong>ol 15, 390–397.L<strong>in</strong>coln G, Andersson H, Loudon A, 2003b: Clock genes <strong>in</strong>calendar cells as the basis of annual timekeep<strong>in</strong>g <strong>in</strong>mammals – a unify<strong>in</strong>g hypothesis. J Endocr<strong>in</strong>ol 179, 1–13.Lu S, Chang H, Du L, Tsunoda K, Sun W, Yang Z, Chang G,Ji D, 2005: Phylogenetic relationships of sheep populationsfrom coastal areas of East Asia. Biochem Genet 42, 251–260.Mart<strong>in</strong> GB, Oldham CM, Cognie Y, Pearce DT, 1986: Thephysiological responses of anovulatory ewes to the<strong>in</strong>troduction of rams – a review. Livest Prod Sci 15,219–247.McNatty KP, Galloway SM, Wilson T, Smith P, Hudson NL,O’Connell A, Bibby AH, Heath DA, Davis GH, HanrahanJP, Juengel JL, 2005: Physiological effects of major genesaffect<strong>in</strong>g ovulation rate <strong>in</strong> sheep. Genet Sel Evol 37(Suppl. 1),25–38.Messer LA, Wang L, Tuggle CK, Yerle M, Chardon P, PompD, Womack JE, Barendse W, Crawford AM, Notter DR,Rothschild MF, 1997: Mapp<strong>in</strong>g of the melaton<strong>in</strong> receptor1a (MTNR1A) gene <strong>in</strong> pigs, sheep, and cattle. MammGenome 8, 368–370.Notter DR, 2002: Opportunities to reduce seasonality ofbreed<strong>in</strong>g <strong>in</strong> sheep by selection. Sheep Goat Res J 17, 20–32.Notter DR, Cockett NE, 2005: Opportunities for detectionand use of QTL <strong>in</strong>fluenc<strong>in</strong>g seasonal reproduction <strong>in</strong> sheep:a review. Genet Sel Evol 37(Suppl. 1), 39–53.Notter DR, Al-Shorepy SA, V<strong>in</strong>cent JN, McQuown EC, 1998:Selection to improve fertility <strong>in</strong> fall lamb<strong>in</strong>g. <strong>in</strong>: Proceed<strong>in</strong>gsof the 6th World Congress on Genetics Applied to LivestockProduction, vol 27, pp. 43–46, NSW, Australia.Notter DR, Cockett NE, Hadfield TS, 2003: Evaluation ofmelaton<strong>in</strong> receptor 1a as a candidate gene <strong>in</strong>fluenc<strong>in</strong>greproduction <strong>in</strong> a fall-lamb<strong>in</strong>g sheep flock. J Anim Sci 81,912–917.Pelletier J, Bod<strong>in</strong> L, Hanocq E, Malpaux B, Teyssier J,Thimonier J, Chem<strong>in</strong>eau P, 2000: Association betweenexpression of reproductive seasonality and alleles of the genefor Mel 1a receptor <strong>in</strong> the ewe. Biol Reprod 62, 1096–1101.Reppert SM, Weaver DR, 2001: Molecular analysis of mammaliancircadian rhythms. Annu Rev Physiol 63, 647–676.Revel FG, Saboureau M, Masson-Pevet M, Pevet P, MikkelsenJD, Simmonneaux V, 2006: KiSS-1: a likely candidatefor the photoperiodic control of reproduction <strong>in</strong> seasonalbreeders. Chronobiol Int 23, 277–287.Safari A, Fogarty NM, Gilmour AR, 2005: A review of geneticparameter estimates for wool, growth, meat and reproductiontraits <strong>in</strong> sheep. Livest Prod Sci 92, 271–289.Smith JT, 2008: Kisspept<strong>in</strong> signal<strong>in</strong>g <strong>in</strong> the bra<strong>in</strong>: steroidregulation <strong>in</strong> the rodent and ewe. Bra<strong>in</strong> Res Rev 57, 288–298.Smith JT, Clay CM, Caraty A, Clarke IJ, 2007: KiSS-1messenger ribonucleic acid expression <strong>in</strong> the hypothalamusof the ewe is regulated by sex steroids and season.Endocr<strong>in</strong>ology 148, 1150–1157.Turner HN, 1982: Orig<strong>in</strong>s of the CSIRO Booroola. <strong>in</strong>: PiperLR, B<strong>in</strong>don BM, Nethery RD (eds), The Booroola Mer<strong>in</strong>o,Proceed<strong>in</strong>gs of a Workshop. CSIRO, pp. 1–7, Melbourne,Australia.Vanimisetti HB, 2006. Genetic evaluation of ewe productivityand its component traits <strong>in</strong> Katahd<strong>in</strong> and Polypay sheep.Ph.D. Dissertation, Virg<strong>in</strong>ia Polytechnic Institute and StateUniversity, Blacksburg, VA, USA.V<strong>in</strong>cent JN, McQuown EC, Notter DR, 2000: Duration of theseasonal anestrus <strong>in</strong> sheep selected for fertility <strong>in</strong> falllamb<strong>in</strong>g. J Anim Sci 78, 1149–1154.Wagner GC, Johnston JD, Clarke IJ, L<strong>in</strong>coln GA, HazleriggDG, 2008: Redef<strong>in</strong><strong>in</strong>g the limits of day length responsiveness<strong>in</strong> a seasonal mammal. Endocr<strong>in</strong>ology 149, 32–39.Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


128 DR NotterWilson T, Wu X-Y, Juengel JL, Ross IK, Lumsden JM, LordEA, Dodds KG, Wall<strong>in</strong>g GA, McEwan JC, O’Connell AR,McNatty KP, Montgomery GW, 2001: Highly prolificBooroola sheep have a mutation <strong>in</strong> the <strong>in</strong>tracellular k<strong>in</strong>asedoma<strong>in</strong> of bone morphogenic prote<strong>in</strong> IB receptor (ALK-6)that is expressed <strong>in</strong> both oocytes and granulose cells. BiolReprod 64, 1225–1235.Xie CX, 1985: The History of Cattle, Sheep, and Goat Rais<strong>in</strong>g<strong>in</strong> Ch<strong>in</strong>a Attached with the History of Deer Rais<strong>in</strong>g.Agricultural Publish<strong>in</strong>g House of Ch<strong>in</strong>a, Beij<strong>in</strong>g.Author’s address (for correspondence): David R Notter, Departmentof Animal and Poultry Sciences, Virg<strong>in</strong>ia Polytechnic Institute andState University, Blacksburg, VA 24061, USA. E-mail: drnotter@vt.eduConflict of <strong>in</strong>terest: The author declares no conflict of <strong>in</strong>terest.Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 129–136 (2008); doi: 10.1111/j.1439-0531.2008.01152.xISSN 0936-6768The Importance of Interactions Among Nutrition, Seasonality and Socio-sexualFactors <strong>in</strong> the Development of Hormone-free Methods for Controll<strong>in</strong>g FertilityRJ Scaramuzzi 1,2 and GB Mart<strong>in</strong> 31 UMRPhysiologie de la <strong>Reproduction</strong> et des Comportements, L’Institut National de la <strong>Reproduction</strong>, Nouzilly, France; 2 Department of Veter<strong>in</strong>aryBasic Sciences, Royal Veter<strong>in</strong>ary College, Hertfordshire, UK; 3 Animal Production Systems, UWA Institute of Agriculture, The University ofWestern Australia, Crawley, WA, AustraliaContentsAround the world, consumers are demand<strong>in</strong>g animal productsthat are produced to agreed standards for human health,environmental management and animal welfare. This has led tothe development <strong>in</strong> Australia of the concept of ‘clean, green andethical’ (CGE) animal production based on the manipulation ofnutrition (‘focus feed<strong>in</strong>g’) and the application of phenomena,such as the ‘male effect’, to provide ‘natural’ methods formanag<strong>in</strong>g small rum<strong>in</strong>ant production systems. With respect tothe management of fertility, CGE <strong>in</strong>volves utilization of the<strong>in</strong>herited responses of animals to environmental factors tomanipulate their reproductive processes. The successfuldevelopment and implementation of this new generation ofmanagement tools depends on a thorough yet holisticunderstand<strong>in</strong>g of the <strong>in</strong>teractions among environmental factorsand the ways these <strong>in</strong>teractions affect reproductive physiologyand behaviour of the animal. For sheep and goats, a centralaspect of CGE management is the way <strong>in</strong> which ovarian functionis affected by three major factors (nutrition, photoperiod andsocio-sexual signals) and by <strong>in</strong>teractions among them. Nutritioncan exert two profound yet contrast<strong>in</strong>g types of effect on ovarianactivity: (i) the complete <strong>in</strong>hibition of reproduction by undernutritionthrough the hypothalamic mechanism that controlsovulation and (ii) the enhancement of fecundity by nutritionalsupplementation, through a direct ovarian mechanism, <strong>in</strong>females that are already ovulat<strong>in</strong>g. A similarly profound controlover ovarian function <strong>in</strong> female sheep and goats is exerted by thewell-known endocr<strong>in</strong>e responses to photoperiod (seasonality)and to male socio-sexual signals. The ‘male effect’ already has along history as a valuable technique for <strong>in</strong>duc<strong>in</strong>g a synchronizedfertile ovulation dur<strong>in</strong>g seasonal and post-partum anoestrus <strong>in</strong>sheep and goats. Importantly, experimentation has shown thatthese three major environmental factors <strong>in</strong>teract, synergisticallyand antagonistically, but the precise nature of these <strong>in</strong>teractionsand their significance to reproductive outcomes are not wellunderstood. Most research to date has been with smallrum<strong>in</strong>ants but CGE pr<strong>in</strong>ciples can be applied to any species <strong>in</strong>a managed environment. For example, a male effect has beenreported for lactat<strong>in</strong>g cattle and, <strong>in</strong> the horse, the pattern ofseasonality of oestrus can be altered by nutrition. Well-fed mareshave a longer breed<strong>in</strong>g season and some animals become nonseasonal.Similar observations have been reported for sheep andgoats. By work<strong>in</strong>g towards a holistic perspective of the physiology,nutrition, genetics and behaviour of our animals, we willbe able to formulate ways to manipulate the animals’environment that will improve management, productivity andprofitability and, simultaneously, promote a CGE <strong>in</strong>dustry.IntroductionAnimal <strong>in</strong>dustries around the world are be<strong>in</strong>g challengedby chang<strong>in</strong>g attitudes <strong>in</strong> the market place, andconsumers are <strong>in</strong>creas<strong>in</strong>gly demand<strong>in</strong>g products that areproduced to agreed standards for human health,environmental management and animal ethics andwelfare. This is particularly evident <strong>in</strong> high-profit,discern<strong>in</strong>g markets that are <strong>in</strong>fluenced by discretionaryspend<strong>in</strong>g power. Our challenge is to turn these developmentsto advantage, improve productivity and profitabilityand, simultaneously, promote ‘clean, green and ethical(CGE) production’ (Mart<strong>in</strong> et al. 2004a; Kadokawa andMart<strong>in</strong> 2006; Mart<strong>in</strong> and Kadokawa 2006). Importantly,these issues need not pose difficulties or <strong>in</strong>crease costs – onthe contrary, they offer excellent opportunities. Forextensively managed sheep and goats, we are develop<strong>in</strong>gmanagement tools, such as ‘focus feed<strong>in</strong>g’ and the ‘maleeffect’, for controll<strong>in</strong>g reproduction and quantitativebreed<strong>in</strong>g values for improv<strong>in</strong>g temperament (Mart<strong>in</strong>et al. 2004a).This paper reviews current <strong>in</strong>formation, ma<strong>in</strong>ly forsheep and goats, on a central aspect of the CGE system –the way <strong>in</strong> which ovarian function is affected by threemajor factors (nutrition, photoperiod and socio-sexualsignals) and by <strong>in</strong>teractions among them. We review theway <strong>in</strong> which nutrition can <strong>in</strong>fluence the ‘decision toreproduce’ through the hypothalamic mechanism thatcontrols ovulation and the ‘decision for fecundity’through a direct ovarian mechanism <strong>in</strong> females that arealready ovulat<strong>in</strong>g. In addition, we review the ways <strong>in</strong>which the endocr<strong>in</strong>e responses to photoperiod(seasonality) and to male socio-sexual signals <strong>in</strong>teract,synergistically and antagonistically, with nutritional<strong>in</strong>puts. It is <strong>in</strong>creas<strong>in</strong>gly clear that the multi-dimensionalnature of these <strong>in</strong>teractions are often far more importantthan the responses to the <strong>in</strong>dividual environmental <strong>in</strong>put(Blache et al. 2007).Most research to date has been with small rum<strong>in</strong>antsbut the ‘CGE pr<strong>in</strong>ciple’, <strong>in</strong> which manipulation of theenvironment is used to control reproductive processes,can be applied to all species. For example, a male effecthas been reported for cattle (review: Ungerfeld 2007)and, <strong>in</strong> this review, we will describe nutrition–reproduction<strong>in</strong>teractions <strong>in</strong> the horse.Thus, the aim of this review is to outl<strong>in</strong>e the state ofour knowledge on the <strong>in</strong>teractions among environmentalfacts that affect reproductive processes, and to po<strong>in</strong>tout research that is needed for ref<strong>in</strong><strong>in</strong>g CGE managementof farm animals, particularly with respect tomanagement of the nutritional <strong>in</strong>puts.Nutrition and Ovarian FunctionAll major measures of reproductive performance (prolificacy,fertility and fecundity) are affected by geneticsÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


130 RJ Scaramuzzi and GB Mart<strong>in</strong>and by a variety of environmental factors <strong>in</strong>clud<strong>in</strong>g:(i) nutrition, energy balance and diet; (ii) photoperiodand seasonality; (iii) socio-sexual factors; (iv) temperatureand relative humidity and (v) stress. Nutritionappears to have bi-directional effects on ovarian functionand the nature of these effects we suggest are <strong>in</strong>hibitory ata hypothalamic level (allow<strong>in</strong>g or prevent<strong>in</strong>g ovulation)and stimulatory at an ovarian level (affect<strong>in</strong>g ovulationrate) and, furthermore, may differ between rum<strong>in</strong>ant andmonogastric species. With respect to prolificacy, smallrum<strong>in</strong>ants are generally regarded as seasonal monocotousspecies, although natural tw<strong>in</strong>n<strong>in</strong>g is not uncommon,and there is considerable variation <strong>in</strong> the <strong>in</strong>cidence oftw<strong>in</strong> ovulations and tw<strong>in</strong> births among and with<strong>in</strong> breeds.The component of the diet that is probably the mostimportant with respect to ovarian function is energyparticularly that derived from glucose, although theevidence <strong>in</strong> favour of any particular nutrient is stillcontroversial and the precise <strong>in</strong>terrelationships betweenovarian function and the components of the diet areobscure.Nutritional effects on ovarian function are bi-directionalNutritional deprivation of the female, whether as aresult of an <strong>in</strong>sufficient supply of energy <strong>in</strong> the diet, or areduced availability of energy because of excessivedemands for energy (extreme physical activity, energydemand<strong>in</strong>gprocesses such as lactation), <strong>in</strong>hibits hypothalamicGnRH release and thus leads to reducedsecretion of pituitary LH and eventually to anovulationand anoestrus. The hypothalamic GnRH pulse-generat<strong>in</strong>gsystem <strong>in</strong> monogastric species (rats, primates andhumans) appears to be more sensitive to nutritionaldeprivation than <strong>in</strong> rum<strong>in</strong>ants (sheep, goats and cattle).In monogastric species, undernutrition and associatedhypoglycaemia quickly leads to suppression of theGnRH system and a cessation of LH pulsatility (Bronson1988; Cagampang et al. 1990). Farmed rum<strong>in</strong>antsrarely develop hypoglycaemia and thus appear to besomewhat protected aga<strong>in</strong>st nutritional anovulation,although LH pulsatility is <strong>in</strong>hibited when they are madehypoglycaemic under experimental conditions (Down<strong>in</strong>gand Scaramuzzi 1997; Szymanski et al. 2007) or as aresult of lactation (Rhodes et al. 1995). On the contrary,for animals <strong>in</strong> normal body condition, nutritionalsupplementation appears to have little direct <strong>in</strong>fluenceon hypothalamic GnRH secretion or pituitary gonadotroph<strong>in</strong>secretion <strong>in</strong> the female and there is littleunambiguous evidence to show that nutritional supplementationstimulates gonadotroph<strong>in</strong> secretion by aprimary hypothalamic mechanism or pituitary mechanism.Thus, for ovulation, primarily a GnRH-dependentprocess, the effects of nutrient supplementation are notsimply the opposite to the effects of nutrient deprivation.On the contrary, nutritional supplementation can stimulatefolliculogenesis (Webb et al. 2004; Scaramuzziet al. 2006) and thus <strong>in</strong>crease prolificacy (Knight et al.1975; Wang et al. 2004; Gask<strong>in</strong>s et al. 2005), but thisappears to <strong>in</strong>volve direct nutritional <strong>in</strong>fluences on thefollicle itself. Follicular responses to <strong>in</strong>creased nutrientsupply are not always positive and although <strong>in</strong>creasedfolliculogenesis is associated with either short-termnutritional supplementation or moderate <strong>in</strong>creases <strong>in</strong>body weight, extreme <strong>in</strong>creases <strong>in</strong> body weight (obesity)and rapid fluctuations <strong>in</strong> nutrient supply, such asrepeated cycles of ‘b<strong>in</strong>ge-eat<strong>in</strong>g’ followed by starvation,have <strong>in</strong>hibitory effects on ovarian function and reducefertility (Hartz et al. 1979; Lake et al. 1997; Wolfe 2005).Thus, there appears to be a nutritional thresholdbelow which reproduction is <strong>in</strong>hibited because of thesuppression of the GnRH pulse-generat<strong>in</strong>g system to alevel of activity below that required to ma<strong>in</strong>ta<strong>in</strong> regularovarian cycles. At the hypothalamic level, nutritional<strong>in</strong>fluences on reproduction are qualitative determ<strong>in</strong><strong>in</strong>gfertility by assess<strong>in</strong>g whether an animal ovulates or not.However, once this threshold is reached and ovariancycles are occurr<strong>in</strong>g, nutritional regulation becomesquantitative and operates at the level of the follicle todeterm<strong>in</strong>e the rate of reproduction – ovulation rate,prolificacy or litter size.Dietary supplementation, flush<strong>in</strong>g and dietary signalsFlush<strong>in</strong>g, a form of dietary supplementation, is apractice that has been used <strong>in</strong> rum<strong>in</strong>ant productionsystems s<strong>in</strong>ce at least the beg<strong>in</strong>n<strong>in</strong>g of the 19th century(Youatt 1837). Despite, or perhaps because of, itswidespread use, there appears to be a variety ofnutritional practices covered by the term, ‘flush<strong>in</strong>g’. Inthe absence of a precise def<strong>in</strong>ition of flush<strong>in</strong>g, the<strong>in</strong>terpretation of published <strong>in</strong>formation and attempts toelucidate the mechanism of nutritional <strong>in</strong>fluences onovarian function have been problematic. The metabolicoutcomes of a period of short-term (3–7 days) nutritionalsupplementation are very different to those for a6- to 8-week period of supplementation. Similarly, themetabolic consequences of dietary supplementation ofanimals <strong>in</strong> negative energy balance will be very differentfrom those for animals <strong>in</strong> positive energy balance. It istherefore very likely that the effects on ovarian folliculogenesisand the GnRH pulse-generat<strong>in</strong>g system willalso depend on the metabolic state of the animal and thenature of the supplementation. For example, Nottleet al. (1997a) reported an experiment with ewes that hadbeen previously managed for 6 months as separateflocks on either a high or low plane of nutrition. The twoflocks were recomb<strong>in</strong>ed 17 days before mat<strong>in</strong>g and givenshort-term nutritional supplementation with lup<strong>in</strong> gra<strong>in</strong>for the 10 days prior to mat<strong>in</strong>g. The short-term supplement<strong>in</strong>creased the ovulation rate by 54% <strong>in</strong> theunderfed group, compared with only 23% <strong>in</strong> the wellfedgroup, and the <strong>in</strong>teraction between previous nutritionalhistory and short-term supplementation wassignificant (Nottle et al. 1997a). Another experiment(Leury et al. 1990) reported an opposite f<strong>in</strong>d<strong>in</strong>g; lup<strong>in</strong>gra<strong>in</strong> supplementation <strong>in</strong> ewes fed 1.2 times ma<strong>in</strong>tenance<strong>in</strong>creased ovulation rate by 22% compared withewes fed at 0.8 times ma<strong>in</strong>tenance where lup<strong>in</strong> gra<strong>in</strong>supplementation <strong>in</strong>creased ovulation rate by 10%. Thereasons for this difference are not immediately apparentand additional <strong>in</strong>vestigation is obviously required.There has been considerable speculation and someresearch attempt<strong>in</strong>g to identify the critical componentsof the diet that <strong>in</strong>fluence ovarian function. The diet ofÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Nutritional Interactions and <strong>Reproduction</strong> 131mammals is made up of complex mixtures of prote<strong>in</strong>s,fats and simple and complex carbohydrates as well astrace elements, vitam<strong>in</strong>s and other micronutrients, andall of these components have the potential to <strong>in</strong>fluencereproduction, either directly or <strong>in</strong>directly. The currentconsensus suggests that dietary energy is a very importantcomponent of the diet with respect to nutritional<strong>in</strong>fluences on ovarian function. The short-term adm<strong>in</strong>istrationof glucose or other energy-yield<strong>in</strong>g substratescan <strong>in</strong>crease ovulation rate <strong>in</strong> ewes (Nottle et al. 1988;Teleni et al. 1989; Down<strong>in</strong>g et al. 1995a,b; V<strong>in</strong>˜ oles et al.2005). However, there are other published reports <strong>in</strong>which the <strong>in</strong>creased supply of glucose did not <strong>in</strong>creaseovulation rate <strong>in</strong> ewes (Iglesias et al. 1996) and furthermore,<strong>in</strong> the male, the published data suggest that the<strong>in</strong>creased spermatogenesis seen <strong>in</strong> response to feed<strong>in</strong>g alup<strong>in</strong> gra<strong>in</strong> supplement is not caused by an <strong>in</strong>creasedavailability of glucose (Blache et al. 2002).From the above remarks it follows that, whenexam<strong>in</strong><strong>in</strong>g the effects of nutrition on ovarian functionand folliculogenesis, there is an obvious need for a moredetailed description of both animals’ diets and metabolicstates than is customary.Sources of metabolic glucose that affect ovarian functionGlucose for the generation of ATP is derived from eitherdietary sources or from gluconeogenesis, either with orwithout accompany<strong>in</strong>g glycogen synthesis. Monogastricspecies derive their glucose pr<strong>in</strong>cipally from dietarysources rather than from gluconeogenesis, whereas therum<strong>in</strong>ant species reduce their dietary glucose <strong>in</strong> theanaerobic conditions of the rumen and derive theirglucose almost exclusively by gluconeogenesis, fromdiet-derived long-cha<strong>in</strong> fatty acids and short-cha<strong>in</strong>volatile fatty acids. Compared with monogastrics, this isprobably a highly significant functional difference withrespect to the effects of nutrition on ovarian functionbecause rum<strong>in</strong>ants, animals with highly efficient gluconeogenesisrarely become hypoglycaemic or, for thatmatter, hyperglycaemic. For example, dairy cows with ahigh genetic merit for milk production do not becomehypoglycaemic at the peak of lactation, although they canbe <strong>in</strong> severe negative energy balance. Indeed, hypoglycaemiais usually only seen <strong>in</strong> rum<strong>in</strong>ants <strong>in</strong> the most extremesituations such as pregnancy toxaemia (tw<strong>in</strong> lambdisease) <strong>in</strong> sheep and <strong>in</strong> lactat<strong>in</strong>g first-calf beef heifersthat are still grow<strong>in</strong>g. Hypoglycaemic beef heifers have anextended period of post-partum anovulation associatedwith reduced LH pulsatility, because the hypoglycaemia<strong>in</strong>hibits the GnRH pulse-generat<strong>in</strong>g system. The situationwith monogastric species appears to be quite differentbecause they depend on the diet for supplies of glucoseand have a limited capacity for gluconeogenesis comparedwith rum<strong>in</strong>ants. Monogastric species readilybecome hypoglycaemic when deprived of dietary energyand the consequence is a reduced GnRH ⁄ LH pulsefrequency and thus anovulation.Seasonality and Ovarian FunctionSeasonal patterns of reproduction are relatively commonamong the mammals <strong>in</strong>clud<strong>in</strong>g the domesticlivestock – sheep, goats, horses and, under somecircumstances, cattle and pigs show seasonal rhythmsof ovarian function that are driven primarily by photoperiod.These patterns and the processes that underp<strong>in</strong>them, especially <strong>in</strong> sheep, have been extensively <strong>in</strong>vestigatedand there are a number of detailed reviews on thesubject (e.g.: Yeates 1949; Hafez 1952; Karsch et al.1984; Malpaux 2006).Interactions between nutrition and seasonalityPhotoperiod and nutrition both exert major <strong>in</strong>fluenceson reproduction so it seems axiomatic that seasonalrhythms <strong>in</strong> ovulation will be <strong>in</strong>fluenced by nutrition. It ishighly probable that photoperiodic and nutritional<strong>in</strong>puts to the hypothalamus <strong>in</strong>teract at the level of theGnRH neuron (Ho¨ tzel et al. 2003; Blache et al. 2007),yet there are relatively few published <strong>in</strong>vestigations ofthe <strong>in</strong>teraction between nutrition and photoperiodism <strong>in</strong>females. There are few controlled studies with ewes<strong>in</strong>vestigat<strong>in</strong>g the relationship between nutrition and thepattern of seasonality, although there are reports ofalterations <strong>in</strong> the seasonal pattern of cyclic ovulatoryactivity associated with nutritional conditions <strong>in</strong> previousseasons (Hunter 1962; Smith 1965; Oldham et al.1990). In one study carried out over 13 months (Huletet al. 1986), ewes at pasture had a lower proportionovulat<strong>in</strong>g <strong>in</strong> the months of anoestrus (May, June andJuly) compared with ewes <strong>in</strong> a drylot that weresupplemented with alfalfa hay (Fig. 1). The pasturefedewes had a higher proportion of anovulatory ewes <strong>in</strong>February suggest<strong>in</strong>g that they were enter<strong>in</strong>g anoestroussooner. Similarly, <strong>in</strong> August, the pasture-fed ewes had alower proportion of anovulatory ewes, aga<strong>in</strong> suggest<strong>in</strong>gthat they were slower to resume ovarian cyclicity at thestart of the new breed<strong>in</strong>g season. In another study, theseasonal pattern of ovarian cyclicity was determ<strong>in</strong>ed <strong>in</strong>Rasa Aragonesa ewes ma<strong>in</strong>ta<strong>in</strong>ed at two levels of bodycondition (Forcada et al. 1992; Forcada and Abecia2006). This study showed that the duration of anoestruswas reduced by approximately 2 months <strong>in</strong> the ewesEwes ovulat<strong>in</strong>g (%)1009080706050403020100FebPastureDry lotMayJun JulTime of the yearsAugSepFig. 1. The proportion of ewes ovulat<strong>in</strong>g <strong>in</strong> February (enter<strong>in</strong>ganoestrus), May to July (dur<strong>in</strong>g anoestrus) and August and September(the start of the next breed<strong>in</strong>g season) <strong>in</strong> two groups of f<strong>in</strong>e-wool ewes.One group was managed on rangeland and the other group wasmanaged with alfalfa hay <strong>in</strong> drylot. The with<strong>in</strong> month treatment effectswere significant for all months except September. Data taken fromHulet et al. (1986)Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


132 RJ Scaramuzzi and GB Mart<strong>in</strong>ma<strong>in</strong>ta<strong>in</strong>ed <strong>in</strong> good body condition (BCS, 2.8) comparedwith ewes ma<strong>in</strong>ta<strong>in</strong>ed <strong>in</strong> poor body condition(BCS, 2.3).More extensive data are available for two otherseasonal species, the horse and the goat. In a study justcompleted at the INRA laboratory at Nouzilly, Salazar-Ortiz and Guillaume studied the effect of nutrition onthe seasonal rhythm of ovarian activity <strong>in</strong> mares keptunder natural photoperiod (Salazar-Ortiz 2006). Thepattern of ovarian cyclicity over three consecutive yearswas determ<strong>in</strong>ed from weekly blood progesterone measurements<strong>in</strong> two groups of mares. One group was wellfed and the other group was received a diet calculated tokeep the mares th<strong>in</strong> but <strong>in</strong> good health. At the start ofthe experiment, the average body weight of the twogroups was approximately 300 kg. The well-fed maresga<strong>in</strong>ed approximately 20 kg <strong>in</strong> the first year after whichtheir mean live weight did not change for the rema<strong>in</strong>derof the experiment. The restricted mares lost approximately65 kg <strong>in</strong> the first year and then their live weightalso stabilized for the rema<strong>in</strong>der of the experiment. Thenutritional regime had a profound effect on the patternof ovarian cyclicity (Fig. 2) – the well-fed mares had amuch longer period of ovarian cyclicity and six of the 10mares had no period of w<strong>in</strong>ter anovulation <strong>in</strong> the 3-yearexperiment. This was <strong>in</strong> contrast with the restrictedgroup <strong>in</strong> which the w<strong>in</strong>ter period of anovulation waslonger and present <strong>in</strong> all the 10 mares.Zarazaga et al. (2005) studied the effect of nutritionon the seasonal pattern of sexual activity <strong>in</strong> femalePayoya goats kept under natural photoperiod over aperiod of 20 months. The does were ma<strong>in</strong>ta<strong>in</strong>ed on twolevels of nutrition: a high group fed 1.5 times theircalculated ma<strong>in</strong>tenance requirements and a low groupfed to ma<strong>in</strong>tenance. The pattern of seasonality was thendeterm<strong>in</strong>ed by daily monitor<strong>in</strong>g for oestrus and forovulation us<strong>in</strong>g progesterone measured <strong>in</strong> blood twice aweek. The length of anoestrus was 32 days shorter <strong>in</strong>does on the high diet and this difference was significantbecause of both a delayed entry <strong>in</strong>to anoestrus and anearlier resumption of ovarian cyclicity at the end ofanoestrus.Although domestic Bos taurus cattle are non-seasonal,one study (Montgomery et al. 1985) reported an <strong>in</strong>teractionbetween season and nutrition on the resumptionof ovarian cyclicity post-partum. In this study, an effectAnovulatory mares (%)100806040200Sum Aut W<strong>in</strong> Spr Sum Aut W<strong>in</strong> Spr Sum Aut W<strong>in</strong> Spr SumSeasonFig. 2. The weekly proportion of anovulatory mares over a 3-yearperiod. The data for the mares on a high plane of nutrition are shownby the solid l<strong>in</strong>e and that for the mares on a low plane of nutrition bythe dashed l<strong>in</strong>e. Redrawn from Salazar-Ortiz (2006)of the season of calv<strong>in</strong>g (w<strong>in</strong>ter versus spr<strong>in</strong>g at latitude45° south) was observed only under conditions of lownutrition. The same seems to apply to laboratory rats, <strong>in</strong>which undernutrition seems to be able to modulatereproductive responses to photoperiod through a p<strong>in</strong>ealdependentpathway (Walker and Bethea 1977).What do all these observations tell us? Most importantly,that there is an <strong>in</strong>teraction <strong>in</strong> which nutritionaffects the seasonal pattern of ovarian cyclicity <strong>in</strong> sheep,goats and horses; the variety of species suggests thatnutritional <strong>in</strong>fluences on patterns of seasonal reproductionare probably present <strong>in</strong> most, if not all, seasonalbreed<strong>in</strong>g mammals, <strong>in</strong>clud<strong>in</strong>g non-seasonal species suchas beef cattle. However, the studies done to date withfemale animals do not shed any light on the mechanismsthat underlie this <strong>in</strong>teraction or on how the <strong>in</strong>teractioncan be usefully <strong>in</strong>corporated <strong>in</strong>to systems of CGEanimal production.Socio-sexual Signall<strong>in</strong>g and Ovarian FunctionThe ‘male effect’ is a well-studied phenomenon <strong>in</strong> sheepand goats and there are a number of excellent recentreviews of the subject (Mart<strong>in</strong> et al. 2004b; Ungerfeldet al. 2004; Chem<strong>in</strong>eau et al. 2006; Ungerfeld 2007). Insummary, sexually active males produce a pheromonethat stimulates the secretion of GnRH pulses and ovarianactivity and can <strong>in</strong>duce anovulatory females to ovulate.Interactions between nutrition and socio-sexual signalsAs with photoperiod, socio-sexual signals are a majorfactor controll<strong>in</strong>g ovulation <strong>in</strong> seasonal species and,because nutrition also has major <strong>in</strong>fluences onovulation, it also seems highly probable that nutritionwould <strong>in</strong>fluence the efficacy of the ‘male effect’ and thatthe site of the <strong>in</strong>teraction is aga<strong>in</strong> the GnRH neuron <strong>in</strong>the hypothalamus (Mart<strong>in</strong> et al. 2004b; Blache et al.2007). When a group of female goats were dividedaccord<strong>in</strong>g to bodyweight, it was observed that theoestrus response to the ‘buck effect’ was reduced <strong>in</strong> thedoes with the lowest body weight compared with thosewith medium and heavy weights (Ve´liz et al. 2006).Similar f<strong>in</strong>d<strong>in</strong>gs have been reported for ewes (Lassouedand Khaldi 1990). Similarly, <strong>in</strong> beef cattle, the sociosexual<strong>in</strong>fluence of bulls can affect growth rate and theage of puberty <strong>in</strong> heifers (Roberson et al. 1991), andnutrition modifies the ability of bulls to reduce theduration of post-partum anoestrus (Monje et al. 1992;Stumpf et al. 1992).In a recent experiment, we have <strong>in</strong>vestigated the<strong>in</strong>teraction between nutrition and the ‘male effect’ <strong>in</strong>anoestrous female goats (De Santiago-Miramontes et al.2008). Two groups of 25 anoestrous female goats grazedvery poor quality natural vegetation from 09:00 to16:00 h daily. Overnight, they were housed <strong>in</strong> pens andone of the groups received a supplement (950 g lucernehay, 290 g rolled corn and 140 g soy bean per animaldaily) for 7 days before exposure to bucks. The proportionof does ovulat<strong>in</strong>g, the proportion <strong>in</strong> oestrus and theovulation rate at the first ovulation detected with<strong>in</strong>5 days of exposure to males were all greater <strong>in</strong> supplementedthan <strong>in</strong> the control females (Fig. 3). The effect ofÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Nutritional Interactions and <strong>Reproduction</strong> 1332.01.610080Ovulat<strong>in</strong>gOestrusOvulation rate1.20.8Response (%)60400.420Fig. 3. The ovulation rate(mean ± SEM; n = 25), <strong>in</strong>terval(mean ± SEM days) from the<strong>in</strong>troduction of bucks to the onsetof oestrus, and the proportions ofdoes ovulat<strong>in</strong>g, show<strong>in</strong>g oestrusand hav<strong>in</strong>g short cycles at the firstovulation follow<strong>in</strong>g buck<strong>in</strong>troduction <strong>in</strong> does that wereeither given nutritional supplementationor not given nutritionalsupplementation for 7 days beforethe <strong>in</strong>troduction of sexually activebucks. All between-treatmentcomparisons are significant. Datataken from De Santiago-Miramontes et al. (2008)Interval to oestrus (days)0.08.06.04.02.00.0Does with short cycles (%)0100806040200Control Supplemented Control Supplementedsupplementation did not persist and the proportion ofdoes <strong>in</strong> oestrus was not different if ovulation occurredafter 5 days (Fig. 4) and neither was the ovulation rate(De Santiago-Miramontes et al. 2008). We concludedthat short-term feed supplementation before the ‘maleeffect’ can <strong>in</strong>crease the proportion of females respond<strong>in</strong>gand their ovulation rate. However, the stimulatory effectof supplementation did not persist and was not observedat the subsequent ovulation. The <strong>in</strong>crease <strong>in</strong> ovulationrate suggests that the supplement promoted the growthof a greater number of ovulatory follicles or reduced therate of atresia among the exist<strong>in</strong>g cohort of follicles.Somewhat surpris<strong>in</strong>gly, supplementation also <strong>in</strong>creasedthe proportion of does show<strong>in</strong>g short cycles after thefirst male-<strong>in</strong>duced ovulation and this response wasassociated with a reduced <strong>in</strong>terval from the <strong>in</strong>troductionof bucks to oestrus (Fig. 3). The effect of nutritionalsupplementation on the proportion of short cycles<strong>in</strong>duced by the male effect could be due to the fact thatDoes <strong>in</strong> oestrus (%)6050403020100a0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15Time relative to the <strong>in</strong>troduction of bucks (days)Fig. 4. The daily proportion of does show<strong>in</strong>g oestrus for 15 days afterthe <strong>in</strong>troduction of sexually active bucks on day 0 and follow<strong>in</strong>g anutritional supplementation for 7 days before the <strong>in</strong>troduction of thebucks. The data for the supplemented group are shown by the dashedl<strong>in</strong>e and for the non-supplemented (control) group by the solid l<strong>in</strong>e; theletter ‘a’ = p < 0.05 compared with the control group at the sametime. Redrawn from De Santiago-Miramontes et al. (2008)more of supplemented does ovulated over the first5 days compared with the non-supplemented doesbecause most does respond<strong>in</strong>g <strong>in</strong> the first 5 days havea short cycle (Chem<strong>in</strong>eau et al. 2006; Delgadillo et al.2006). These f<strong>in</strong>d<strong>in</strong>gs are consistent with the suggestionthat a shorter delay from male <strong>in</strong>troduction to the firstovulation is associated with an <strong>in</strong>creased proportion ofshort cycles (Pearce et al. 1985). The nutritional supplementappears to have accelerated the follicular phase<strong>in</strong>duced by the ‘buck effect’ and the shorter period offollicular development (Fig. 4) leads to either a nonfunctionalCL or, perhaps, to uter<strong>in</strong>e entra<strong>in</strong>ment for anearly luteolytic signal.Somewhat similar f<strong>in</strong>d<strong>in</strong>gs have been reported forewes. In one study, Nottle et al. (1997b) reported thatsupplementation of Mer<strong>in</strong>o ewes daily with 500 g lup<strong>in</strong>gra<strong>in</strong> from Days 12 to 26 after the <strong>in</strong>troduction of ramssignificantly <strong>in</strong>creased the ovulation rate from 1.26 to1.46 at the second ovulation follow<strong>in</strong>g ram <strong>in</strong>troduction.Work<strong>in</strong>g with lactat<strong>in</strong>g Sarda ewes, Molle et al. (1995)also observed an <strong>in</strong>crease <strong>in</strong> ovulation rate follow<strong>in</strong>gsupplementation start<strong>in</strong>g 2 weeks before the ‘ram effect’,but only with a supplement of soya bean meal and notwith whole corn gra<strong>in</strong>. In a later study, Molle et al.(1997) reported that short-term supplementation foronly 7 days before the <strong>in</strong>troduction of rams <strong>in</strong>creasedovulation rate from 1.36 to 1.66 but, <strong>in</strong> this case,the difference was not significant, possibly because of thegroup size of 15 ewes per treatment was too small. Theseoutcomes contrast with those from a factorial study with390 Mer<strong>in</strong>o ewes (Fisher et al. 1993) – there were nosignificant effects of short-term supplementation withlup<strong>in</strong> gra<strong>in</strong> on the proportion of the flock respond<strong>in</strong>g,ovulation rate or the frequency of short cycles <strong>in</strong> ewesstimulated with the ‘ram effect’. This outcome was<strong>in</strong>dependent of the previous nutritional history of theewes, although the percentage of ewes ovulat<strong>in</strong>g washigher <strong>in</strong> ewes <strong>in</strong> good condition than <strong>in</strong> ewes <strong>in</strong> poorÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


134 RJ Scaramuzzi and GB Mart<strong>in</strong>condition. There is no obvious explanation for thedifferences <strong>in</strong> outcomes among these various studies.Nutritional studies are difficult because they requirestrict control of previous nutritional history. The mechanismbeh<strong>in</strong>d the reproductive response to nutritionalsupplementation is still problematic because we do nothave experimental models to expla<strong>in</strong> the variability <strong>in</strong>the response to a nutritional supplementation, the maleeffect and their <strong>in</strong>teraction.Overall, it is clear that the efficacy of the ‘male effect’is <strong>in</strong>fluenced by nutritional and metabolic factors <strong>in</strong> thefemales. Thus, optimization of the efficacy of the ‘maleeffect’ for CGE management requires a very clearunderstand<strong>in</strong>g of how nutrition <strong>in</strong>fluences the responsivenessof females to socio-sexual signals.ConclusionsThis brief review of nutritional <strong>in</strong>teractions with seasonalrhythms and the ‘male effect’ draws attention tothe importance of nutrition as a major factor <strong>in</strong>fluenc<strong>in</strong>greproductive outcomes and po<strong>in</strong>ts to critical areas offuture research. There are some reports, often onlydescriptively document<strong>in</strong>g the nature of these <strong>in</strong>teractionsbut there has been very little research explor<strong>in</strong>g theunderly<strong>in</strong>g processes. We need molecular genetic explanationsof the reproductive, metabolic, behavioural andphysiological processes that underp<strong>in</strong>, for example, the<strong>in</strong>fluence of nutritional <strong>in</strong>puts on the melaton<strong>in</strong>-drivenseasonal rhythms of ovarian cyclicity, and on femaleresponsiveness to the ‘male effect’ at both bra<strong>in</strong> andovarian levels. By work<strong>in</strong>g towards a holistic perspectiveof the physiology, nutrition, genetics and behaviour ofour animals, we will be able to formulate ways tomanipulate their environment that will improve management,productivity and profitability and, simultaneously,promote a CGE <strong>in</strong>dustry.AcknowledgementsRJ Scaramuzzi is the recipient of a Marie Curie Chair of Excellencefrom the European Union (SUSTREPRO: FP6-2005-Mobility-10 ⁄ 42499) and their support is gratefully acknowledged. GB Mart<strong>in</strong>was funded by Meat and Livestock Australia (Project MS.027;‘LambMax’). Thanks are due to Dr B Malpaux and Dr D Guillaumefor their constructive comments on the manuscript.ReferencesBlache D, Adam CL, Mart<strong>in</strong> GB, 2002: The mature malesheep: a model to study the effects of nutrition on thereproductive axis. Reprod Suppl 59, 219–233.Blache D, Chagas LM, Mart<strong>in</strong> GB, 2007: Nutritional <strong>in</strong>puts<strong>in</strong>to the reproductive neuroendocr<strong>in</strong>e control system – amultidimensional perspective. In: Juengel JI, Murray JF,Smith MF (eds), <strong>Reproduction</strong> <strong>in</strong> <strong>Domestic</strong> Rum<strong>in</strong>ants VI.Nott<strong>in</strong>gham University Press, Nott<strong>in</strong>gham, pp. 123–139.Bronson FH, 1988: Effect of food manipulation on the GnRH-LH-estradiol axis of young female rats. Am J Physiol 254,R616–621.Cagampang FR, Maeda K, Yokoyama A, Ota K, 1990: Effectof food deprivation on the pulsatile LH release <strong>in</strong> the cycl<strong>in</strong>gand ovariectomized female rat. Horm Metab Res 22, 269–272.Chem<strong>in</strong>eau P, Pellicer-Rubio MT, Lassoued N, Khaldi G,Monniaux D, 2006: Male-<strong>in</strong>duced short oestrous andovarian cycles <strong>in</strong> sheep and goats: a work<strong>in</strong>g hypothesis.Reprod Nutr Dev 46, 417–429.De Santiago-Miramontes MA, Rivas-Mun˜ oz R, Mun˜ oz-Gutiérrez M, Malpaux B, Scaramuzzi RJ, Delgadillo JA,2008: Ovulation rate <strong>in</strong> female anoestrous goats exposed tothe male effect is <strong>in</strong>creased by nutritional supplementationunder graz<strong>in</strong>g conditions. Anim Reprod Sci 105, 409–416.Delgadillo JA, Flores JA, Véliz FG, Duarte G, Vielma J,Hernandez H, Fernandez IG, 2006: Importance of thesignals provided by the buck for the success of the maleeffect <strong>in</strong> goats. Reprod Nutr Dev 46, 391–400.Down<strong>in</strong>g JA, Scaramuzzi RJ, 1997: The effect of the <strong>in</strong>fusionof <strong>in</strong>sul<strong>in</strong> dur<strong>in</strong>g the luteal phase of the estrous cycle on theovulation rate and on plasma concentrations of LH, FSHand glucose <strong>in</strong> ewes. Theriogenology 47, 747–759.Down<strong>in</strong>g JA, Joss J, Scaramuzzi RJ, 1995a: A mixture of thebranched cha<strong>in</strong> am<strong>in</strong>o acids, leuc<strong>in</strong>e, isoleuc<strong>in</strong>e and val<strong>in</strong>e,<strong>in</strong>creases ovulation rate <strong>in</strong> ewes when <strong>in</strong>fused dur<strong>in</strong>g the lateluteal phase of the oestrous cycle: an effect that may bemediated by <strong>in</strong>sul<strong>in</strong>. J Endocr<strong>in</strong>ol 145, 315–323.Down<strong>in</strong>g JA, Joss J, Scaramuzzi RJ, 1995b: Ovulation rateand the concentrations of gonadotroph<strong>in</strong>s and metabolichormones <strong>in</strong> ewes <strong>in</strong>fused with glucose dur<strong>in</strong>g the lateluteal phase of the oestrous cycle. J Endocr<strong>in</strong>ol 147, 403–410.Fisher JS, Mart<strong>in</strong> GB, Oldham CM, Gray SJ, 1993: Long-termeffects of nutrition on spontaneous ovulation <strong>in</strong> Mer<strong>in</strong>oewes and their responses to the ‘ram effect’. Proc VII WorldConf Anim Prod 2, 32–33.Forcada F, Abecia JA, 2006: The effect of nutrition on theseasonality of reproduction <strong>in</strong> ewes. Reprod Nutr Dev 46,355–365.Forcada F, Abecia JA, Sierra I, 1992: Seasonal changes <strong>in</strong>oestrous activity and ovulation rate <strong>in</strong> Rasa Aragonesa ewesma<strong>in</strong>ta<strong>in</strong>ed at two different body condition levels. SmallRum<strong>in</strong> Res 8, 313–324.Gask<strong>in</strong>s CT, Snowder GD, Westman MK, Evans M, 2005:Influence of body weight, age, and weight ga<strong>in</strong> on fertilityand prolificacy <strong>in</strong> four breeds of ewe lambs. J Anim Sci 83,1680–1689.Hafez ESE, 1952: Studies on the breed<strong>in</strong>g season andreproduction of the ewe. J Agric Sci (Camb) 42, 189–265.Hartz AJ, Barboriak PN, Wong A, Katayama KP, Rimm AA,1979: The association of obesity with <strong>in</strong>fertility and relatedmenstrual abnormalities <strong>in</strong> women. Int J Obes 3, 57–73.Ho¨ tzel MJ, Walkden-Brown SW, Fisher JS, Mart<strong>in</strong> GB, 2003:Determ<strong>in</strong>ants of the annual pattern of reproduction <strong>in</strong>mature male Mer<strong>in</strong>o and Suffolk sheep: responses to anutritional stimulus <strong>in</strong> the breed<strong>in</strong>g and non-breed<strong>in</strong>gseasons. Reprod Fertil Dev 15, 1–9.Hulet CV, Shupe WL, Ross T, Richards W, 1986: Effects ofnutritional environment and ram effect on breed<strong>in</strong>g season<strong>in</strong> range sheep. Theriogenology 25, 317–323.Hunter GL, 1962: Observations on oestrus <strong>in</strong> Mer<strong>in</strong>os. Proc SAfr Soc Anim Prod 1, 67–69.Iglesias RMR, Ciccioli NH, Irazoqui H, Giglioli C, 1996:Ovulation rate <strong>in</strong> ewes after s<strong>in</strong>gle oral glucogenic dosagedur<strong>in</strong>g a ram-<strong>in</strong>duced follicular phase. Anim Reprod Sci 44,211–221.Kadokawa H, Mart<strong>in</strong> GB, 2006: A new perspective onmanagement of reproduction <strong>in</strong> dairy cows: the need fordetailed metabolic <strong>in</strong>formation, an improved selection <strong>in</strong>dexand extended lactation. J Reprod Dev 52, 161–168.Karsch FJ, Bittman EL, Foster DL, Goodman RL, Legan SJ,Rob<strong>in</strong>son JE, 1984: Neuroendocr<strong>in</strong>e basis of seasonalreproduction. Recent Prog Horm Res 40, 185–232.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Nutritional Interactions and <strong>Reproduction</strong> 135Knight TW, Oldham CM, L<strong>in</strong>dsay DR, 1975: Studies <strong>in</strong> ov<strong>in</strong>e<strong>in</strong>fertility <strong>in</strong> agricultural regions <strong>in</strong> Western Australia: the<strong>in</strong>fluence of a supplement of lup<strong>in</strong>s (Lup<strong>in</strong>us angustifolius cv.Uniwhite) at jo<strong>in</strong><strong>in</strong>g on the reproductive performance ofewes. Aust J Agric Res 26, 567–576.Lake JK, Power C, Cole TJ, 1997: Women’s reproductivehealth: the role of body mass <strong>in</strong>dex <strong>in</strong> early and adult life.Int J Obes Relat Metab Disord 21, 432–438.Lassoued N, Khaldi G, 1990: Influence du niveau alimentaireavant et apre` s la mise bas sur la brebis de race Barbar<strong>in</strong>e àl’effet mâle. Ann INRAT Tunisie 63, 1–16.Leury BJ, Murray PJ, Rowe JB, 1990: Effect of nutrition onthe response <strong>in</strong> ovulation arte <strong>in</strong> Mer<strong>in</strong>o ewes follow<strong>in</strong>gshort-term lup<strong>in</strong> supplementation and <strong>in</strong>sul<strong>in</strong> adm<strong>in</strong>istration.Aust J Agric Res 41, 751–759.Malpaux B, 2006: Seasonal regulation of reproduction <strong>in</strong>mammals. In: Neill JD (ed.), Knobil and Neill’s Physiologyof <strong>Reproduction</strong>, 3rd edn. Elsevier Academic Press,Amsterdam, pp. 2231–2281.Mart<strong>in</strong> GB, Kadokawa H, 2006: ‘Clean, green and ethical’animal production. Case study: reproductive efficiency <strong>in</strong>small rum<strong>in</strong>ants. J Reprod Dev 52, 145–152.Mart<strong>in</strong> GB, Milton JT, Davidson RH, Banchero HunzickerGE, L<strong>in</strong>dsay DR, Blache D, 2004a: Natural methods for<strong>in</strong>creas<strong>in</strong>g reproductive efficiency <strong>in</strong> small rum<strong>in</strong>ants. AnimReprod Sci 82–83, 231–245.Mart<strong>in</strong> GB, Rodger J, Blache D, 2004b: Nutritional andenvironmental effects on reproduction <strong>in</strong> small rum<strong>in</strong>ants.Reprod Fertil Dev 16, 491–501.Molle G, Branca A, Ligios S, Sitzia M, Casu S, Landau S,Zoref Z, 1995: Effect of graz<strong>in</strong>g background and flush<strong>in</strong>gsupplementation on reproductive performance <strong>in</strong> Sardaewes. Small Rum<strong>in</strong> Res 17, 245–254.Molle G, Landau S, Branca A, Sitzia M, Fois N, Ligios S,Casu S, 1997: Flush<strong>in</strong>g with soybean meal can improvereproductive performances <strong>in</strong> lactat<strong>in</strong>g Sarda ewes on amature pasture. Small Rum<strong>in</strong> Res 24, 157–165.Monje AR, Alberio R, Schiersmann G, Chedrese J, Carou N,Callejas S, 1992: Male effect on the post-partum sexualactivity of cows ma<strong>in</strong>ta<strong>in</strong>ed on two nutritional levels. AnimReprod Sci 29, 145–156.Montgomery GW, Scott IC, Hudson N, 1985: An <strong>in</strong>teractionbetween season of calv<strong>in</strong>g and nutrition on the resumptionof ovarian cycles <strong>in</strong> post-partum beef cattle. J Reprod Fertil73, 45–50.Nottle MB, Hynd PI, Seamark RF, Setchell BP, 1988:Increases <strong>in</strong> ovulation rate <strong>in</strong> lup<strong>in</strong>-fed ewes are <strong>in</strong>itiatedby <strong>in</strong>creases <strong>in</strong> prote<strong>in</strong> digested post-rum<strong>in</strong>ally. J ReprodFertil 84, 563–566.Nottle MB, Kleemann DO, Seamark RF, 1997a: Effect ofprevious undernutrition on the ovulation rate of mer<strong>in</strong>oewes supplemented with lup<strong>in</strong> gra<strong>in</strong>. Anim Reprod Sci 49,29–36.Nottle MB, Kleemann DO, Grosser TI, Seamark RF, 1997b:Evaluation of a nutritional strategy to <strong>in</strong>crease ovulationrate <strong>in</strong> Mer<strong>in</strong>o ewes mated <strong>in</strong> late spr<strong>in</strong>g-early summer.Anim Reprod Sci 47, 255–261.Oldham CM, L<strong>in</strong>dsay DR, Mart<strong>in</strong> GB, 1990: Effects ofseasonal variation of live weight on the breed<strong>in</strong>g activity ofMer<strong>in</strong>o ewes. In: Oldham CM, Mart<strong>in</strong> GB, Purvis IW(eds), Reproductive Physiology of Mer<strong>in</strong>o Sheep – Conceptsand Consequences. School of Agriculture (AnimalScience), The University of Western Australia, Perth, pp.41–58.Pearce DT, Mart<strong>in</strong> GB, Oldham CM, 1985: Corpora luteawith a short life-span <strong>in</strong>duced by rams <strong>in</strong> seasonallyanovulatory ewes are prevented by progesterone delay<strong>in</strong>gthe preovulatory surge of LH. J Reprod Fertil 75, 79–84.Rhodes FM, Fitzpatrick LA, Entwistle KW, K<strong>in</strong>der JE, 1995:Pulsatile hormone secretion dur<strong>in</strong>g the first ovarian follicularwave <strong>in</strong> Bos <strong>in</strong>dicus heifers. J Reprod Fertil Suppl 49,523–526.Roberson MS, Wolfe MW, Stumpf TT, Werth LA, Cupp AS,Kojima ND, Wolfe PL, Kittok RJ, K<strong>in</strong>der JE, 1991:Influence of growth rate and exposure to bulls on age atpuberty <strong>in</strong> beef heifers. J Anim Sci 69, 2092–2098.Salazar-Ortiz J, 2006: Bilan des effets du niveau d’alimentationsur la reproduction de la jument. Thèse pour obtenir legrade de Docteur de l, Universite´ de Tours, Universite´François Rabelais, Tours.Scaramuzzi RJ, Campbell BK, Down<strong>in</strong>g JA, Kendall NR,Khalid M, Mun˜ oz-Gutiérrez M, Somchit A, 2006: A reviewof the effects of supplementary nutrition <strong>in</strong> the ewe on theconcentrations of reproductive and metabolic hormones andthe mechanisms that regulate folliculogenesis and ovulationrate. Reprod Nutr Dev 46, 339–354.Smith ID, 1965: The <strong>in</strong>fluence of level of nutrition dur<strong>in</strong>gw<strong>in</strong>ter and spr<strong>in</strong>g upon oestrous activity <strong>in</strong> the ewe. WorldRev Anim Prod 4, 95–101.Stumpf TT, Wolfe MW, Wolfe PL, Day ML, Kittok RJ,K<strong>in</strong>der JE, 1992: Weight changes prepartum and presence ofbulls postpartum <strong>in</strong>teract to affect duration of postpartumanestrus <strong>in</strong> cows. J Anim Sci 70, 3133–3137.Szymanski LA, Schneider JE, Friedman MI, Ji H, Kurose Y,Blache D, Rao A, Dunshea FR, Clarke IJ, 2007: Changes <strong>in</strong><strong>in</strong>sul<strong>in</strong>, glucose and ketone bodies, but not lept<strong>in</strong> or body fatcontent precede restoration of lute<strong>in</strong>is<strong>in</strong>g hormone secretion<strong>in</strong> ewes. J Neuroendocr<strong>in</strong>ol 19, 449–460.Teleni E, Rowe JB, Croker KP, Murray PJ, K<strong>in</strong>g WR, 1989:Lup<strong>in</strong>s and energy-yield<strong>in</strong>g nutrients <strong>in</strong> ewes. II.Responses <strong>in</strong> ovulation rate <strong>in</strong> ewes to <strong>in</strong>creased availabilityof glucose, acetate and am<strong>in</strong>o acids. Reprod FertilDev 1, 117–125.Ungerfeld R, 2007: Socio-sexual signall<strong>in</strong>g and gonadalfunction: opportunities for reproductive management <strong>in</strong>domestic rum<strong>in</strong>ants. In: Juengel JI, Murray JF, Smith MF(eds), <strong>Reproduction</strong> <strong>in</strong> <strong>Domestic</strong> Rum<strong>in</strong>ants VI. Nott<strong>in</strong>ghamUniversity Press, Nott<strong>in</strong>gham, pp. 207–221.Ungerfeld R, Forsberg M, Rubianes E, 2004: Overview of theresponse of anoestrous ewes to the ram effect. Reprod FertilDev 16, 479–490.Véliz FG, Po<strong>in</strong>dron P, Malpaux B, Delgadillo JA, 2006:Positive correlation between the body weight of anestrusgoats and their response to the male effect with sexuallyactive bucks. Reprod Nutr Dev 46, 657–661.V<strong>in</strong>˜ oles C, Forsberg M, Mart<strong>in</strong> GB, Cajarville C, Repetto J,Meikle A, 2005: Short-term nutritional supplementation ofewes <strong>in</strong> low body condition affects follicle development dueto an <strong>in</strong>crease <strong>in</strong> glucose and metabolic hormones. <strong>Reproduction</strong>129, 299–309.Walker RF, Bethea CL, 1977: Gonadal function <strong>in</strong> underfedrats. I. Effect of p<strong>in</strong>eal gland and constant light onmaturation and fecundity. Biol Reprod 17, 623–629.Wang JX, Warnes GW, Davies MJ, Norman RJ, 2004:Overweight <strong>in</strong>fertile patients have a higher fecundity thannormal-weight women undergo<strong>in</strong>g controlled ovarianhyperstimulation with <strong>in</strong>trauter<strong>in</strong>e <strong>in</strong>sem<strong>in</strong>ation. FertilSteril 81, 1710–1712.Webb R, Garnsworthy PC, Gong JG, Armstrong DG, 2004:Control of follicular growth: local <strong>in</strong>teractions and nutritional<strong>in</strong>fluences. J Anim Sci 82 (E-Suppl), E63–74.Wolfe BE, 2005: Reproductive health <strong>in</strong> women witheat<strong>in</strong>g disorders. J Obstet Gynecol Neonatal Nurs 34,255–263.Yeates NTM, 1949: The breed<strong>in</strong>g season of the sheep withparticular reference to modification by artificial means us<strong>in</strong>glight. J Agric Sci (Camb) 39, 1–43.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


136 RJ Scaramuzzi and GB Mart<strong>in</strong>Youatt W, 1837: Sheep, Their Breeds, Management andDiseases. Baldw<strong>in</strong> & Craddock, London.Zarazaga LA, Guzma´n JL, Domı´nguez C, Pérez MC, PrietoR, 2005: Effect of plane of nutrition on seasonality ofreproduction <strong>in</strong> Spanish Payoya goats. Anim Reprod Sci87, 253–267.Author’s address (for correspondence): RJ Scaramuzzi, UMR Physiologiede la <strong>Reproduction</strong> et des Comportements, Centre INRA deTours, 37380 Nouzilly, France. E-mail: rscara@rvc.ac.ukConflicts of <strong>in</strong>terest: Much of GBM’s research is funded by Meat &Livestock, Australia; RJS has not declared any conflict of <strong>in</strong>terests.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 137–143 (2008); doi: 10.1111/j.1439-0531.2008.01153.xISSN 0936-6768Developmental Capabilities of Embryos Produced In Vitro from Prepubertal LambOocytesKM MortonThe Camel <strong>Reproduction</strong> Centre, Dubai, United Arab EmiratesContentsBreed<strong>in</strong>g from prepubertal females, known as juvenile <strong>in</strong> vitroembryo transfer (JIVET), reduces the generation <strong>in</strong>terval and<strong>in</strong>creases the rate of genetic ga<strong>in</strong> <strong>in</strong> animal breed<strong>in</strong>g programs.While the birth of the first lambs from prepubertal ewesoccurred nearly 30 years ago; and there is considerable <strong>in</strong>terest<strong>in</strong> the commercialization of this technology, its efficiencyrema<strong>in</strong>s too low. The advent of <strong>in</strong> vitro production (IVP) ofembryo resulted <strong>in</strong> the more widespread use of JIVET.Morphologic and metabolic differences coupled with reduced<strong>in</strong> vitro and <strong>in</strong> vivo development of oocytes derived fromprepubertal animals have been reported. Research has beenundertaken to optimize donor selection and hormone stimulationmethods <strong>in</strong> an attempt to reduce the variability and<strong>in</strong>crease the proportion of donors respond<strong>in</strong>g to hormonestimulation and <strong>in</strong>crease oocyte developmental competence.Yet, this variation persists and the development of oocytes andembryos from prepubertal animals rema<strong>in</strong>s reduced whencompared with adults. Recent improvements to JIVET,result<strong>in</strong>g from a modified hormone stimulation regime, haveelim<strong>in</strong>ated the failure of donors to respond to hormonestimulation, and <strong>in</strong>creased both the number and developmentalcompetence of oocytes harvested from very young prepubertallambs. This <strong>in</strong>creased efficiency has facilitated the<strong>in</strong>corporation of other reproductive technologies such assperm sex<strong>in</strong>g with JIVET, result<strong>in</strong>g <strong>in</strong> the birth of lambs of apre-determ<strong>in</strong>ed sex from prepubertal lambs. Increased rates ofgenetic ga<strong>in</strong> <strong>in</strong> sheep breed<strong>in</strong>g programs can be achieved bycomb<strong>in</strong><strong>in</strong>g sexed sperm with oocytes obta<strong>in</strong>ed from lambs asyoung as 3–4 weeks of age. Cont<strong>in</strong>ued <strong>in</strong>creases <strong>in</strong> theefficiency of JIVET result<strong>in</strong>g from further improvements tohormone stimulation regimes and an <strong>in</strong>creased understand<strong>in</strong>gof the differences between oocytes from adult and prepubertalanimals will result <strong>in</strong> the commercialization of this technology.IntroductionThe use of prepubertal ewes for breed<strong>in</strong>g programsprovides an excit<strong>in</strong>g opportunity for sheep producers toreduce the generation <strong>in</strong>terval and thereby, <strong>in</strong>crease therate of genetic ga<strong>in</strong> <strong>in</strong> their flocks (Nicholas 1996;Armstrong et al. 1997). This can be performed us<strong>in</strong>g aform of assisted reproductive technology, known asjuvenile <strong>in</strong> vitro embryo transfer (JIVET), to produceoffspr<strong>in</strong>g after the transfer of <strong>in</strong> vitro produced (IVP)embryos derived from oocytes obta<strong>in</strong>ed from prepubertal(juvenile) animals.Traditional sheep multiple ovulation and embryotransfer (MOET) schemes, which use adult ewes, result<strong>in</strong> a generation <strong>in</strong>terval of at least 12 months. Us<strong>in</strong>g 8–12-week-old lambs <strong>in</strong> a JIVET scheme, reduces thegeneration <strong>in</strong>terval to 7 months (van der Werf 2005).Us<strong>in</strong>g oocytes obta<strong>in</strong>ed from 3- to 4-week-old lambsfurther reduces this generation <strong>in</strong>terval to 6 months, andoffers producers a substantial <strong>in</strong>crease <strong>in</strong> the rate ofgenetic ga<strong>in</strong> when compared with other reproductivetechnologies such as MOET. Further <strong>in</strong>creases <strong>in</strong> therate of genetic ga<strong>in</strong> (of approximately 5%) can beachieved by comb<strong>in</strong><strong>in</strong>g sexed sperm with JIVET, whichis most advantageous at the commercial level (van derWerf 2005).Dur<strong>in</strong>g the early post-natal period <strong>in</strong> sheep, there isremarkable growth of the reproductive tract. Ovarianweight <strong>in</strong>creased to 6.8 and 10.8 times the weight at birthby 4 and 8 weeks of age, respectively, and the number ofgrow<strong>in</strong>g ovarian follicles <strong>in</strong>creased significantly by4 weeks of age (Kennedy et al. 1974). Both the numberof grow<strong>in</strong>g follicles and ovarian weight decl<strong>in</strong><strong>in</strong>g by 8–12 weeks of age rema<strong>in</strong>ed constant until 33 weeks of age(Kennedy et al. 1974). Ovarian follicles from 4-week-oldlambs began to show signs of atresia whilst the numberof follicles display<strong>in</strong>g signs of advanced atresia werehigher from 8- to 10-week-old lambs (Tassell et al. 1978).These results suggest this post-natal flourish of folliculargrowth provides a ‘w<strong>in</strong>dow of opportunity’ to collectlarge numbers of oocytes from non-atretic follicles whenlambs are aged between 3 and 8 weeks of age.Initial attempts to produce embryos from prepubertallambs were achieved us<strong>in</strong>g MOET and, while prepubertallambs were capable of respond<strong>in</strong>g to exogenousgonadotroph<strong>in</strong>s (Tassell et al. 1978; Worth<strong>in</strong>gton andKennedy 1979), the results were highly variable and thesuperovulatory response was generally low. In addition,embryos which were recovered from prepubertal lambsdisplayed a reduced development dur<strong>in</strong>g <strong>in</strong> vitro culture(Wright et al. 1976) and after transfer to recipients(Quirke and Hanrahan 1977; McMillan and McDonald1985) when compared with those obta<strong>in</strong>ed from adultdonors. Despite these difficulties, a lamb was successfullyborn after the transfer of embryos derived from 10-to 16-week-old Welsh Mounta<strong>in</strong> ewe lambs (Trounsonet al. 1977). The development of IVP, comb<strong>in</strong>ed withjuvenile oocytes resulted <strong>in</strong> the birth of a calf (Armstronget al. 1992) followed closely by the birth of lambs(Armstrong et al. 1994; Earl et al. 1994).The use of JIVET was considerably expanded by thedevelopment of IVP and <strong>in</strong>creased knowledge about thedifferences between oocytes from prepubertal and adultanimals. Despite considerable research, the efficiency ofJIVET has rema<strong>in</strong>ed below commercially acceptablelevels, and <strong>in</strong>creases <strong>in</strong> efficiency have been h<strong>in</strong>dered bythe high proportion of lambs which do not respond tohormone stimulation, the highly variable response tohormone stimulation, the low proportion of oocytes thatreach the blastocyst stage and the high rates of foetalloss and malformations observed after the transfer ofIVP embryos derived from prepubertal animals (PtakÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


138 KM Mortonet al. 1999). Increases <strong>in</strong> both the number and <strong>in</strong> vitrodevelopmental competence of oocytes derived from veryyoung donors have been reported recently (Mortonet al. 2005c). This improvement has been attributed tothe hormone stimulation regime and an optimized IVPsystem, and has resulted <strong>in</strong> the birth of offspr<strong>in</strong>g fromIVP embryos derived from prepubertal lambs as youngas 3–4 weeks of age (Morton et al. 2004a, 2005d) andthe first pre-sexed JIVET offspr<strong>in</strong>g (Morton et al.2004a). The current status of JIVET and a number offactors affect<strong>in</strong>g its efficiency <strong>in</strong>clud<strong>in</strong>g selection of thedonor, hormone stimulation prior to oocyte collectionas well as the developmental capabilities of prepubertaloocytes will be described.Donor SelectionThe ideal donor lamb will produce large numbers offollicles and developmentally competent oocytes <strong>in</strong>response to hormone stimulation. Prepubertal lambs,like their adult counterparts, display a wide variation <strong>in</strong>the number of ovarian follicles (both pre- and posthormonestimulation) and <strong>in</strong> the number of oocytescollected (O’Brien et al. 1997a; Morton et al. 2005b; c,d). Approximately, 20% of lambs failed to respond tohormone stimulation (Ptak et al. 1999, 2003), which issimilar to adult animals (Cognie 1999). Oocyte developmentto the blastocyst stage is highly dependant on<strong>in</strong>dividual lambs (Ptak et al. 2003; Morton et al. 2005c),with the number of embryos produced per lamb rang<strong>in</strong>gfrom 0 to 55 (Morton et al. 2005b, c). Thus, selection oflambs which produce large numbers of follicles andoocytes with a high developmental potential becomesparamount.Various methods have been studied for select<strong>in</strong>glambs although most have been <strong>in</strong>conclusive. Bodyweight prior to hormone stimulation and growth rateare not related to ovarian weight or the number ofovarian follicles after hormone stimulation (Mortonet al. 2005c). Donors have been selected on the basis ofovarian follicular status assessed by laparoscopy priorto hormone stimulation (Earl et al. 1995b); the numberof follicles observed was highly related (R = 0.98) tothe number post-hormone stimulation. Yet, this requirestwo laporscopic procedures with<strong>in</strong> a fortnight and thereis no correlation between the number of ovarian folliclesand the <strong>in</strong> vitro developmental capabilities of theseoocytes. Ptak et al. (2003) observed similarities betweenfive sibl<strong>in</strong>g lambs derived from a lamb with oocytes of ahigh developmental capability, highlight<strong>in</strong>g the role ofgenotype <strong>in</strong> oocyte developmental competence.Age of oocyte donorsArmstrong et al. (1997) suggest that the ideal age tocollect oocytes from prepubertal lambs is between 4 and6 weeks of age as this is the time of most follicularresponsiveness. Yet, <strong>in</strong> cattle oocytes obta<strong>in</strong>ed fromolder donors display an <strong>in</strong>creased response to hormonestimulation (Armstrong et al. 1992, 1994), oocyte development<strong>in</strong> vitro (Presicce et al. 1995; Tervit et al. 1997)and pregnancy rate (Yang et al. 1997) when comparedwith oocytes from younger animals. Earl et al. (1995a)reported similar rates of <strong>in</strong> vitro development for oocytesfrom 8- to 9-week-hormone stimulated and 4-monthunstimulatedlambs and O’Brien et al. (1997a) reportedthat development of oocytes <strong>in</strong> vitro was higher from3- to 6-week-old hormone stimulated lambs than 16–24-week-old unstimulated lambs. Yet, Morton et al.(2005d) observed similar rates of oocyte developmentto the blastocyst stage for oocytes derived from 16- to24-week-unstimulated prepubertal lambs (29.0%) andadult ewes (39.3%) but was reduced for oocytes derivedfrom 3- to 6-week-hormone stimulated lambs (27.8%).These studies compared the development of oocytesfrom younger hormone stimulated lambs with olderunstimulated lambs, thereby preclud<strong>in</strong>g the identificationof the relative contributions of lamb age andhormone stimulation.Compar<strong>in</strong>g hormone stimulated lambs of differ<strong>in</strong>gages (3–4 and 6–7 weeks), Morton et al. (2005b)observed that lamb age affected uter<strong>in</strong>e weight, whileovarian weight, the number of ovarian follicles, thenumber of oocytes recovered or the proportion ofoocytes suitable for IVP were not affected. Yet, oocytedevelopment to the blastocyst stage was higher for 6–7 week (27.9%) than 3–4 week (20.4%) lambs demonstrat<strong>in</strong>gthe <strong>in</strong>crease <strong>in</strong> oocyte development with donorage. Despite these results and those of Morton et al.(2005d), the greater follicular responsiveness and numberof oocytes obta<strong>in</strong>ed from younger lambs (4–6 weeksof age) outweights the <strong>in</strong>crease <strong>in</strong> development whenoocytes are obta<strong>in</strong>ed from older lambs agree<strong>in</strong>g withArmstrong et al. (1997).In summary, a w<strong>in</strong>dow of opportunity which existswhen lambs are aged between 3 and 6 weeks, to collectlarge numbers of oocytes which are capable of fulldevelopment <strong>in</strong> vitro and <strong>in</strong> vivo. Yet, the lack of efficientmethods for select<strong>in</strong>g oocyte donors lambs prevents fullexploitation of this opportunity and further research isrequired to develop an efficient method of select<strong>in</strong>g 3–6-week-old lambs for JIVET.Hormone Stimulation of DonorsHormone stimulation prior to oocyte collection is usedto <strong>in</strong>crease the number of ovarian follicles and oocytesharvested (O’Brien et al. 1997a; Ledda et al. 1999;Morton et al. 2005b), collect oocytes at a synchronousstage of development (Armstrong et al. 1997) and<strong>in</strong>crease oocyte development <strong>in</strong> vitro (O’Brien et al.1997a; Morton et al. 2005b).While the precise mechanisms responsible for this<strong>in</strong>crease <strong>in</strong> developmental competence are not known,the effects are not mediated solely through an <strong>in</strong>crease <strong>in</strong>follicle size, but also by the activation of biosyntheticprocesses with<strong>in</strong> the oocyte that are required forsuccessful embryo development (Armstrong 2001). Biochemicalchanges <strong>in</strong> the follicle cells and <strong>in</strong>trafollicularenvironment that would not have otherwise occurreduntil after puberty are also <strong>in</strong>duced by hormonetreatment (O’Brien et al. 1997a), for example, <strong>in</strong>creas<strong>in</strong>gthe production of gonadotroph<strong>in</strong> receptors <strong>in</strong> granulosacells result<strong>in</strong>g <strong>in</strong> an enhanced ability of the follicle torespond to gonadotroph<strong>in</strong> stimulation, thereby improv<strong>in</strong>goocyte quality and function.Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Developmental Capabilities of Prepubertal Lamb Embryos 139Most researchers do <strong>in</strong>clude hormone stimulation <strong>in</strong>their JIVET regimes for lambs (Ptak et al. 1999, 2003,2006; Kelly et al. 2005a,b, 2007; Morton et al. 2005a; b,c, d) although Ledda et al. (1999) reported no difference<strong>in</strong> developmental competence of oocytes from unstimulatedor stimulated oocyte donors. The birth ofoffspr<strong>in</strong>g after <strong>in</strong> vitro maturated-<strong>in</strong> vitro fertilised(IVM-IVF) of oocytes from untreated 4–6-week-oldlambs (Ledda et al. 1999) demonstrates that hormonestimulation is not mandatory for such young donorsalthough it is usually applied for the aforementionedreasons.Considerable research has been undertaken to optimizethe regime for hormone stimulation of prepubertallambs. Hormone stimulation regimes used with prepubertalanimals have been based on adult superovulationregimes and consist of treatment over 3–4 days withFSH (s<strong>in</strong>gle or multiple doses) or gonadotroph<strong>in</strong>s with along half-life [i.e. equ<strong>in</strong>e chorionic gonadotroph<strong>in</strong>(eCG); Armstrong et al. 1997].S<strong>in</strong>gle vs multiple <strong>in</strong>jections of FSH did not affect thenumber of oocytes recovered and cultured per lamb(Armstrong et al. 1994; Ptak et al. 1999). Yet, multipleFSH <strong>in</strong>jections <strong>in</strong>creased oocyte development to theblastocyst stage (Ptak et al. 1999). Furthermore, blastocystdevelopment was <strong>in</strong>creased when oocytes werecollected 48 and 72 h after the <strong>in</strong>itial FSH <strong>in</strong>jection andlambs received multiple FSH <strong>in</strong>jections when comparedwith a s<strong>in</strong>gle FSH <strong>in</strong>jection (Kelly et al. 2005b).Hormone stimulation of prepubertal donors hasrelied on lower doses of gonadotroph<strong>in</strong>s than adultsbecause of their lower body mass and greater follicularresponsiveness before the <strong>in</strong>traovarian regulatory mechanismsdesigned to limit ovulation rate have come <strong>in</strong>toplay (Armstrong et al. 1997). Given that oocytes derivedfrom prepubertal animals have a relatively lower exposureto gonadotroph<strong>in</strong>s when compared with oocytesderived from adults, and exposure to <strong>in</strong>creased FSHdur<strong>in</strong>g IVM <strong>in</strong>creases oocyte developmental competence<strong>in</strong> vitro (Morton et al. 2004b), larger gonadotroph<strong>in</strong>doses may confirm advantageous for cytoplasmic maturationof prepubertal oocytes. Recently, the use oflarger doses of FSH (130–160 mg) has resulted <strong>in</strong> an<strong>in</strong>crease <strong>in</strong> the efficiency of JIVET (Morton et al. 2004a,2005a; b, c, d; Kelly et al. 2005b, 2007).The <strong>in</strong>clusion of eCG <strong>in</strong> the regime did not <strong>in</strong>creasethe number of oocytes collected but oocyte developmentto the blastocyst stage was <strong>in</strong>creased (42.4% vs 43.9% ofoocytes; Kelly et al. 2005b) as did the adm<strong>in</strong>istration ofeCG with the last FSH <strong>in</strong>jection (29.6% vs 18.9%; Kellyet al. 2005b). Yet, Morton et al. (2005a,b,c,d) reportedsimilar rates of oocyte development <strong>in</strong> vitro when eCGwas adm<strong>in</strong>istered with first FSH <strong>in</strong>jection.Hormone stimulation regimes consist<strong>in</strong>g of bothsteroids and gonadotroph<strong>in</strong>s are known to <strong>in</strong>crease theyield and <strong>in</strong> vitro development of oocytes (O’Brien et al.1997a; Morton et al. 2005b) although the relativecontribution of steroids to this <strong>in</strong>crease rema<strong>in</strong>sunknown and the majority of regimes do not utilizesteroids. Progestagen treatment for 7 days, followed byits withdrawal, resulted <strong>in</strong> a twofold <strong>in</strong>crease <strong>in</strong> thenumber of ovarian follicles when compared with calvesand lambs where progestagen was not withdrawn(Armstrong et al. 1994), suggest<strong>in</strong>g that folliculargrowth is suppressed <strong>in</strong> the presence of progestagen.Yet, oocytes subsequently collected after progestagenwithdrawal were observed to be at different stages ofmaturation suggest<strong>in</strong>g that the <strong>in</strong>crease <strong>in</strong> the number ofovarian follicles may be negated by a reduction <strong>in</strong>oocyte quality.While, progesterone treatment did not affect thenumber of oocytes collected, the efficiency of blastocystproduction dependent on the time of oocyte collection(48 h vs 60 h) and type of FSH adm<strong>in</strong>istration(1 · 160 mg dose vs 4 · 40 mg dose; Kelly et al.2005b). These authors reported the highest oocytedevelopment to the blastocyst stage after no progesteronetreatment, four 40 mg <strong>in</strong>jections of FSH, andcollection of oocytes 48 h after the last FSH treatment.While these results suggest that progesterone treatmentis not necessary, it has been successfully used byprevious authors (O’Brien et al. 1997a; Ptak et al.1999, 2003; Morton et al. 2004a, 2005a; b, c, d) withhigh ovarian responses reported (approximately 120–140 follicles per lamb; Morton et al. 2005b).GnRH treatment of lambs prior to oocyte collection,to facilitate the collection of <strong>in</strong> vivo matured oocytes, didnot alter the number of oocytes collected, oocytemorphology or development to the blastocyst stage(Kelly et al. 2007). Previously, Armstrong et al. (1997)adm<strong>in</strong>istered GnRH to lambs and reported difficulty <strong>in</strong>accurately controll<strong>in</strong>g the tim<strong>in</strong>g of the endogenous LHsurge and asynchronous stages of maturity both with<strong>in</strong>and between donors for the oocytes collected. For thesereasons, GnRH is not rout<strong>in</strong>ely <strong>in</strong>cluded <strong>in</strong> hormonestimulation regimes as most researchers aim to collectimmature oocytes and mature them <strong>in</strong> vitro.In summary, hormone stimulation <strong>in</strong>creases thenumber of oocytes collected as well as oocyte developmentalcompetence, but the optimal regime for hormonestimulation of prepubertal lambs and the effects of therelative components of hormone stimulation regime onoocyte developmental competence have yet to be determ<strong>in</strong>ed.Response to hormone stimulationThe ovarian response to hormone stimulation variesconsiderably between <strong>in</strong>dividual prepubertal donors(Earl et al. 1994; Ptak et al. 1999, 2003). Ptak et al.(2003) found that 50–72% of lambs aged 2–7 monthsdid not have a sufficient response to hormone stimulation(number and size of follicles) for embryo production.Morton et al. (2005b), on the contrary, observedthat all lambs responded to hormone stimulation and24.0% (6 ⁄ 25), 44.0% (11 ⁄ 25) and 32.0% (8 ⁄ 25) of 3–4-week-old lambs had low (100) responses, respectively,and a similar distribution was observed for 6–7-week-old lambs.Ptak et al. (1999, 2003) observed that oocytes fromlambs with a low response to hormone stimulation didnot develop to the blastocyst stage <strong>in</strong> vitro. On thecontrary, Morton et al. (2005b) reported that oocytesfrom low respond<strong>in</strong>g 3–4 and 6–7-week-old lambs werecapable of full development <strong>in</strong> vitro. Furthermore, whileÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


140 KM Mortonthe number of oocytes suitable for IVP was reduced tolow when compared with medium and high respond<strong>in</strong>glambs, the percentage of oocytes suitable for culture (%of total recovered) did not differ between low andmedium respond<strong>in</strong>g lambs, but was reduced <strong>in</strong> highrespond<strong>in</strong>g lambs (Morton et al. 2005b). Oocyte cleavageand blastocyst formation were not affected byresponse to hormone stimulation <strong>in</strong> 3–4-week-oldlambs, but oocytes from 6- to 7-week-old lambs with ahigh response to hormone stimulation displayed reducedrates of cleavage (Morton et al. 2005b). It rema<strong>in</strong>s to beseen whether this result was related to <strong>in</strong>dividual lambs,or the early development of the negative feedbackmechanisms normally established after puberty to limitovulation rate.The effect of hormone stimulation regime on thevariation <strong>in</strong> response to stimulation rema<strong>in</strong>s unknown.Most authors report considerable variation <strong>in</strong> theresponse to hormone stimulation regardless of theregime used. This variation is also accompanied by ahigh proportion of lambs which do not respond tostimulation and by poor <strong>in</strong> vivo survival of embryos.Yet, a modified regime, consist<strong>in</strong>g of progestagenprim<strong>in</strong>g, steroids and gonadotroph<strong>in</strong>s (eCG and FSH)yielded a high average response to stimulation and high<strong>in</strong> vivo survival of IVP embryos (Morton et al. 2004a,2005a, b, c, d).In summary, modifications to hormone stimulationregimes have reduce the proportion of prepubertallambs which do not respond to stimulation, thereby<strong>in</strong>creas<strong>in</strong>g the efficiency of JIVET. Us<strong>in</strong>g the modifiedhormone stimulation regime, the majority of lambsdisplayed a medium to high response to hormonestimulation which does not reduce oocyte development<strong>in</strong> vitro for 3–4-week-old lambs. Despite these improvements,the variation <strong>in</strong> response to hormone stimulationbetween lambs persists.Morphologic and Metabolic Characteristics ofPrepubertal OocytesMorphological differences have been observed <strong>in</strong> theorganelles of adult and prepubertal oocytes. Oocytesfrom prepubertal animals are smaller than those derivedfrom adult animals but <strong>in</strong> GV stage oocytes derivedfrom prepubertal and adult animals, the morphologywas similar for cytoplasmic organelles such as themitochondria, Golgi complexes, endoplasmic reticulaand lipid droplets (Ledda et al. 1997). Yet, oocytesderived from prepubertal compared with adult ewesconta<strong>in</strong>ed a lower number of corona cell foot projections(Ledda et al. 2001) and there was a delay <strong>in</strong> themigration of the cortical granules <strong>in</strong> IVM oocytes(O’Brien et al. 1996).Prote<strong>in</strong> synthesis dur<strong>in</strong>g oocyte maturation, which isdependant on the correct coupl<strong>in</strong>g of the cumulus cellsand the oocyte, is essential for normal meiotic progressionand embryo development (Moor and Crosby 1986).Lower am<strong>in</strong>o acid uptake (Ledda et al. 1996b, 2001;Kochhar et al. 2002) and prote<strong>in</strong> synthesis (Kochharet al. 2002) dur<strong>in</strong>g IVM, and different times of peakprote<strong>in</strong> synthesis have been reported for prepubertalwhen compared with adult oocytes (Kochhar et al.2002), and is related to the defective cumulus cell-oocytecoupl<strong>in</strong>g reported by Ledda et al. (1996b, 2001). Inaddition, the level and pattern of maturation promot<strong>in</strong>gfactor (MPF) activity was similar <strong>in</strong> adult and prepubertaloocytes but after maturation, MPF activity waslower <strong>in</strong> oocytes from prepubertal than adult animals(Ledda et al. 2001).Metabolic differences between adult and prepubertaloocytes have also been reported. Glutam<strong>in</strong>e metabolismdur<strong>in</strong>g IVM was lower for prepubertal than adultoocytes while glucose and pyruvate metabolism wassimilar (O’Brien et al. 1996). These results po<strong>in</strong>t tomorphologic, metabolic and ultrastructural differencesbetween oocytes from adult and prepubertal animals,although it must be remembered that oocytes used <strong>in</strong>these studies were obta<strong>in</strong>ed from both unstimulated andhormone stimulated prepubertal lambs, rang<strong>in</strong>g <strong>in</strong> agefrom 40 days to 6 months, which may have <strong>in</strong>fluencedthe f<strong>in</strong>d<strong>in</strong>gs.In summary, there are clear morphological and metabolicdifferences between oocytes derived from adultand prepubertal animals. Yet, comparisons betweenstudies are confounded by differ<strong>in</strong>g ages of the prepubertaldonors (rang<strong>in</strong>g from days to many months) andwhether the donors were hormone stimulated prior tooocyte collection. The extend of these differences rema<strong>in</strong>sunclear and studies describ<strong>in</strong>g the changes <strong>in</strong> morphologicand metabolic characteristics of oocytes from veryyoung (3–4 week) to older (16–24 week) are required.Development of Oocytes from PrepubertalanimalsCompared with adult oocytes, those from prepubertaldonors display similar rates of meiotic maturation(O’Brien et al. 1996, 1997b; Ledda et al. 2001) but ahigher prevalence of fertilization abnormalities (Leddaet al. 1996a, 1997; O’Brien et al. 1996, 1997b; Kochharet al. 2002) and lower development to the blastocyststage dur<strong>in</strong>g <strong>in</strong> vitro culture (O’Brien et al. 1996, 1997b;Ptak et al. 1999; Kochhar et al. 2002; Leoni et al.2006b), attributed to perturbed cytoplasmic maturation(discussed below).Blastocysts produced <strong>in</strong> vitro, derived from prepubertaloocytes, are morphologically <strong>in</strong>dist<strong>in</strong>guishable(O’Brien et al. 1997b), conta<strong>in</strong> similar numbers of cells(O’Brien et al. 1996; Ledda et al. 1997; Kochhar et al.2002; Leoni et al. 2006b) and have a similar <strong>in</strong>ner cellmass to total cell number ratio (Kochhar et al. 2002;Leoni et al. 2006b), when compared with those derivedfrom adult oocytes. Yet, altered speed of developmenthas been reported for oocytes derived from prepubertalsheep. Kochhar et al. (2002) reported altered k<strong>in</strong>etics ofIVM, observ<strong>in</strong>g that lamb oocytes required an additional2 h of IVM to ‘catch up’, but other studies havereported no difference (O’Brien et al. 1996; Ledda et al.1997). Delays <strong>in</strong> development of embryos derived fromprepubertal oocytes have been observed previously atthe two-cell (Leoni et al. 2006b), four-cell (Ptak et al.1999) and blastocyst stage (O’Brien et al. 1997b; Ptaket al. 1999; Leoni et al. 2006b). Yet, Morton et al.(2005d) reported no difference <strong>in</strong> the speed of embryodevelopment which may be related to higher FSH dosesÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Developmental Capabilities of Prepubertal Lamb Embryos 141dur<strong>in</strong>g hormone stimulation, or differences <strong>in</strong> therespective culture systems, <strong>in</strong>clud<strong>in</strong>g media (KM Mortonand SL Catt, unpublished data) and oxygen tension(Leoni et al. 2007).The k<strong>in</strong>etics of both oocyte maturation (Dom<strong>in</strong>ko andFirst 1997) and embryo development (Van Soom et al.1997) <strong>in</strong>dicate the developmental capabilities of embryos.The reduced speed of development of prepubertal derivedembryos may expla<strong>in</strong> the low, or reduced <strong>in</strong> vivo survivalreported by numerous workers (Quirke and Hanrahan1977; McMillan and McDonald 1985; O’Brien et al.1997a; Ptak et al. 1999, 2003, 2006; Kelly et al. 2005b). Inaddition to this low survival rate, remarkably high ratesof embryonic ⁄ foetal loss (O’Brien et al. 1997a; Ptak et al.1999, 2003, 2006) and mummification (Ptak et al. 1999)have been reported. Ptak et al. (2006) reported severely<strong>in</strong>terrupted pregnancy (between days 40 and 60) andfoetal losses (days 80–100) with only 6% of the 628embryos transferred surviv<strong>in</strong>g until full gestation.This reduced development is generally attributed to<strong>in</strong>complete or perturbed cytoplasmic maturation, whichmay be expressed at many stages of embryonic development.Perturbed cytoplasmic maturation commonlypresents as a failure of sperm penetration and decondensation,<strong>in</strong>ability to form normal male pronuclei,failure of the block to polyspermy, early cleavagefailure, failure to reach or survive the transition frommaternal to embryonic genome expression, and developmentalfailure lead<strong>in</strong>g to embryonic losses at laterpre-implantation and post-implantation stages of development(Armstrong 2001). All of these have beenreported for oocytes and embryos derived from prepubertalanimals, and the evidence regard<strong>in</strong>g perturbedcytoplasmic maturation <strong>in</strong> prepubertal oocytes is nowoverwhelm<strong>in</strong>g.The perturbed cytoplasmic maturation is generallyattributed to the age of the donors and the lack of timeto sequester factors <strong>in</strong>to the cytoplasm (Armstrong2001). Yet, Ptak et al. (2006) also demonstrated <strong>in</strong>completenuclear maturation of prepubertal lamb oocytes,and the authors suggest that the previous def<strong>in</strong>ition ofnuclear competence (i.e. the ability to complete meiosis)was <strong>in</strong>adequate. Furthermore, the authors observeddiscordance between follicle and oocyte growth, lead<strong>in</strong>gto <strong>in</strong>complete or perturbed oocyte growth. This, accord<strong>in</strong>gto Ptak et al. (2006), resulted <strong>in</strong> <strong>in</strong>complete developmentof both the nuclear and cytoplasmiccompartments, which was responsible for the reduceddevelopmental competence of oocytes derived fromprepubertal animals.Reduction <strong>in</strong> global genome methylation (Ptak et al.2006) and mRNA storage (Leoni et al. 2006a) havebeen reported for oocytes derived from prepubertallambs. Ptak et al. (2006) observed that genome-widemethylation status (% of methylated vs total DNA)was lower for oocytes derived from prepubertal lambswhen compared with adult animals. The authorshypothesize that the reduced global methylation affectscerta<strong>in</strong> impr<strong>in</strong>ted genes as the patterns of foetal lossobserved by Ptak et al. (1999, 2006) are similar to micelack<strong>in</strong>g oocyte-specific DNA methyltransferase-1(Dnmt1o) where the majority of foetus die <strong>in</strong> the lastthird of gestation (Howell et al. 2001). The reducedmethylation, mRNA storage and perturbed growth ofoocytes from prepubertal animals, provides strongevidence for the contribution of nuclear immaturityto their reduced developmental competence. Furtherstudies aim<strong>in</strong>g to locate the specific sites where thedifferences <strong>in</strong> methylation occur will provide molecularexplanations for the differences <strong>in</strong> the developmentalcapabilities of oocytes from adult and prepubertalanimals.Most workers, most notably Ptak et al. (1999, 2006),have reported low rates of <strong>in</strong> vitro development forprepubertal oocytes accompanied by substantial embryonicand foetal loss. In other studies, by contrast,prepubertal oocytes have displayed a high developmentalcompetence <strong>in</strong> vitro not dissimilar to that of oocytesfrom adult sheep, suggest<strong>in</strong>g that the use of large FSHdoses dur<strong>in</strong>g hormone stimulation may attenuate manyof the problems associated with JIVET, specifically thehigh proportion of lambs which fail to respond tohormone stimulation, high rates of fertilization abnormalities,reduced and delayed development to the blastocyststage and compromised embryo survival aftertransfer to recipient ewes (Morton et al. 2005b; c, d).While Ptak et al. (1999, 2006) and Morton et al.(2005b,c,d) utilized oocytes from lambs of a similar age(4 weeks), the different breeds of the donors, hormonestimulation regimes (<strong>in</strong> particular, the FSH dose: 2.7 or7 mg and 130 mg pFSH, respectively) and IVP systemsconfound comparisons between these studies. The role of<strong>in</strong> vitro culture on the perturbation of embryonic geneexpression is well-documented (Wrenzycki et al. 2005).Yet, the effects of different IVC systems and hormonestimulation with a high FSH regime dur<strong>in</strong>g oocytegrowth on gene expression, have yet to be <strong>in</strong>vestigatedfor oocytes from prepubertal animals.Despite the reduced developmental competence ofoocytes from prepubertal animals, offspr<strong>in</strong>g have beenproduced from both fresh and frozen-thawed embryosderived from lambs as young as 3–4 weeks of age(Morton et al. 2004a, 2005d) and the improvements tothe efficiency of JIVET technology have facilitated the<strong>in</strong>corporation of JIVET with other emerg<strong>in</strong>g reproductivetechnologies such as sperm sex<strong>in</strong>g (Maxwell et al.2004) for the production of pre-sexed embryos andoffspr<strong>in</strong>g.Future Directions and PerspectivesLimitations to the commercial application of JIVEThave <strong>in</strong>cluded the high proportion of donors that fail torespond to hormone stimulation, the highly variableresponse to hormone stimulation and the reduceddevelopmental competence of oocytes derived fromprepubertal animals. Us<strong>in</strong>g a high FSH stimulationregime (Morton et al. 2004a, 2005a, b, c, d) all lambsresponded to hormone stimulation and oocyte developmentalcompetence was significantly improved. Developmentrate was similar for oocytes derived fromprepubertal and adult ewes, and <strong>in</strong> vivo survival afterembryo transfer was high and free from the highmalformation rates reported by other authors.The advent of genomic technologies has further<strong>in</strong>creased our knowledge on the differences betweenÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


142 KM Mortonoocytes derived from adult and prepubertal animals.Alterations <strong>in</strong> the expression of several developmentallyimportant genes have been observed <strong>in</strong> embryos derivedfrom prepubertal cattle (Oropeza et al. 2004) and sheep(Leoni et al. 2006a). Gene expression and epigeneticstudies will undoubtedly elucidate differences <strong>in</strong> oocytesand embryos derived from adult and prepubertalanimals. Furthermore, this technology will be used tore-evaluate hormone stimulation and IVP systems, sothat prepubertal oocytes and embryos may haveimproved competence.Two new techniques have great potential for comb<strong>in</strong>ationwith JIVET. Further reductions <strong>in</strong> the generation<strong>in</strong>terval on the female side could be achieved by <strong>in</strong>corporat<strong>in</strong>gfoetal oocytes <strong>in</strong>to IVP systems. Yet, foetal calfoocytes display lower rates of maturation, fertilization,cleavage and embryo development compared with oocytesfrom adult animals (Chohan and Hunter 2004) andfurther research is required. Utilization of gametes fromprepubertal male animals could dramatically reduce thegeneration <strong>in</strong>terval on the male side. Spermatogenesiscan be <strong>in</strong>itiated <strong>in</strong> prepubertal ram lambs by adm<strong>in</strong>ister<strong>in</strong>geCG (Morton et al. 2004c). The <strong>in</strong>jection of gametesfrom prepubertal ram lambs <strong>in</strong>to IVM oocytes fromadult and prepubertal lambs resulted <strong>in</strong> the production of16–32 cell stage embryos (KM Morton, SL Catt, WMCMaxwell and G Evans, unpublished data). This technologyoffers, for the first time, a chance for producers toreduce the generation <strong>in</strong>terval on the paternal side. Whilethe potential of this method has been demonstrated, thebirth of lambs produced from both prepubertal ram andewe lambs, and further ref<strong>in</strong>ement of treatment regimesfor ram lambs are required.In conclusion, oocytes from prepubertal animalsdisplay a reduced developmental competence whencompared with oocytes derived from their adult counterparts,result<strong>in</strong>g from perturbations to cytoplasmic andnuclear maturation. Despite these perturbations, oocytesfrom prepubertal lambs as young as 3–4 weeks of age arecapable of develop<strong>in</strong>g <strong>in</strong>to viable offspr<strong>in</strong>g after transferto recipients. Recent advances <strong>in</strong> JIVET technology,which have significantly improved its efficiency, havebrought its commercialization closer and facilitated<strong>in</strong>corporation with other emerg<strong>in</strong>g reproductive technologiessuch as sperm sex<strong>in</strong>g. With the developments <strong>in</strong>genomic technologies cont<strong>in</strong>u<strong>in</strong>g to add to knowledgeregard<strong>in</strong>g the differences between oocytes derived fromadult and prepubertal animals and the improvements <strong>in</strong>the efficiency of JIVET this will br<strong>in</strong>g, commercializationof this technology is foreseeable.AcknowledgementsThe Australian Research Council (ARC), XY Inc, Bioniche AnimalHealth Australasia and Sydney IVF for fund<strong>in</strong>g and research support.Professor W.M.C. Maxwell is thanked for his <strong>in</strong>valuable editorialassistance.ReferencesArmstrong DT, 2001: Effects of maternal age on oocytedevelopmental competence. Theriogenology 55, 1303–1322.Armstrong DT, Holm P, Irv<strong>in</strong>e B, Petersen BA, Stubb<strong>in</strong>gs RB,McLean D, Stevens R, Seamark RF, 1992: Pregnancies andlive birth from <strong>in</strong> vitro fertilization of calf oocytes collectedby laparoscopic follicular aspiration. Theriogenology 38,667–678.Armstrong DT, Irv<strong>in</strong>e BJ, Earl CR, 1994: In vitro fertilisationof follicular oocytes from juvenile lambs and their developmentalcompetence <strong>in</strong> vitro and <strong>in</strong> vivo. Biol Reprod50(Suppl.), 189.Armstrong DT, Kotaras PJ, Earl CR, 1997: Advances <strong>in</strong> theproduction of embryos <strong>in</strong> vitro from juvenile and prepubertaloocytes from the calf and lamb. Reprod Fertil Dev 9,333–339.Chohan KR, Hunter AG, 2004: In vitro maturation, fertilizationand early cleavage rates of bov<strong>in</strong>e fetal oocytes.Theriogenology 61, 373–380.Cognie Y, 1999: State of the art <strong>in</strong> sheep-goat embryo transfer.Theriogenology 51, 105–116.Dom<strong>in</strong>ko T, First NL, 1997: Tim<strong>in</strong>g of meiotic progression <strong>in</strong>bov<strong>in</strong>e oocytes and its effect of early embryonic development.Mol Reprod Dev 47, 456–467.Earl CR, Irv<strong>in</strong>e B, Armstrong DT, 1994: Development oftechniques for the production of viable embryos from sixto seven week old lambs. Proc Aust Soc Anim Prod 20,428.Earl CR, Irv<strong>in</strong>e BJ, Kelly JM, Rowe J, Armstrong DT, 1995a:Ovarian stimulation protocols for oocyte collection and<strong>in</strong> vitro embryo production from 8 to 9 week old lambs.Theriogenology 43, 203.Earl CR, Kotaras PJ, Rowe J, Kelly JM, DeBarro TM, 1995b:Laparoscopic selection of juvenile donors prior to stimulation.Proc Aust Soc Reprod Biol 26, 2.Howell CY, Bestor TH, D<strong>in</strong>g F, Latham KE, Mert<strong>in</strong>eit C,Trasler JM, Chaillet JR, 2001: Genomic impr<strong>in</strong>t<strong>in</strong>g disruptedby a maternal effect mutation <strong>in</strong> the Dnmt1 gene.Cell 104, 829–838.Kelly JM, Kleeman DO, Walker SK, 2005a: The effect ofnutrition dur<strong>in</strong>g pregnancy on the <strong>in</strong> vitro production ofembryos from result<strong>in</strong>g lambs. Theriogenology 63, 2020–2031.Kelly JM, Kleeman DO, Walker SK, 2005b: Enhancedefficiency <strong>in</strong> the production of offspr<strong>in</strong>g from 4- to 8-weekoldlambs. Theriogenology 63, 1876–1890.Kelly JM, Kleeman DO, Maxwell WMC, Walker SK, 2007:Effect of GnRH treatment on the maturation and <strong>in</strong> vitrodevelopment of oocytes collected from 4- to 6-week-oldMer<strong>in</strong>o lambs. Reprod Fertil Dev 19, 947–953.Kennedy JP, Worth<strong>in</strong>gton CA, Cole ER, 1974: The postnataldevelopment of the ovary and uterus <strong>in</strong> the Mer<strong>in</strong>o lamb. JReprod Fertil 36, 275–282.Kochhar HP, Wu B, Morris LH, Buckrell BC, Pollard JW,Basrur PK, K<strong>in</strong>g WA, 2002: Maturation status, prote<strong>in</strong>synthesis and developmental competence of oocytes derivedfrom lambs and ewes. Reprod Domest Anim 37, 19–25.Ledda S, Bogliolo L, Calvia P, Leoni G, Naitana S, 1996a:Developmental competence of follicular oocytes from juvenilelambs matured <strong>in</strong> vitro <strong>in</strong> different conditions. J ReprodFertil Abstr Ser 17, 28.Ledda S, Bogliolo L, Leoni G, Calvia P, Naitana S, 1996b:Influence of vasoactive <strong>in</strong>test<strong>in</strong>al peptide (VIP), atrialnatiuretic peptide (ANP) and <strong>in</strong>sul<strong>in</strong>-like growth factors-I(IGF-I) on <strong>in</strong> vitro maturation of prepubertal and adultsheep oocytes. Zygote 4, 343–348.Ledda S, Bogliolo L, Calvia P, Leoni G, Naitana S, 1997:Meiotic progression and developmental competence ofoocytes collected from juvenile and adult ewes. J ReprodFertil 109, 73–78.Ledda S, Bogliolo L, Leoni G, Naitana S, 1999: Productionand lamb<strong>in</strong>g rate of blastocysts derived from <strong>in</strong> vitromatured oocytes after gonadotrop<strong>in</strong> treatment of prepubertalewes. J Anim Sci 77, 2234–2239.Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Developmental Capabilities of Prepubertal Lamb Embryos 143Ledda S, Bogliolo L, Leoni G, Naitana S, 2001: Cell coupl<strong>in</strong>gand maturation-promot<strong>in</strong>g factor activity <strong>in</strong> <strong>in</strong> vitromaturedprepubertal and adult sheep oocytes. Biol Reprod65, 247–252.Leoni GG, Bebbere D, Succu S, Berl<strong>in</strong>guer F, Mossa F,Galioto M, Bogliolo L, Ledda S, Naitana S, 2006a:Relations between relative mRNA abundance and developmentalcompetence of ov<strong>in</strong>e oocytes. Mol Reprod Dev 74,249–257.Leoni GG, Succu S, Berl<strong>in</strong>guer F, Rosati I, Bebbere D,Bogliolo L, Ledda S, Naitana S, 2006b: Delay on the <strong>in</strong> vitrok<strong>in</strong>etic development of prepubertal ov<strong>in</strong>e embryos. AnimReprod Sci 92, 373–383.Leoni GG, Rosati I, Succu S, Bogliolo L, Bebbere L,Berl<strong>in</strong>guer F, Ledda S, Naitana S, 2007: A low oxygenatmosphere dur<strong>in</strong>g IVF accelerates the k<strong>in</strong>etic of formationof <strong>in</strong> vitro produced ov<strong>in</strong>e blastocysts. Reprod DomestAnim 42, 299–304.Maxwell WMC, Evans G, Holl<strong>in</strong>shead FK, Bathgate R, deGraaf SP, Eriksson B, Gillan L, Morton KM, O’Brien JK,2004: Integration of sperm sex<strong>in</strong>g <strong>in</strong>to the ART toolbox.Anim Reprod Sci 82–83, 79–95.McMillan WH, McDonald MF, 1985: Survival of fertilisedova from ewe lambs and adult ewes <strong>in</strong> the uteri of ewelambs. Anim Reprod Sci 8, 235–240.Moor RM, Crosby IM, 1986: Prote<strong>in</strong> requirements forgerm<strong>in</strong>al vesicle breakdown <strong>in</strong> ov<strong>in</strong>e oocytes. J EmbryolExp Morph 94, 207–220.Morton KM, Catt SL, Holl<strong>in</strong>shead FK, Maxwell WMC,Evans G, 2004a: Production of lambs after the transfer offresh and cryopreserved <strong>in</strong> vitro produced embryos fromprepubertal lamb oocytes and unsorted and sex-sortedfrozen-thawed spermatozoa. Reprod Domest Anim 39,454–461.Morton KM, Maxwell WMC, Evans G, 2004b: The effect ofFSH concentration dur<strong>in</strong>g IVM and gamete co-<strong>in</strong>cubationlength dur<strong>in</strong>g IVF on the development of unstimulatedprepubertal ewe oocytes. Reprod Fertil Dev 16, 205.Morton KM, Maxwell WMC, Evans G, 2004c: The effect ofPMSG adm<strong>in</strong>istration on the growth and development ofthe reproductive tract of prepubertal ram lambs. In:Proceed<strong>in</strong>gs of the 15th International Congress on Animal<strong>Reproduction</strong> Porto Seguro, Brazil, 1, 233.Morton KM, Catt SL, Holl<strong>in</strong>shead FK, Maxwell WMC,Evans G, 2005a: The effect of gamete co-<strong>in</strong>cubation timedur<strong>in</strong>g <strong>in</strong> vitro fertilisation with frozen-thawed unsorted andsex-sorted ram spermatozoa on the development of <strong>in</strong> vitromatured adult and prepubertal ewe oocytes. Theriogenology64, 363–377.Morton KM, Catt SL, Maxwell WMC, Evans G, 2005b:Effects of lamb age, hormone stimulation and response tohormone stimulation on the yield and <strong>in</strong> vitro developmentalcompetence of prepubertal lamb oocytes. Reprod FertilDev 17, 593–601.Morton KM, Catt SL, Maxwell WMC, Evans G, 2005c: Anefficient method of ovarian stimulation and <strong>in</strong> vitro embryoproduction from prepubertal lambs. Reprod Fertil Dev 17,701–706.Morton KM, Catt SL, Maxwell WMC, Evans G, 2005d: Invitro and <strong>in</strong> vivo developmental capabilities and k<strong>in</strong>etics of<strong>in</strong> vitro development of <strong>in</strong> vitro matured oocytes from adult,unstimulated and hormone stimulated prepubertal ewes.Theriogenology 64, 1320–1332.Nicholas FW, 1996: Genetic improvement through reproductivetechnology. Anim Reprod Sci 42, 205–214.O’Brien JK, Dwarte D, Ryan JP, Maxwell WMC, Evans G,1996: Developmental capacity, energy metabolism andultrastructure of mature oocytes from prepubertal and adultsheep. Reprod Fertil Dev 8, 1029–1037.O’Brien JK, Beck NFG, Maxwell WMC, Evans G, 1997a:Effect of hormone pre-treatment of prepubertal sheep on theproduction and developmental capacity of oocytes <strong>in</strong> vitroand <strong>in</strong> vivo. Reprod Fertil Dev 9, 625–631.O’Brien JK, Catt SL, Ireland KA, Maxwell WMC, Evans G,1997b: In vitro and <strong>in</strong> vivo developmental capacity ofoocytes from prepubertal and adult sheep. Theriogenology47, 1433–1443.Oropeza A, Wrenzycki C, Herrmann D, Hadeler K-G,Niemann H, 2004: Improvement of the developmentalcapacity of oocytes from prepubertal cattle by <strong>in</strong>traovarianIGF-I application. Biol Reprod 70, 1634–1643.Presicce GA, Jiang S, Simk<strong>in</strong> M, Yang X, 1995: Oocyte qualityand embryo development <strong>in</strong> prepubertal calves. Biol Reprod52(Suppl.), 127.Ptak G, Loi P, Dattena M, Tischner M, Cappai P, 1999:Offspr<strong>in</strong>g from one-month-old lambs: Studies on the developmentalcapability of prepubertal oocytes. Biol Reprod 61,1568–1574.Ptak G, Tischner M, Bernabo N, Loi P, 2003: Donordependantdevelopmental competence of oocytes fromlambs subjected to repeated hormonal stimulation. BiolReprod 69, 278–285.Ptak G, Matsukawa K, Palmieri C, Salda LD, Scapolo PA,Loi P, 2006: Developmental and functional evidence ofnuclear immaturity <strong>in</strong> prepubertal oocytes. Hum Reprod 21,2228–2237.Quirke JF, Hanrahan JP, 1977: Comparison of the survival <strong>in</strong>the uteri of adult ewes of cleaved ova from adult ewes andewe lambs. J Reprod Fertil 51, 487–489.Tassell R, Chamley AW, Kennedy JP, 1978: Gonadotroph<strong>in</strong>levels and ovarian development <strong>in</strong> the neonatal ewe lamb.Aust J Biol Sci 31, 267–273.Tervit HR, McMillan WH, McGowan LT, Smith JF, HallDR, Donnison MJ, 1997: Effect of juvenile calf age onfollicular dynamics and <strong>in</strong> vitro embryo production. Theriogenology47, 300.Trounson AO, Willadsen SM, Moor RM, 1977: Reproductivefunction <strong>in</strong> prepubertal lambs: ovulation, embryo developmentand ovarian steroidogenesis. J Reprod Fertil 49, 69–75.Van Soom A, Ysebaert MT, de Kruif A, 1997: Relationshipbetween tim<strong>in</strong>g of development, morula morphology andcell allocation to <strong>in</strong>ner cell mass and trophectoderm <strong>in</strong> <strong>in</strong>vitro produced bov<strong>in</strong>e embryos. Mol Reprod Dev 47, 47–56.van der Werf J, 2005: Apply<strong>in</strong>g new technologies <strong>in</strong> sheepbreed<strong>in</strong>g programs. Mer<strong>in</strong>otech Best Practice Sheep Breed<strong>in</strong>gForum Kojonup, Western Australia, 17–23.Worth<strong>in</strong>gton CA, Kennedy JP, 1979: Ovarian response toexogenous hormones <strong>in</strong> six-week-old lambs. Aust J Biol Sci32, 91–95.Wrenzycki C, Herrmann D, Lucas-Hahn A, Korsawe K,Lemme E, Niemann H, 2005: Messenger RNA expressionpatterns <strong>in</strong> bov<strong>in</strong>e embryos derived from <strong>in</strong> vitro proceduresand their implications for development. Reprod Fertil Dev17, 23–35.Wright RW, Anderson GB, Cupps PT, Drost M, BradforsGE, 1976: In vitro culture of embryos from adult andprepubertal ewes. J Anim Sci 42, 912–917.Yang X, Presicce GA, Du F, Jiang S, 1997: Pregnancies andcalves derived from pre-pubertal calf oocytes. Theriogenology47, 163.Author’s address (for correspondence): Kather<strong>in</strong>e M. Morton, TheCamel <strong>Reproduction</strong> Centre, P.O. Box 79914, Dubai, United ArabEmirates. E-mail: kmorton@vetsci.usyd.edu.auConflict of <strong>in</strong>terest: The author declares no conflict of <strong>in</strong>terest.Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 144–149 (2008); doi: 10.1111/j.1439-0531.2008.01154.xISSN 0936-6768Updates on Reproductive Physiology, Genital Diseases and Artificial Insem<strong>in</strong>ation <strong>in</strong>the <strong>Domestic</strong> CatE Axne´rDivision of <strong>Reproduction</strong>, Department of Cl<strong>in</strong>ical Sciences, Swedish University of Agricultural Sciences, Uppsala, SwedenContents<strong>Reproduction</strong> <strong>in</strong> the domestic cat is characterized by large<strong>in</strong>dividual variations <strong>in</strong> the female oestrous cycle and <strong>in</strong> malesemen quality. The female cat reproduction is strongly<strong>in</strong>fluenced by season, and new data suggest that it is possiblethat season also affects male fertility. Repeated periods ofoestrus and spontaneous ovulation may lead to degenerativeendometrial changes caus<strong>in</strong>g <strong>in</strong>fertility. For artificial <strong>in</strong>sem<strong>in</strong>ation(AI), <strong>in</strong>duction of ovulation is necessary <strong>in</strong> the absenceof the mat<strong>in</strong>g stimuli. The large variation <strong>in</strong> the relationshipbetween follicular growth and tim<strong>in</strong>g of expression of oestruscomplicates, however, tim<strong>in</strong>g of ovulation <strong>in</strong>duction. Stressmay lead to progesterone secretion by the adrenal glands andpossibly have a negative impact on early pregnancy. The large<strong>in</strong>dividual variation <strong>in</strong> semen quality makes fertility evaluationand fel<strong>in</strong>e semen conservation a challenge. For AI, the semencan be deposited <strong>in</strong> the cranial vag<strong>in</strong>a, the uterus or <strong>in</strong> theuter<strong>in</strong>e tubes. Intrauter<strong>in</strong>e <strong>in</strong>sem<strong>in</strong>ation results <strong>in</strong> higherpregnancy rates than <strong>in</strong>travag<strong>in</strong>al but is more complicated.Surgical <strong>in</strong>trauter<strong>in</strong>e or <strong>in</strong>tratubal <strong>in</strong>sem<strong>in</strong>ation has resulted <strong>in</strong>the birth of kittens, but is an <strong>in</strong>vasive procedure that is notallowed <strong>in</strong> all countries. Transcervical <strong>in</strong>trauter<strong>in</strong>e <strong>in</strong>sem<strong>in</strong>ationwith frozen-thawed semen has, however, recently resulted<strong>in</strong> the birth of kittens.IntroductionThe domestic cat has been shown to be an <strong>in</strong>terest<strong>in</strong>gmodel for the development of reproductive biotechnologies<strong>in</strong> wild felids with the aim to conserve geneticvariation <strong>in</strong> small populations. There is, however, also apotential for the use or semen preservation and artificial<strong>in</strong>sem<strong>in</strong>ation (AI) <strong>in</strong> the breed<strong>in</strong>g of domestic cats. Inthe dog, these techniques can be considered rout<strong>in</strong>etoday. In contrast, more research is necessary before AI,and semen conservation can be considered rout<strong>in</strong>eprocedures <strong>in</strong> the domestic cat. To be able to successfullyuse reproductive biotechnologies, a thoroughknowledge about reproductive physiology and pathologyis necessary.Physiology of the Female CatThe oestrous cycleThe female cat is seasonally polyoestrous with changes<strong>in</strong> daylength (<strong>in</strong>creas<strong>in</strong>g number of light hours) regulat<strong>in</strong>gthe seasonality (Leyva et al. 1989; Tsutsui et al.2004a). Characteristic for reproduction <strong>in</strong> the domesticcat is the large normal variation <strong>in</strong> the length of theoestrous cycle, the length of the reproductive season andthe number of cycles ⁄ year between queens (Shille et al.1979; Tsutsui et al. 2004a). The presence of matureoocytes <strong>in</strong> the ovaries and the effect of ovulation<strong>in</strong>duction seem to be dependent on the day of the onsetof physiological oestrus dur<strong>in</strong>g the follicular phase(Banks and Stabenfeldt 1982; Glover et al. 1985).Because there is a variation between queens <strong>in</strong> whichday dur<strong>in</strong>g the follicular phase they will start to showoestrous behaviour, the day of oestrous behaviour <strong>in</strong>relation to the progress of oocyte maturation is likely todiffer between queens (Shille et al. 1979; Malanda<strong>in</strong>et al. 2002). Most queens will respond with an LHrelease,lead<strong>in</strong>g to ovulation of good quality oocytesafter a mat<strong>in</strong>g <strong>in</strong> midoestrus, while mat<strong>in</strong>g <strong>in</strong> earlyoestrus is not always followed by ovulation (Banks andStabenfeldt 1982; Glover et al. 1985). Expression ofFSH- and LH receptors <strong>in</strong> the ovarian follicles variesdepend<strong>in</strong>g on the stage of follicular growth or atresia(Sa<strong>in</strong>t-Dizier et al. 2007).Cyclical changes <strong>in</strong> the female tubular genitalia <strong>in</strong> relationto hysterographic appearance and cervical patencyThe outer diameter of the uterus and the appearance ofthe endometrial l<strong>in</strong><strong>in</strong>g will vary depend<strong>in</strong>g on the stageof the oestrous cycle. Dur<strong>in</strong>g <strong>in</strong>active stages, a s<strong>in</strong>glelayer of cuboidal cells l<strong>in</strong>es the endometrium, andthe endometrial glands are <strong>in</strong>active. When the queenenters the follicular stage, the endometrial and myometrialdiameters <strong>in</strong>crease and the endometrial glandsproliferate. The luteal phase endometrium is characterizedby a high number of elongated and active glands(Chatdarong et al. 2005). The shape of the uter<strong>in</strong>e cavitycan be studied with hysterography. In the healthy cat,the endometrial outl<strong>in</strong><strong>in</strong>g is smooth. Dur<strong>in</strong>g the <strong>in</strong>activephase of the cycle, the uter<strong>in</strong>e horns are straight.Because of the endometrial growth and <strong>in</strong>creasedmuscular activity, the uter<strong>in</strong>e horns become curved witha wavy lum<strong>in</strong>al cavity dur<strong>in</strong>g the follicular phase. Thelum<strong>in</strong>al shape dur<strong>in</strong>g the luteal phase varies fromstraight to irregular, wavy and coiled (Chatdarong et al.2005). The cervix is normally only patent (physiologicallyopen cervical canal) dur<strong>in</strong>g oestrus and theduration of cervical patency does not depend onwhether or not the queen ovulates (Chatdarong et al.2002, 2006a). The period of cervical patency usuallyco<strong>in</strong>cides with the period when the vag<strong>in</strong>al smear iscornified (Chatdarong et al. 2002).Sperm distribution after mat<strong>in</strong>gSperm deposition dur<strong>in</strong>g mat<strong>in</strong>g is vag<strong>in</strong>al. Initial spermtransport <strong>in</strong> the female reproductive tract is very rapid.Spermatozoa can be found <strong>in</strong> the uter<strong>in</strong>e tube 30 m<strong>in</strong>after mat<strong>in</strong>g. The uterotubal junction and uter<strong>in</strong>e cryptsserve as <strong>in</strong>itial sperm reservoirs but spermatozoa areÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Reproductive Physiology, Pathology and AI <strong>in</strong> Cat 145subsequently redistributed to the isthmus (Chatdaronget al. 2004), and the oocytes are fertilized <strong>in</strong> the oviductaround the time of ovulation (Swanson et al. 1994). As as<strong>in</strong>gle mat<strong>in</strong>g or AI at the time of ovulation <strong>in</strong>ductionmay result <strong>in</strong> conception, the fertile life of cat spermatozoamust be at least the same as the time betweenmat<strong>in</strong>g and ovulation (Chatdarong et al. 2007).Adrenal progesterone secretionProgesterone is produced by the corpora lutea afterovulation but can also be released from the adrenalglands after stress or <strong>in</strong>jection with ACTH (Chatdaronget al. 2006b). A serum progesterone concentration of16.3 nmol ⁄ l has been observed after ACTH <strong>in</strong>jection ofan ovariohysterectomized female, <strong>in</strong>dicat<strong>in</strong>g that thefel<strong>in</strong>e adrenals may produce substantial amounts ofprogesterone (Chatdarong et al. 2006b). ACTH adm<strong>in</strong>istrationto sows has been shown to cause an alteredprogesterone profile, a possible decrease <strong>in</strong> the time fortransport of oocytes ⁄ embryos through the oviduct andloss of embryos (Brandt et al. 2007). Therefore, onecannot exclude that stress may have negative effects alsoon early pregnancy <strong>in</strong> cats, a phenomenon that warrantsfurther research <strong>in</strong> this species.Disorders of the Female Genital OrgansInfertility <strong>in</strong> the cycl<strong>in</strong>g queenInfertility <strong>in</strong> the queen with normal ovarian activitypresents a diagnostic challenge for the cl<strong>in</strong>ician. In thequeen with normal cycles, normal mat<strong>in</strong>g and mat<strong>in</strong>g<strong>in</strong>ducedovulation, CEH or low-grade endometritis maycause <strong>in</strong>fertility without any other obvious cl<strong>in</strong>ical signs.CEH is believed to be the result of cumulative changes<strong>in</strong> the endometrium <strong>in</strong>duced by oestradiol and progesterone(Perez et al. 1999; Misirlioglu et al. 2006). Noefficient treatment for CEH except ovariohysterectomyhas been reported. CEH can be diagnosed with ultrasoundor hysterography (Chatdarong et al. 2005). Lowgradeendometritis is extremely difficult to diagnose.Uter<strong>in</strong>e biopsy is an <strong>in</strong>vasive procedure and does notguarantee that the whole endometrium is accessed andmay therefore give a false negative result if the pathologicalchanges are focal. Vag<strong>in</strong>al discharge is an<strong>in</strong>dication of genital pathology, but may not be presentif the cervix is closed, the <strong>in</strong>fection is mild, or <strong>in</strong> thepresence of a chronic <strong>in</strong>flammation. Leukocytes <strong>in</strong> avag<strong>in</strong>al swab <strong>in</strong>dicate an acute <strong>in</strong>flammation, butleukocytes may be few or absent even if the queen hasa uter<strong>in</strong>e <strong>in</strong>fection. Vag<strong>in</strong>al bacterial culture is notdiagnostic because the normal vag<strong>in</strong>al bacterial floracannot be dist<strong>in</strong>guished from that of the flora associatedwith uter<strong>in</strong>e pathologies, such as endometritis (Stro¨mHolst et al. 2003). If an endometritis is suspected,bacterial culture can, however, be used to select anappropriate antibiotic. The likelihood of tak<strong>in</strong>g asample from the vag<strong>in</strong>a that is truly representative ofthe uter<strong>in</strong>e flora (i.e. f<strong>in</strong>d<strong>in</strong>g the same bacteria <strong>in</strong> avag<strong>in</strong>al swab as those that are present <strong>in</strong> the uterus), isprobably higher dur<strong>in</strong>g oestrus when the cervix is open,than at other stages of the cycle, although this has neverbeen confirmed. To decrease the risk of select<strong>in</strong>gbacterial stra<strong>in</strong>s resistant to antibiotics, the decision toput an <strong>in</strong>fertile queen on antibiotics should only bemade after a thorough cl<strong>in</strong>ical exam<strong>in</strong>ation to rule outother causes of <strong>in</strong>fertility. Mild degenerative changes ofthe endometrium may be difficult to detect on ultrasoundbut can be a reason for treatment failure (Axne´ret al., 2008).Reproductive Physiology of the Male CatIn tomcats, spermatogenesis is established at 6–8 months of age, at which time an <strong>in</strong>crease <strong>in</strong> testicularweight and testosterone production can be observed.The spermatogenic function <strong>in</strong> young toms is usuallynot comparable to that of mature males until after8 months of age (Tsutsui et al. 2004b; Siemieniuch andWoclawek-Potocka 2007). The daily sperm productionhas been estimated to be 16 · 10 6 spermatozoa ⁄ testis(Franc¸a and God<strong>in</strong>ho 2003). The effect of season onreproductive function <strong>in</strong> the male domestic cat is<strong>in</strong>sufficiently explored and reports are contradictory(Johnstone et al. 1984; Sp<strong>in</strong>dler and Wildt 1999). In aretrospective study on sperm morphology <strong>in</strong> privatelyowned cats, we found that the percentage of normalspermatozoa was higher <strong>in</strong> ejaculates collected from catsdur<strong>in</strong>g the breed<strong>in</strong>g season than dur<strong>in</strong>g the non-breed<strong>in</strong>gseason, <strong>in</strong>dicat<strong>in</strong>g a possible effect of season on malefertility <strong>in</strong> cats (Axnér and L<strong>in</strong>de Forsberg 2007).Blottner and Jewgenow (2007) found seasonal variations<strong>in</strong> epididymal sperm quality and testosterone productionbetween spr<strong>in</strong>g and autumn. These recent f<strong>in</strong>d<strong>in</strong>gs<strong>in</strong>dicate that there might be seasonal variation also <strong>in</strong>domestic cat male fertility.Semen qualitySemen quality displays large <strong>in</strong>dividual variations <strong>in</strong> thecat. In a retrospective study, the median percentage ofmorphologically normal spermatozoa <strong>in</strong> an unselectedpopulation of cats was 44% (n = 48), (Axnér and L<strong>in</strong>deForsberg 2007). Of all cats with


146 E Axnérbetween, but also with<strong>in</strong> <strong>in</strong>dividual cats, presents achallenge for the cl<strong>in</strong>ician wish<strong>in</strong>g to perform a fertilityexam<strong>in</strong>ation or to conserve spermatozoa from anyparticular cat.Infertility <strong>in</strong> the Male CatThe causes of <strong>in</strong>fertility <strong>in</strong> the male cat can be classifiedas developmental disorders, poor libido, testiculardegeneration, testicular hypoplasia and miscellaneous.These categories are, however, not always clearlydist<strong>in</strong>guished from each other. Chromosomal abnormalitiesare, for example, developmental disorders thatmay result <strong>in</strong> testicular hypoplasia, as seen <strong>in</strong> tortoiseshellmale cats with the Kl<strong>in</strong>efelter’s syndrome (Axnéret al. 1996). Testicular degeneration is usually anacquired non-hereditary condition, and therefore has abetter prognosis than testicular hypoplasia, which is agenetic condition. Infertile cats often have a low spermconcentration and a high percentage of different spermdefects, or are completely azoospermic (Axne´r et al.1996; Axne´r and L<strong>in</strong>de Forsberg 2007). Both testicularhypoplasia and testicular degeneration will result <strong>in</strong> adecrease <strong>in</strong> semen quality but are not always possible todifferentiate between <strong>in</strong> a cl<strong>in</strong>ical situation. To make adef<strong>in</strong>itive diagnosis of <strong>in</strong>fertility, a semen sample isusually needed. Repeated samples may, however, benecessary as fertility sometimes fluctuates over time(Axnér et al. 1996; Axne´r and L<strong>in</strong>de Forsberg 2007).Sperm Collection, Sperm Conservation andArtificial Insem<strong>in</strong>ationSperm collectionEjaculated spermatozoa can be collected by an artificialvag<strong>in</strong>a or by electroejaculation (Sojka et al. 1970; Platzand Seager 1978; Platz et al. 1978; Howard et al. 1990).Collection by an artificial vag<strong>in</strong>a generally yields ahigher total number of spermatozoa and allows repeatedsemen collections with<strong>in</strong> short <strong>in</strong>tervals. However,because the use of an artificial vag<strong>in</strong>a is not alwayssuccessful, such use requires tra<strong>in</strong><strong>in</strong>g and is unlikely tobe convenient <strong>in</strong> a cl<strong>in</strong>ical situation; electroejaculation isusually the method of choice (Platz and Seager 1978).Electroejaculation requires a surgical plane of anaesthesiabecause reactions <strong>in</strong> unanaesthethized animals <strong>in</strong>dicatethat the procedure is associated with pa<strong>in</strong> (Platzand Seager 1978; Stafford 1995). In wild felids, althoughketam<strong>in</strong>e alone has been used previously, comb<strong>in</strong>edregimens for anaesthesia are now preferred as theyprovide better sedation and analgesia (Swanson et al.2007). In our laboratory, we use a comb<strong>in</strong>ation ofmedetomid<strong>in</strong>e (80 lg ⁄ kg bw) and ketam<strong>in</strong>e–HCl(5 mg ⁄ kg bw) (Axne´r et al. 1998). Propofol at a doseof 10 mg ⁄ kg bw is a suitable alternative for anaesthesiafor electroejaculation <strong>in</strong> domestic cats (Chatdaronget al. 2006c). Medetomid<strong>in</strong>e, an a-adrenergic agent,was shown to <strong>in</strong>crease sperm outflow when used at adose of 130–140 lg ⁄ kg bw (Zambelli et al. 2007). Thenumber of spermatozoa <strong>in</strong> an ejaculate collected byelectroejaculation, rarely exceeds the number of spermatozoathat are needed for one <strong>in</strong>sem<strong>in</strong>ation only withthe techniques and knowledge available today (Sojkaet al. 1970; Platz et al. 1978; Howard et al. 1992;Tanaka et al. 2000; Tsutsui 2006; Chatdarong et al.2007). Ref<strong>in</strong>ement of methods for sperm cryopreservation,ovulation <strong>in</strong>duction and AI allow<strong>in</strong>g lower spermnumbers to be used for AI would <strong>in</strong>crease the usefulnessof reproductive biotechnologies <strong>in</strong> cat breed<strong>in</strong>g.As the domestic cat is rout<strong>in</strong>ely subjected to castration,studies on domestic cat epididymal spermatozoaare useful as a model for epididymal sperm preservationfrom wild felids threatened with ext<strong>in</strong>ction. Epididymalsperm preservation has, however, also a potential to beuseful <strong>in</strong> breed<strong>in</strong>g programmes for domestic cats (Tsutsuiet al. 2003; Hermansson and Axne´r 2007). It has notbeen fully elucidated <strong>in</strong> the cat whether epididymalspermatozoa are comparable to ejaculated spermatozoa<strong>in</strong> their function and reaction to sperm conservationprotocols. In other mammalian species, sem<strong>in</strong>al plasmaconta<strong>in</strong>s, for example, decapacitation factors, which canb<strong>in</strong>d to the spermatozoa and <strong>in</strong>hibit or reverse capacitation,but also factors that enhance capacitation(Oliphant et al. 1985; Boue´ et al. 1996; Therien et al.1998).It has been shown <strong>in</strong> several species that ejaculatedspermatozoa are more susceptible to cold shock thanepididymal spermatozoa (Gilmore et al. 1998). Spermmotility and viability of frozen-thawed epididymalcat spermatozoa was, however, similar to that ofejaculated spermatozoa (Tsutsui et al. 2003). Althoughejaculated spermatozoa had higher <strong>in</strong>itial motility andplasma membrane <strong>in</strong>tegrity, there was no effect of spermsource (ejaculated or epididymal) <strong>in</strong> the sensitivity tocold shock at the cool<strong>in</strong>g rates usually used <strong>in</strong> spermpreservation protocols (Hermansson and Axne´r 2007).The first birth of kittens after <strong>in</strong>sem<strong>in</strong>ation with frozenthawedepididymal cat spermatozoa was reported byTsutsui et al. (2003).Semen conservationSemen can be used fresh, chilled or frozen-thawed forAI. Usually, the use of conserved semen is of more<strong>in</strong>terest than the use of fresh semen as it allows forstorage for extended time periods. Each step <strong>in</strong> semenpreservation protocols may cause damage to the spermatozoa,thereby reduc<strong>in</strong>g their fertiliz<strong>in</strong>g potential andprobably also their longevity. Therefore, a highernumber of spermatozoa is needed for <strong>in</strong>sem<strong>in</strong>ation withfrozen-thawed semen than with fresh spermatozoa(Tsutsui 2006). Cat spermatozoa do not seem to besusceptible to cold shock at the cool<strong>in</strong>g rates usuallyapplied for sperm conservation (Hermansson and Axne´r2007) although there are conflict<strong>in</strong>g reports regard<strong>in</strong>gthe effect of cool<strong>in</strong>g on acrosomal <strong>in</strong>tegrity (Pukazhenthiet al. 1999). Freez<strong>in</strong>g, on the contrary, <strong>in</strong>duces moresevere sperm damage (Axnér et al. 2004; Luvoni 2006).Different cryopreservation protocols have been published.In our laboratory, we use a protocol that hasbeen modified from a protocol that was orig<strong>in</strong>allydeveloped for preservation of can<strong>in</strong>e spermatozoa (Rotaet al. 1997; Axne´r et al. 2004). Transcervical AI withejaculated spermatozoa preserved with this protocol hasresulted <strong>in</strong> the birth of live kittens demonstrat<strong>in</strong>g thefertiliz<strong>in</strong>g capacity of spermatozoa cryopreserved withÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Reproductive Physiology, Pathology and AI <strong>in</strong> Cat 147this method (Chatdarong et al. 2007). Addition ofEquex STM (Nova Chemical Sales, Scituate Inc., MA,USA) paste to a Tris–egg yolk extender reduces acrosomedamage after freez<strong>in</strong>g-thaw<strong>in</strong>g, but may have atoxic effect dur<strong>in</strong>g <strong>in</strong>cubation, lead<strong>in</strong>g to a fasterdecrease <strong>in</strong> sperm viability (Axnér et al. 2004). Wash<strong>in</strong>gfrozen-thawed spermatozoa by centrifugation to removethe freez<strong>in</strong>g extender <strong>in</strong>clud<strong>in</strong>g the Equex STM pastehad, however, no beneficial effect on sperm longevity <strong>in</strong>a prelim<strong>in</strong>ary study (Axne´r et al. 2006). Addition ofantioxidants to the freez<strong>in</strong>g buffer has been shown toimprove post-thaw cat epididymal sperm quality(Luvoni et al. 2002; Thuwanut et al., 2008).Ovulation <strong>in</strong>duction and tim<strong>in</strong>g of <strong>in</strong>sem<strong>in</strong>ationIn the absence of the mat<strong>in</strong>g stimuli, ovulation must beartificially <strong>in</strong>duced before AI can be carried out. Thelarge variation <strong>in</strong> the female oestrous cycle complicatesdeterm<strong>in</strong>ation of the optimal time for ovulation <strong>in</strong>duction.Ultrasound evaluation may be a reliable method tofollow follicular maturation (Malanda<strong>in</strong> et al. 2002).The occurrence of spontaneous ovulation may contributeto difficulties <strong>in</strong> tim<strong>in</strong>g the AI (Lawler et al. 1993;Chatdarong et al. 2007). In order to control the oestrouscycle, studies on different hormonal therapies have beenperformed (Pelican et al. 2006). Pharmacological <strong>in</strong>ductionof oestrus and ovulation cause, however, abnormalhormonal profiles that may <strong>in</strong>terfere with fertility(Pelican et al. 2006). Ovulation <strong>in</strong>duction is usuallycarried out with one or two <strong>in</strong>jections of hCG <strong>in</strong>midoestrus (Pelican et al. 2006; Tsutsui 2006; Chatdaronget al. 2007). For <strong>in</strong>duction of oestrus, a comb<strong>in</strong>ationof eCG with an <strong>in</strong>jection of hCG 80–84 h later isoften used (Pelican et al. 2006). Both hCG and eCGstimulate the production of antibodies, hence, frequentadm<strong>in</strong>istration can lead to decreased treatment success(Pelican et al. 2006). The optimal protocol for ovulation<strong>in</strong>duction and tim<strong>in</strong>g of ovulation <strong>in</strong>duction rema<strong>in</strong>s tobe elucidated.Spermatozoa should be <strong>in</strong>sem<strong>in</strong>ated with<strong>in</strong> 49 hafter ovulation <strong>in</strong>duction as no fertilizations have beenobserved to occur after this time (Sojka et al. 1970;Howard et al. 1992). Fel<strong>in</strong>e spermatozoa probably donot require a long time for capacitation. Niwa et al.(1985) reported that 100% of homologous oocyteswere penetrated by epididymal spermatozoa with<strong>in</strong>30 m<strong>in</strong> after <strong>in</strong>sem<strong>in</strong>ation, while ejaculated cat spermatozoaseem to need 2.5–3 h for <strong>in</strong> vitro capacitation(Long et al. 1996). Consider<strong>in</strong>g the reduced longevityof frozen-thawed spermatozoa, they should probablybe <strong>in</strong>sem<strong>in</strong>ated close to ovulation. The optimal timefor AI <strong>in</strong> relation to ovulation has, however, not beenfully elucidated although some studies report AI atdifferent times with some conflict<strong>in</strong>g results concern<strong>in</strong>gthe effect of pre-ovulatory anaesthesia on ovulation(Howard et al. 1992; Tsutsui 2006; Chatdarong et al.2007).Techniques of semen depositionPregnancies have been achieved with <strong>in</strong>travag<strong>in</strong>al as wellas with surgical or laparoscopic <strong>in</strong>trauter<strong>in</strong>e or <strong>in</strong>tratubalsemen deposition (Sojka et al. 1970; Platz et al.1978; Howard et al. 1992; Tsutsui 2006; Tsutsui et al.2001). In most species as well as <strong>in</strong> the cat, <strong>in</strong>trauter<strong>in</strong>esemen deposition yields better pregnancy results thanvag<strong>in</strong>al deposition although the cat spermatozoa aredeposited <strong>in</strong> the vag<strong>in</strong>a dur<strong>in</strong>g natural coitus (L<strong>in</strong>de-Forsberg et al. 1999; Tanaka et al. 2000; Thomassenet al. 2006; Tsutsui 2006; Chatdarong et al. 2007).Surgical <strong>in</strong>sem<strong>in</strong>ation is an <strong>in</strong>vasive procedure that isnot allowed <strong>in</strong> all countries. Therefore, studies ontranscervical AI have been performed. Transcervicalcatheterization <strong>in</strong> the cat is, however, associated withdifficulties because of the vag<strong>in</strong>al anatomy. Abdom<strong>in</strong>alfixation of the cervix as carried out with the Norwegiantranscervical catheter for the bitch is not possible <strong>in</strong> thequeen because of the position of the cervix and thenarrow cranial vag<strong>in</strong>a makes endoscopy-guided transcervicalpassage impossible (L<strong>in</strong>de-Forsberg et al. 1999;Chatdarong et al. 2002; Zambelli et al. 2004; Thomassenet al. 2006). Zambelli and Castagnetti (2001)performed rectally guided transcervical catheterizationus<strong>in</strong>g a rounded tip needle <strong>in</strong>serted at the cut end of a 3-Fr tomcat catheter.Birth of live kittens has resulted after transcervical<strong>in</strong>trauter<strong>in</strong>e deposition of frozen-thawed semen with thehelp of a catheter modified from a transcervical catheterfirst described by Swanson and Godke (1994) (Chatdaronget al. 2005, 2007). The result with birth of livekittens is promis<strong>in</strong>g for the use of non-surgical AI <strong>in</strong>small felids, although the protocol still needs to beref<strong>in</strong>ed to improve pregnancy results and to allow theuse of a lower sperm number (Chatdarong et al. 2007).In conclusion, although more studies on evaluation offollicular maturation, protocols for artificial ovulation<strong>in</strong>duction, semen conservation and techniques of AI areneeded before an optimal protocol for rout<strong>in</strong>e AI <strong>in</strong> thecat can be developed, some promis<strong>in</strong>g progress has beenachieved <strong>in</strong> the development of semen conservation andAI <strong>in</strong> the cat.ReferencesAxne´r E, L<strong>in</strong>de Forsberg C, 2007: Sperm morphology <strong>in</strong> thedomestic cat, and its relation with fertility: a retrospectivestudy. Reprod Domest Anim 42, 282–291.Axne´r E, Stro¨m B, L<strong>in</strong>de-Forsberg C, Gustavsson I, L<strong>in</strong>dbladK, Wallgren M, 1996: Reproductive disorders <strong>in</strong> 10 domesticmale cats. J Small Anim Pract 37, 394–401.Axne´r E, Stro¨m Holst B, L<strong>in</strong>de Forsberg C, 1998: Morphologyof spermatozoa <strong>in</strong> the cauda epididymidis beforeand after electroejaculation and a comparison with ejaculatedspermatozoa <strong>in</strong> the domestic cat. Theriogenology50, 973–979.Axne´r E, Hermansson U, L<strong>in</strong>de-Forsberg C, 2004: The effectof Equex STM paste and sperm morphology on post-thawsurvival of cat epididymal spermatozoa. Anim Reprod Sci84, 179–191.Axne´r E, Kronsell A, L<strong>in</strong>de-Forsberg C, 2006: Effect of postthawcentrifugation on viability of epididymal cat spermatozoa.<strong>in</strong>: Luvoni GC, Thuro´czy J (eds), Proc. 5th BiannualEVSSAR Conf. Budapest, April, P06.Axne´r E,A˚ gren E, Ba˚ verud V, Stro¨m Holst B. Infertility <strong>in</strong> thecycl<strong>in</strong>g queen: seven cases. Journal of fel<strong>in</strong>e medic<strong>in</strong>e andsurgery, 2008: DOI: 10.1016/j.jfms.2008.04.005.Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


148 E AxnérBanks DH, Stabenfeldt G, 1982: Lute<strong>in</strong>iz<strong>in</strong>g hormone release<strong>in</strong> the cat <strong>in</strong> response to coitus on consecutive days of estrus.Biol Reprod 26, 603–611.Blottner S, Jewgenow K, 2007: Moderate seasonality <strong>in</strong> testisfunction of domestic cat. Reprod Domest Anim 42, 536–540.Boue´ F, Blais J, Sullivan R, 1996: Surface localisation ofP34H, an epididymal prote<strong>in</strong>, dur<strong>in</strong>g maturation, capacitation,and acrosome reaction of human spermatozoa. BiolReprod 54, 1009–1017.Brandt Y, Madej A, Rodrı´guez-Martı´nez H, E<strong>in</strong>arsson S,2007: Effects of exogenous ACTH dur<strong>in</strong>g oestrus on earlyembryo development and oviductal transport <strong>in</strong> the sow.Reprod Domest Anim 42, 118–125.Chatdarong K, Rungsipipat A, Axne´r E, L<strong>in</strong>de-Forsberg C,2002: Investigation of cervical patency and uter<strong>in</strong>e appearance<strong>in</strong> domestic cats by fluoroscopy and sc<strong>in</strong>tigraphy.Reprod Domest Anim 37, 275–282.Chatdarong K, Lohachit C, L<strong>in</strong>de Forsberg C, 2004: Distributionof spermatozoa <strong>in</strong> the female reproductive tract ofthe domestic cat <strong>in</strong> relation to ovulation <strong>in</strong>duced by naturalmat<strong>in</strong>g. Theriogenology 62, 1027–1041.Chatdarong K, Rungsipipat A, Axne´r E, L<strong>in</strong>de Forsber C,2005: Hysterographic appearance and uter<strong>in</strong>e histology atdifferent stages of the reproductive cycle and after progestagentreatment <strong>in</strong> the domestic cat. Theriogenology 64, 12–29.Chatdarong K, Lohachit C, Kiartmanakul S, Axne´r E, L<strong>in</strong>deForsberg C, 2006a: Cervical patency dur<strong>in</strong>g non-ovulatoryand ovulatory estrus cycles <strong>in</strong> domestic cats. Theriogenology66, 804–810.Chatdarong K, Ponglowhapan S, Karlsson A, L<strong>in</strong>de-ForsbergC, 2006b: Exogenous ACTH <strong>in</strong>duced progesterone levels <strong>in</strong>female domestic cats. Theriogenology 66, 1482–1487.Chatdarong K, Ponglowhapan S, Manee-<strong>in</strong> S, Pongphet K,2006c: The use of propofol for electroejaculation <strong>in</strong> domesticcats. Theriogenology 66, 615–617.Chatdarong K, Axne´r E, Manee-In S, Thuwanut P, L<strong>in</strong>de-Forsberg C, 2007: Pregnancy <strong>in</strong> the domestic cat aftervag<strong>in</strong>al or transcervical <strong>in</strong>sem<strong>in</strong>ation with fresh and frozensemen. Theriogenology 68, 1326–1333.França LR, God<strong>in</strong>ho CL, 2003: Testis morphometry, sem<strong>in</strong>iferousepithelium cycle length, and daily sperm production<strong>in</strong> domestic cats (Felis catus). Biol Reprod 68, 1554–1561.Gilmore JA, McGann LE, Ashworth E, Acker JP, Raath JP,Bush M, Critser JK, 1998: Fundamental cryobiology ofselected African mammalian spermatozoa and its role <strong>in</strong>biodiversity preservation through the development ofgenome resource bank<strong>in</strong>g. Anim Reprod Sci 53, 277–298.Glover TE, Watson PF, Bonney RC, 1985: Observations onvariability <strong>in</strong> LH release and fertility dur<strong>in</strong>g oestrus <strong>in</strong> thedomestic cat (Felis catus). J Reprod Fertil 75, 145–152.Hermansson U, Axne´r E, 2007: Epididymal and ejaculated catspermatozoa are resistant to cold shock but egg yolkpromotes sperm longevity dur<strong>in</strong>g cold storage at 4°C.Theriogenology 67, 1239–1248.Howard JG, Brown JL, Bush M, Wildt DE, 1990: Teratospermicand normospermic domestic cats: ejaculate traits,pituitary-gonadal hormones, and improvement of spermatozoalmotility and morphology after swim-up process<strong>in</strong>g. JAndrol 11, 204–215.Howard JG, Barone MA, Donoghue AM, Wildt DE, 1992:The effect of pre-ovulatory anesthesia on ovulation <strong>in</strong>laparoscopically <strong>in</strong>sem<strong>in</strong>ated domestic cat. J Reprod Fertil96, 175–186.Johnstone IP, Bancroft BJ, McFarlane JR, 1984: Testosteroneand androstenedione profiles <strong>in</strong> the blood of domestic tomcats. Anim Reprod Sci 7, 363–375.Lawler DF, Johnston SD, Hegstad RL, Keltner DG, OwensSF, 1993: Ovulation without cervical stimulation <strong>in</strong> domesticcats. J Reprod Fertil Suppl 47, 57–61.Leyva H, Madley T, Stabenfeldt GH, 1989: Effect of lightmanipulation on ovarian activity and melaton<strong>in</strong> andprolact<strong>in</strong> secretion <strong>in</strong> the domestic cat. J Reprod FertilSuppl 39, 125–133.L<strong>in</strong>de-Forsberg C, Stro¨m Holst B, Govette G, 1999: Comparisonof fertility data from vag<strong>in</strong>al vs <strong>in</strong>trauter<strong>in</strong>e <strong>in</strong>sem<strong>in</strong>ationof frozen-thawed dog semen: a retrospective study.Theriogenology 52, 11–23.Long JA, Wild DE, Wolfe BA, Critser JK, DeRossi RV,Howard JG, 1996: Sperm capacitation and the acrosomereaction are compromised <strong>in</strong> teratospermic domestic cats.Biol Reprod 54, 638–646.Luvoni GC, 2006: Gamete cryopreservation <strong>in</strong> the domesticcat. Theriogenology 66, 101–111.Luvoni GC, Guggiero C, Mar<strong>in</strong>oni C, Kalchsmidt E, 2002:Effect of taur<strong>in</strong>e-conta<strong>in</strong><strong>in</strong>g diluent for cryopreservation ofdomestic cat epididymal spermatozoa. Theriogenology 57,466.Malanda<strong>in</strong> E, Rault D, Froment E, Ficheux C, Pichard JP,Fontbonne A, Begon D, Chastan-Maillard S, 2002: Followupof follicular growth dur<strong>in</strong>g non ovulatory estral phases <strong>in</strong>queen by ultrasonography and vag<strong>in</strong>al cytology. <strong>in</strong>: VerstegenJ, Oncl<strong>in</strong> K, L<strong>in</strong>de Forsberg C (eds), Proc. 3rd EVSSARCongr. Lie` ge, 10–12 May, p. 141.Misirlioglu D, Nak D, Sevimli A, Nak Y, Ozyigit O, Akkoc A,Cangul IT, 2006: Steroid receptor expression and HER-2 ⁄ neu (c-erB-2) oncoprote<strong>in</strong> <strong>in</strong> the uterus of cats with cysticendometrial hyperplasia-pyometra complex. J Vet Med A53, 225–229.Niwa K, Oˆ hara K, Hosoi Y, Iritani A, 1985: Early events of <strong>in</strong>vitrofertilization of cat eggs by epididymal spermatozoa. JReprod Fertil 74, 657–660.Oliphant G, Reynold AB, Thomas TS, 1985: Sperm surfacecomponents <strong>in</strong>volved <strong>in</strong> the control of the acrosomereaction. Am J Anat 174, 269–283.Pelican KM, Wildt DE, Pukazhenthi B, Howard JG, 2006:Ovarian control for assisted reproduction <strong>in</strong> the domesticcat and wild felids. Theriogenology 66, 37–48.Perez JF, Conley AJ, Dieter JA, Sanz-Ortega J, Lasley BL,1999: Studies on the orig<strong>in</strong> of ovarian <strong>in</strong>terstitial tissue andthe <strong>in</strong>cidence of endometrial hyperplasia <strong>in</strong> domestic andferal cats. Gen Comp Endocr<strong>in</strong>ol 116, 10–20.Platz CC, Seager SWJ, 1978: Semen collection by electroejaculation<strong>in</strong> the domestic cat. J Am Vet Med Assoc 173,1353–1355.Platz CC, Wildt DE, Seager SW, 1978: Pregnancy <strong>in</strong> thedomestic cat after artificial <strong>in</strong>sem<strong>in</strong>ation with previouslyfrozen spermatozoa. J Reprod Fertil 52, 279–282.Pukazhenthi B, Pelican K, Wildt DE, Howard JG, 1999:Sensitivity of domestic cat (Felis catus) sperm from normospermicversus teratospermic donors to cold-<strong>in</strong>duced acrosomaldamage. Biol Reprod 61, 135–141.Rota A, Stro¨m B, L<strong>in</strong>de-Forsberg C, Rodriguez-Mart<strong>in</strong>ez H,1997: Effects of equex STM paste on viability of frozenthawed dog spermatozoa dur<strong>in</strong>g <strong>in</strong> vitro <strong>in</strong>cubation at 38°C.Theriogenology 47, 1093–1101.Sa<strong>in</strong>t-Dizier M, Malanda<strong>in</strong> E, Thoumire S, Remy B, Chastan-Maillard S, 2007: Expression of follicle stimulat<strong>in</strong>g hormoneand lute<strong>in</strong>is<strong>in</strong>g hormone receptors dur<strong>in</strong>g follicular growth<strong>in</strong> the domestic cat ovary. Mol Reprod Dev 74, 989–996.Shille VM, Lundstro¨m KE, Stabenfeldt GH, 1979: Follicularfunction <strong>in</strong> the domestic cat as determ<strong>in</strong>ed by estradiol-17bconcentrations <strong>in</strong> plasma: relation to estrous behaviour andcornification of exfoliated vag<strong>in</strong>al epithelium. Biol Reprod21, 953–963.Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Reproductive Physiology, Pathology and AI <strong>in</strong> Cat 149Siemieniuch MJ, Woclawek-Potocka I, 2007: Morphologicalfeatures of the sem<strong>in</strong>iferous epithelium <strong>in</strong> cat (Felis catus, L.1758) testes. J Reprod Dev 53, 1125–1130.Sojka NJ, Jenn<strong>in</strong>gs LL, Hamner CE, 1970: Artificial <strong>in</strong>sem<strong>in</strong>ation<strong>in</strong> the cat (Felis catus L). Lab Anim Care 20, 198–204.Sp<strong>in</strong>dler RE, Wildt DE, 1999: Circannual variations <strong>in</strong><strong>in</strong>traovarian oocyte but not epididymal sperm quality <strong>in</strong>the domestic cat. Biol Reprod 61, 188–194.Stafford KJ, 1995: Electroejaculation: a welfare issue. Surveillance22, 14–17.Stro¨m Holst B, Bergstro¨m A, Lagerstedt AS, Karlstam E,Englund L, Ba˚ verud V, 2003: Characterization of thebacterial population of the genital tract of adult cats. AmJ Vet Res 64, 963–968.Swanson WF, Godke RA, 1994: Transcervical embryo transfer<strong>in</strong> the domestic cat. Lab Anim Sci 44, 228–291.Swanson WF, Roth TL, Wildt DE, 1994: In vivo embryogenesis,embryo migration, and embryonic mortality <strong>in</strong> thedomestic cat. Biol Reprod 51, 452–464.Swanson WF, Magarey GM, Herrick JR, 2007: Spermcryopreservation <strong>in</strong> endangered felids: develop<strong>in</strong>g l<strong>in</strong>kageof <strong>in</strong> situ–ex situ populations. Soc Reprod Fertil Suppl 65,417–432.Tanaka A, Takagi Y, Nakagawa K, Fujimoto Y, Hori T,Tsutsui T, 2000: Artificial <strong>in</strong>travag<strong>in</strong>al <strong>in</strong>sem<strong>in</strong>ation us<strong>in</strong>gfresh semen <strong>in</strong> cats. J Vet Med Sci 62, 1163–1167.Therien I, Soubeyrand S, Manjunath P, 1998: Major prote<strong>in</strong>sof bov<strong>in</strong>e sem<strong>in</strong>al plasma and high-density lipoprote<strong>in</strong><strong>in</strong>duce cholesterol efflux from epdidiymal sperm. BiolReprod 59, 768–776.Thomassen R, Sanson G, Krogenaes A, Fougner JA, BergKA, Farstad W, 2006: Artificial <strong>in</strong>sem<strong>in</strong>ation with frozensemen <strong>in</strong> dogs: a retrospective study of 10 years us<strong>in</strong>g a nonsurgicalapproach. Theriogenology 66, 1645–1650.Thuwanut P, Chatdarong K, Techakumphu M, Axne´r E,2008: The effect of antioxidants on motility, viability,acrosome <strong>in</strong>tegrity and DNA <strong>in</strong>tegrity of frozen-thawedepididymal cat spermatozoa. Theriogenology, doi: 10.1016/j.theriogenology.2008.04.005.Tsutsui T, 2006: Artificial <strong>in</strong>sem<strong>in</strong>ation <strong>in</strong> domestic cats (Feliscatus). Theriogenology 66, 122–125.Tsutsui T, Tanaka A, Hori T, 2001: Intratubal <strong>in</strong>sem<strong>in</strong>ationwith fresh semen <strong>in</strong> cats. J Reprod Fertil Suppl 57, 347–351.Tsutsui T, Wada M, Anzai M, Hori T, 2003: Artificial<strong>in</strong>sem<strong>in</strong>ation with frozen-thawed epididymal sperm <strong>in</strong> cats. JVet Med Sci 65, 397–399.Tsutsui T, Nakagawa K, Hirano T, Nagakubo K, Sh<strong>in</strong>omiyaM, Yamamoto K, Hori T, 2004a: Breed<strong>in</strong>g season <strong>in</strong> femalecats acclimated under a natural photoperiod and <strong>in</strong>tervaluntil puberty. J Vet Med Sci 66, 1129–1132.Tsutsui T, Kuwabara S, Kuwabara K, Kugota Y, K<strong>in</strong>jo T,Hori T, 2004b: Development of spermatogenic function <strong>in</strong>the sex maturation process <strong>in</strong> male cats. J Vet Med Sci 66,1125–1127.Zambelli D, Castagnetti C, 2001: Transcervical <strong>in</strong>sem<strong>in</strong>ationwith fresh or frozen semen <strong>in</strong> the domestic cat: newtechnique and prelim<strong>in</strong>ary results. In: Proc. 5th AnnualConf. ESDAR. Vienna, September, p. 34.Zambelli D, Buccioli M, Castagnetti C, Belluzzi S, 2004:Vag<strong>in</strong>al cervical anatomic modification dur<strong>in</strong>g the oestruscycle <strong>in</strong> relation to transcervical catheterization <strong>in</strong> thedomestic cat. Reprod Domest Anim 39, 76–80.Zambelli D, Cunto M, Prati F, Merlo B, 2007: Effects ofketam<strong>in</strong>e or medetomid<strong>in</strong>e adm<strong>in</strong>istration on quality ofelectroejaculated sperm and on sperm flow <strong>in</strong> the domesticcat. Theriogenology 68, 796–803.Author’s address (for correspondence): Eva Axne´r, Division of<strong>Reproduction</strong>, Department of Cl<strong>in</strong>ical Sciences, Swedish Universityof Agricultural Sciences, Uppsala, Sweden. E-mail: eva.axner@og.slu.seConflict of <strong>in</strong>terest: The author declares no conflict of <strong>in</strong>terest.Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 150–156 (2008); doi: 10.1111/j.1439-0531.2008.01155.xISSN 0936-6768<strong>Reproduction</strong> of the Female Ferret (Mustela putorius furo)H L<strong>in</strong>debergUniversity of Kuopio, Department of Biosciences, Kuopio, F<strong>in</strong>landContentsThe domestic ferret is a seasonally polyoestrous species.Females reach puberty at the age of 8–12 months. Femalesexhibit a constant oestrus between late March and earlyAugust if they are not bred. Increas<strong>in</strong>g tumescence <strong>in</strong> thep<strong>in</strong>k-coloured vulva is a sign of pro-oestrus. Oestrus canpersist for up to 5 months, but once ovulation is <strong>in</strong>duced,either pregnancy or pseudopregnancy ensues. Folliculardevelopment and atresia overlap <strong>in</strong> such a manner that thereis a recent cohort of follicles available for ovulation whenevercopulation might occur. Copulation may last from 15 m<strong>in</strong> to3 h, the average be<strong>in</strong>g 1 h. Ovulation is <strong>in</strong>duced by pressureon the cervix associated with copulation. After sufficient LHrelease, the pre-ovulatory follicles mature and an average of12 oocytes (5–13) per female are ovulated 30–40 h aftercopulation <strong>in</strong>to the ovarian bursa. The ferret oocytes aremost capable of be<strong>in</strong>g fertilized up to 12 h after ovulation,i.e. 42–52 h after copulation. Ferret oocytes ovulate at themetaphase of the second meiotic division (MII) embedded <strong>in</strong>three layers of corona radiata cells. Embryos enter the uterusover a period of several days start<strong>in</strong>g on day 5 after mat<strong>in</strong>g.Between days 12 and 13 after mat<strong>in</strong>g, the embryos havebecome implanted <strong>in</strong> the endometrium. Implantation <strong>in</strong> theferret is central with rapid <strong>in</strong>vasion of the uter<strong>in</strong>e epitheliumby the trophoblast over a broad area that eventually becomesa zonary band of endotheliochorial placenta. Gestationlength is 41 days (39–42 days). The domestic ferret givesbirth to an average of eight kits (1–18 kits), which weigh6–12 g at birth.IntroductionMustelids are polytocous carnivorous species demonstrat<strong>in</strong>ga variety of unique reproductive characteristics.The reproductive physiology of some species has beenquite thoroughly studied, while less is known aboutother species. This literature review was partly publishedas the PhD thesis of the author (L<strong>in</strong>deberg 2003), and itconcentrates on the results obta<strong>in</strong>ed with the domesticferret. For comparative purposes, when necessary,results concern<strong>in</strong>g other mustelids and carnivores arealso <strong>in</strong>cluded.Photoperiods and seasonalityThe domestic ferret is considered to be a seasonallypolyoestrous species, but females exhibit a constantoestrus between late March and early August if they arenot bred (Mead 1989). Marshall (1904) classified the ferretas a mono-oestrous species. Offspr<strong>in</strong>g born the previoussummer reach puberty by the follow<strong>in</strong>g spr<strong>in</strong>g at the ageof 8–12 months (Fox and Bell 1998). Initiation of thegonadal activity is totally dependent on the light-darkcycle, which stimulates or <strong>in</strong>hibits reproduction throughtransmission of <strong>in</strong>formation about the day length to thebra<strong>in</strong> (Bissonnette 1932; Turek and Van Cauter 1988).The ferret’s gonadal response to a given photoperioddepends both on the duration of the photoperiod andon the previous photoperiodic experience. Ferrets, likeother ‘long-day’ seasonal breeders, need alternat<strong>in</strong>gperiods of <strong>in</strong>creas<strong>in</strong>g duration of daylight (long days),at which time they are sensitive to light, and days ofdecreas<strong>in</strong>g duration of daylight (short days), dur<strong>in</strong>gwhich time they are <strong>in</strong>sensitive to light, so that theannual cycle recurs normally (Herbert 1989). The stateof temporary unresponsiveness to photic stimuli isreferred to as the ‘photorefractory’ condition (Elliottand Goldman 1981), and it is required to <strong>in</strong>duce areturn to the photosensitive state (Forsberg 1992).Ferrets exposed to artificially chang<strong>in</strong>g long and shortdaylight conditions (long days: 16 h light and 8 hdarkness; short days: 8 h light and 16 h darkness) startshow<strong>in</strong>g oestrus approximately 3 weeks after the changefrom short days to long days (Fox and Bell 1998), andcease show<strong>in</strong>g oestrus about the same time after achange from long days to short days. The repeatedchange from long days to short days after every6 months causes one period of gonadal activity peryear similar to a natural breed<strong>in</strong>g season when theanimals are exposed to natural outdoor light conditions.A change <strong>in</strong> light conditions every 4 or 2 months causestwo or three periods of gonadal activity a year,respectively (Herbert 1989). Through the use of reverselight cycles, ferrets can be <strong>in</strong>duced to breed any timedur<strong>in</strong>g the year (Harvey and MacFarlane 1958).Oestrous cycle and vag<strong>in</strong>al cytologyThe <strong>in</strong>creas<strong>in</strong>g tumescence <strong>in</strong> the p<strong>in</strong>k-coloured vulva isa sign of pro-oestrus <strong>in</strong> the ferret. The vulva enlarges upto 50 times its normal size dur<strong>in</strong>g a 2- to 3-week periodat the beg<strong>in</strong>n<strong>in</strong>g of the breed<strong>in</strong>g season (Hammond andMarshall 1930), which extends from March to August(Marshall 1904). No change <strong>in</strong> the turgidity of the vulvaoccurs up to 36 h after copulation (Hammond andWalton 1934), but 3 or 4 days after mat<strong>in</strong>g, the vulvastarts regress<strong>in</strong>g and rega<strong>in</strong>s its normal size <strong>in</strong>2–3 weeks. If the vulva does not recede, ovulation hasprobably not taken place, and the female may need to beremated (Lagerqvist 1992). Oestrus can persist for up to5 months, but once ovulation is <strong>in</strong>duced, either pregnancyor pseudopregnancy ensues (Hammond andMarshall 1930).In conjunction with the vulval swell<strong>in</strong>g dur<strong>in</strong>g oestrus,there is a thicken<strong>in</strong>g of the uter<strong>in</strong>e endometrium, andfollicles develop <strong>in</strong> the ovaries. The pattern of folliculardevelopment dur<strong>in</strong>g a prolonged oestrus is unknown,but it is assumed to be cont<strong>in</strong>uous (Rob<strong>in</strong>son 1918). Inthe absence of copulation, which results <strong>in</strong> a prolongedÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


<strong>Reproduction</strong> of <strong>Domestic</strong> Ferret 151oestrus, a cohort of follicles develops, degenerates and isreplaced by a new cohort of follicles. Folliculardevelopment and atresia overlap <strong>in</strong> such a manner thatthere is a recent cohort of follicles available forovulation whenever copulation might occur (Murphy1989). After copulation, some pre-ovulatory folliclesmay persist <strong>in</strong> the ovaries and lute<strong>in</strong>ise, but they do notrupture (Rob<strong>in</strong>son 1918). The <strong>in</strong>terval required fordevelopment from primordial to pre-ovulatory folliclehas not been reported for any mustelid. It has beenproposed that the time required for a wave of follicles todevelop prior to <strong>in</strong>duced ovulation <strong>in</strong> ferrets isapproximately 6 days (Murphy 1989).Oestradiol secreted by the follicles controls vulvalswell<strong>in</strong>g, uter<strong>in</strong>e development, sexual receptivity andchanges <strong>in</strong> vag<strong>in</strong>al cells (Hamilton and Gould 1940).Ferrets with vulval diameter greater than 1 cm havemated successfully (Murphy 1989). Vag<strong>in</strong>al cytology iscommonly used to detect oestrus <strong>in</strong> some domesticcarnivores, especially dogs (Olson et al. 1984), and thetechnique has been described for domestic ferrets(Hamilton and Gould 1940; Williams et al. 1992), m<strong>in</strong>k(Mustela vison) (Hansson 1947) and silver foxes (Vulpesvulpes) (Jalkanen et al. 1988). In ferrets, pro-oestrus ischaracterized by an <strong>in</strong>creas<strong>in</strong>g percentage of superficialepithelial cells together with enlargement of the vulva.Dur<strong>in</strong>g oestrus, usually >90% of epithelial cells aresuperficial cells, and after several days these cells arefully kerat<strong>in</strong>ized. Neutrophils are common dur<strong>in</strong>g allstages of the oestrous cycle (Williams et al. 1992).Follow<strong>in</strong>g copulation, which may last for 15 m<strong>in</strong> up to3 h (Hammond and Walton 1934), the average be<strong>in</strong>g 1 h(Fox and Bell 1998), the percentage of superficial cells <strong>in</strong>the vag<strong>in</strong>a decl<strong>in</strong>es with vulval swell<strong>in</strong>g. Dur<strong>in</strong>g mat<strong>in</strong>g,the male grips the female’s neck and makes repeatedpelvic thrusts until <strong>in</strong>tromission is achieved (Carrollet al. 1985).OvulationOvulation is non-spontaneous, i.e. it is <strong>in</strong>duced bypressure on the cervix associated with natural copulation,lead<strong>in</strong>g to endogenous LH release (Carroll et al.1985), or may be <strong>in</strong>duced artificially by hCG treatment(Mead et al. 1988a). The <strong>in</strong>crease <strong>in</strong> LH concentrationafter copulation is modest (only 3- to 4-fold elevation),and dur<strong>in</strong>g oestrus, LH does not exhibit the usualmammalian pattern of a pulsatile release (Carroll et al.1985; Tritt 1986). After sufficient LH release, the preovulatoryfollicles mature and an average of 12 oocytes(5–13) per female are ovulated <strong>in</strong>to the ovarian bursa30–40 h after copulation (Hammond and Walton 1934;Chang and Yanagimachi 1963; Carroll et al. 1985).The ferret ovary is encapsulated <strong>in</strong> a fatty bursacompletely enclos<strong>in</strong>g the ovary, so that the ovulatedoocytes cannot be shed <strong>in</strong>to the abdom<strong>in</strong>al cavity(Marshall 1904). The oocytes rema<strong>in</strong> capable of be<strong>in</strong>gfertilized for a period of 30–36 h after ovulation(Hammond and Walton 1934; Chang and Yanagimachi1963), but mat<strong>in</strong>g dur<strong>in</strong>g a period of 18–30 h afterovulation produce small litters (1–3 kits) (Hammondand Walton 1934). If the number of penetrated oocytesis considered as evidence of fertilization, the ferretoocytes are most capable of be<strong>in</strong>g fertilized up to 12 hafter ovulation, i.e. 42–52 h after copulation (Changand Yanagimachi 1963). Spermatozoa are found <strong>in</strong> theovarian bursa 6 hours after copulation <strong>in</strong> the ferret(Hammond and Walton 1934) or one hour aftercopulation <strong>in</strong> the stoat (Mustela erm<strong>in</strong>ea) (Amstislavsky,personal communication), but they require 3.5–11.5 h of capacitation time <strong>in</strong> the female reproductivetract to become fertile (Chang and Yanagimachi 1963).The spermatozoa apparently reta<strong>in</strong> their capability to<strong>in</strong>duce fertilization for up to 126 h after copulation(Chang 1965b), but the rate of fertilization depends onthe short lifespan of the oocytes. The exact site wherefertilization takes place is not known (Murphy 1989),although the middle-third of the oviduct (Rob<strong>in</strong>son1918) has been suggested. Accord<strong>in</strong>g to Chang andYanagimachi (1963), ferrets may sometimes ovulatespontaneously when they are handled. Generally,however, pre-<strong>in</strong>tromission events do not lead to any<strong>in</strong>crease <strong>in</strong> LH, and thus do not <strong>in</strong>duce ovulation(Carroll et al. 1985).Oocyte maturation and fertilizationAfter copulation, a nuclear maturation process is<strong>in</strong>itiated, and the formation of the first polar body andthe appearance of the second maturation sp<strong>in</strong>dle occur(Hamilton 1934). Without coital stimuli, ferret oocytesrema<strong>in</strong> immature <strong>in</strong> the germ<strong>in</strong>al vesicle stage until theydegenerate. Ferret oocytes ovulate at the metaphase ofthe second meiotic division (MII) (Ma<strong>in</strong>land 1931; Pilttiet al. 2003) embedded <strong>in</strong> three layers of corona radiatacells (Chang 1950), which detach after fertilization. Apositive correlation between the naked appearance offerret oocytes and fertilization has been reported (Chang1965b). Parthenogenetic cleavage of oocytes, i.e. theformation and division of blastomeres <strong>in</strong> the absence offertilization, is common up to 2–6 cells, and may occur<strong>in</strong> 43–60% of the unfertilized oocytes, most likely as aresult of oocyte age<strong>in</strong>g (Chang 1950, 1957). Themajority of unfertilized oocytes are embedded <strong>in</strong> thecorona radiata even 4–5 days after sterile mat<strong>in</strong>g(Chang and Yanagimachi 1963). If the oocyte isfertilized, the entire spermatozoon (head together withtail) passes through the zona pellucida (Hamilton 1934).The first pronuclei are discovered 40 ¾–43½ h aftercopulation (Ma<strong>in</strong>land 1930; Hamilton 1934; Chang andYanagimachi 1963; Piltti et al. 2003). The second polarbody is extruded soon after oocyte penetration by aspermatozoon, as <strong>in</strong> most other mammals (Hamilton1934, Farstad et al. 2001). In the ferret, the morphologicaldifferences between the first and the second polarbody are not dist<strong>in</strong>ctive (Chang 1965a). The number ofpolar bodies that may change their location <strong>in</strong> thedist<strong>in</strong>ct perivitell<strong>in</strong>e space (Hamilton 1934) varies fromtwo to four (Ma<strong>in</strong>land 1931; Chang and Yanagimachi1963), <strong>in</strong>dicat<strong>in</strong>g that <strong>in</strong> both fertilized and unfertilizedoocytes the first polar body may either frequently divide(Chang and Yanagimachi 1963) or become fragmented.The thickness of the zona pellucida varies from 2 to20 lm (Ma<strong>in</strong>land 1932; Chang 1950). The size of anunfertilized oocyte without a corona radiata has beenreported to be between 140 and 160 lm (Hamilton 1934;Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


152 H L<strong>in</strong>debergChang 1950). The oocyte is spherical or ovoid; itsnucleus is large, round and eccentric with one or, rarely,two nucleoli (Rob<strong>in</strong>son 1918). It is rich <strong>in</strong> lipid particles,which <strong>in</strong> one-half of the oocyte are less numerous than<strong>in</strong> the other half, mak<strong>in</strong>g the oocyte look less dense <strong>in</strong>this half (Hamilton 1934) and mak<strong>in</strong>g it easier tovisualize the nuclear structures on the less dense half ofthe oocyte. This seems to be particular to the ferretoocyte compared with other lipid-rich oocytes observed<strong>in</strong> other carnivorous species, such as the can<strong>in</strong>e andfel<strong>in</strong>e oocytes.Early embryonic developmentThe first cleavage produces two blastomeres that aresimilar <strong>in</strong> appearance but unequal <strong>in</strong> size. The first twoblastomeres divide asynchronously, so that furtherstages of uneven numbers of blastomeres are produced(Hamilton 1934). Asynchronous divisions are a commonfeature for carnivore embryos (Amstislavsky et al. 1993;L<strong>in</strong>deberg et al. 1993) but not for embryos of rum<strong>in</strong>ants(Betteridge and Fle´ chon 1988). A rough estimation ofthe <strong>in</strong>tervals between the first and second, and thesecond and third cleavages was 10–16 h for each stage(Chang 1965b). Asynchronous division of the cells anddifferences <strong>in</strong> sizes are detected up to the 16-cell stage,but no morphological differences can be detectedbetween the central (<strong>in</strong>ner cell mass) cells and thesurround<strong>in</strong>g (trophoectoderm) cells at the morula stage,<strong>in</strong> which the cells are grouped closely together and adist<strong>in</strong>ct perivitell<strong>in</strong>e space is present. Fat is equallydistributed among the cells (Hamilton 1934).The degree of development of embryos of the sameage varies considerably (Rider and Heap 1986). At thesame time after mat<strong>in</strong>g, embryos from one ferret can beat the blastocyst stage, whereas those of another ferretmay still be at the morula stage (Chang 1969).Blastocysts may be further developed than those <strong>in</strong>which a longer time <strong>in</strong>terval has elapsed after mat<strong>in</strong>g.Blastocysts appear to be almost spherical and completelyfill the zona pellucida, which has become th<strong>in</strong>ned bythe presence of the expand<strong>in</strong>g blastocyst. The <strong>in</strong>ner cellmass appears as a dark mass at one pole of theblastocyst. The flattened cells of the trophoectodermand the <strong>in</strong>ner cell mass are dotted with fatty granules(Rob<strong>in</strong>son 1925; Hamilton 1934). Uter<strong>in</strong>e blastocystsexpand from a size of 200 lm <strong>in</strong> diameter to more than2 mm <strong>in</strong> diameter dur<strong>in</strong>g their pre-implantation development(Daniel 1970; Enders and Schlafke 1972).Progesterone has been reported to support blastocystexpansion up to a diameter of 1 mm, but for furtherblastocyst expansion additional ovarian factors arerequired (McRae 1992). Without the presence ofmaternal progesterone, neither cleavage of embryos <strong>in</strong>the oviducts or <strong>in</strong> the uterus (Rider and Heap 1986) norexpansion of blastocysts <strong>in</strong> the uterus take place(Buchanan 1969; McRae 1992).Oviductal passageFerret embryos enter the uterus over a period of severaldays start<strong>in</strong>g from day 5 after mat<strong>in</strong>g. The embryosstart enter<strong>in</strong>g the uterus on day 4, if females are treatedwith hCG (Chang 1969), and almost all embryos arefound <strong>in</strong> the uterus by day 7 as described <strong>in</strong> studies ofRob<strong>in</strong>son (1918), Hammond and Walton (1934), whoanalyzed Rob<strong>in</strong>son’s (1918) data and Hamilton’s(1934). L<strong>in</strong>deberg and Ja¨ rv<strong>in</strong>en (2003) reported thatuntil day 5 after the first mat<strong>in</strong>g, embryos of farmedEuropean polecats (Mustela putorius) were recoveredonly from the oviducts. Between days 6 and 9, embryoswere recovered both from the oviducts and the uteri.From day 10 onwards, the embryos were recoveredonly from the uteri. Ferret pre-implantation embryos(Fig. 1a and b) experience a prolonged period ofoviductal residence – a phenomenon that has also beendemonstrated <strong>in</strong> the cat (Swanson et al. 1994), the bluefox (Valtonen et al. 1985; Valtonen and Jalkanen 1993),the silver fox (Jalkanen 1992), the dog (Holst andPhemister 1971; Renton et al. 1991) and the horse(Betteridge et al. 1982). After unilateral or bilateralembryo transfer, ferret pre-implantation embryos havebeen reported to migrate from one horn to another (Liet al. 2006).ImplantationThree types of pregnancy have been identified <strong>in</strong> theMustelidae (Mead 1989; Ternovsky and Ternovskaya1994). All polecat species (Mustela putorius, Mustelaputorius furo, Mustela eversmanni, Mustela lutreola)have a short period of pregnancy of constant duration(37–44 days). Species such as the stoat (Mustelaerm<strong>in</strong>ea) and the sable (Martes zibell<strong>in</strong>a) exhibit anobligatory embryonic diapause at the blastocyst stageand a long gestation period (7–10 months) (Amstislavskyand Ternovskaya 2000). In the American m<strong>in</strong>k(Mustela vison), the gestation period is short butvariable (range 45–61 days), and may or may not<strong>in</strong>clude implantation delay (Mead 1989).Implantation <strong>in</strong> the ferret is central with rapid<strong>in</strong>vasion of the uter<strong>in</strong>e epithelium by the trophoblast(a)(b)Fig. 1. (a) Freshly flushed day-9blastocysts; (b) Frozen-thawedday-9 blastocysts <strong>in</strong> the farmedEuropean polecatÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


<strong>Reproduction</strong> of <strong>Domestic</strong> Ferret 153over a broad area that eventually becomes a zonaryband of endotheliochorial placenta (Strahl and Ballman1915; cited <strong>in</strong> Enders and Schlafke 1972). The endotheliochorialplacenta has three foetal (endothelium, connectivetissue, trophoblast) but only two maternal layers(connective tissue, endothelium) because the maternalepithelium is lost. The foetal trophoblast <strong>in</strong>vades theendometrial epithelium, but does not destroy theendothelium of the maternal capillaries (Mossman1987; Valtonen 1992). Between days 12 and 13 aftermat<strong>in</strong>g, the embryos are implanted <strong>in</strong> the endometrium(Enders and Schlafke 1972; Mead et al. 1988b).Prior to implantation, the trophoblast differentiatesrapidly and gives rise to patches of syncytial trophoblast(Gulamhuse<strong>in</strong> and Beck 1973). The first feature ofpenetration is the projection of a th<strong>in</strong> fold of syncytialtrophoblast between adjacent epithelial cells. As implantationprogresses, more of the blastocyst is <strong>in</strong>volved<strong>in</strong> implantation, and by day 14, a cont<strong>in</strong>uous sheet,either penetrat<strong>in</strong>g or overly<strong>in</strong>g the area of the wall of theuterus, constitutes two-thirds of the circumference of thefuture zonary band placenta (Enders and Schlafke1972). At the cervical and ovarian ends of the chorionicvesicle and <strong>in</strong> the mesometrial region, the trophoectodermis non-<strong>in</strong>vasive (Gulamhuse<strong>in</strong> and Beck 1975). Inthese non-attached regions, the trophoectodermic cellsabsorb uter<strong>in</strong>e milk (Gulamhuse<strong>in</strong> and Beck 1973). Inthe immediate vic<strong>in</strong>ity of implant<strong>in</strong>g chorionic vesicles, ahighly localized <strong>in</strong>crease <strong>in</strong> the permeability of uter<strong>in</strong>eblood vessels is associated with the f<strong>in</strong>al stage ofattachment to the uter<strong>in</strong>e epithelium, this be<strong>in</strong>g firstdetectable <strong>in</strong> the morn<strong>in</strong>g of day 12 after mat<strong>in</strong>g (Meadet al. 1988b). Prostagland<strong>in</strong>s are proposed to play animportant role <strong>in</strong> the process of implantation, but theprocess is unrelated to decidual formation because theferret is an adeciduate species (Mead et al. 1988b); thatis, ferret endometrium is not known to be capable ofdecidual transformation dur<strong>in</strong>g implantation (Beck1974) <strong>in</strong> contrast to the situation of rodents and humansthat have a primary decidualization reaction beforeblastocyst(s) start penetrat<strong>in</strong>g the uter<strong>in</strong>e epithelium(Johnson and Everitt 2000). Follow<strong>in</strong>g implantation, awave of epithelial hypertrophy sweeps progressivelyfrom the uter<strong>in</strong>e lumen towards the bases of the glands,and epithelial cells become extraord<strong>in</strong>arily enlarged withnuclei as large as 90–100 lm <strong>in</strong> diameter. Most of thelum<strong>in</strong>al cells lose their <strong>in</strong>tegrity and form massesconta<strong>in</strong><strong>in</strong>g whole or fragmented nuclei. This degeneratetissue is termed a symplasma because, although technicallyit is a syncytium, it is not an active tissue (Amoroso1952; Buchanan 1966). This degenerated tissue probablycontributes to the histiotrophe because it is <strong>in</strong>gested bythe syncytiotrophoblast (Gulamhuse<strong>in</strong> and Beck 1975).In the ferret, the placental labyr<strong>in</strong>th is fully developedat day 18, when the greatly hypertrophied maternalcapillaries are completely surrounded by a layer ofsyncytiotrophoblast (Gulamhuse<strong>in</strong> and Beck 1975). Atthe same time, accumulations of maternal blood, whichvary considerably <strong>in</strong> size and location and constitute the‘haemophagous organ’ (Creed and Biggers 1964),appear <strong>in</strong> the antimesometrial region between theplacental discs. This organ is fully formed by day 28,and it ma<strong>in</strong>ta<strong>in</strong>s its size almost to term (Gulamhuse<strong>in</strong>and Beck 1975). It is thought to act as an alternativesource of iron for the foetuses (Baker and Morgan1973).Gestation and function of corpora luteaGestation length is 41 days (39–42 days) <strong>in</strong> the domesticferret (Hammond and Marshall 1930; Ternovsky andTernovskaya 1994; Fox and Bell 1998). If fertilizationdoes not occur, pseudopregnancy last<strong>in</strong>g 40–42 daysensues, the functional life of corpora lutea (CL) be<strong>in</strong>gsimilar to that <strong>in</strong> normal pregnancy (Hammond andMarshall 1930; Chang and Yanagimachi 1963). FerretCL start secret<strong>in</strong>g progesterone immediately afterovulation, and the progesterone concentrations risecont<strong>in</strong>uously to peak at approximately days 12–14dur<strong>in</strong>g the period of implantation (Daniel 1976), thendecrease steadily after approximately day 15 and leveloff by day 24 of pregnancy (Heap and Hammond 1974).Ferret CL consist of small (25 lm) luteal cells, with the smaller cells predom<strong>in</strong>at<strong>in</strong>gon day 6. A shift towards larger sizes is observed bydays 13 and 24 of pregnancy, and concurrently thepercentage of smaller cells decl<strong>in</strong>es (Joseph and Mead1988). Progesterone concentrations decl<strong>in</strong>e cont<strong>in</strong>uouslybetween day 24 and parturition at day 42 (Blatchley andDonovan 1976). Mustelids and other carnivores displaya protracted decl<strong>in</strong>e <strong>in</strong> circulat<strong>in</strong>g progesterone, andprogesterone levels reach basal concentrations ‡1 weekafter parturition (Møller 1973). The conceptuses haveno effect on the duration of the luteal phase, becausepregnancy and pseudopregnancy are <strong>in</strong>dist<strong>in</strong>guishable(Hammond and Marshall 1930; Heap and Hammond1974; Agu et al. 1986). Removal of the uterus dur<strong>in</strong>g theluteal phase has no effect on the life span of the CL(Deanesly 1967). Factors caus<strong>in</strong>g luteal regression <strong>in</strong> theferret are not known.The functional ferret CL secrete glucose-6-phosphateisomerase on days 6–9 of pregnancy, the time at whichimplantation-promot<strong>in</strong>g activity has been found <strong>in</strong>corpora lutea. This isomerase has been identified to benecessary for embryo implantation <strong>in</strong> the domestic ferret(Schulz and Bahr 2003). It seems that oestrogen is notessential for implantation <strong>in</strong> the ferret (Foresman andMead 1978; Mead and McRae 1982). Experimentally<strong>in</strong>duced maternal pregnancy reactions (for <strong>in</strong>stance,traumatization with an <strong>in</strong>trauter<strong>in</strong>e thread that results<strong>in</strong> the formation and subsequent necrosis of symplasmalnests of endometrial epithelial cells and hypertrophy ofthe maternal capillary endothelium) show that theendometrium is sensitive to implantation between days9 and 14 (Gulamhuse<strong>in</strong> and Beck 1977).Parturition, rebreed<strong>in</strong>g, <strong>in</strong>duction of oestrus andsuperovulationThe domestic ferret gives birth to an average of eightkits (1–18 kits), which weigh 6–12 g at birth (Ternovskyand Ternovskaya 1994; Fox and Bell 1998). Females willreturn to oestrus with<strong>in</strong> 2 weeks after wean<strong>in</strong>g if theyare exposed to a stimulatory photoperiod. If the kits areremoved at birth, the mothers return to oestrus 8 weeksafter mat<strong>in</strong>g, as do pseudopregnant animals and femalesÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


154 H L<strong>in</strong>debergthat have resorbed their foetuses (Marston and Kelly1969). If females give birth to only a small number ofkits (e.g. 1–5), they may return to oestrus while nurs<strong>in</strong>g(Fox and Bell 1998). Induction of oestrus <strong>in</strong> anoestrousferrets has been accomplished by treatment with crudepituitary extracts (Hill and Parkes 1930). Most anoestrousfemales ma<strong>in</strong>ta<strong>in</strong>ed on a non-stimulatory lightcycle (10 h light and 14 h dark) showed oestrous signs,and were bred 6–13 days after treatment with 0.25 mgof FSH, adm<strong>in</strong>istered twice daily (Mead and Neir<strong>in</strong>ckx1989). For successful superovulation, ferret females havebeen treated with eCG followed by hCG 72 h later (Liet al. 2001, 2002).ReferencesAgu GO, Rajkumar K, Murphy BD, 1986: Evidence fordopam<strong>in</strong>ergic regulation of prolact<strong>in</strong> and a luteotropiccomplex <strong>in</strong> the ferret. Biol Reprod 35, 508–515.Amoroso EC, 1952: Placentation. In: Parkes AS (ed.),Marshall’s Physiology of <strong>Reproduction</strong>, vol 2. Longman,London, pp. 127–311.Amstislavsky SY, Ternovskaya Y, 2000: <strong>Reproduction</strong> <strong>in</strong>mustelids. Anim Reprod Sci 60–61, 571–581.Amstislavsky SY, Maksimovsky LF, Ternovsky YG, TernovskyDV, 1993: Erm<strong>in</strong>e reproduction and embryodevelopment (Mustela erm<strong>in</strong>ea). Scientifur 17, 293–298.Baker E, Morgan EH, 1973: Placental iron transfer <strong>in</strong> the cat.J Physiol 232, 485–501.Beck F, 1974: The development of a maternal pregnancyreaction <strong>in</strong> the ferret. J Reprod Fertil 40, 61–69.Betteridge KJ, Fle´ chon JE, 1988: The anatomy and physiologyof pre-attachment bov<strong>in</strong>e embryos. Theriogenology 29, 155–187.Betteridge KJ, Eaglesome MD, Mitchell D, Flood PF, Be´ riaultR, 1982: Development of horse embryos up to twenty daysafter ovulation: observations on fresh specimens. J Anat135, 191–209.Bissonnette TH, 1932: Modification of mammalian sexualcycles; reactions of ferrets (Putorius vulgaris) of both sexes toelectric light added after dark <strong>in</strong> November and December.Proc R Soc Lond 110, 322–336.Blatchley FR, Donovan BT, 1976: Progesterone secretiondur<strong>in</strong>g pregnancy and pseudopregnancy <strong>in</strong> the ferret.J Reprod Fertil 46, 455–456.Buchanan GD, 1966: <strong>Reproduction</strong> <strong>in</strong> the ferret (Mustela furo)I. Uter<strong>in</strong>e histology and histochemistry dur<strong>in</strong>g pregnancyand pseudopregnancy. Am J Anat 118, 195–216.Buchanan GD, 1969: <strong>Reproduction</strong> <strong>in</strong> the ferret (Mustela furo)II. Changes follow<strong>in</strong>g ovariectomy dur<strong>in</strong>g early pregnancy.J Reprod Fertil 18, 305–316.Carroll RS, Ersk<strong>in</strong>e MS, Doherty PC, Lundell LA, Baum MJ,1985: Coital stimuli controll<strong>in</strong>g lute<strong>in</strong>iz<strong>in</strong>g hormone secretionand ovulation <strong>in</strong> the female ferret. Biol Reprod 32, 925–933.Chang MC, 1950: Cleavage of unfertilised ova <strong>in</strong> immatureferrets. Anat Rec 108, 31–44.Chang MC, 1957: Natural occurrence and artificial <strong>in</strong>ductionof parthenogenetic cleavage of ferret ova. Anat Rec 128,187–200.Chang MC, 1965a: Implantation of ferret ova fertilized bym<strong>in</strong>k sperm. J Exp Zool 160, 67–80.Chang MC, 1965b: Fertiliz<strong>in</strong>g life of ferret sperm <strong>in</strong> the femaletract. J Exp Zool 158, 87–100.Chang MC, 1969: Development of transferred ferret eggs <strong>in</strong>relation to the age of corpora lutea. J Exp Zool 171, 459–464.Chang MC, Yanagimachi R, 1963: Fertilization of ferret ovaby deposition of epididymal sperm <strong>in</strong>to the ovarian capsulewith special reference to the fertilizable life of ova and thecapasitation of sperm. J Exp Zool 154, 175–188.Creed RFS, Biggers JD, 1964: Placental haemophagous organs<strong>in</strong> the Procyonides and Mustelidae. J Reprod Fertil 8, 133–137.Daniel JC Jr, 1970: Co<strong>in</strong>cidence of embryonic growth anduter<strong>in</strong>e prote<strong>in</strong> <strong>in</strong> the ferret. J Embryol Exp Morphol 24,305–312.Daniel JC Jr, 1976: Plasma progesterone levels before and atthe time of implantation <strong>in</strong> the ferret. J Reprod Fertil 48,437–438.Deanesly R, 1967: Experimental observations on the ferretcorpus luteum of pregnancy. J Reprod Fertil 13, 183–185.Elliott JA, Goldman BD, 1981: Seasonal reproduction:photoperiodism and biological clocks. In: Adler NT (ed.),Neuroendocr<strong>in</strong>ology of <strong>Reproduction</strong>, Physiology andBehaviour. Plenum Press, New York, pp. 377–423.Enders AC, Schlafke S, 1972: Implantation <strong>in</strong> the ferret:epithelial penetration. Am J Anat 133, 291–316.Farstad W, Hyttel P, Hafne AL, Nielsen J, 2001: Maturationand fertilization of blue fox (Alopex lagopus) oocytes <strong>in</strong>vitro. J Reprod Fertil Suppl 57, 161–165.Foresman KR, Mead RA, 1978: Luteal control of nidation <strong>in</strong>the ferret (Mustela putorius). Biol Reprod 18, 490–496.Forsberg M, 1992: Seasonal breed<strong>in</strong>g and the significance oflight. In: Tauson A-H, Valtonen M (eds), <strong>Reproduction</strong> <strong>in</strong>Carnivorous Fur Bear<strong>in</strong>g <strong>Animals</strong>. Nordiska JordbruksforskaresFo¨ ren<strong>in</strong>g, Rapport Nr. 75, Jordbrugsforlaget,Nordisk Kulturfond, Copenhagen, pp. 17–38.Fox JG, Bell JA, 1998: Growth, reproduction and breed<strong>in</strong>g.In: Fox JG (ed.), Biology and Diseases of the Ferret, 2ndedn. Williams and Wilk<strong>in</strong>s, Baltimore, pp. 211–227.Gulamhuse<strong>in</strong> AP, Beck F, 1973: Light and electron microscopicobservations at the pre- and early post-implantationstages <strong>in</strong> the ferret uterus. J Anat 115, 159–174.Gulamhuse<strong>in</strong> AP, Beck F, 1975: Development and structure ofthe extra-embryonic membranes of the ferret. A lightmicroscopic and ultrastructural study. J Anat 120, 349–365.Gulamhuse<strong>in</strong> AP, Beck F, 1977: Determ<strong>in</strong>ation of a sensitiveperiod for the <strong>in</strong>duction of the maternal pregnancy reaction<strong>in</strong> the ferret. J Reprod Fertil 49, 127–128.Hamilton WJ, 1934: The early stages <strong>in</strong> the development of theferret: fertilisation to the formation of the prochordal plate.Trans R Soc Ed<strong>in</strong>b B 58, 251–278.Hamilton WJ, Gould GH, 1940: The normal oestrous cycle ofthe ferret: the correlation of the vag<strong>in</strong>al smear and thehistology of the genital tract, with notes on the distributionof glycogen, the <strong>in</strong>cidence of growth, and the reaction to<strong>in</strong>travitam sta<strong>in</strong><strong>in</strong>g by trypan blue. Trans R Soc Ed<strong>in</strong>b B60, 87–106.Hammond J, Marshall FHA, 1930: Estrus and pseudopregnancy<strong>in</strong> ferret. Proc R Soc Lond B Biol Sci 105, 607–630.Hammond J, Walton A, 1934: Notes on ovulation andfertilization <strong>in</strong> the ferret. J Exp Biol 14, 307–319.Hansson A, 1947: The physiology of reproduction <strong>in</strong> m<strong>in</strong>k(Mustela vison Schreb) with special reference to delayedimplantation. Acta Zool (Stockh) 28, 1–136.Harvey NE, MacFarlane WV, 1958: The effects of day lengthon the coat-shedd<strong>in</strong>g cycles, body weight, and reproductionof the ferret. Aust J Biol Sci 11, 187–199.Heap RB, Hammond J Jr, 1974: Plasma progesterone levels <strong>in</strong>pregnant and pseudopregnant ferret. J Reprod Fertil 39,149–152.Herbert J, 1989: Light as a multiple control system onreproduction <strong>in</strong> mustelids. In: Seal US, Thorne ET, BoganM, Anderson S (eds), Conservation Biology of theÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


<strong>Reproduction</strong> of <strong>Domestic</strong> Ferret 155Black-footed Ferret. Yale University Press, New Haven,CT, pp. 138–159.Hill M, Parkes AS, 1930: On the relation between the anteriorpituitary body and the gonads. Part II. The <strong>in</strong>duction ofovulation <strong>in</strong> the anoestrous ferret. Proc R Soc Lond B 107,39–49.Holst PA, Phemister RD, 1971: The prenatal development ofthe dog: preimplantation events. Biol Reprod 5, 194–206.Jalkanen L, 1992: Embryonal development and embryo lossesdur<strong>in</strong>g the preimplantation period <strong>in</strong> the silver fox. Norw JAgric Sci 9, 108–114.Jalkanen L, Valtonen M, Lukola A, 1988: Electrical resistanceand cytology of the vag<strong>in</strong>al tract <strong>in</strong> relation to ovarianhormone levels <strong>in</strong> the silver fox dur<strong>in</strong>g estrus. In: Proceed<strong>in</strong>gsof the 4th International Congress Fur Animal Production,Ontorio, Canada, pp. 5–9 .Johnson MH, Everitt BJ, 2000: Essential <strong>Reproduction</strong>, 5thedn. Blackwell Science Ltd, London, pp. 173–192.Joseph MM, Mead RA, 1988: Size distribution of ferret lutealcells dur<strong>in</strong>g pregnancy. Biol Reprod 39, 1159–1169.Lagerqvist G, 1992: Reproductive features <strong>in</strong> the ferret(Mustela putorius). In: Tauson A-H, Valtonen M (eds),<strong>Reproduction</strong> <strong>in</strong> Carnivorous Fur Bear<strong>in</strong>g <strong>Animals</strong>.Nordiska Jordbruksforskares Fo¨ ren<strong>in</strong>g, Rapport Nr. 75.Jordbrugsforlaget, Nordisk Kulturfond Copenhagen, pp.87–95.Li ZY, Jiang Q, Zhang YL, Liu XM, Engelhardt JF, 2001:Successful production of offspr<strong>in</strong>g after superovulation and<strong>in</strong> vitro culture of embryos from domestic ferrets (Mustelaputorius furos). <strong>Reproduction</strong> 122, 611–618.Li ZY, Jiang Q, Sabet MR, Zhang Y, Ritchie TC,Engelhardt JF, 2002: Conditions for <strong>in</strong> vitro maturationand artificial activation of ferret oocytes. Biol Reprod 66,1380–1386.Li ZY, Sun X, Chen J, Leno GH, Engelhardt JF, 2006:Factors affect<strong>in</strong>g the efficiency of embryo transfer <strong>in</strong> thedomestic ferret (Mustela putorius furo). Theriogenology 66,183–190.L<strong>in</strong>derberg H, 2003: Embryo technology <strong>in</strong> the farmedEuropean Povelat (Mustela Putorius). PhD thesis, Universityof Kuopio, pp. 1–110.L<strong>in</strong>deberg H, Ja¨ rv<strong>in</strong>en M, 2003: Early embryonic developmentand <strong>in</strong> vitro culture of <strong>in</strong> vivo produced embryos <strong>in</strong> thefarmed European polecat (Mustela putorius). Theriogenology60, 965–975.L<strong>in</strong>deberg H, Jalkanen L, Savola<strong>in</strong>en R, 1993: In vitro cultureof silver fox embryos. Theriogenology 40, 779–788.Ma<strong>in</strong>land D, 1930: The early development of the ferret: thePronuclei. J Anat 64, 262–287.Ma<strong>in</strong>land D, 1931: A quantitative study of the polar body ofthe ferret, with a note on the second polar sp<strong>in</strong>dle. AmJ Anat 47, 195–240.Ma<strong>in</strong>land D, 1932: The early development of the ferret: thezona granulosa, zona pellucida and associated structures.J Anat 66, 586–601.Marshall FHA, 1904: The estrous cycle <strong>in</strong> the common ferret.Quart J Microsc Sci 48, 323–345.Marston JH, Kelly WA., 1969: Contraceptive action of <strong>in</strong>trauter<strong>in</strong>edevices <strong>in</strong> the ferret. J Reprod Fertil 18, 419–429.McRae AC, 1992: Effect of ovariectomy on blastocystexpansion and survival <strong>in</strong> ferrets (Mustela putorius furo).Reprod Fertil Dev 4, 239–247.Mead RA, 1989: <strong>Reproduction</strong> <strong>in</strong> mustelids. In: Seal US,Thorne ET, Bogan MA, Anderson SH (eds), ConservationBiology and the Black-footed Ferret. Yale University Press,New Haven, CT, pp. 124–137.Mead RA, McRae M, 1982: Is estrogen required forimplantation <strong>in</strong> the ferret? Biol Reprod 27, 540–547.Mead RA, Neir<strong>in</strong>ckx S, 1989: Hormonal <strong>in</strong>duction of oestrusand pregnancy <strong>in</strong> anoestrous ferrets (Mustela putorius furo).J Reprod Fertil 86, 309–314.Mead RA, Joseph MM, Neir<strong>in</strong>ckx S, 1988a: Optimal dose ofhuman chorionic gonadotroph<strong>in</strong> for <strong>in</strong>duc<strong>in</strong>g ovulation <strong>in</strong>the ferret. Zoo Biol 7, 263–267.Mead RA, Bremner S, Murphy BD, 1988b: Changes <strong>in</strong>endometrial vascular permeability dur<strong>in</strong>g the periimplantationperiod <strong>in</strong> the ferret (Mustela putorius). J Reprod Fertil82, 293–298.Møller OM, 1973: The f<strong>in</strong>e structure of the lute<strong>in</strong> cells <strong>in</strong> them<strong>in</strong>k (Mustela vison) with special reference to the secretoryactivity dur<strong>in</strong>g pregnancy. Z Zellforsc Mikrosk Anat 138,523–544.Mossman HW, 1987: Vertebrate Fetal Membranes. RutgersUniversity Press, New Brunswick, NJ, pp. 250–262.Murphy BD, 1989: Reproductive physiology of femalemustelids. In: Seal US, Thorne ET, Bogan MA, AndersonSH (eds), Conservation Biology and the Black-footedFerret. Yale University Press, New Haven, CT, pp. 107–123.Olson PN, Thrall MA, Wykes PM, Husted PW, Nett TM,Sawyer HR, 1984: Vag<strong>in</strong>al cytology. Part I. A useful tool forstag<strong>in</strong>g the can<strong>in</strong>e estrous cycle. Compend Cont<strong>in</strong> EducPract 6, 288–297.Piltti K, Aalto J, Ja¨ rv<strong>in</strong>en M, Korhonen H, Kuronen V,L<strong>in</strong>deberg H, Amstislavsky S, Ternovskaya Y, Valtonen M,Halmekyto¨ M, 2003: Nuclear maturation of Europeanpolecat (Mustela putorius) <strong>in</strong> vivo oocytes and success <strong>in</strong>us<strong>in</strong>g mustelid hybrids as recipients for embryo transfer.Theriogenology 59, 403. (abstract).Renton JP, Boyd JS, Eckersall PD, Ferguson JM, HarveyMJA, Mullaney J, Perry B, 1991: Ovulation, fertilizationand early embryonic development <strong>in</strong> the bitch (Canisfamiliaris). J Reprod Fertil 93, 1–10.Rider V, Heap RB, 1986: Heterologous anti-progesteronemonoclonal antibody arrest early embryonic developmentand implantation <strong>in</strong> the ferret (Mustela putorius). J ReprodFertil 76, 459–470.Rob<strong>in</strong>son A, 1918: The formation, rupture, and closure ofovarian follicles <strong>in</strong> ferrets and ferret polecat hybrids, andsome associated phenomena. Trans R Soc Ed<strong>in</strong>b B 52, 303–362.Rob<strong>in</strong>son A, 1925: Lipoids <strong>in</strong> mammalian ova and theirzygotes. Proceed<strong>in</strong>gs of the Anatomical Society of GreatBrita<strong>in</strong> and Ireland; J Anat 59, 109–111.Schulz LC, Bahr JM, 2003: Glucose-6-phosphate isomerase isnecessary for embryo implantation <strong>in</strong> the domestic ferret.Proc Natl Acad Sci USA 100, 8561–8566.Strahl H, Ballman E, 1915: Embryonalhu¨ llen und Plazentavon Putorius furo. Abhand Ko¨ nigl Akademie derWissenschaften Physik-Mathem Classe Nr. 4. Berl<strong>in</strong>, pp.1–69Swanson WF, Roth TL, Wildt DE, 1994: In vivo embryogenesis,embryo migration, and embryonic mortality <strong>in</strong> thedomestic cat. Biol Reprod 51, 452–464.Ternovsky DV, Ternovskaya YG, 1994: Ecology of Mustelids.Scientific Siberian Press, Novosibirsk Nauka, <strong>in</strong> Russian.Tritt SH, 1986: Regulation of lute<strong>in</strong>iz<strong>in</strong>g hormone (LH)secretion <strong>in</strong> the estrous ferret. Biol Reprod 34 (Suppl. 1) 198.Turek FW, Van Cauter E, 1988: Rhythms <strong>in</strong> reproduction. In:Knobil E, Neill JD (eds), The Physiology of <strong>Reproduction</strong>.Raven Press, New York, pp. 1789–1830.Valtonen M, 1992: Comparative aspects of reproductivephysiology <strong>in</strong> domestic animals. In: Tauson A-H, ValtonenM(eds), <strong>Reproduction</strong> <strong>in</strong> Carnivorous Fur Bear<strong>in</strong>g <strong>Animals</strong>.Nordiska Jordbruksforskares Fo¨ ren<strong>in</strong>g, Rapport Nr. 75.Jordbrugsforlaget, Nordisk Kulturfond Copenhagen,pp. 1–15.Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


156 H L<strong>in</strong>debergValtonen M, Jalkanen L, 1993: Species-specific features ofoestrous development and blastogenesis <strong>in</strong> domestic can<strong>in</strong>especies. J Reprod Fertil Suppl 47, 133–137.Valtonen M, K<strong>in</strong>g WA, Gustavson I, Ma¨ k<strong>in</strong>en A, 1985:Embryonic development <strong>in</strong> the blue fox. Nord Vet Med 37,243–248.Williams ES, Thorne ET, Kwiatkowski DR, Lutz K,Anderson SL, 1992: Comparative vag<strong>in</strong>al cytology of theestrous cycle of black-footed ferrets (Mustela nigripes),Siberian polecats (M. eversmanni), and domestic ferret(M. putorius furo). J Vet Diagn Invest 4, 38–44.Author’s address (for correspondence): H L<strong>in</strong>deberg, University ofKuopio, Department of Biosciences, P.O. Box 1627, FIN-70211Kuopio, F<strong>in</strong>land. E-mail: l<strong>in</strong>deber@messi.uku.fiConflict of <strong>in</strong>terest: The author declares no conflict of <strong>in</strong>terest.Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 157–164 (2008); doi: 10.1111/j.1439-0531.2008.01156.xISSN 0936-6768Physiology of the Can<strong>in</strong>e Anoestrus and Methods for Manipulation of Its LengthJ. de Gier, NJ Beijer<strong>in</strong>k, HS Kooistra and AC OkkensDepartment of Cl<strong>in</strong>ical Sciences of Companion <strong>Animals</strong>, Faculty of Veter<strong>in</strong>ary Medic<strong>in</strong>e, University of Utrecht, Yalelaan, Utrecht, The NetherlandsContentsProgression from early to late anoestrus is characterized by theappearance of a larger number of gonadotroph<strong>in</strong>-releas<strong>in</strong>ghormone (GnRH) pulses with a higher amplitude, an <strong>in</strong>crease<strong>in</strong> the sensitivity of the pituitary to GnRH, an <strong>in</strong>crease <strong>in</strong>ovarian responsiveness to gonadotroph<strong>in</strong>s, and an <strong>in</strong>crease <strong>in</strong>basal plasma follicle-stimulat<strong>in</strong>g hormone (FSH) concentration.A period of <strong>in</strong>creased lute<strong>in</strong>iz<strong>in</strong>g hormone (LH) pulsatilityhas been observed shortly before the onset of pro-oestrus.Apart from these changes <strong>in</strong> the hypothalamus-pituitary-ovaryaxis, the <strong>in</strong>itiation of a new follicular phase <strong>in</strong> the bitch is alsostimulated by dopam<strong>in</strong>ergic <strong>in</strong>fluences other than the accompany<strong>in</strong>gplasma prolact<strong>in</strong> decrease. Metergol<strong>in</strong>e, a drug which<strong>in</strong> a low dosage lowers the plasma prolact<strong>in</strong> concentration viaa seroton<strong>in</strong>-antagonistic pathway, does not shorten theanoestrus; while bromocript<strong>in</strong>e, <strong>in</strong> a dosage <strong>in</strong>sufficient tocause a decrease <strong>in</strong> the plasma prolact<strong>in</strong> concentration, doesprematurely <strong>in</strong>duce a follicular phase. These observations<strong>in</strong>dicate that it is not the decrease <strong>in</strong> the plasma prolact<strong>in</strong>concentration, but another dopam<strong>in</strong>e-agonistic <strong>in</strong>fluence thatplays a crucial role <strong>in</strong> the transition to a new follicular phase.The dopam<strong>in</strong>e-agonist <strong>in</strong>duced oestrus is associated with arapid rise <strong>in</strong> the basal plasma FSH concentration, similar towhat is observed dur<strong>in</strong>g the physiological late anoestrus.Adm<strong>in</strong>istration of GnRH, eCG and oestrogens may also beused to <strong>in</strong>duce oestrus but with variable results. Oestrus can beprevented surgically or medically, for which purpose progestagensare the most important drugs. The mechanism is stillunclear, although it has been demonstrated that with cont<strong>in</strong>u<strong>in</strong>gmedroxyprogesterone acetate (MPA) treatment the FSHresponse to GnRH stimulation decreases and changes occur <strong>in</strong>the pulsatile release of the gonadotroph<strong>in</strong>s. In general, LHpulses co<strong>in</strong>cide with a FSH pulse, but dur<strong>in</strong>g MPA treatment,LH pulses were observed while there was such a small <strong>in</strong>crease<strong>in</strong> FSH that it was not recognized as significant FSH pulse.Physiology of the Can<strong>in</strong>e AnoestrusHypothalamus-pituitary-ovary axisThe duration of the oestrous cycle of the bitch isconsiderably longer than that of most other domesticanimals. The follicular phase and spontaneous ovulationsare followed by a luteal phase hav<strong>in</strong>g an average durationof approximately 2 months, irrespective of pregnancy. Anon-seasonal anoestrus, with a duration of 2 to10 months, follows each oestrous cycle (Schaefers-Okkens1996). Follicle-stimulat<strong>in</strong>g hormone (FSH) andlute<strong>in</strong>iz<strong>in</strong>g hormone (LH) play an essential role <strong>in</strong> the<strong>in</strong>duction of folliculogenesis and ovulation. The ma<strong>in</strong>regulator of FSH and LH secretion is gonadotroph<strong>in</strong>releas<strong>in</strong>ghormone (GnRH). The spontaneous GnRHrelease from excised hypothalamic fragments, that<strong>in</strong>clude the ‘mediobasal hypothalamic-preoptic areasuprachiasmaticnucleus units’ derived from beaglebitches at different stages of the oestrous cycle, is pulsatilethroughout all stages of the oestrous cycle. Progressionfrom early to late anoestrus <strong>in</strong> the bitch is characterized byan <strong>in</strong>creased release of GnRH by the hypothalamus.Especially dur<strong>in</strong>g late anoestrus GnRH pulse frequency issignificantly <strong>in</strong>creased (Tani et al. 1996). The sensitivityof the pituitary to GnRH and the <strong>in</strong>direct response of theovary to GnRH of bitches <strong>in</strong> early and late anoestrus havebeen <strong>in</strong>vestigated by way of the <strong>in</strong>travenous adm<strong>in</strong>istrationof graded doses of GnRH. The responses, expressedas circulat<strong>in</strong>g LH and oestradiol concentration profilesover time, were significantly dose-dependent and higher<strong>in</strong> late anoestrus than <strong>in</strong> early anoestrus. In addition,GnRH-<strong>in</strong>duced LH and oestradiol profiles were positivelycorrelated (van Haaften et al. 1994). These results<strong>in</strong>dicate that dur<strong>in</strong>g the course of anoestrus, there is an<strong>in</strong>crease <strong>in</strong> the sensitivity of the pituitary to GnRH and <strong>in</strong>ovarian responsiveness to gonadotroph<strong>in</strong>s (Jeffcoate1993; van Haaften et al. 1994).In order to study the role of gonadotroph<strong>in</strong>s dur<strong>in</strong>gthe transition from anoestrus to the follicular phase, thepulsatile plasma profiles of LH and FSH were determ<strong>in</strong>eddur<strong>in</strong>g early-, mid- and late anoestrus <strong>in</strong> beaglebitches (Kooistra et al. 1999a). Dur<strong>in</strong>g anoestrus, eachFSH pulse co<strong>in</strong>cided with a LH pulse. The mean plasmaLH concentration of the smoothed basel<strong>in</strong>e and themean area under the curve (AUC) for LH did not differsignificantly when the different phases of anoestrus werecompared. In contrast, the mean plasma FSH concentrationof the smoothed basel<strong>in</strong>e and the AUC for FSHdur<strong>in</strong>g late anoestrus was significantly higher than thosedur<strong>in</strong>g mid- and early anoestrus (Kooistra et al. 1999a;Oncl<strong>in</strong> et al. 2001). These observations suggest that an<strong>in</strong>crease <strong>in</strong> circulat<strong>in</strong>g FSH levels is a key event <strong>in</strong>ovarian folliculogenesis <strong>in</strong> the dog and consequently forthe term<strong>in</strong>ation of anoestrus. Indeed, <strong>in</strong> most mammalsstudied, FSH is regarded as the most important factor <strong>in</strong>the early stages of follicular development, whereas LH isregarded as the primary regulatory factor <strong>in</strong> the moremature follicles (Moyle and Campbell 1995; Monniauxet al. 1997). Consequently, it may be hypothesized thatat a certa<strong>in</strong> moment dur<strong>in</strong>g anoestrus, the ris<strong>in</strong>g plasmaFSH concentration will exceed the threshold value of themost sensitive follicles of the can<strong>in</strong>e ovarian antralfollicle pool lead<strong>in</strong>g to an enhancement of the developmentof these follicles. One of the ma<strong>in</strong> effects of FSH isthe acquisition of LH receptors <strong>in</strong> the granulosa cells.Beyond this stage, LH is progressively able to replaceFSH <strong>in</strong> support<strong>in</strong>g follicular maturation (Monniauxet al. 1997). Although FSH pulses appear to occurconcomitantly with LH pulses <strong>in</strong> all stages of the can<strong>in</strong>eoestrous cycle and anoestrus (Kooistra et al. 1999a),differential regulation of FSH and LH has also beenreported <strong>in</strong> this species, both dur<strong>in</strong>g anoestrus, dur<strong>in</strong>gthe follicular phase and dur<strong>in</strong>g the period of ovulationÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


158 J de Gier, NJ Beijer<strong>in</strong>k, HS Kooistra and AC Okkensand fertilization (Kooistra et al. 1999a; de Gier et al.2006). Differential regulation of FSH and LH secretioncan at least partly be expla<strong>in</strong>ed by the frequency andamplitude of GnRH pulses (Haisenleder et al. 1991;Vizcarra et al. 1997). In addition, gonadal feedback(Mann et al. 1992; Shupnik 1996) and a specific hypothalamicFSH-releas<strong>in</strong>g factor (Yu et al. 1997) may playa role <strong>in</strong> the differential or non-parallel secretion of FSHand LH. Furthermore, the <strong>in</strong>tracellular mechanisms forthe storage and release differ for FSH and LH (Chowdhuryand Ste<strong>in</strong>berger 1975; Moyle and Campbell 1995;Nicol et al. 2004).In addition to a rise <strong>in</strong> hypothalamic GnRH release, an<strong>in</strong>crease <strong>in</strong> basal plasma FSH concentration, and<strong>in</strong>creased sensitivity of the pituitary and ovaries dur<strong>in</strong>gprogression of anoestrus, several other factors that maybe <strong>in</strong>volved <strong>in</strong> the <strong>in</strong>itiation of folliculogenesis andterm<strong>in</strong>ation of anoestrus have been reported. In somebitches, an <strong>in</strong>creased LH pulsatility has been observedshortly before the start of pro-oestrus (Concannon et al.1986; Kooistra et al. 1999b; Tani et al. 1999; Beijer<strong>in</strong>ket al. 2004). The exact role of <strong>in</strong>creased LH pulsatility <strong>in</strong>the term<strong>in</strong>ation of anoestrus <strong>in</strong> the bitch rema<strong>in</strong>s elusive.One of the ma<strong>in</strong> effects of the ris<strong>in</strong>g FSH level is theacquisition of LH receptors <strong>in</strong> the granulosa cells(Monniaux et al. 1997). It is therefore possible that the<strong>in</strong>crease <strong>in</strong> LH pulsatility at the end of anoestrus providesa stimulus to follicles which are no longer receptive toFSH but have acquired sufficient LH receptors.In addition, enhancement of the expression of thegenes encod<strong>in</strong>g for the oestrogen receptor (Tani et al.1997) and P450 aromatase (which catalyses oestrogenbiosynthesis <strong>in</strong> the can<strong>in</strong>e hypothalamus) dur<strong>in</strong>g anoestrushas been reported <strong>in</strong> dogs (Inaba et al. 2002).However, although sporadic elevations are observed,plasma oestradiol concentrations are usually low dur<strong>in</strong>ganoestrus and do not beg<strong>in</strong> to rise until approximately amonth before the LH peak (Jeffcoate 1993). Furthermore,there are no manifestations of oestrogen <strong>in</strong>fluenceson the reproductive tract or on sexual behaviourdur<strong>in</strong>g anoestrus, and vag<strong>in</strong>al endoscopy or cytologyreveals no evidence of oestrogenic stimulation until lateanoestrus.In addition, there is some evidence that factorscaus<strong>in</strong>g a decrease <strong>in</strong> opioidergic activity promote LHrelease and the term<strong>in</strong>ation of anoestrus (Concannon1993). Treatment with naloxone, an opioid antagonist,stimulated LH release at nearly all stages of the oestrouscycle (Concannon and Temple 1988). F<strong>in</strong>ally, nochanges <strong>in</strong> the expression of the FSH receptor havebeen found dur<strong>in</strong>g can<strong>in</strong>e anoestrus (McBride et al.2001). This study demonstrated that anoestrus <strong>in</strong> bitchesis neither due to failure of expression of the can<strong>in</strong>e FSHreceptor nor due to a change <strong>in</strong> splic<strong>in</strong>g pattern to an<strong>in</strong>active form.Dopam<strong>in</strong>ergic <strong>in</strong>fluences and <strong>in</strong>duction of a prematureoestrusIn addition to the earlier-mentioned changes <strong>in</strong> thehypothalamus-pituitary-ovary axis, there is evidence of<strong>in</strong>volvement of dopam<strong>in</strong>ergic <strong>in</strong>fluences <strong>in</strong> the <strong>in</strong>itiationof a new follicular phase <strong>in</strong> the bitch. Adm<strong>in</strong>istration ofthe dopam<strong>in</strong>e-agonists bromocript<strong>in</strong>e and cabergol<strong>in</strong>e isassociated with both <strong>in</strong>hibition of prolact<strong>in</strong> release andshorten<strong>in</strong>g of the <strong>in</strong>teroestrous <strong>in</strong>terval (Okkens et al.1985; van Haaften et al. 1989; Concannon 1993; Oncl<strong>in</strong>et al. 1995; Kooistra et al. 1999b; Verstegen et al. 1999;Gobello et al. 2002; Beijer<strong>in</strong>k et al. 2003). If bromocript<strong>in</strong>etreatment is started dur<strong>in</strong>g the luteal phase,shorten<strong>in</strong>g of the <strong>in</strong>teroestrous <strong>in</strong>terval is primarily theresult of a shorten<strong>in</strong>g of the anoestrus (Okkens et al.1985), but is also due to shorten<strong>in</strong>g of the luteal phase(Okkens et al. 1985, 1990). The shorten<strong>in</strong>g of the lutealphase is probably caused by a decrease <strong>in</strong> the secretionof prolact<strong>in</strong>, the ma<strong>in</strong> luteotrophic factor <strong>in</strong> the bitch(Okkens et al. 1990; Oncl<strong>in</strong> and Verstegen 1997).It has been hypothesized that the shorten<strong>in</strong>g of theanoestrus by dopam<strong>in</strong>e-agonists is also the result of thesuppression of prolact<strong>in</strong> secretion, as prolact<strong>in</strong> may<strong>in</strong>hibit gonadotroph<strong>in</strong> release. Indeed, it has beendemonstrated <strong>in</strong> various mammalian species that highcirculat<strong>in</strong>g levels of prolact<strong>in</strong> <strong>in</strong> different pathologicalsituations <strong>in</strong>hibit LH pulsatility (Sauder et al. 1984;Yazigi et al. 1997) or are associated with decreased LHsecretion (Park et al. 1993). In addition, decreasedplasma LH levels were observed dur<strong>in</strong>g physiologicalhyperprolact<strong>in</strong>aemia <strong>in</strong> lactat<strong>in</strong>g sows, while lower<strong>in</strong>g ofthe plasma prolact<strong>in</strong> concentration by bromocript<strong>in</strong>eadm<strong>in</strong>istration led to a rise <strong>in</strong> plasma LH levels <strong>in</strong> theseanimals (Bevers et al. 1983). Yet, under physiologicalconditions plasma prolact<strong>in</strong> concentrations are lowdur<strong>in</strong>g can<strong>in</strong>e anoestrus (Olson et al. 1982; Kooistraand Okkens 2001), and no obvious changes <strong>in</strong> plasmaprolact<strong>in</strong> concentration have been observed dur<strong>in</strong>g thetransition from anoestrus to the follicular phase <strong>in</strong> thebitch (Olson et al. 1982). Furthermore, anoestrus wasnot shortened <strong>in</strong> dogs treated with low dosages of theseroton<strong>in</strong> receptor antagonist metergol<strong>in</strong>e (Okkens et al.1997), although the plasma prolact<strong>in</strong> levels were lowerthan <strong>in</strong> bromocript<strong>in</strong>e-treated dogs. The results of thelatter study suggest that the <strong>in</strong>duction of the follicularphase is not <strong>in</strong>itiated by suppression of prolact<strong>in</strong>secretion. This raised the question whether adm<strong>in</strong>istrationof a dopam<strong>in</strong>e agonist <strong>in</strong> a dosage that is too low tosuppress prolact<strong>in</strong> secretion still will result <strong>in</strong> a shorten<strong>in</strong>gof anoestrus <strong>in</strong> the bitch. To <strong>in</strong>vestigate thishypothesis, bitches were daily treated twice with 5(5-group), 20 (20-group), or 50 (50-group) microgramsof bromocript<strong>in</strong>e per kg body weight orally, start<strong>in</strong>g28 days after ovulation (Beijer<strong>in</strong>k et al. 2003). In thebitches receiv<strong>in</strong>g 5 lg bromocript<strong>in</strong>e per kg body weighttwice daily, there was no difference between the averageplasma prolact<strong>in</strong> concentration prior to and dur<strong>in</strong>gtreatment. This is <strong>in</strong> contrast with the bitches whichreceived 20 or 50 lg bromocript<strong>in</strong>e per kg body weighttwice daily, <strong>in</strong> which the average plasma prolact<strong>in</strong>concentration prior to bromocript<strong>in</strong>e treatment wassignificantly higher than that dur<strong>in</strong>g treatment. Themean ± SEM <strong>in</strong>teroestrous <strong>in</strong>terval was 136 ± 16 days<strong>in</strong> the 5-group, 96 ± 6 days <strong>in</strong> the 20-group, and92 ± 11 days <strong>in</strong> the 50-group. Each of these <strong>in</strong>tervalswas significantly shorter than the mean <strong>in</strong>teroestrous<strong>in</strong>terval <strong>in</strong> control cycles, 216 ± 9 days. The mean<strong>in</strong>teroestrous <strong>in</strong>tervals <strong>in</strong> the 20- and 50-groups weresimilar and significantly shorter than that of theÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Can<strong>in</strong>e Anoestrus, Oestrous Induction and Prevention 1595-group. This study provided further evidence for theassumption that a premature oestrus is not <strong>in</strong>duced by adecrease <strong>in</strong> plasma prolact<strong>in</strong> concentration.Besides an <strong>in</strong>hibition of prolact<strong>in</strong> release, the bromocript<strong>in</strong>e-<strong>in</strong>ducedshorten<strong>in</strong>g of anoestrus is also associatedwith a quick rise <strong>in</strong> the basal plasma FSHconcentration without a concomitant <strong>in</strong>crease <strong>in</strong> thebasal plasma LH concentration (Fig. 1) (Kooistra et al.1999b). These results give further support to the notionthat <strong>in</strong> the bitch, an <strong>in</strong>crease <strong>in</strong> circulat<strong>in</strong>g FSH shouldbe considered to be a critical event required for ovarianfolliculogenesis. Treatment with bromocript<strong>in</strong>e maycause an <strong>in</strong>crease <strong>in</strong> plasma FSH concentration to alevel that enhances development of follicles. This issimilar to the endocr<strong>in</strong>e events dur<strong>in</strong>g late anoestrus <strong>in</strong>non-treated bitches (Kooistra et al. 1999a).A role for dopam<strong>in</strong>e <strong>in</strong> the control of reproductionhas been demonstrated <strong>in</strong> different mammalian species,although the effects of dopam<strong>in</strong>e <strong>in</strong> other species differfrom those <strong>in</strong> the bitch (Beck et al. 1978; Havern et al.1994; Besognet et al. 1997). In species other than thedog, dopam<strong>in</strong>e-agonists may <strong>in</strong>hibit gonadotroph<strong>in</strong>secretion dur<strong>in</strong>g anoestrus and dopam<strong>in</strong>e-antagonistsmay <strong>in</strong>duce reproductive activity; whereas <strong>in</strong> the bitch,dopam<strong>in</strong>e-agonists <strong>in</strong>duce the onset of oestrus.As opposed to dopam<strong>in</strong>e agonists, low dosages of theseroton<strong>in</strong> receptor antagonist metergol<strong>in</strong>e do not result<strong>in</strong> shorten<strong>in</strong>g of anoestrus (Okkens et al. 1997).Therefore, it may be expected that the changes <strong>in</strong>gonadotroph<strong>in</strong> release associated with dopam<strong>in</strong>eagonist-<strong>in</strong>duced shorten<strong>in</strong>g of anoestrus will not beobserved dur<strong>in</strong>g treatment with low dosages of theseroton<strong>in</strong> receptor antagonist metergol<strong>in</strong>e. To<strong>in</strong>vestigate the effects of a low dose of a seroton<strong>in</strong>antagonist on the pulsatile secretion patterns of FSH andLH, 0.1 mg metergol<strong>in</strong>e per kg body weight orally twicedaily, was adm<strong>in</strong>istered to bitches, start<strong>in</strong>g 100 days afterovulation (Beijer<strong>in</strong>k et al. 2004). The mean <strong>in</strong>teroestrous<strong>in</strong>terval <strong>in</strong> the eight bitches treated with the seroton<strong>in</strong>antagonist metergol<strong>in</strong>e was not shortened as expecteddespite a decreased plasma prolact<strong>in</strong> concentration.Moreover, dur<strong>in</strong>g the first weeks of treatment with theseroton<strong>in</strong> antagonist metergol<strong>in</strong>e, there were no significantchanges <strong>in</strong> the pulsatile plasma profiles of FSH orLH. These f<strong>in</strong>d<strong>in</strong>gs <strong>in</strong>dicate that seroton<strong>in</strong> antagonist<strong>in</strong>ducedlower<strong>in</strong>g of plasma prolact<strong>in</strong> does not lead to<strong>in</strong>creased secretion of FSH.In addition to adm<strong>in</strong>istration of dopam<strong>in</strong>e-agonists,<strong>in</strong>duction of a follicular phase may also occur via theadm<strong>in</strong>istration of GnRH, gonadotroph<strong>in</strong>s, eCG, hCG,porc<strong>in</strong>e LH and oestrogens (for review see Kutzler2007). Although many protocols exist for oestrous<strong>in</strong>duction <strong>in</strong> bitches, the fertility results, with theexception of dopam<strong>in</strong>e agonists, are variable andgenerally poor. Some methods are also too costly orlabour <strong>in</strong>tensive to be suitable for veter<strong>in</strong>ary cl<strong>in</strong>icalpractice. In addition, aglepristone, a progesteronereceptorantagonist and prostagland<strong>in</strong> F 2a shorten the<strong>in</strong>teroestrous <strong>in</strong>terval, if adm<strong>in</strong>istered dur<strong>in</strong>g the lutealphase (Romagnoli et al. 1993; Galac et al. 2004).Oestrus preventionOestrus can be prevented medically or surgically.Ovariectomy has several advantages. It is effective afterone procedure. It considerably lowers the risk formammary cancer if performed before approximately2.5 years after the first oestrus. It also prevents thedevelopment of pyometra and progesterone-<strong>in</strong>ducedgrowth hormone excess. There are, however, also severaldisadvantages such as the risk of complications dur<strong>in</strong>ganaesthesia and surgery, and the irreversibility of theprocedure. In addition, there is also the possibility ofside-effects such as changes <strong>in</strong> the coat and ur<strong>in</strong>ary<strong>in</strong>cont<strong>in</strong>ence. There are <strong>in</strong>dications that the risk forur<strong>in</strong>ary <strong>in</strong>cont<strong>in</strong>ence is greater if the procedure isperformed prior to the first oestrus. Furthermore, ithas been shown that early-age gonadectomy is associatedwith an <strong>in</strong>creased rate of cystitis and that age atgonadectomy is negatively correlated with the rate ofur<strong>in</strong>ary <strong>in</strong>cont<strong>in</strong>ence (Spa<strong>in</strong> et al. 2004). Ur<strong>in</strong>ary <strong>in</strong>cont<strong>in</strong>enceoccurs ma<strong>in</strong>ly <strong>in</strong> large breeds and some breedse.g. the Boxer, Dobermann, Bouvier des Flandres, GiantSchnauzer, Irish Setter, M<strong>in</strong>iature Poodle, Old EnglishSheepdog, Weimaraner and Rottweiler appear to beespecially at risk for develop<strong>in</strong>g ur<strong>in</strong>ary <strong>in</strong>cont<strong>in</strong>ence(Thrushfield et al. 1998).Medical oestrous prevention can be accomplishedwith several types of drugs. The progestagens are themost important drugs for oestrous prevention.Fig. 1. The mean (± SEM) plasma FSH and LH concentrations of the smoothed basel<strong>in</strong>e <strong>in</strong> six beagle bitches the day before and 14, 28 and42 days after the start of bromocript<strong>in</strong>e treatment (* <strong>in</strong>dicates significant difference) (Kooistra et al. 1999b)Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


160 J de Gier, NJ Beijer<strong>in</strong>k, HS Kooistra and AC OkkensAndrogens can also be used for this purpose, butprimarily for short-term prevention. As androgens arenot currently registered for this use, they are not<strong>in</strong>cluded <strong>in</strong> this paper.GnRH agonists or GnRH antagonistsGnRH agonists or GnRH antagonists may also be usedfor oestrus prevention. At this stage, the GnRHantagonists are unfit for cl<strong>in</strong>ical use, because ofeconomic and delivery problems for long-term use(Vickery et al. 1989). Gonadotroph<strong>in</strong>-releas<strong>in</strong>g hormoneagonists adm<strong>in</strong>istered at high dosages over a longperiod of time prevent oestrus by pituitary downregulation.However, the early stimulatory effect ofGnRH analogues may cause signs of oestrus, if adm<strong>in</strong>istereddur<strong>in</strong>g anoestrus and sometimes even if adm<strong>in</strong>istereddur<strong>in</strong>g the luteal phase. It is possible to preventthe occurrence of oestrus by adm<strong>in</strong>ister<strong>in</strong>g a GnRHagonist <strong>in</strong> pre-pubertal bitches at approximately4 months of age, whereas implantation of a GnRHagonist at 7 months of age does not prevent theoccurrence of an oestrous cycle (Rubion et al. 2006;Trigg et al. 2006). However, <strong>in</strong> the latter dogs, oestrus isdelayed for a long time. Rubion et al. (2006) showedthat GnRH agonist implants adm<strong>in</strong>istered beforepuberty prevented reproductive function for 1 year <strong>in</strong>pre-pubertal bitches. Follow<strong>in</strong>g removal of the implant,oestrus occurred naturally <strong>in</strong> 7 of 10 bitches and was<strong>in</strong>duced <strong>in</strong> three bitches after 1.2–14.3 months. Anotherstrategy to prevent oestrous <strong>in</strong>duction dur<strong>in</strong>g anoestrusafter implantation of a GnRH agonist is adm<strong>in</strong>istrationof a progestagen before implantation. The efficacy ofthis <strong>in</strong>itial treatment, however, appears to depend on the<strong>in</strong>terval between start of progestagen treatment andimplantation, the stage of anoestrus, the dosage of theused progestagen or the type of GnRH analogue(Wright et al. 2001; Sung et al. 2006). Furthermore,add<strong>in</strong>g progestagens to prevent the <strong>in</strong>itial stimulation ofGnRH agonists cancels the advantage of not adm<strong>in</strong>ister<strong>in</strong>gprogestagens for oestrous prevention. Additionally,it is sometimes a problem to remove these implants,because they cannot be found.ProgestagensThe mechanism of the contraceptive activity of progestagensis still unclear. In many species, there is evidencethat contraceptive progestagens reduce serum concentrationsof gonadotroph<strong>in</strong>s. However, high doses ofmedroxyprogesterone acetate (MPA) adm<strong>in</strong>istered tobeagle bitches for several months did not reduce the<strong>in</strong>creased circulat<strong>in</strong>g concentrations of LH <strong>in</strong> ovariectomizedbitches, nor did it lower LH concentrations <strong>in</strong><strong>in</strong>tact bitches (McCann et al. 1987). In another study,high contraceptive doses of megestrol acetate (MA) didnot suppress basal gonadotroph<strong>in</strong> secretion dur<strong>in</strong>ganoestrus, nor was the pituitary hypersecretion of LHand FSH that occurs <strong>in</strong> ovariectomized bitches suppressed(Colon et al. 1993). Beijer<strong>in</strong>k (2007) exam<strong>in</strong>ed 6-h plasma profiles of FSH and LH <strong>in</strong> five bitches beforeand 3, 6, 9 and 12 months after the start of MPAtreatment. The results of this study demonstrate thattreatment with MPA affects the hypothalamic-pituitaryovarianaxis. Oestrus, ovulation and a subsequent lutealphase did not occur <strong>in</strong> any of the bitches dur<strong>in</strong>gtreatment with MPA. However, the prevention ofoestrus by MPA could not be ascribed to a significantreduction <strong>in</strong> circulat<strong>in</strong>g levels of either FSH or LH. Onthe contrary, dur<strong>in</strong>g the first months of MPA treatment,there was an <strong>in</strong>crease <strong>in</strong> basal plasma FSH and LHconcentrations. This progestagen-<strong>in</strong>duced <strong>in</strong>crease <strong>in</strong>gonadotroph<strong>in</strong> concentration was not observed <strong>in</strong> thestudies of McCann et al. (1987) and Colon et al. (1993),and its recognition may be expla<strong>in</strong>ed by the repeatedsampl<strong>in</strong>g employed <strong>in</strong> the studies by Beijer<strong>in</strong>k (2007).The elevated plasma gonadotroph<strong>in</strong> levels dur<strong>in</strong>g thefirst months of MPA treatment may be due to a direct<strong>in</strong>hibitory effect of MPA at the ovarian level, result<strong>in</strong>g <strong>in</strong>suppression of the ovarian secretion of oestradiol or<strong>in</strong>hib<strong>in</strong> (Mann et al. 1992; Shupnik 1996).With cont<strong>in</strong>u<strong>in</strong>g MPA treatment, basal plasmagonadotroph<strong>in</strong> concentrations returned to pre-treatmentlevels. In addition, the pituitary FSH response toGnRH stimulation decreased, suggest<strong>in</strong>g that MPAtreatment attenuated pituitary FSH sensitivity to endogenousGnRH (Fig. 2) (Beijer<strong>in</strong>k et al. 2007). PulsatileFSH and LH release was ma<strong>in</strong>ta<strong>in</strong>ed dur<strong>in</strong>g MPAtreatment, but there were <strong>in</strong>dications that changesoccurred <strong>in</strong> the pulsatile release of the gonadotroph<strong>in</strong>s.In general, LH pulses co<strong>in</strong>cide with a FSH pulse, butdur<strong>in</strong>g MPA treatment several LH pulses were accompaniedby such small <strong>in</strong>creases <strong>in</strong> FSH that these<strong>in</strong>creases were not recognized as significant (Fig. 3)(Beijer<strong>in</strong>k 2007).The progestagens most frequently used for oestrousprevention <strong>in</strong> the dog are proligestone and MPA. Thes<strong>in</strong>gle subcutaneous <strong>in</strong>jection dosage recommended byLH (µg/l)FSH (µg/l)12010080604020050403020100Before 2 5 8 11Months of treatmentBefore 2 5 8 11Months of treatmentFig. 2. Plasma LH and FSH responses (mean ± SEM) <strong>in</strong> five dogs <strong>in</strong>a comb<strong>in</strong>ed anterior pituitary function test accord<strong>in</strong>g to Meij et al.(1996) before and subsequently 2, 5, 8 and 11 months after the start ofthe treatment with MPA (10 lg ⁄ kg, every 4 weeks). Blood sampleswere collected at -15, 0, 5, 10, 20, 30 and 45 m<strong>in</strong> follow<strong>in</strong>g the <strong>in</strong>jectionof the releas<strong>in</strong>g hormones at 0 m<strong>in</strong> (arrow) (Beijer<strong>in</strong>k et al. 2007)Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Can<strong>in</strong>e Anoestrus, Oestrous Induction and Prevention 161Fig. 3. The 6-h plasma profiles of FSH (black squares) and LH (white triangles) <strong>in</strong> a 3-year-old beagle bitch before, and 3, 6, 9 and 12 monthsafter the start of treatment with MPA (10 lg ⁄ kg, every 4 weeks). *Significant pulses of both FSH and LH identified by the Pulsar programme;^significant pulse of LH without a concurrent significant pulse of FSH (Beijer<strong>in</strong>k 2007)the manufacturer for proligestone 1 ranges from10 mg ⁄ kg for a dog of approximately 60 kg, to30 mg ⁄ kg for one of 3 kg and for MPA 2 , the s<strong>in</strong>gle1 DelvosteronÒ, Mycofarm, Delft, The Netherlands2 DepopromoneÒ, Upjohn, Ede, The Netherlands; PerlutexÒ, LeoPharma, Ballerup, Denmarksubcutaneous <strong>in</strong>jection dosage is 2 mg ⁄ kg (maximum60 mg per animal) (Schaefers-Okkens 1996). Thesedrugs should be adm<strong>in</strong>istered dur<strong>in</strong>g anoestrus approximately1 month before the onset of the expectedfollicular phase. The first oestrus after the use ofproligestone can be expected <strong>in</strong> the majority of bitcheswith<strong>in</strong> 9–12 months, us<strong>in</strong>g MPA it may take up toÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


162 J de Gier, NJ Beijer<strong>in</strong>k, HS Kooistra and AC Okkens2–3 years. For that reason, the manufacturer does notrecommend us<strong>in</strong>g MPA <strong>in</strong>tended for breed<strong>in</strong>g at a laterdate. Medroxyprogesterone acetate 3 can also be adm<strong>in</strong>isteredorally, 5 mg once daily (10 mg for large dogsdur<strong>in</strong>g the first 5 days) for a maximum of 21 days. Thereturn of oestrus may vary from 2 to 9 months.Use of progestagens for oestrus prevention may leadto the follow<strong>in</strong>g side-effects:• Development of cystic endometrial hyperplasia(CEH)-endometritis (Sokolowski and Zimbelman1974).• Prolonged pregnancy. This occurs if progestagensare adm<strong>in</strong>istered subcutaneously at the onset of thefollicular phase and the bitch becomes pregnant. Thegestation will be prolonged, and a caesarian sectionmay be needed.• Hypersecretion of growth hormone, which maylead to diabetes mellitus (Selman et al. 1997). Thishypersecretion of growth hormone as a result ofprogestagen adm<strong>in</strong>istration can be successfully treatedby the progesterone receptor blocker aglepristone(Bhatti et al. 2006).• An <strong>in</strong>creased risk of neoplastic transformation ofmammary tissue. This may range from hyperplasia,adenomatous hyperplasia and adenomas to malignanttumours. The progestagen-<strong>in</strong>duced neoplastic transformationof mammary tissue starts with the proliferationof undifferentiated term<strong>in</strong>al ductal structures,so-called term<strong>in</strong>al end buds (Russo and Russo 1991).This proliferation <strong>in</strong>creases the susceptibility of themammary tissue to malignant transformation.The occurrence of these side-effects is, with theexception of ‘prolonged pregnancy’, largely dependentupon the total progestagen exposure. With the adviseddosage regimens, the exposure may be higher with MPAand MA than with proligestone, the latter be<strong>in</strong>g a ratherweak progestagen.3 ProveraÒ, Upjohn, Ede, The Netherlands; OvuconÒ, Intervet,Boxmeer, The NetherlandsReferencesBeck W, Hancke JL, Wuttke W, 1978: Increased sensitivity ofdopam<strong>in</strong>ergic <strong>in</strong>hibition of lute<strong>in</strong>iz<strong>in</strong>g hormone release <strong>in</strong>immature and castrated female rats. Endocr<strong>in</strong>ology 102,837–843.Beijer<strong>in</strong>k NJ, 2007: Endocr<strong>in</strong>ology of physiological andprogest<strong>in</strong>-<strong>in</strong>duced can<strong>in</strong>e anoestrus. Ch. 8 Pulsatile plasmaprofiles of FSH and LH before and dur<strong>in</strong>g medroxyprogesteroneacetatetreatment <strong>in</strong> the bitch. PhD Thesis, UtrechtUniversity.Beijer<strong>in</strong>k NJ, Dieleman SJ, Kooistra HS, Okkens AC, 2003:Low doses of bromocript<strong>in</strong>e shorten the <strong>in</strong>terestrous <strong>in</strong>terval<strong>in</strong> the bitch without lower<strong>in</strong>g plasma prolact<strong>in</strong> concentration.Theriogenology 60, 1397–1386.Beijer<strong>in</strong>k NJ, Kooistra HS, Dieleman SJ, Okkens AC, 2004:Seroton<strong>in</strong>-antagonist-<strong>in</strong>duced lower<strong>in</strong>g of prolact<strong>in</strong> secretiondoes not affect the pattern of pulsatile secretion offollicle-stimulat<strong>in</strong>g hormone and lute<strong>in</strong>iz<strong>in</strong>g hormone <strong>in</strong> thebitch. <strong>Reproduction</strong> 128, 181–188.Beijer<strong>in</strong>k NJ, Bhatti SFM, Okkens AC, Dieleman SJ, Mol JA,Duchateau L, van Ham LML, Kooistra HS, 2007: Adenohypophysealfunction <strong>in</strong> bitches treated with medroxyprogesteroneacetate. Domest Anim Endocr<strong>in</strong>ol 32, 63–78.Besognet B, Hansen S, Daels PF, 1997: Induction of reproductivefunction <strong>in</strong> anestrous mares us<strong>in</strong>g a dopam<strong>in</strong>eantagonist. Theriogenology 47, 467–480.Bevers MM, Willemse AH, Kruip TAM1983: The effect ofbromocript<strong>in</strong>e on lute<strong>in</strong>iz<strong>in</strong>g hormone levels <strong>in</strong> the lactat<strong>in</strong>gsow: evidence for a suppressive action by prolact<strong>in</strong> and thesuckl<strong>in</strong>g stimulus. Acta Endocr<strong>in</strong>ol 104, 261–265.Bhatti SFM, Duchateau L, Okkens AC, van Ham LML, MolJA, Kooistra HS, 2006: Treatment of growth hormoneexcess <strong>in</strong> dogs with the progesterone receptor antagonistaglépristone. Theriogenology 66, 797–803.Chowdhury M, Ste<strong>in</strong>berger E1975: Biosynthesis and secretionof follicle-stimulat<strong>in</strong>g hormone. Endocr Rev 4, 369–374.Colon J, Kimball M, Hansen B, Concannon PW, 1993: Effectsof contraceptive doses of the progestagen megestrol acetateon lute<strong>in</strong>iz<strong>in</strong>g hormone and follicle-stimulat<strong>in</strong>g hormonesecretion <strong>in</strong> female dogs. J Reprod Fertil Suppl 47, 519–521.Concannon PW, 1993: Biology of gonadotroph<strong>in</strong> secretion <strong>in</strong>adult and prepubertal female dogs. J Reprod Fertil Suppl47, 3–27.Concannon PW, Temple M, 1988: Periovulatory decl<strong>in</strong>e andlate anestrus <strong>in</strong>crease <strong>in</strong> naloxone-<strong>in</strong>ducible LH release <strong>in</strong>adult domestic bitches. Biol Reprod Suppl 36, 101.Concannon PW, Whaley S, Anderson SP, 1986: Increased LHpulse frequency associated with term<strong>in</strong>ation of anestrusdur<strong>in</strong>g the ovarian cycle of the dog. Biol Reprod Suppl 34,119 (Abstract).Galac S, Kooistra HS, Dieleman SJ, Cestnik V, Okkens AC,2004: Effects of aglépristone, a progesterone receptorantagonist, adm<strong>in</strong>istered dur<strong>in</strong>g the early luteal phase <strong>in</strong>non-pregnant bitches. Theriogenology 66, 494–500.de Gier J, Kooistra HS, Djajad<strong>in</strong><strong>in</strong>grat-Laanen SC, DielemanSJ, Okkens AC, 2006: Differential regulation of the secretionof lute<strong>in</strong>iz<strong>in</strong>g hormone and follicle-stimulat<strong>in</strong>g hormonearound the time of ovulation <strong>in</strong> the bitch. Theriogenology66, 1419–1422.Gobello C, Castex G, Corrada Y, 2002: Use of cabergol<strong>in</strong>e totreat primary and secondary anestrus <strong>in</strong> dogs. J Am VetMed Assoc 11, 1653–1654.van Haaften B, Dieleman SJ, Okkens AC, Bevers MM,Willemse AH, 1989: Induction of oestrus and ovulation <strong>in</strong>dogs by treatment with PMSG and ⁄ or bromocript<strong>in</strong>e.J Reprod Fertil Suppl 39, 330–331.van Haaften B, Bevers MM, van den Brom WE, Okkens AC,van Sluijs FJ, Willemse AH, Dieleman SJ, 1994: Increas<strong>in</strong>gsensitivity of the pituitary to GnRH from early to lateanoestrus <strong>in</strong> the beagle bitch. J Reprod Fertil 101, 221–225.Haisenleder DJ, Dalk<strong>in</strong> AC, Ortolano GA, Marshall JC,Shupnik MA, 1991: A pulsatile gonadotrop<strong>in</strong>-releas<strong>in</strong>ghormone stimulus is required to <strong>in</strong>crease transcription ofthe gonadotrop<strong>in</strong> subunit genes: evidence for differentialregulation of transcription by pulse frequency <strong>in</strong> vivo.Endocr<strong>in</strong>ology 128, 509–517.Havern RL, Whisnant CS, Goodman RL, 1994: Dopam<strong>in</strong>ergicstructures <strong>in</strong> the ov<strong>in</strong>e hypothalamus mediat<strong>in</strong>g estradiolnegative feedback <strong>in</strong> anestrous ewes. Endocr<strong>in</strong>ology 134,1905–1914.Inaba T, Namura T, Tani H, Matsuyama S, Torii R, KawataN, Tamada H, Hatoya S, Kumagai D, Sugiura K, SawadaT, 2002: Enhancement of aromatase gene expression <strong>in</strong> themediobasal hypothalamus dur<strong>in</strong>g anestrus <strong>in</strong> the beaglebitch. Neurosci Lett 333, 107–110.Jeffcoate IA, 1993: Endocr<strong>in</strong>ology of anoestrous bitches.J Reprod Fertil Suppl 47, 69–76.Kooistra HS, Okkens AC, 2001: Secretion of prolact<strong>in</strong> andgrowth hormone <strong>in</strong> relation to ovarian activity <strong>in</strong> the dog.Reprod Domest Anim 36, 115–119.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Can<strong>in</strong>e Anoestrus, Oestrous Induction and Prevention 163Kooistra HS, Okkens AC, Bevers MM, Popp-Snijders C, vanHaaften B, Dieleman SJ, Schoemaker J, 1999a: Concurrentpulsatile secretion of lute<strong>in</strong>iz<strong>in</strong>g hormone and folliclestimulat<strong>in</strong>ghormone dur<strong>in</strong>g different phases of the estrouscycle and anestrus <strong>in</strong> beagle bitches. Biol Reprod 60, 65–71.Kooistra HS, Okkens AC, Bevers MM, Popp-Snijders C, vanHaaften B, Dieleman SJ, Schoemaker J, 1999b: Bromocript<strong>in</strong>e-<strong>in</strong>ducedpremature oestrus is associated with changes <strong>in</strong>the pulsatile secretion pattern of follicle-stimulat<strong>in</strong>g hormone<strong>in</strong> beagle bitches. J Reprod Fertil 117, 387–393.Kutzler MA, 2007: Estrus <strong>in</strong>duction and synchronization <strong>in</strong>canids and felids. Theriogenology 68, 354–374.Mann GE, Campbell BK, McNeilly AS, Baird DT, 1992: Therole of <strong>in</strong>hib<strong>in</strong> and oestradiol <strong>in</strong> the control of FSHsecretion <strong>in</strong> the sheep. J Endocr<strong>in</strong>ol 133, 381–391.McBride MW, Aughey E, O’Shaughnessy PJ, JeffcoateIA2001: Ovarian function and FSH receptor characteristicsdur<strong>in</strong>g can<strong>in</strong>e anoestrus. J Reprod Fertil Suppl 57, 3–10.McCann JP, Altszuler N, Hampshire J, Concannon PW, 1987:Growth hormone, <strong>in</strong>sul<strong>in</strong>, glucose, cortisol, lute<strong>in</strong>iz<strong>in</strong>ghormone, and diabetes <strong>in</strong> beagle bitches treated withmedroxyprogesterone acetate. Acta Endocr<strong>in</strong>ol 116, 73–80.Meij BP, Mol JA, Hazew<strong>in</strong>kel HAW, Bevers MM, Rijnberk A,1996: Assessment of a comb<strong>in</strong>ed anterior pituitary functiontest <strong>in</strong> beagle dogs: rapid sequential <strong>in</strong>travenous adm<strong>in</strong>istrationof four hypothalamic releas<strong>in</strong>g hormones. DomAnim Endocr<strong>in</strong>ol 13, 161–170.Monniaux D, Huet C, Besnard N, Cle´ment F, Mariana JC,Bosc M, Monget P, 1997: Follicular growth and ovariandynamics <strong>in</strong> mammals. J Reprod Fertil Suppl 51, 3–23.Moyle WR, Campbell RK1995: Gonadotrop<strong>in</strong>s. In: AdashiEY, Rock JA, Rosenwaks Z(eds), Reproductive Endocr<strong>in</strong>ology,Surgery and Technology. Lipp<strong>in</strong>cott-Raven Publishers,Philadelphia, pp. 683–724.Nicol L, McNeilly JR, Stridsberg M, McNeilly AS, 2004:Differential secretion of gonadotroph<strong>in</strong>s: <strong>in</strong>vestigation ofthe role of secretogran<strong>in</strong> II and chromogran<strong>in</strong> A <strong>in</strong> therelease of LH and FSH <strong>in</strong> LbetaT2 cells. J Mol Endocr<strong>in</strong>ol32, 467–480.Okkens AC, Bevers MM, Dieleman SJ, Willemse AH, 1985:Shorten<strong>in</strong>g of the <strong>in</strong>teroestrus <strong>in</strong>terval and the lifespan ofthe corpus luteum of the cyclic dog by bromocript<strong>in</strong>etreatment. Vet Quart 7, 173–176.Okkens AC, Bevers MM, Dieleman SJ, Willemse AH, 1990:Evidence for prolact<strong>in</strong> as the ma<strong>in</strong> luteotrophic factor <strong>in</strong> thecyclic dog. Vet Quart 12, 193–203.Okkens AC, Kooistra HS, Dieleman SJ, Bevers MM, 1997:Dopam<strong>in</strong>e agonistic effects as opposed to prolact<strong>in</strong> concentrations<strong>in</strong> plasma as the <strong>in</strong>fluenc<strong>in</strong>g factor on duration ofanoestrus <strong>in</strong> bitches. J Reprod Fertil Suppl 51, 55–58.Olson PN, Bowen RA, Behrendt MD, Olson JD, Nett TM,1982: Concentrations of reproductive hormones <strong>in</strong> can<strong>in</strong>eserum throughout late anestrus, proestrus and estrus. BiolReprod 27, 1196–1206.Oncl<strong>in</strong> K, Verstegen JP1997: In vivo <strong>in</strong>vestigation of lutealfunction <strong>in</strong> dogs: effects of cabergol<strong>in</strong>e, a dopam<strong>in</strong>e agonist,and prolact<strong>in</strong> on progesterone secretion dur<strong>in</strong>g mid-pregnancyand -diestrus. Dom Anim Endocr<strong>in</strong>ol 14, 25–38.Oncl<strong>in</strong> K, Verstegen J, Silva LDM, Concannon P1995:Patterns of circulat<strong>in</strong>g prolact<strong>in</strong>, LH and FSH dur<strong>in</strong>gdopam<strong>in</strong>e-agonist <strong>in</strong>duced term<strong>in</strong>ation of anestrus <strong>in</strong> beagledogs. Biol Reprod Suppl 52, 314.Oncl<strong>in</strong> K, Lauwers F, Verstegen JP, 2001: FSH secretionpatterns dur<strong>in</strong>g pregnant and nonpregnant luteal periodsand 24 h secretion patterns <strong>in</strong> male and female dogs. JReprod Fertil Suppl 57, 15–21.Park SK, Keenan MW, Selmanoff M, 1993: Graded hyperprolact<strong>in</strong>emiafirst suppresses LH pulse frequency and thepulse amplitude <strong>in</strong> castrated male rats. Neuroendocr<strong>in</strong>ology58, 448–453.Romagnoli SE, Camillo F, Cela M, Johnston SD, Grassi F,Ferdegh<strong>in</strong>i M, Aria G, 1993: Cl<strong>in</strong>ical use of prostagland<strong>in</strong>F2a to <strong>in</strong>duce early abortion <strong>in</strong> bitches: serum progesterone,treatment outcome and <strong>in</strong>terval to subsequent oestrus. JReprod Fertil Suppl 47, 425–431.Rubion S, Desmoul<strong>in</strong>s PO, Riviere-Godet E, K<strong>in</strong>ziger M,Salavert F, Rutten F, Flochlay-Sigognault A, DriancourtMA, 2006: Treatment with a subcutaneous GnRH agonistconta<strong>in</strong><strong>in</strong>g controlled release device reversibly preventspuberty <strong>in</strong> bitches. Theriogenology 66, 1651–1654.Russo IH, Russo J1991: Progestagens and mammary glanddevelopment: Differentiation versus carc<strong>in</strong>ogenesis. ActaEndocr<strong>in</strong>ol 125 (Suppl. 1) 7–12.Sauder SE, Frager M, Case GD, Kelch RP, Marshall JC, 1984:Abnormal patterns of pulsatile lute<strong>in</strong>iz<strong>in</strong>g hormone secretion<strong>in</strong> women with hyperprolact<strong>in</strong>emia and amenorrhea:responses to bromocript<strong>in</strong>e. J Cl<strong>in</strong> Endocr<strong>in</strong>ol Metab 59,941–948.Schaefers-Okkens AC1996: Ovaries. In: Rijnberk A(ed.),Cl<strong>in</strong>ical Endocr<strong>in</strong>ology of Dogs and Cats. Kluwer AcademicPublishers, Dordrecht, pp. 131–156.Selman PJ, Mol JA, Rutteman GR, van Garderen E, van denIngh TSGAM, Rijnberk A1997: Effects of progest<strong>in</strong> adm<strong>in</strong>istrationon the hypothalamic-pituitary-adrenal axis andglucose-homeostasis <strong>in</strong> dogs. J Reprod Fertil Suppl 51, 345–354.Shupnik MA, 1996: Gonadal hormone feedback on pituitarygonadotrop<strong>in</strong> genes. Trends Endocr<strong>in</strong>ol Metab 7, 272–276.Sokolowski JH, Zimbelman RG, 1974: Can<strong>in</strong>e reproductioneffects of multiple treatments of medroxyprogesteroneacetate on reproductive organs of the bitch. Am J Vet Res35, 1285–1287.Spa<strong>in</strong> CV, Scarlett JM, Houpt KA, 2004: Long-term risks andbenefits of early-age gonadectomy <strong>in</strong> dogs. J Am Vet MedAssoc 224, 380–386.Sung M, Armour AF, Wright PJ, 2006: The <strong>in</strong>fluence ofexogenous progest<strong>in</strong> on the occurrence of proestrous orestrous signs, plasma concentrations of lute<strong>in</strong>iz<strong>in</strong>g hormoneand estradiol <strong>in</strong> deslorel<strong>in</strong> (GnRH agonist) treated anestrousbitches. Theriogenology 66, 1513–1517.Tani H, Inaba T, Tamada H, Sawada T, Mori J, Torii R, 1996:Increas<strong>in</strong>g gonadotrop<strong>in</strong>-releas<strong>in</strong>g hormone release by perifusedhypothalamus form early to late anestrus <strong>in</strong> the beaglebitch. Neurosci Lett 207, 1–4.Tani H, Inaba T, Matsuyama S, Takamor Y, Torii R, TakanoH, Tamada H, Sawada T, 1997: Enhancement of estrogenreceptor gene expression <strong>in</strong> the mediobasal hypothalamusdur<strong>in</strong>g anestrus <strong>in</strong> the beagle bitch. Neurosci Lett 227,149–152.Tani H, Inaba T, Nonami M, Natsuyama S, Takamor Y, ToriR, Tamada H, Kawate N, Sawada T, 1999: Increased LHpulse frequency and estrogen secretion associated withterm<strong>in</strong>ation of anestrus followed by enhancement of uter<strong>in</strong>eestrogen receptor gene expression <strong>in</strong> the beagle bitch.Theriogenology 52, 593–607.Thrushfield MV, Holt PE, Muirhead RH, 1998: Acquiredur<strong>in</strong>ary <strong>in</strong>cont<strong>in</strong>ence <strong>in</strong> bitches: its <strong>in</strong>cidence and relationshipto neuter<strong>in</strong>g practices. J Small Anim Pract 39, 559–566.Trigg TE, Doyle AG, Walsh JD, Swangchan-uthai T, 2006: Areview of advances <strong>in</strong> the use of the GnRH agonistdeslorel<strong>in</strong> <strong>in</strong> control of reproduction. Theriogenology 66,1507–1512.Verstegen JP, Oncl<strong>in</strong> K, Silva LD, Concannon PW, 1999:Effect of stage of anestrus on the <strong>in</strong>duction of estrus by thedopam<strong>in</strong>e agonist cabergol<strong>in</strong>e <strong>in</strong> dogs. Theriogenology 51,597–611.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


164 J de Gier, NJ Beijer<strong>in</strong>k, HS Kooistra and AC OkkensVickery BH, McRae GI, Goodpasture JC, Sanders LM, 1989:Use of potent LHRH analogues for chronic contraceptionand pregnancy term<strong>in</strong>ation <strong>in</strong> dogs. J Reprod Fertil Suppl39, 175–187.Vizcarra JA, Wettemann RP, Braden TD, Turzillo AM, NettTM, 1997: Effect of gonadotrop<strong>in</strong>-releas<strong>in</strong>g hormone(GnRH) pulse frequency on serum and pituitaryconcentrations of lute<strong>in</strong>iz<strong>in</strong>g hormone and follicle-stimulat<strong>in</strong>ghormone, GnRH receptors, and messenger ribonucleicacid for gonadotrop<strong>in</strong> subunits <strong>in</strong> cows. Endocr<strong>in</strong>ology 138,594–601.Wright PJ, Verstegen JP, Oncl<strong>in</strong> K, Jo¨ chle W, Armour AF,Mart<strong>in</strong> GB, Trigg TE, 2001: Supression of the oestrusresponses <strong>in</strong> bitches to the GnRH analogue deslorel<strong>in</strong> byprogest<strong>in</strong>. J Reprod Fertil Suppl 57, 263–268.Yazigi RA, Wu<strong>in</strong>tero CH, Salameh WA1997: Prolact<strong>in</strong>disorders. Fertil Steril 67, 215–225.Yu WH, Karanth S, Walczewska A, Sower SA, McCann SM,1997: A hypothalamic follicle-stimulat<strong>in</strong>g hormone-releas<strong>in</strong>gdecapeptide <strong>in</strong> the rat. Proc Natl Acad Sci 94,9499–9503.Author’s address (for correspondence): Auke C. Okkens, Departmentof Cl<strong>in</strong>ical Sciences of Companion <strong>Animals</strong>, Faculty of Veter<strong>in</strong>aryMedic<strong>in</strong>e, University of Utrecht, Yalelaan, Utrecht, The Netherlands.E-mail: a.c.schaefers-okkens@uu.nlConflict of <strong>in</strong>terest: The authors declare no conflict of <strong>in</strong>terests.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 165–171 (2008); doi: 10.1111/j.1439-0531.2008.01157.xISSN 0936-6768The Ethics and Role of AI with Fresh and Frozen Semen <strong>in</strong> DogsGCW England 1 and KM Millar 21 School of Veter<strong>in</strong>ary Medic<strong>in</strong>e and Science; 2 Centre for Applied Bioethics, School of Biosciences, University of Nott<strong>in</strong>gham, Nott<strong>in</strong>gham, UKContentsThe use of artificial <strong>in</strong>sem<strong>in</strong>ation (AI) with fresh semen hasresulted <strong>in</strong> many benefits for the management of dog breed<strong>in</strong>g,but there are disadvantages that can sometimes be overlooked.Furthermore, poorer quality semen aris<strong>in</strong>g as a result ofcryopreservation necessitates uter<strong>in</strong>e <strong>in</strong>sem<strong>in</strong>ation, whichraises the potential for surgical <strong>in</strong>sem<strong>in</strong>ation. A number ofsignificant ethical concerns have been raised by key stakeholders(such as The Kennel Club and the Royal College ofVeter<strong>in</strong>ary Surgeons) about AI per se, but particularly aboutthe use of surgical <strong>in</strong>sem<strong>in</strong>ation. This paper discusses thetechnological development of AI and explores a number of theethical issues raised by its application to dog breed<strong>in</strong>g. AnEthical Matrix method is used to map the potential ethicalissues for key <strong>in</strong>terest groups, namely dogs, breeders, owners,veter<strong>in</strong>arians and wider society. There are national variations<strong>in</strong> the way <strong>in</strong> which <strong>in</strong>stitutions have evaluated potentialethical impacts, and this is reflected <strong>in</strong> the different regulatoryframeworks govern<strong>in</strong>g the use of AI <strong>in</strong> dogs. In order tofacilitate decision-mak<strong>in</strong>g and reduce some of the ethical risksassociated with this technology, the veter<strong>in</strong>ary research communitycould take several proactive steps <strong>in</strong>clud<strong>in</strong>g: (i)clarify<strong>in</strong>g cl<strong>in</strong>ical decision-mak<strong>in</strong>g processes, (ii) enhanc<strong>in</strong>g<strong>in</strong>formed choice among clients and (iii) <strong>in</strong>creas<strong>in</strong>g the knowledge-baseof potential impacts of AI.IntroductionArtificial <strong>in</strong>sem<strong>in</strong>ation (AI) is the process of plac<strong>in</strong>gsemen <strong>in</strong>to the female reproductive tract without anatural mat<strong>in</strong>g. AI has become popular as a result ofthe import<strong>in</strong>g and export<strong>in</strong>g of frozen dog semen,however, semen can be <strong>in</strong>sem<strong>in</strong>ated fresh, or cooled andstored for a few days prior to <strong>in</strong>sem<strong>in</strong>ation, or frozenand stored <strong>in</strong>def<strong>in</strong>itely before be<strong>in</strong>g thawed prior to use.The first specific report of mammalian AI was <strong>in</strong> a dog<strong>in</strong> the 1700s, and the first AI follow<strong>in</strong>g storage of dogsemen was reported <strong>in</strong> the 1950s. In the ensu<strong>in</strong>g50 years, there have been huge developments <strong>in</strong> reproductivebiology and biotechnology enabl<strong>in</strong>g the use,and potential misuse, of AI. There are national variations<strong>in</strong> the ethical and legislative frameworks govern<strong>in</strong>gthe use of AI <strong>in</strong> dogs, and the purpose of this paperis to review the techniques and consider some of thepotential ethical questions posed by <strong>in</strong>creas<strong>in</strong>g use ofAI <strong>in</strong> this species.Role of Artificial Insem<strong>in</strong>ation <strong>in</strong> DogsAdvantages and disadvantages of AIThere is no doubt that AI with fresh semen has anumber of potential advantages <strong>in</strong> the management ofbreed<strong>in</strong>g <strong>in</strong> dogs <strong>in</strong>clud<strong>in</strong>g that it: (i) may allow the useof males or females that are unable to breed because ofanatomical or pathological reasons, (ii) may overcomerefusal to breed because of psychological reasons, (iii)may allow the splitt<strong>in</strong>g of an ejaculate so that morefemales can be bred, (iv) is an acceptable method of l<strong>in</strong>ebreed<strong>in</strong>g, (v) can be highly efficient at facilitat<strong>in</strong>g geneticimprovement, (vi) may allow the control of some<strong>in</strong>fectious diseases either by remov<strong>in</strong>g physicalcontact between animals or by allow<strong>in</strong>g treatment ofsemen prior to <strong>in</strong>sem<strong>in</strong>ation, (vii) enables exam<strong>in</strong>ationof semen quality prior to <strong>in</strong>sem<strong>in</strong>ation and if necessarythe selection of an alternative stud, (viii) can be aconvenient and rapid method of breed<strong>in</strong>g, and (ix) bycryopreservation enables conservation and storage ofvaluable genes from male animals almost <strong>in</strong>def<strong>in</strong>itely.When comb<strong>in</strong>ed with semen preservation, there areadditional potential advantages <strong>in</strong>clud<strong>in</strong>g; (i) mak<strong>in</strong>gshipp<strong>in</strong>g of semen possible such that genetic material isavailable from outside of a breed<strong>in</strong>g colony, (ii) overcom<strong>in</strong>gquarant<strong>in</strong>e restrictions, (iii) overcom<strong>in</strong>g theneed to transport the animal (reduc<strong>in</strong>g transportationstress ⁄ disease risks), and (iv) the ability to utilize semenafter death of the male. For breeders produc<strong>in</strong>g dogs forprofessional purposes, such as bomb ⁄ m<strong>in</strong>e detectiondogs, drug detection dogs and guide dogs for the bl<strong>in</strong>d,etc., semen freez<strong>in</strong>g can enable castration of males at anearly age but ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g availability of their geneswhile their performance can be evaluated. The need tokeep males <strong>in</strong> the breed<strong>in</strong>g kennels is also reduced,which will reduce costs.Despite these clear benefits, the possible veter<strong>in</strong>arydisadvantages are often overlooked. These can <strong>in</strong>clude;(i) caus<strong>in</strong>g physical or psychological trauma dur<strong>in</strong>g theAI process, (ii) undertak<strong>in</strong>g AI for <strong>in</strong>appropriatereasons (e.g. where reluctance to breed is a manifestationof underly<strong>in</strong>g hereditary disease such as hipdysplasia or anatomical abnormality of the reproductivetract), (iii) potential for <strong>in</strong>troduction of heritablediseases or abnormalities, (iv) potentially allow<strong>in</strong>goveruse of a male with<strong>in</strong> a programme or breed, and(v) possibly allow<strong>in</strong>g confusion of parentage (for reviewsee L<strong>in</strong>de-Forsberg 2002). In all the cases, it would beprudent to ensure a controlled and certified process<strong>in</strong>volv<strong>in</strong>g cl<strong>in</strong>ical and ⁄ or molecular exam<strong>in</strong>ation ofmales to ensure that they do no spread genetic or<strong>in</strong>fectious disease.Technique of <strong>in</strong>sem<strong>in</strong>ationAt the time of natural mat<strong>in</strong>g, semen is deposited with<strong>in</strong>the vag<strong>in</strong>a but sperm are transported <strong>in</strong>to the uterusbecause of significant vag<strong>in</strong>al and active uter<strong>in</strong>e contractions(Evans 1933). While vag<strong>in</strong>al <strong>in</strong>sem<strong>in</strong>ation[AI(V)] is a simple procedure that is well tolerated <strong>in</strong>the bitch (Seager et al. 1975), vag<strong>in</strong>al contractions areÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


166 GCW England and KM Millarscant even when efforts are made to cause vag<strong>in</strong>aldistension, and active uter<strong>in</strong>e contractions do notappear to develop at least not as to the same extent aswith natural mat<strong>in</strong>g (GCW England, unpublishedobservations). The lack of a bitch’s physiological responseat the time of AI results <strong>in</strong> relatively few spermbe<strong>in</strong>g transported <strong>in</strong>to the uterus and this situation isworse for cryopreserved semen, because the spermthemselves have poor motility. Furthermore, cryopreservedsperm have a short longevity compared withfreshly ejaculated sperm (Olar 1984). These two effectsprobably expla<strong>in</strong> the relatively poor fertility observedwhen cryopreserved semen is <strong>in</strong>sem<strong>in</strong>ated <strong>in</strong>to thevag<strong>in</strong>a (Olar 1984).For these reasons, a number of techniques have beendeveloped to place the semen at the cervix or <strong>in</strong>to theuterus. This is difficult <strong>in</strong> the bitch, because the vag<strong>in</strong>a islong and narrow and the cervix is placed at anunusual angle (L<strong>in</strong>dsay 1983). However, Takeishi et al.(1976) reported success from <strong>in</strong>tra-cervical <strong>in</strong>sem<strong>in</strong>ation[AI(C)], and various commercial devices have beenproduced to achieve AI(C) whereby semen is forced <strong>in</strong>tothe cervix us<strong>in</strong>g a special catheter, which forms a tightseal at the cranial vag<strong>in</strong>a. The catheter can be left <strong>in</strong>place to simulate a copulatory ‘tie’.Transcervical <strong>in</strong>trauter<strong>in</strong>e <strong>in</strong>sem<strong>in</strong>ation [AI(TC)] ispossible us<strong>in</strong>g a method described <strong>in</strong>itially by Fougneret al. (1973). An outer catheter sheath is placed <strong>in</strong>to thevag<strong>in</strong>a, the cervix can then be realigned by palpat<strong>in</strong>gthrough the abdom<strong>in</strong>al wall and a central catheter is<strong>in</strong>serted through the cervix. Relatively few reports detailthe success rates <strong>in</strong> achiev<strong>in</strong>g AI(TC) or the <strong>in</strong>cidence ofcomplications (England and Verstegen 1996); however,although overall the technique is thought to be m<strong>in</strong>imally<strong>in</strong>vasive, it is reported that the transabdom<strong>in</strong>alpalpation is resented by a significant proportion ofbitches (Wilson 1993, 2001). The AI(TC) was furtherdeveloped by Wilson (1993) with a rigid endoscopicmethod and us<strong>in</strong>g a wire to guide a catheter through thecervix. Wilson (1993) reported that 97% of bitches couldbe relatively easily catheterized us<strong>in</strong>g this method, andwhile it is clear that significant tra<strong>in</strong><strong>in</strong>g is required, todate no reports of adverse effects have been reported(Wilson 2001).A simple way to overcome the requirement fortra<strong>in</strong><strong>in</strong>g and to ensure that uter<strong>in</strong>e AI can be reliablyperformed is to undertake surgical <strong>in</strong>trauter<strong>in</strong>e <strong>in</strong>sem<strong>in</strong>ation[AI(S)] at laparotomy or laparoscopy (Smith1984; Wildt 1986). Clearly, these methods are <strong>in</strong>vasiveand require general anaesthesia, and although there iswide applicability <strong>in</strong> some countries, especially the USA,the ethics of a surgical <strong>in</strong>sem<strong>in</strong>ation has been the subjectof some debate (Royal College of Veter<strong>in</strong>ary Surgeons(RCVS) 2005). Interest<strong>in</strong>gly, there are no published dataon complications of surgical <strong>in</strong>sem<strong>in</strong>ation.Success rates of artificial <strong>in</strong>sem<strong>in</strong>ationThere are many reports of pregnancy rates after AI, butwith a few notable exceptions (L<strong>in</strong>de-Forsberg andForsberg 1993; Thomassen et al. 2006), most <strong>in</strong>volvevery small numbers of animals and lack adequatecontrol groups. Furthermore, variations <strong>in</strong> pregnancyrates may be the result of differences <strong>in</strong> tim<strong>in</strong>g of<strong>in</strong>sem<strong>in</strong>ation, quality of the semen <strong>in</strong>sem<strong>in</strong>ated, site ofsemen deposition, number of <strong>in</strong>sem<strong>in</strong>ations and the<strong>in</strong>herent fertility of the female and the male (<strong>in</strong>clud<strong>in</strong>geffects of age). L<strong>in</strong>de-Forsberg and Forsberg (1993)developed a simple scor<strong>in</strong>g scheme <strong>in</strong> an attempt toquantify these variables, but application of their methodto other published studies is difficult as many authorsfail to report these important factors. Nevertheless, it iscommonly agreed that; (i) fresh semen AI has a greatersuccess rate than cryopreserved semen AI regardless ofthe site of <strong>in</strong>sem<strong>in</strong>ation, (ii) <strong>in</strong>creas<strong>in</strong>g the number ofsperm <strong>in</strong>sem<strong>in</strong>ated improves the success rate regardlessof the site of AI, (iii) multiple AIs have a greater successrate than s<strong>in</strong>gle AIs, (iv) AI(TC) and AI(S) producehigher pregnancy rates than AI(V), especially whenus<strong>in</strong>g frozen-thawed semen (see Fontbonne and Bad<strong>in</strong>and1993; L<strong>in</strong>de-Forsberg and Forsberg 1993; Thomassenet al. 2006).The success with fresh semen AI depends upon itsquality and the fertility of the bitch, but pregnancy ratesare approximately 80 ± 16 (SD)% for AI(V) and97 ± 4 (SD)% for AI(TC) and AI(S). Reported pregnancyrates for chilled semen <strong>in</strong>sem<strong>in</strong>ations are onaverage 47 ± 9 (SD)% for AI(V) and 81 ± 19 (SD)%for AI(TC). F<strong>in</strong>ally, pregnancy rates for frozen-thawedsemen are on average: 45 ± 24 (SD)% for AI(V);60 ± 15 (SD)% for AI(C); 70 ± 11 (SD)% for AI(TC);and 95 ± 7 (SD)% for AI(S) (data collatedfrom: Seager et al. 1975; Olar 1984; Smith 1984;Fontbonne and Bad<strong>in</strong>and 1993; L<strong>in</strong>de-Forsberg andForsberg 1993; Wilson 2001; Thomassen et al. 2006). Itis important that these data are <strong>in</strong>terpreted cautiouslybecause of significant variations <strong>in</strong> methodology and thesmall numbers of animals used <strong>in</strong> many of the studies.Conduct<strong>in</strong>g an Ethical AnalysisWith technological improvements and an <strong>in</strong>creas<strong>in</strong>g<strong>in</strong>terest <strong>in</strong> the use of AI(TC) and AI(S), a number ofsignificant ethical concerns have been raised (RCVS2005). One approach for explor<strong>in</strong>g these ethical issuesis to conduct a structured ethical analysis. A numberof methods have been developed to facilitate ethicalanalysis and stakeholder engagement, <strong>in</strong>clud<strong>in</strong>g theEthical Matrix (EM) method (Mepham 2000). TheEM is applied to facilitate the assessment of a proposedstrategy (i.e. the use of reproductive technology) <strong>in</strong>terms of respect (or lack of respect) for three ethicalpr<strong>in</strong>ciples; wellbe<strong>in</strong>g, autonomy and fairness, as appliedto a def<strong>in</strong>ed set of <strong>in</strong>terest groups. The ‘weight’ orsignificance assigned to each ethical impact is determ<strong>in</strong>edby the evaluation of evidence. The EM methodhas been previously applied to a number of biotechnologycases (e.g. Mepham 2000; Mepham et al. 2006;Millar and Tomk<strong>in</strong>s 2007). It should be noted that thismethod is not prescriptive and therefore will notproduce ‘an answer’, but the method can make theethically relevant issues transparent and thus facilitate<strong>in</strong>formed decision-mak<strong>in</strong>g. The value of the approach isthat, it makes explicit the evidence used to justify aposition and encourages ethical reflection on the impactsfor all ethically relevant <strong>in</strong>terest groups. The method canÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


The Ethics and Role of AI <strong>in</strong> Dogs 167Modified ethical matrix(Translation of the ethical pr<strong>in</strong>ciples for the correspond<strong>in</strong>g <strong>in</strong>terest group)WELLBEING AUTONOMY FAIRNESSDOGS(dog, bitch andpuppies)Welfare(DW)Behaviouralfreedom(DA)Intr<strong>in</strong>sicvalue(DF)BREEDERSSatisfactory <strong>in</strong>comeand work<strong>in</strong>gconditions (BW)Managerialfreedom(BA)Fair regulations andtrade(BF)OWNERSSafety andquality of life(OW)Choice(OA)Affordability ofproducts(OF)VETERINARIANSSatisfactory <strong>in</strong>comeand work<strong>in</strong>gconditions(VW)Professionalfreedom(VA)Equitable standardsof practice(VF)Fig. 1. Modified ethical matrixSOCIETYSafety and socialharmony(SW)Democratic choice(SA)Fair resourceallocation(SF)also act as a start<strong>in</strong>g po<strong>in</strong>t for ethical deliberation <strong>in</strong>public policy decision-mak<strong>in</strong>g.When consider<strong>in</strong>g the ethical issues raised by the useof AI <strong>in</strong> the dog, an adapted form of the Matrix methodcan be applied. This adapted EM (Fig. 1) will be appliedto map the issues raised and to analyse whether theapplication of AI approaches might <strong>in</strong>fr<strong>in</strong>ge upon orsupport broadly-def<strong>in</strong>ed ethical pr<strong>in</strong>ciples for the specified<strong>in</strong>terest groups. With<strong>in</strong> the limits of this paper it isnot possible to comprehensively explore all of the ethicalissues raised by the various forms of AI; however, thismethod can be used to highlight some of the moreprom<strong>in</strong>ent ethical considerations and clarify whereconflicts may arise. It should be noted that ethicalanalysis will highlight differences between the differentforms of AI (with fresh or frozen semen).The modified Matrix, which will be applied here,<strong>in</strong>corporates five <strong>in</strong>terest groups; dogs, breeders, owners,veter<strong>in</strong>arians and wider society. Here, breeders aredef<strong>in</strong>ed as those <strong>in</strong>dividuals responsible for the mat<strong>in</strong>gof the bitch and dog, whereas owners are the recipientsof the pups. In a number of cases, this can be the sameperson, but this dist<strong>in</strong>ction is important when consider<strong>in</strong>gthe ethical issues raised.Ethical Analysis of the Use of AI by InterestGroupDogs (dog, bitch and puppies)Welfare (DW)It is necessary to consider two welfare aspects whenassess<strong>in</strong>g the use of AI <strong>in</strong> dogs; firstly, what are thegeneric benefits and risks for the bitch or ⁄ and dog? andsecondly, what are the risks associated with the differentAI methods?Many organizations classify natural breed<strong>in</strong>g as thepreferable means of produc<strong>in</strong>g a pregnancy. In order torespect the wellbe<strong>in</strong>g particularly of the bitch, anydecision to <strong>in</strong>tervene must be carried out <strong>in</strong> her best<strong>in</strong>terests. Some benefits can be conferred from <strong>in</strong>tervention;specifically, forced mat<strong>in</strong>g may be traumatic <strong>in</strong>itself and can lead to physical and psychological harm.In such cases, the use of AI can respect animal wellbe<strong>in</strong>gand prevent harm, yet, the selection of an alternativesexual partner would represent greater respect. Wellbe<strong>in</strong>gmay be respected if the use of AI protects aga<strong>in</strong>st thetransmission of <strong>in</strong>fectious disease. However, it mightbe argued that the existence of a disease risk may bejustification <strong>in</strong> itself not to breed from these animals. Incountries where import regulations are strict andwhere the breed<strong>in</strong>g population is very small, AI maybe necessary to prevent <strong>in</strong>breed<strong>in</strong>g, which may compromisehealth.It has been claimed that the bitch ga<strong>in</strong>s no welfarebenefit from becom<strong>in</strong>g pregnant. However, beyond abehavioural need, some authors contend that bitchesmay ga<strong>in</strong> a number of physiological benefits frompregnancy and lactation such as reduc<strong>in</strong>g the risk ofdevelop<strong>in</strong>g pyometra and mammary neoplasia, althoughevidence to support these assertions is very anecdotal.Even if reproductive <strong>in</strong>tervention is not considered <strong>in</strong>the best <strong>in</strong>terest of the dog, <strong>in</strong>tervention may representonly a neutral or very m<strong>in</strong>or <strong>in</strong>fr<strong>in</strong>gement of wellbe<strong>in</strong>g(although any <strong>in</strong>tervention must be expla<strong>in</strong>ed andclearly justified). It is, therefore, important to ascerta<strong>in</strong>the documented impact on animal wellbe<strong>in</strong>g and theperceived risks from the three forms of AI.Complications from AI appear to be rare and thewelfare risks associated with AI(V) often relate only tothe need to physically restra<strong>in</strong> the bitch <strong>in</strong> order to<strong>in</strong>sem<strong>in</strong>ate. It might be suggested that any form ofuter<strong>in</strong>e AI has a greater welfare risk than AI(V). Of themethods available, AI(TC) is by its nature a reducedwelfare burden than AI(S). However, the use ofAI(TC) is seen as a procedure requir<strong>in</strong>g a skilledoperator and <strong>in</strong> <strong>in</strong>experienced hands may result <strong>in</strong> localÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


168 GCW England and KM Millartrauma or a prolonged and potentially stressful procedure.The AI(S) is an <strong>in</strong>fr<strong>in</strong>gement of wellbe<strong>in</strong>g even ifthe procedure is conducted without complications.Justification for the use of AI(S) is that, adverse effectsare m<strong>in</strong>imal and therefore the welfare risk is equallysmall; however, there is limited data to support orrefute this statement. Some have also claimed thatAI(S) may be less stressful to the animal than a difficultAI(TC), and that AI(TC) may not be possible <strong>in</strong> somesmaller breeds as well as <strong>in</strong> some large or obese bitches;however, both these difficulties may be due to operator<strong>in</strong>experience or <strong>in</strong>appropriate equipment (i.e. endoscopesize). The use of low-dose sedatives, may reducehandl<strong>in</strong>g stress.When exam<strong>in</strong><strong>in</strong>g the risks of AI and the animalwelfare burden, the use of non-surgical approaches,namely AI(V) and AI(TC), respectively, appear torepresent far smaller welfare risks than AI(S).Behavioural freedom (DA)When AI is used over natural mat<strong>in</strong>g, dogs and bitchesare denied the ability to engage <strong>in</strong> normal sexualbehaviour (a negative <strong>in</strong>fr<strong>in</strong>gement). Studies have <strong>in</strong>dicatedthat some bitches can be highly selective <strong>in</strong> theirmat<strong>in</strong>g choices. If breeder <strong>in</strong>tervention occurs, eitherforced mat<strong>in</strong>g or <strong>in</strong>sem<strong>in</strong>ation removes the ability toself-determ<strong>in</strong>e the mate and this <strong>in</strong>fr<strong>in</strong>ges the <strong>in</strong>st<strong>in</strong>ctivebehavioural pattern. In addition, forced mat<strong>in</strong>g can betraumatic for an unreceptive bitch. Cont<strong>in</strong>ued attemptsto mate could re<strong>in</strong>force a negative behavioural experience.In these circumstances, the use of AI would be theonly option for breed<strong>in</strong>g.Intr<strong>in</strong>sic value (DF)When consider<strong>in</strong>g whether the use of AI may <strong>in</strong>fr<strong>in</strong>geor respect the notion of <strong>in</strong>tr<strong>in</strong>sic value of the animal, itis important to consider whether the application ofthese technologies is potentially <strong>in</strong>creas<strong>in</strong>g the objectificationof the dog or bitch. It could be argued thatthe need to produce pups results <strong>in</strong> the dog be<strong>in</strong>g seenmerely as a ‘reproductive vessel’. This may <strong>in</strong> turndetract from the relationship between dog and owner⁄ breeder and deny the dog respect as an animal thathas value <strong>in</strong> its own right, beyond its <strong>in</strong>strumentalvalue, for example, as a work<strong>in</strong>g dog or breed<strong>in</strong>g dog.However, it may be argued that the dog has an <strong>in</strong>natedrive to reproduce and raise young and that bydeny<strong>in</strong>g the bitch the ability to do so would be an<strong>in</strong>fr<strong>in</strong>gement of her ‘telos’ or <strong>in</strong>nate purpose (l<strong>in</strong>ked torespect for behavioural freedom). There is no <strong>in</strong>dicationfrom the literature or from anedoctal experienceby dog breeders that, bitches that are artificially<strong>in</strong>sem<strong>in</strong>ated as opposed to naturally mated femalesshow less maternal behaviour towards their pups.BreedersSatisfactory <strong>in</strong>come and work<strong>in</strong>g conditions (BW)Breeders are not a homogenous group; many breeders,particularly <strong>in</strong> the UK, are <strong>in</strong>dividuals who operate ona non-commercial basis and are driven by theiradmiration of a specific breed. In contrast, a numberof breeders focus on economic factors and a need toproduce pedigree dogs for sale. However, when consider<strong>in</strong>gthe conditions that drive requests to veter<strong>in</strong>ariansto use AI, a number of overarch<strong>in</strong>g aspects can beidentified for all breeders as well as specific aspects thatrelate exclusively to commercially-oriented breeders.The availability of assisted reproductive technologies(ARTs) allows breeders to manage their breed<strong>in</strong>gprogrammes and maximize fertility rates for their bestbitches. The availability of AI prevents breed<strong>in</strong>g delaysand should not reduce litter size, which might prohibituse. In addition, breeders may wish to <strong>in</strong>troducebreed<strong>in</strong>g programmes for animals that have exaggeratedphysical or temperamental features, which canmake natural mat<strong>in</strong>g difficult. However, this may result<strong>in</strong> breeders susta<strong>in</strong><strong>in</strong>g problem behaviour or undesirablephysical features with<strong>in</strong> a l<strong>in</strong>e. The use of AIcould, therefore, <strong>in</strong>advertently perpetuate the selectionof undesirable traits with<strong>in</strong> a breed which wouldnegatively impact on breeders’ wellbe<strong>in</strong>g as well asbe<strong>in</strong>g detrimental for the breed. In contrast, thetechnology could allow the <strong>in</strong>troduction of desirabletraits and ensure the preservation of endangered breedswith the secondary benefit of potentially enhanc<strong>in</strong>g thewellbe<strong>in</strong>g of breeders.Managerial freedom (BA)Breeders are free to apply and optimize all availablereproductive technologies. Some breeders may have nooption other than to request AI, as quality breed<strong>in</strong>gdogs may only realistically be available from otherbreeders outside of their geographic region. AI alsoallows breeders to use those dogs that are not accustomedto mat<strong>in</strong>g bitches (e.g. <strong>in</strong>experienced dogs) or thedogs that are rejected by the bitch. The technology canensure that these quality animals are still able to breedand produce litters. However, the free availability of thistechnology may result <strong>in</strong> breeders opt<strong>in</strong>g for AI forvirg<strong>in</strong> dogs and bitches, rather than <strong>in</strong>vest<strong>in</strong>g time <strong>in</strong>tra<strong>in</strong><strong>in</strong>g these animals to mate naturally. This may leadto an ‘<strong>in</strong>tervention treadmill’ where AI is alwaysrequested for high value animals to ensure timelypregnancies.Fair regulations and trade (BF)The <strong>in</strong>creas<strong>in</strong>g availability of new reproductive technologiesmay underm<strong>in</strong>e the ability of smaller breeders totrade <strong>in</strong> an <strong>in</strong>creas<strong>in</strong>gly competitive environment, asonly large commercial breeders will have the f<strong>in</strong>ancialmeans to access these technologies. This differencebetween commercial and leisure breeders may be positivelyre<strong>in</strong>forc<strong>in</strong>g over time and could eventually changethe composition of the breed<strong>in</strong>g community. However,this argument could be advanced for all new <strong>in</strong>novationssuch as nutritional advances, veter<strong>in</strong>ary treatment, etc.,therefore represent<strong>in</strong>g a m<strong>in</strong>or or neutral <strong>in</strong>fr<strong>in</strong>gementof the pr<strong>in</strong>ciple of justice for all breeders. The breed<strong>in</strong>gof certa<strong>in</strong> dogs, particularly sport<strong>in</strong>g dogs and servicedogs, may be a highly commercial enterprise, andtherefore, improv<strong>in</strong>g breed<strong>in</strong>g efficiency is a naturalÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


The Ethics and Role of AI <strong>in</strong> Dogs 169progression for an <strong>in</strong>dustry that is develop<strong>in</strong>g with<strong>in</strong> amarket economy.Owners (recipients of pups)Safety and quality of life (OW)Us<strong>in</strong>g AI may reduce the risk of disease transmissionthat will not only benefit the dog but could also havepotentially positive impacts on the puppies’ owner(s).Use of AI may ensure a high-quality supply of pupsand allow owners to receive a pup on demand, ratherthan be<strong>in</strong>g affected by breed<strong>in</strong>g difficulties. However,AI use with problem dogs (male and female) mayresult <strong>in</strong> progeny that are unsuitable as companionanimals (e.g. behavioural problems) or which will notreproduce themselves without <strong>in</strong>tervention (e.g. anatomicalor physiological limitations). The overuse ofmales through AI may also facilitate the <strong>in</strong>corporationof undesirable traits with<strong>in</strong> a l<strong>in</strong>e that are notapparent until later generations. Any form of ARTuse may be seen by some to <strong>in</strong>fr<strong>in</strong>ge the dog–ownerrelationship. The risks to animal welfare associatedwith the use of AI and the perceived view of theunnaturalness of the process may be seen to have anegative impact on the companion animal–humanrelationship and, hence, <strong>in</strong>fr<strong>in</strong>ge the wellbe<strong>in</strong>g of theowner.Choice (OA)In order to respect the pr<strong>in</strong>ciple of autonomy forpotential owners, it is important that all techniquesand procedures applied to produce pups are disclosed.This will allow owners to make an <strong>in</strong>formed decisionabout whether they want a pup that has beenconceived us<strong>in</strong>g AI, particularly AI(S); as this maybe an important differentiation for some owners.Potential owners should also be privy to the justificationfor us<strong>in</strong>g AI, i.e. such as for behavioural oranatomical reasons. In order to respect the autonomyof potential owners, this <strong>in</strong>formation should be logged<strong>in</strong> the breed<strong>in</strong>g record and should be offered (i.e. thisis a positive duty) to owners rather than be<strong>in</strong>g madeavailable on request. The use of AI may compromisethe reliability of the parentage <strong>in</strong>formation as semenmay be ‘mixed’ or the orig<strong>in</strong> may be difficult to verify.However, this risk could be managed through currentsystems such as breeder certification and veter<strong>in</strong>arysupervision of AI (e.g. the use of stud books andmandatory DNA sampl<strong>in</strong>g at the time of semencollection etc.).Affordability of products (OF)Recently, the International Association of Human-Animal Interaction Organizations declared that ‘it is auniversal, natural and basic human right to benefit fromthe presence of animals’. The <strong>in</strong>troduction of AI technologiescould result <strong>in</strong> a number of specialized breedsbe<strong>in</strong>g prohibitively expensive for some members ofsociety. If that was the case, an <strong>in</strong>crease <strong>in</strong> the use of thistechnology with<strong>in</strong> the dog-breed<strong>in</strong>g community may beseen as an <strong>in</strong>fr<strong>in</strong>gement, and therefore, unfair to someeconomically disadvantaged members of the dog-own<strong>in</strong>gcommunity.Veter<strong>in</strong>ariansSatisfactory <strong>in</strong>come and work<strong>in</strong>g conditions (VW)Unlike human health professionals where services aresupported via public fund<strong>in</strong>g, veter<strong>in</strong>arians operate <strong>in</strong> amarket environment where the quality and diversity oftheir services <strong>in</strong>fluence their <strong>in</strong>come streams. Veter<strong>in</strong>arianswho <strong>in</strong>vest <strong>in</strong> new skills and are able to offeradditional services can enhance the profitability of theirpractice and their personal <strong>in</strong>come. In a marketenvironment, this can result <strong>in</strong> a competitive advantageover rival practices. The ability to respond to clients’needs and offer new services, such as AI, enhances(respects) veter<strong>in</strong>arians’ wellbe<strong>in</strong>g.Professional freedom (VA)Freedom to <strong>in</strong>novate is an important driver of change <strong>in</strong>many fields and this is no less the case for the veter<strong>in</strong>aryprofession. By develop<strong>in</strong>g and apply<strong>in</strong>g new diagnosticmethods, surgical techniques and veter<strong>in</strong>ary products,veter<strong>in</strong>arians have improved the wellbe<strong>in</strong>g of theirpatients as well as their clients (e.g. dog owners). Theopportunity to use AI allows veter<strong>in</strong>arians to determ<strong>in</strong>ethe best course of treatment for their patients sorespect<strong>in</strong>g their professional autonomy, particularly ifthe cause of the bitches’ <strong>in</strong>fertility is a barrier that canonly be overcome by AI. Artificial <strong>in</strong>sem<strong>in</strong>ation mayprovide the only option for treat<strong>in</strong>g dogs that arecompromised because of anatomical, physiological orbehavioural problems.Equitable standards of practice (VF)It may be claimed that the availability of noveltechnologies, which are cost-effective but perhaps poserisks for animal welfare (e.g. surgical risks), couldunduly <strong>in</strong>fluence the market and decrease the use ofother non-<strong>in</strong>vasive techniques that may require additionalskills or be more time-demand<strong>in</strong>g. In order toensure the pr<strong>in</strong>ciple of justice is not <strong>in</strong>fr<strong>in</strong>ged, adequate<strong>in</strong>formation would need to be provided to breeders andowners on the options for treatment. This could <strong>in</strong>cludeadvis<strong>in</strong>g aga<strong>in</strong>st the use of an <strong>in</strong>tervention method orpossibly even advis<strong>in</strong>g aga<strong>in</strong>st breed<strong>in</strong>g from a particulardog. In addition, when consider<strong>in</strong>g the use of thedifferent AI methods, the risks of each technique shouldbe clearly stated and, if necessary, referral to otherqualified veter<strong>in</strong>arians should be offered. This wouldensure that trad<strong>in</strong>g and professional standards were notunduly <strong>in</strong>fluenced by lack of transparency <strong>in</strong> the market.SocietySafety and social harmony (SW)The rout<strong>in</strong>e use of reproductive technologies may affectthe human–dog relationship by <strong>in</strong>creas<strong>in</strong>g society’s<strong>in</strong>strumental view of companion animals. However,the cl<strong>in</strong>ical ability to <strong>in</strong>tervene and facilitate a successfulpregnancy may further enhance the positive nature ofÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


170 GCW England and KM Millarthe animal–human bond. The impact on society’swellbe<strong>in</strong>g will be <strong>in</strong>fluenced by the justification putforward for us<strong>in</strong>g the technique and whether the stated‘reasons’ enhance or <strong>in</strong>fr<strong>in</strong>ge the human–dog relationship.Democratic choice (SA)The development and application of these techniquesmay result <strong>in</strong> the ability to conserve valuable genetictraits for the can<strong>in</strong>e species and this could be seen as apositive impact. This will ensure society’s ability tochoose the breed traits that it wishes to preserve andpotentially allow future generations a degree of autonomythat may not have been possible without theapplication of these technologies. However, this argumentis relevant to the use of the technique only whenapplied to specific breed<strong>in</strong>g strategies, rather than forrout<strong>in</strong>e treatment purposes.Fair resource allocation (SF)Because the f<strong>in</strong>ancial cost of us<strong>in</strong>g any form of AI willbe born by the breeder and the potential owner of thenew pup, the impact for the pr<strong>in</strong>ciple of justice forsociety will be neutral.Ethical Evaluation and DiscussionAlthough, because of the limits of this paper, the ethicalanalysis has only <strong>in</strong>itially mapped the possible impactsof the application of AI technologies <strong>in</strong> dogs, theanalysis has highlighted a number of important issues.In order to come to a f<strong>in</strong>al judgement or position, theimpacts for the various <strong>in</strong>terest groups need to beweighed aga<strong>in</strong>st each other (ethical evaluation). It is thisweigh<strong>in</strong>g that can help identify the key areas ofdisagreement or value conflict between stakeholders.This process can also help identify knowledge gaps andareas of uncerta<strong>in</strong>ty. For some groups, the use of ascor<strong>in</strong>g system (e.g. +1, 0, )1) can aid this weigh<strong>in</strong>gprocess, but it should be noted that the EM is a decisionsupportframework and not a decision-mak<strong>in</strong>g tool. Theuse of numerical weigh<strong>in</strong>g <strong>in</strong> ethical evaluation has itslimitations.It is clear from the analysis that the use of AI may<strong>in</strong>fr<strong>in</strong>ge ethical pr<strong>in</strong>ciples, particularly for the affectedanimal, but that it may respect other pr<strong>in</strong>ciples forbreeders, owners and veter<strong>in</strong>arians. Any veter<strong>in</strong>aryprocedure has a risk of <strong>in</strong>fr<strong>in</strong>g<strong>in</strong>g an animal’s wellbe<strong>in</strong>gand autonomy as well as potentially be<strong>in</strong>g unjust.However, veter<strong>in</strong>arian <strong>in</strong>tervention is repeatedly justifiedon the basis of cl<strong>in</strong>ical ‘need’ (i.e. <strong>in</strong>flict<strong>in</strong>g acutepa<strong>in</strong> for long-term benefit). One of the key issues <strong>in</strong> thedebate that surrounds the use of AI is the <strong>in</strong>terpretationof ‘need’, as any ‘unnecessary procedure’ would representan <strong>in</strong>fr<strong>in</strong>gement of the dog’s wellbe<strong>in</strong>g and wouldnot be an ethically acceptable <strong>in</strong>tervention under themajority of EU veter<strong>in</strong>ary codes of practice. This impliesthat before AI is considered appropriate a comprehensivereproductive assessment must be carried out by aveter<strong>in</strong>arian. It also implies that a sequential approachshould be applied with the justification for each decisionstep [i.e. rul<strong>in</strong>g out natural service and proceed<strong>in</strong>g to theuse of AI(V)]. This <strong>in</strong>formation should then beproactively offered to future puppy owners and kennelclubs, etc.While the use of AI can result <strong>in</strong> a number of positiveethical impacts (for example for disease control orpreservation of genetic material), which outweigh potentialrisks, such benefits appears to be predicated onfour conditions: (i) a sequential decision-mak<strong>in</strong>g process,which ensures that the use of AI is applied after naturalmat<strong>in</strong>g options are ruled out for cl<strong>in</strong>ical reasons, (ii)<strong>in</strong>formed choice for breeder and owner is ensuredthrough proactive <strong>in</strong>formation provision and appropriaterecord keep<strong>in</strong>g, (iii) veter<strong>in</strong>arian competence <strong>in</strong> use ofAI technologies is ensured, and (iv) the welfare consequencesfor the bitch are measured as negligible.If AI per se is acceptable under these conditions, italso appears that the judgment on whether surgical AI isacceptable (when all other options are excluded) ismodulated by evidence on (i) the <strong>in</strong>cidence and nature ofthe reproductive failure and (ii) welfare consequencesfor the bitch. What appears to confound the assessmentof these two issues is a paucity of data. The literatureconta<strong>in</strong>s very few reports on the consequences for thebitch and there are even fewer studies that record thefrequency of use and the form of the decision-mak<strong>in</strong>gprocess. There is a significant need for peer-reviewedevidence <strong>in</strong> these areas <strong>in</strong> order to facilitate an <strong>in</strong>formedethical evaluation.Without explicitly conduct<strong>in</strong>g an ethical analysis, anumber of national bodies and professional organizationshave articulated their approach to ‘weigh<strong>in</strong>g’ theconflict<strong>in</strong>g impacts and set out their ethical positions(e.g. through guidel<strong>in</strong>es, regulations, etc.). It is usefulhere to exam<strong>in</strong>e a few of these positions and to reflect onthe risks associated with these strategies. In the UK, theRoyal College of Veter<strong>in</strong>ary Surgeons (RCVS) set outtheir position (RCVS, 2005), stat<strong>in</strong>g that, although theuse of surgical AI ‘is unlikely to be carried out <strong>in</strong> the best<strong>in</strong>terests of any particular dog’, veter<strong>in</strong>ary surgeons mayperform the procedure when justified (e.g. ‘for example,the <strong>in</strong>corporation of new genetic traits’). The RCVS<strong>in</strong>dicated that the reasons for not us<strong>in</strong>g other approaches,e.g. transcervical <strong>in</strong>sem<strong>in</strong>ation, should berecorded. The RCVS approach is more conservativethan that applied <strong>in</strong> the USA where AI(S) is morecommonly used. It is important to note that the RCVS’sethical position is based on the assumption that, <strong>in</strong>exceptional circumstances, the potential welfare consequencesfor the dog are acceptable only if a sequentialdecision-mak<strong>in</strong>g process and good record keep<strong>in</strong>goccur. However, no explicit advice is given on ensur<strong>in</strong>g<strong>in</strong>formed choice [i.e. <strong>in</strong>formation provision on theavailability of AI(TC)] or whether the ‘recorded justification’for the procedure will be audited and reviewedby the RCVS. Because the surgical procedure is <strong>in</strong>vasive,there may also be a proactive duty of care for theattend<strong>in</strong>g veter<strong>in</strong>arian to monitor and report on thewelfare outcomes for the bitch. These three aspects mayrepresent a significance risk to the RCVS’s pr<strong>in</strong>cipledapproach (ethical position) on the use of AI(S).Some EU countries, for example, Sweden, haveprohibited the use of AI(S). In the light of uncerta<strong>in</strong>tyÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


The Ethics and Role of AI <strong>in</strong> Dogs 171regard<strong>in</strong>g the welfare consequences for the dog, thesecountries appear to have applied a much moreprecautionary approach and have advanced theirresponsibility to the dog (duty of care) over theautonomy of breeders and veter<strong>in</strong>arians. The decisionalso appears to be predicated on the availability ofviable alternatives. Although some cl<strong>in</strong>icians haveargued that this type of ‘ban’ is unworkable, as theavailability of viable alternatives is extremely limited(i.e. there are few surgeons who can perform theAI(TC) procedure successfully), others have arguedthat such a position may enhance veter<strong>in</strong>arianwellbe<strong>in</strong>g and autonomy <strong>in</strong> the long-term by encourag<strong>in</strong>g:(i) <strong>in</strong>novation through research (i.e. developmentof new techniques and enhanced semenpreservation) and (ii) personal <strong>in</strong>vestment <strong>in</strong> <strong>in</strong>sem<strong>in</strong>ationtra<strong>in</strong><strong>in</strong>g.In conclusion, the use of AI raises some importantethical questions which cannot be fully analysed andevaluated here, however, the veter<strong>in</strong>ary and researchcommunity can take several proactive steps to reducethe ethical risks associated with this reproductivetechnology by: (i) clarify<strong>in</strong>g cl<strong>in</strong>ical decision-mak<strong>in</strong>g,(ii) enhanc<strong>in</strong>g <strong>in</strong>formed choice among clients and (iii)<strong>in</strong>creas<strong>in</strong>g the knowledge-base on potential impacts andthe use of all methods of AI.AcknowledgementsThe authors would like to thank Prof. Cathar<strong>in</strong>a L<strong>in</strong>de Forsberg(Swedish University of Agricultural Sciences) and Sandy Tomk<strong>in</strong>s(University of Nott<strong>in</strong>gham) for their provision of valuable literaturethat aided the development of this paper.ReferencesEngland GCW, Verstegen JP, 1996: Radiographic contrastmedium for uter<strong>in</strong>e <strong>in</strong>sem<strong>in</strong>ation <strong>in</strong> the bitch, and its effectupon the quality and fertility of fresh dog semen. Theriogenology46, 1233–1241.Evans EI, 1933: The transport of spermatozoa <strong>in</strong> the dog. AmJ Physiol 105, 287–293.Fontbonne A, Bad<strong>in</strong>and F, 1993: Can<strong>in</strong>e artificial <strong>in</strong>sem<strong>in</strong>ationwith frozen semen: comparison of <strong>in</strong>travag<strong>in</strong>al and<strong>in</strong>trauter<strong>in</strong>e deposition of semen. J Reprod Fertil Suppl 47,325–327.Fougner JA, Aamdal J, Andersen K, 1973: Intrauter<strong>in</strong>e<strong>in</strong>sem<strong>in</strong>ation with frozen semen <strong>in</strong> the Blue Fox. NordicVet Med 25, 144–149.L<strong>in</strong>de-Forsberg C, 2002: Ethical aspects of artificial <strong>in</strong>sem<strong>in</strong>ation(AI). In: Proceed<strong>in</strong>gs of the 3rd EVSSAR Congress,Liege, Belgium, pp. 41–42.L<strong>in</strong>de-Forsberg C, Forsberg M, 1993: Results of 527 controlledartificial <strong>in</strong>sem<strong>in</strong>ations <strong>in</strong> dogs. J Reprod FertilSuppl 47, 313–323.L<strong>in</strong>dsay FEF, 1983: The normal endoscopic appearance of thecaudal reproductive tract of the cyclic and non-cyclic bitch:post-uter<strong>in</strong>e endoscopy. J Small Anim Pract 24, 1–15.Mepham TB, 2000: A framework for the ethical evaluation ofnovel foods: the Ethical Matrix. J Agric Environ Ethics 12,165–176.Mepham TB, Kaiser M, Thorstensen E, Tomk<strong>in</strong>s S, Millar K,2006: Ethical Matrix Manual. Agricultural EconomicsResearch Institute (LEI), The Netherlands.Millar K, Tomk<strong>in</strong>s S, 2007: Ethical analysis of the use of GM<strong>in</strong> aquaculture: emerg<strong>in</strong>g issues for aquaculture development.J Agric Environ Ethics 20, 437–453.Olar TT, 1984: Cryopreservation of dog spermatozoa. PhDThesis, Colorado State University.Royal College of Veter<strong>in</strong>ary Surgeons (RCVS), 2005: AdviceNote 8 – Can<strong>in</strong>e Surgical Artificial Insem<strong>in</strong>ation. RCVS,London.Seager SWJ, Platz C, Fletcher WS, 1975: Conception rates andrelated data us<strong>in</strong>g frozen dog semen. J Reprod Fertil 45,189–192.Smith FO, 1984: Cryopreservation of can<strong>in</strong>e semen: techniqueand performance. PhD Thesis, University of M<strong>in</strong>nesota.Takeishi M, Mikami T, Kodama Y, Tsunekane T, Iwaki T,1976: Studies on reproduction <strong>in</strong> the dog. VII Artificial <strong>in</strong>sem<strong>in</strong>ationus<strong>in</strong>g frozen semen. Jpn J Anim Reprod 22, 28–33.Thomassen R, Sanson G, Krogenaes A, Fougner J, Berg K,Farstad W, 2006: Artificial <strong>in</strong>sem<strong>in</strong>ation with frozen semen<strong>in</strong> dogs: a retrospective study of 10 years us<strong>in</strong>g non-surgicalapproach. Theriogenology 66, 1645–1650.Wildt DE, 1986: Diagnostic procedures, laparoscopy. In:Burke TJ (ed.), Small Animal <strong>Reproduction</strong> and Infertility,A Cl<strong>in</strong>ical Approach to Diagnosis and Treatment. Lea &Febiger, Philadelphia, pp. 121–140.Wilson MS, 1993: Non-surgical <strong>in</strong>trauter<strong>in</strong>e artificial <strong>in</strong>sem<strong>in</strong>ation<strong>in</strong> bitches us<strong>in</strong>g frozen semen. J Reprod Fertil Suppl47, 307–311.Wilson MS, 2001: Transcervical <strong>in</strong>sem<strong>in</strong>ation techniques <strong>in</strong> thebitch. Vet Cl<strong>in</strong> North America 31, 291–303.Author’s address (for correspondence): Gary CW England, Dean,School of Veter<strong>in</strong>ary Medic<strong>in</strong>e and Science, The University ofNott<strong>in</strong>gham, Sutton Bon<strong>in</strong>gton Campus, Loughborough, LE125RD, UK. E-mail: Gary.England@nott<strong>in</strong>gham.ac.ukConflict of <strong>in</strong>terest: The authors declare no conflict of <strong>in</strong>terests.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 172–178 (2008); doi: 10.1111/j.1439-0531.2008.01158.xISSN 0936-6768Basic Aspects of the Control of GnRH and LH Secretions by Kisspept<strong>in</strong>: PotentialApplications for Better Control of Fertility <strong>in</strong> FemalesA Caraty and I Francesch<strong>in</strong>iPhysiologie de la reproduction et des comportements, INRA, Nouzilly, FranceContentsThe neuronal control of fertility and sterility has been a subjectof research for years. However, nowadays, <strong>in</strong> spite ofconsiderable literature about GnRH dur<strong>in</strong>g the last fewdecades, the precise cellular and molecular mechanismswhereby gonadal steroids and other peripheral signals converge<strong>in</strong> the bra<strong>in</strong> to achieve the f<strong>in</strong>e regulation of GnRHsecretion rema<strong>in</strong>s partially unknown. In this scenario, a majorbreakthrough <strong>in</strong> our understand<strong>in</strong>g of the neuronal signalsgovern<strong>in</strong>g reproduction took place <strong>in</strong> 2003 with the discoveryof metast<strong>in</strong> ⁄ kisspept<strong>in</strong> as a major player <strong>in</strong> the control ofGnRH secretion. This molecule, first described as hav<strong>in</strong>g acrucial role <strong>in</strong> trigger<strong>in</strong>g the onset of puberty, is <strong>in</strong>volved <strong>in</strong> allphases of reproductive life and hence has attracted the <strong>in</strong>terestof many reproductive neuroendocr<strong>in</strong>ologists. Adm<strong>in</strong>isteredeither centrally or peripherally, kisspept<strong>in</strong> strongly <strong>in</strong>duces thesecretion of gonadotrop<strong>in</strong> <strong>in</strong> many species, ma<strong>in</strong>ly throughstimulation of GnRH secretion. Kisspept<strong>in</strong> cells <strong>in</strong>volved <strong>in</strong>the control of GnRH secretion are located <strong>in</strong> two regions ofthe bra<strong>in</strong>: the preoptic area and the arcuate nucleus. Carry<strong>in</strong>goestradiol receptor alpha, kisspept<strong>in</strong> cells of these regionsappear to be the ma<strong>in</strong> <strong>in</strong>tegration centres for the expression ofboth the positive and negative feedback of steroid on GnRHsecretion. More recently, this molecule has been shown to beable to synchronize preovulatory surges <strong>in</strong> cyclic ewes andcause ovulation <strong>in</strong> seasonally acyclic ewes. This reviewsummarizes the most relevant aspects of the role of kisspept<strong>in</strong><strong>in</strong> GnRH ⁄ LH release and the potential application of thismolecule <strong>in</strong> new strategies for controll<strong>in</strong>g female fertility.IntroductionSuccessful reproduction requires the completion of acomplex cascade of events, which are the substrate of a‘f<strong>in</strong>e dialog’ between the bra<strong>in</strong>, the pituitary and thegonads. Early studies on the regulation of the hypothalamo-pituitary-gonadotropicaxis have emphasizedthe pivotal role of pulsatile GnRH release fromhypothalamic neurons for the secretion of the gonadotrop<strong>in</strong>sLH and FSH by the pituitary, and then theproduction of sexual hormones and gametogenesis bythe gonads (see review by Knobil 2005). Follow<strong>in</strong>g<strong>in</strong>tense research, our knowledge of GnRH regulationhas greatly improved <strong>in</strong> recent years, but the neuroendocr<strong>in</strong>emechanisms lead<strong>in</strong>g to the f<strong>in</strong>e modulationof GnRH pulsatility rema<strong>in</strong> to be def<strong>in</strong>ed. Forexample, <strong>in</strong> most if not all mammals, it is clear thatoestradiol <strong>in</strong>duces a strong <strong>in</strong>crease <strong>in</strong> GnRH releasedur<strong>in</strong>g the follicular phase of the cycle, lead<strong>in</strong>g to apreovulatory GnRH surge (rat: Sarkar et al. 1976; ewe:Clarke et al. 1989; Moenter et al. 1991; monkey: Xiaet al. 1992), but the pathways and the hierarchy of themechanisms <strong>in</strong>volved <strong>in</strong> this regulation are onlypartially understood.In 2003, this field of research was given new impetusby a conceptual breakthrough, with the discovery of anew central player, the Kiss-1 ⁄ GPR54 system, operat<strong>in</strong>gupstream of the GnRH structure. The unsuspected l<strong>in</strong>kbetween Kiss-1 ⁄ GPR54 and reproductive physiologywas identified <strong>in</strong> two <strong>in</strong>dependent studies, report<strong>in</strong>g thatisolated hypogonadotropic hypogonadism <strong>in</strong> humans isassociated with a genetic defect of the GPR54 gene, andthat the loss-of-function by a mutation of the GPR54gene <strong>in</strong> mice leads to a deficiency <strong>in</strong> sexual maturationand hence <strong>in</strong>fertility (de Roux et al. 2003; Sem<strong>in</strong>araet al. 2003). This discovery of a l<strong>in</strong>k between GPR54(a G-prote<strong>in</strong>-coupled receptor) and sexual maturationled to immediate <strong>in</strong>terest <strong>in</strong> its ligand. This molecule,first identified as a metastasis suppressor molecule, wasnamed metast<strong>in</strong> or kisspept<strong>in</strong> (Kotani et al. 2001; Muiret al. 2001; Ohtaki et al. 2001). Kisspept<strong>in</strong> is the productof the Kiss-1 gene, which encodes a 145-am<strong>in</strong>o acidpeptide that is further processed to generate biologicallyactive peptides of various lengths (10–54 am<strong>in</strong>o acids)termed kisspept<strong>in</strong>s. These appear to be well conserved <strong>in</strong>different species, with only m<strong>in</strong>or structural changes forthe last 10 am<strong>in</strong>o acids of the C-term<strong>in</strong>al end, whichbear nearly all the biological activity of the molecule<strong>in</strong> vivo (Gottsch et al. 2004) and <strong>in</strong> vitro (Ors<strong>in</strong>i et al.2007). Kisspept<strong>in</strong>s b<strong>in</strong>d to a receptor previouslydescribed as a galan<strong>in</strong>-related orphan receptor, GPR54(Lee et al. 1999), and display similar high-aff<strong>in</strong>ityb<strong>in</strong>d<strong>in</strong>g <strong>in</strong> heterelogous cell systems (Kotani et al.2001). There have been considerable advances <strong>in</strong> kisspept<strong>in</strong>research, and this peptide family is now recognizedas a major player <strong>in</strong> the control of thehypothalamic-pituitary-gonadal axis.Kisspept<strong>in</strong>s Stimulate GnRH⁄LH ReleaseThe very potent stimulatory effect of kisspept<strong>in</strong> ongonadotrop<strong>in</strong> secretion is unequivocal. First, kisspept<strong>in</strong>was shown to elicit the release of gonadotrop<strong>in</strong> when<strong>in</strong>jected <strong>in</strong>to the cerebral ventricle of the mouse bra<strong>in</strong>(Gottsch et al. 2004). Us<strong>in</strong>g either <strong>in</strong>tracerebroventricularor peripheral <strong>in</strong>jections (<strong>in</strong>travenous, subcutaneous…) of kisspept<strong>in</strong>, this f<strong>in</strong>d<strong>in</strong>g was widely confirmed <strong>in</strong>many species: the rat (Matsui et al. 2004; Navarro et al.2005), sheep (Messager et al. 2005), pig (Lents and Barb2007), bov<strong>in</strong>e (Kadokawa et al. 2008a), monkeys (Shahabet al. 2005) and humans (Dhillo et al. 2005). Thestimulatory effect of kisspept<strong>in</strong> on gonadotrop<strong>in</strong> secretionis mostly mediated by a stimulatory effect onGnRH release, as evidenced <strong>in</strong> the follow<strong>in</strong>g studies: (1)local adm<strong>in</strong>istration of kisspept<strong>in</strong> near the GnRH cellÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Control of Fertility <strong>in</strong> Females by Kisspept<strong>in</strong> 173Fig. 1. Effect of central kisspept<strong>in</strong> adm<strong>in</strong>istration (50 nmol given as acont<strong>in</strong>uous adm<strong>in</strong>istration <strong>in</strong>to the lateral ventricle over 4 h) onGnRH release <strong>in</strong>to the cerebrosp<strong>in</strong>al fluid of the third ventricle (filledsquare) and LH release <strong>in</strong> the peripheral blood (open lozenge) of anovariectomized oestradiol-treated ewe dur<strong>in</strong>g the anoestrous season.Redrawn from Messager et al. (2005)bodies <strong>in</strong> the medial preoptic area stimulated LH release<strong>in</strong> the rat (Patterson et al. 2006); (2) <strong>in</strong>tracerebroventricularadm<strong>in</strong>istration of kisspept<strong>in</strong> <strong>in</strong> sheep <strong>in</strong>duced alarge and susta<strong>in</strong>ed release of GnRH <strong>in</strong>to the cerebrosp<strong>in</strong>alfluid (Messager et al. 2005; Fig. 1); and (3) thestimulatory effect on gonadotrop<strong>in</strong> secretion has beenfound to be totally blocked <strong>in</strong> the presence of a GnRHantagonist (Gottsch et al. 2004; Irwig et al. 2004;Matsui et al. 2004). This hypothesis is further supportedby evidence that a large proportion of GnRH neuronsexpress GPR54 (Irwig et al. 2004; Han et al. 2005), thatkisspept<strong>in</strong>-immunoreactive fibres have been observed <strong>in</strong>close apposition to GnRH cell bodies (K<strong>in</strong>oshita et al.2005; Clarkson and Herbison 2006), and that adm<strong>in</strong>istrationof kisspept<strong>in</strong> <strong>in</strong>creases excitability of GnRHneurons (Han et al. 2005).While the action of kisspept<strong>in</strong> <strong>in</strong> regulat<strong>in</strong>g GnRHrelease at the level of the hypothalamus is unequivocal,the possible role of this peptide at the pituitary levelrema<strong>in</strong>s a matter of controversy. Kisspept<strong>in</strong> has beenshown to stimulate LH and FSH release from ratpituitary explants (Navarro et al. 2005), LH releasefrom bov<strong>in</strong>e and porc<strong>in</strong>e pituitary cells (Suzuki et al.2007), and dose-related LH and growth hormonesecretory responses from dispersed pituitary cells (rat:Gutierrez-Pascual et al. 2007; bov<strong>in</strong>e: Kadokawa et al.2008b). In contrast, other studies have found no effect ofkisspept<strong>in</strong> on pituitary response (Matsui et al. 2004;Thompson et al. 2004) or <strong>in</strong> alter<strong>in</strong>g GnRH-stimulatedLH secretion <strong>in</strong> hypothalamo-pituitary disconnectedewes (Smith et al. 2007a). On a morphological basis,GPR54 is expressed <strong>in</strong> the human pituitary (Kotaniet al. 2001; Muir et al. 2001), and kisspept<strong>in</strong>-immunoreactivefibres are located <strong>in</strong> the external zone of themedian em<strong>in</strong>ence <strong>in</strong> sheep (Francesch<strong>in</strong>i et al. 2006;Pompolo et al. 2006). Moreover, as kisspept<strong>in</strong> isreleased <strong>in</strong>to the hypophyseal portal blood (Smith et al.2007a), it is more than likely that kisspept<strong>in</strong> has a role atthe pituitary level, even if this is not presently known. Ina recent study, peripheral adm<strong>in</strong>istration of kisspept<strong>in</strong>was clearly demonstrated to lead to a differentialresponse <strong>in</strong> terms of LH and FSH release (Caraty et al.2007). After rapid <strong>in</strong>itial <strong>in</strong>crease <strong>in</strong> both LH and FSHrelease <strong>in</strong> response to the kisspept<strong>in</strong>-<strong>in</strong>duced GnRHsecretion, plasma FSH levels were found to rema<strong>in</strong> highFig. 2. Effect of peripheral adm<strong>in</strong>istration of kisspept<strong>in</strong> (100 nmolgiven iv) on LH (filled lozenge) and FSH release (open square) <strong>in</strong> theperipheral blood of an ovariectomized oestradiol-treated ewe dur<strong>in</strong>gthe anoestrous season. LH and FSH concentration <strong>in</strong>crease with<strong>in</strong>10 m<strong>in</strong> and then beg<strong>in</strong> to decrease accord<strong>in</strong>g to the half-life of eachhormone. After 2 h, FSH reaches a plateau and stays above the pre<strong>in</strong>jectionlevel until the end of the sampl<strong>in</strong>g period. Redrawn fromCaraty et al. (2007)for several hours (Fig. 2), suggest<strong>in</strong>g a secondary effectof the peptide at the gonadotrope level enhanc<strong>in</strong>gconstitutive FSH release. Further studies are clearlyneeded to clarify whether and how the pituitarycontributes to the full gonadotrop<strong>in</strong> response to kisspept<strong>in</strong>adm<strong>in</strong>istration.Kisspept<strong>in</strong> Neurons <strong>in</strong> the Bra<strong>in</strong>, Distributionand Regulation per Sex SteroidsIn the mouse, Kiss-1 mRNA is found <strong>in</strong> the anteroventralperiventricular nucleus (AVPV), the periventricularnucleus, the anterodorsal preoptic nucleus, themedial amygdala and the arcuate nucleus (ARC)(Gottsch et al. 2004). In the ewe, the distribution ofthe Kiss-1 mRNA cells is similar, with a densepopulation <strong>in</strong> the ARC and an additional populationof positive cells <strong>in</strong> the POA (Smith et al. 2007b). Inl<strong>in</strong>e with these mRNA data, immunocytochemistryus<strong>in</strong>g an antiserum raised aga<strong>in</strong>st the am<strong>in</strong>o acidresidues 43–52 of mouse kisspept<strong>in</strong> (Francesch<strong>in</strong>i et al.2006) identified kisspept<strong>in</strong>-immunoreactive cells <strong>in</strong> theAVPV and ARC <strong>in</strong> the mouse (Clarkson and Herbison2006) and <strong>in</strong> the POA and ARC <strong>in</strong> the ewe(Francesch<strong>in</strong>i et al. 2006; Fig. 3). In the mouse andsheep, a few immunoreactive cells for kisspept<strong>in</strong> havealso been localized <strong>in</strong> the dorsomedial hypothalamicnucleus, an area devoid of Kiss-1 mRNA (Smith et al.2007b). This may <strong>in</strong>dicate a slight cross reactivity ofthe antisera with other members of the relatedC-term<strong>in</strong>al RF amide family.Kiss-1 mRNA expression appears to be stronglyregulated by sex steroids. In the rat, mouse and ewe,steroid removal by ovariectomy greatly <strong>in</strong>creasesmRNA expression <strong>in</strong> the ARC, an effect reversed byoestradiol replacement (Smith et al. 2005, 2006, 2007b;Maeda et al. 2007). Accord<strong>in</strong>gly, Kiss-1 mRNA expressionis found to be lower at proestrus and oestrus thanat dioestrus 1 (Navarro et al. 2004). This picture differsclearly from what happens at the POA level. A decreaseof Kiss-1 mRNA expression <strong>in</strong> the AVPV occurs afterovariectomy <strong>in</strong> the female rat and mouse, an effectwhich is reversed by oestradiol adm<strong>in</strong>istration (SmithÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


174 A Caraty and I Francesch<strong>in</strong>iFig. 3. Two ma<strong>in</strong> populations of kisspept<strong>in</strong> cells are found <strong>in</strong> thehypothalamus of the ewe: one is located <strong>in</strong>to the preoptic area (POA)and the other one is located <strong>in</strong> the arcuate nucleus (ARC). Cells <strong>in</strong> thePOA are distributed close to the wall of the third ventricle (V). Thedensity of the cells is higher <strong>in</strong> the caudal part of the ARC, where theyare surrounded by a dense network of varicose fibres. Redrawn fromFrancesch<strong>in</strong>i et al. (2006)et al. 2005; Maeda et al. 2007). In contrast, no similarchanges are observed <strong>in</strong> the POA of the ewe (Smith et al.2007b).With regard to the regulation of peptide expression,few data are available to date. In sheep, ovariectomygreatly <strong>in</strong>creases the number of kisspept<strong>in</strong> cells of theARC (Pompolo et al. 2006), which is <strong>in</strong> l<strong>in</strong>e with <strong>in</strong> situhybridization studies. Similarly <strong>in</strong> rat, the changes <strong>in</strong>number of kisspept<strong>in</strong> cells observed <strong>in</strong> the AVPV andthe ARC by <strong>in</strong> situ hybridization between ovariectomizedand ovariectomized oestradiol-treated females aremirrored by similar changes <strong>in</strong> the number of cellsobserved by immunocytochemistry (Maeda et al. 2007).Yet, the changes <strong>in</strong> prote<strong>in</strong> are not always parallel withthe changes <strong>in</strong> mRNA expression of kisspept<strong>in</strong>. Forexample, Kiss-1 mRNA level is lower at proestrus andoestrus than at dioestrus 1 while the number ofkisspept<strong>in</strong> neurons as well as the number of kisspept<strong>in</strong>neurons express<strong>in</strong>g Fos are higher at proestrus than atdioestrus (K<strong>in</strong>oshita et al. 2005).As sex steroids have a profound impact on Kiss-1mRNA expression, it is not surpris<strong>in</strong>g that most, if notall, kisspept<strong>in</strong> cells have been found to express oestradiolreceptor alpha (ERa) <strong>in</strong> the mouse and rat (Smithet al. 2005, 2006). In the ewe, the majority of kisspept<strong>in</strong>cells of the ARC express ERa (Francesch<strong>in</strong>i et al. 2006)as well as the progesterone receptor (Smith et al. 2007b).In contrast to rodents, only 50% of the kisspept<strong>in</strong> cellsexpress ERa <strong>in</strong> the POA, suggest<strong>in</strong>g that functionallydist<strong>in</strong>ct subpopulations might exist <strong>in</strong> this species(Fig. 3), and this could be related to the positivefeedback effect of oestradiol on GnRH secretion (anissue addressed <strong>in</strong> the follow<strong>in</strong>g sections).Kisspept<strong>in</strong> and the Regulation of FemaleCyclicityThere is compell<strong>in</strong>g evidence that the AVPV kisspept<strong>in</strong>cell population <strong>in</strong> the rat and mouse is critical forgenerat<strong>in</strong>g the preovulatory GnRH surge. The role ofAVPV <strong>in</strong> the <strong>in</strong>duction of a GnRH ⁄ LH surge <strong>in</strong> rodentsis unequivocal (see review of Herbison 2007). Thekisspept<strong>in</strong> cell population of this nucleus is highlysexually dimorphic. Both kisspept<strong>in</strong> prote<strong>in</strong>s and Kiss-1mRNA levels are higher <strong>in</strong> the female than the male(Clarkson and Herbison 2006; Gottsch et al. 2006;Kauffman et al. 2007). If female rats are neonatallytreated with testosterone dur<strong>in</strong>g the critical period ofbra<strong>in</strong> differentiation, the number of Kiss-1 mRNA <strong>in</strong>this nucleus is low, similar to that of male rats(Kauffman et al. 2007). In contrast, no similar sexuallydimorphic expression is observed for the kisspept<strong>in</strong>population of the ARC. Lesions of the AVPV block theLH surge, as does immunoneutralization of kisspept<strong>in</strong><strong>in</strong> the POA by a specific monoclonal antibody (K<strong>in</strong>oshitaet al. 2005). More importantly, expression of Kiss-1mRNA is up-regulated concomitantly with the preovulatoryGnRH ⁄ LH surge (Smith et al. 2006), and amarked <strong>in</strong>crease <strong>in</strong> c-Fos expression occurs at that time<strong>in</strong> these cells, prov<strong>in</strong>g transcriptional activation (Smithet al. 2006). The simplest and most plausible conclusionthat can be drawn from these data is that kisspept<strong>in</strong>neurons of the AVPV, bear<strong>in</strong>g ERa and hav<strong>in</strong>g directcontact with GnRH cell bodies <strong>in</strong> the POA, are a majortarget for oestradiol to provoke preovulatoryGnRH ⁄ LH release. Unfortunately, the picture is notso simple. First, it is important to bear <strong>in</strong> m<strong>in</strong>d thatthere is a circadian component <strong>in</strong> the <strong>in</strong>duction of theGnRH ⁄ LH surge <strong>in</strong> rodents and that circadian signalsfrom the suprachiasmatic nucleus form additionalregulatory elements of the kisspept<strong>in</strong> cells (Gu andSimerly 1997). Secondly, <strong>in</strong> ovariectomized oestradioltreatedfemale rats, Kiss-1 mRNA <strong>in</strong> AVPV is high,while LH is strongly suppressed (Smith et al. 2005).F<strong>in</strong>ally, it has recently been shown that oestradiol<strong>in</strong>duces Fos expression <strong>in</strong> GnRH neurons and producesa GnRH-dependent LH surge <strong>in</strong> GPR54 KO mice(Dungan et al. 2007). This latter result may reflect acompensatory mechanism <strong>in</strong> GPR54 KO mice. Nevertheless,this f<strong>in</strong>d<strong>in</strong>g questions the role of the kisspept<strong>in</strong>cells of the AVPV <strong>in</strong> the oestradiol-<strong>in</strong>duced GnRHsurge mechanism.The picture is different for ewes, because oestradiolacts to <strong>in</strong>duce a preovulatory GnRH ⁄ LH surge <strong>in</strong> themedio-basal hypothalamus and not <strong>in</strong> the POA (Blacheet al. 1991; Caraty et al. 1998). As mentioned above,immunocytochemistry has demonstrated that ovariectomyup-regulates the level of kisspept<strong>in</strong> <strong>in</strong> the ARC ofthe ewe (Pompolo et al. 2006). More recently, upregulationof Kiss-1 mRNA has been found to occur <strong>in</strong>the caudal region of the ARC dur<strong>in</strong>g the preovulatoryperiod (Estrada et al. 2006), when peripheral oestradiolconcentration is high. Overall, these data suggest thatdifferent kisspept<strong>in</strong> cell populations with<strong>in</strong> the ARC are<strong>in</strong>volved <strong>in</strong> negative and positive feedback regulation ofGnRH. Yet, we have recently found that Fos <strong>in</strong> <strong>in</strong>tactcyclic ewes is activated <strong>in</strong> kisspept<strong>in</strong> cells of the POAjust prior to the preovulatory GnRH ⁄ LH surge, andnot at the ARC level (Hoffman, personal communication).This transcriptional activation of the kisspept<strong>in</strong>cells of the POA may be secondary to previousactivation of cells <strong>in</strong> the ARC. Clearly, more experimentsare needed to determ<strong>in</strong>e by which mechanism(s),oestradiol act<strong>in</strong>g <strong>in</strong> the medio-basal hypothalamusactivates GnRH cells to provoke the preovulatoryGnRH release <strong>in</strong> ewe.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Control of Fertility <strong>in</strong> Females by Kisspept<strong>in</strong> 175Potential Applications of Kisspept<strong>in</strong> <strong>in</strong> theControl of Fertility <strong>in</strong> FemalesThe unequivocal demonstration of a strong stimulatoryeffect of kisspept<strong>in</strong> on GnRH and gonadotrop<strong>in</strong> secretionhas already generated research to determ<strong>in</strong>e howthis type of molecule could provide new strategies forcontroll<strong>in</strong>g the reproductive axis.An earlier study has shown that a s<strong>in</strong>gle subcutaneous<strong>in</strong>jection of kisspept<strong>in</strong> <strong>in</strong> prepubertal female rats is ableto <strong>in</strong>duce ovulation (Matsui et al. 2004). However, itmust be po<strong>in</strong>ted out that these animals had been primedwith pregnant mare’s serum gonadotrop<strong>in</strong>, which stimulatesgonadal function and causes ovulation <strong>in</strong> sheepand goats (Ritar et al. 1984). A recent detailed analysisof the pattern of LH and FSH release <strong>in</strong> response tosystemic kisspept<strong>in</strong> adm<strong>in</strong>istration <strong>in</strong> ewes has def<strong>in</strong>ed aprotocol caus<strong>in</strong>g gonadotrop<strong>in</strong> secretion similar to thatobserved dur<strong>in</strong>g a preovulatory gonadotrop<strong>in</strong> surge <strong>in</strong>larger animals (Caraty et al. 2007). Acute <strong>in</strong>travenous orsubcutaneous adm<strong>in</strong>istration of kisspept<strong>in</strong> was observedto <strong>in</strong>duce only a short-lived stimulation of LH release,with little relationship between the duration of thestimulation and the dose given. It must be recalled thaton a molecular weight basis, the potency to release LHfor kisspept<strong>in</strong> is at least 10 times lower than for GnRHboth <strong>in</strong> vitro and <strong>in</strong> vivo (Thompson et al. 2004). Incontrast to acute kisspept<strong>in</strong> adm<strong>in</strong>istration, a low-dose<strong>in</strong>fusion of the peptide was shown to provide prolongedgonadotrop<strong>in</strong> release. This protocol was then tested <strong>in</strong>ewes dur<strong>in</strong>g the breed<strong>in</strong>g season, with the aim of<strong>in</strong>duc<strong>in</strong>g well-timed ovulation. Ewes show<strong>in</strong>g regularoestrous cycles were synchronized by the adm<strong>in</strong>istrationof prostagland<strong>in</strong> followed by <strong>in</strong>sertion of an <strong>in</strong>travag<strong>in</strong>alprogesterone implant (CIDR) for 14 days, and aperfusion of kisspept<strong>in</strong> (or of the vehicle as control) wasgiven 30 h after removal of the progesterone CIDR (afew hours before the time when the earliest spontaneousLH surge is usually observed). As illustrated <strong>in</strong> Fig. 4,an LH surge was found to occur with<strong>in</strong> 2 h of the startof <strong>in</strong>fusion <strong>in</strong> all kisspept<strong>in</strong>-treated animals, whereas theLH surges were observed later and more widelyFig. 4. Left panel: dur<strong>in</strong>g the breed<strong>in</strong>g season, constant iv <strong>in</strong>fusion ofkisspept<strong>in</strong> (3.84 lmol for 8 h), start<strong>in</strong>g 30 h after progesterone CIDRremoval <strong>in</strong> cyclic ewes, <strong>in</strong>duces with<strong>in</strong> 2 h an LH surge <strong>in</strong> all animals(9 ⁄ 9; closed circle). In animals receiv<strong>in</strong>g the vehicle, the LH surgeswere latter and more widely dispersed (open circle), hence only five outof n<strong>in</strong>e animals show an LH surge dur<strong>in</strong>g the period of bloodsampl<strong>in</strong>g. Right panel: progesterone plasma concentration <strong>in</strong>creasesfor all animals over the next 5 days follow<strong>in</strong>g the <strong>in</strong>fusion, but the riseis significantly earlier for the kisspept<strong>in</strong>-treated ewes. *p < 0.05,**p < 0.01. Redrawn from Caraty et al. (2007)dispersed <strong>in</strong> the vehicle-treated animals (from 42 h tomore than 65 h follow<strong>in</strong>g progesterone removal). Theseperfectly timed preovulatory LH surges represent apharmacological <strong>in</strong>tervention send<strong>in</strong>g a kisspept<strong>in</strong><strong>in</strong>ducedGnRH surge signal to an oestrogen-primedpituitary. This result is <strong>in</strong> l<strong>in</strong>e with the observation thatkisspept<strong>in</strong> stimulates gonadotrop<strong>in</strong> release most potentlydur<strong>in</strong>g the preovulatory phase of the menstrualcycle <strong>in</strong> women (Dhillo et al. 2007). This explosive LHrelease <strong>in</strong>dicates that there is a w<strong>in</strong>dow of time dur<strong>in</strong>gthe late follicular phase when kisspept<strong>in</strong> synchronizesovulation perfectly and could therefore be used toenhance the efficacy of artificial <strong>in</strong>sem<strong>in</strong>ation or <strong>in</strong> vitrofecundation programmes.The kisspept<strong>in</strong> <strong>in</strong>fusion protocol was also used tore<strong>in</strong>itiate cyclicity <strong>in</strong> animals with a quiescent gonadalaxis. A low-dose <strong>in</strong>fusion of the peptide was given over48 h to anoestrus ewes. This treatment was carried out<strong>in</strong> two breeds of sheep and was shown to stimulategonadotrop<strong>in</strong> secretion reliably and to <strong>in</strong>duce LHsurges followed by successful ovulation <strong>in</strong> more than80% of the acyclic females. As illustrated <strong>in</strong> Fig. 5, an<strong>in</strong>itial LH response was seen with<strong>in</strong> 1 h of start<strong>in</strong>g the<strong>in</strong>fusion, but the elevated LH response was not ma<strong>in</strong>ta<strong>in</strong>edwith cont<strong>in</strong>uous <strong>in</strong>fusion. This gonadotrop<strong>in</strong><strong>in</strong>crease was, however, sufficient to ‘activate’ the systemas evidenced by a rise <strong>in</strong> peripheral oestradiol concentrations,and ovulatory surges were found to occurapproximately 20 h after the start of treatment, which isconsistent with positive oestrogen feedback. Accord<strong>in</strong>gly,it can be concluded that constant low-doseadm<strong>in</strong>istration of kisspept<strong>in</strong> to acyclic females is sufficientto activate the hypothalamo-pituitary axis andcause ovulation. This opens a very promis<strong>in</strong>g avenuewith regard to problems of fertility <strong>in</strong> large farmanimals such as the difficulty of obta<strong>in</strong><strong>in</strong>g well-timedovulation <strong>in</strong> equ<strong>in</strong>e species or chronic <strong>in</strong>fertility <strong>in</strong>postpartum dairy cows.ConclusionAdvances <strong>in</strong> kisspept<strong>in</strong> research have been rapid, and ithas become evident that kisspept<strong>in</strong> is a major player <strong>in</strong>the control of the hypothalamo-pituitary-gonadal axis<strong>in</strong> numerous species. Kisspept<strong>in</strong> is the most powerfulknown secretagogue of GnRH, and kisspept<strong>in</strong> cells <strong>in</strong>the hypothalamus are more than likely to be key players<strong>in</strong> the steroid feedback control of GnRH secretion.Kisspept<strong>in</strong> appears to be <strong>in</strong>volved <strong>in</strong> most if not alltransitional steps of reproductive life, such as the onsetof puberty, <strong>in</strong>itiation of the breed<strong>in</strong>g season and thedynamic changes of gonadotrop<strong>in</strong> secretion throughoutthe oestrous cycle. Us<strong>in</strong>g kisspept<strong>in</strong> to manipulate thegonadotrop<strong>in</strong> axis therefore appears to offer a verypromis<strong>in</strong>g avenue. The demonstration that cont<strong>in</strong>uous<strong>in</strong>fusion of kisspept<strong>in</strong> can synchronize LH surges <strong>in</strong>progesterone-primed cyclic ewes and re<strong>in</strong>itiate cyclicity<strong>in</strong> anoestrous animals suggests that other means ofcont<strong>in</strong>uous delivery of kisspept<strong>in</strong> will be useful forsynchroniz<strong>in</strong>g ovulation. This will assist the design ofnew protocols for optimiz<strong>in</strong>g artificial <strong>in</strong>sem<strong>in</strong>ationand improv<strong>in</strong>g fertility <strong>in</strong> mammals. More than30 years after the discovery of GnRH, the possibilityÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


176 A Caraty and I Francesch<strong>in</strong>iFig. 5. LH secretion and ensu<strong>in</strong>govulation <strong>in</strong> ewes treated dur<strong>in</strong>gthe non-breed<strong>in</strong>g season with an<strong>in</strong>travenous <strong>in</strong>fusion of kisspept<strong>in</strong>(20 lg ⁄ h for 48 h). The upperpanels show two examples of plasmaLH levels <strong>in</strong> kisspept<strong>in</strong>-<strong>in</strong>fusedewes, <strong>in</strong>dicat<strong>in</strong>g that the treatmentled to <strong>in</strong>creased secretion but thepreovulatory surge did not occuruntil 25 h later. This strongly suggeststhat the <strong>in</strong>fusion <strong>in</strong>itiated anartificial follicular phase and thatthe <strong>in</strong>creased oestrogen result<strong>in</strong>gfrom this activated a positivefeedback response. The experimentwas repeated <strong>in</strong> France and <strong>in</strong>Australia with similar results.Approximately 80% of the ewestreated with kisspept<strong>in</strong> ovulated, asevidenced by result<strong>in</strong>g corporalutea on the ovaries. Adapted fromCaraty et al. (2007)of us<strong>in</strong>g kisspept<strong>in</strong> for the control of the gonadotrop<strong>in</strong>axis provides hope for resolv<strong>in</strong>g all fertility problems.Who knows if this is the end of the story, or perhaps anew molecule activat<strong>in</strong>g kisspept<strong>in</strong> might be discovered<strong>in</strong> another 30 years’ time!ReferencesBlache D, Fabre-Nys CJ, Venier G, 1991: Ventromedialhypothalamus as a target for oestradiol action on proceptivity,receptivity and lute<strong>in</strong>iz<strong>in</strong>g hormone surge of the ewe.Bra<strong>in</strong> Res 546, 241–249.Caraty A, Fabre-Nys C, Delaleu B, Locatelli A, Bruneau G,Karsch FJ, Herbison A, 1998: Evidence that the mediobasalhypothalamus is the primary site of action of estradiol <strong>in</strong><strong>in</strong>duc<strong>in</strong>g the preovulatory gonadotrop<strong>in</strong> releas<strong>in</strong>g hormonesurge <strong>in</strong> the ewe. Endocr<strong>in</strong>ology 139, 1752–1760.Caraty A, Smith JT, Lomet D, Ben Said S, Morrissey A,Cognie J, Doughton B, Baril G, Briant C, Clarke IJ, 2007:Kisspept<strong>in</strong> synchronizes preovulatory surges <strong>in</strong> cyclical ewesand causes ovulation <strong>in</strong> seasonally acyclic ewes. Endocr<strong>in</strong>ology148, 5258–5267.Clarke IJ, Cumm<strong>in</strong>s JT, Jenk<strong>in</strong> M, Phillips DJ, 1989: Theoestrogen-<strong>in</strong>duced surge of LH requires a ‘signal’ pattern ofgonadotroph<strong>in</strong>-releas<strong>in</strong>g hormone <strong>in</strong>put to the pituitarygland <strong>in</strong> the ewe. J Endocr<strong>in</strong>ol 122, 127–134.Clarkson J, Herbison AE, 2006: Postnatal development ofkisspept<strong>in</strong> neurons <strong>in</strong> mouse hypothalamus; sexual dimorphismand projections to gonadotrop<strong>in</strong>-releas<strong>in</strong>g hormoneneurons. Endocr<strong>in</strong>ology 147, 5817–5825.Dhillo WS, Chaudhri OB, Patterson M, Thompson EL,Murphy KG, Badman MK, McGowan BM, Amber V,Patel S, Ghatei MA, Bloom SR, 2005: Kisspept<strong>in</strong>-54stimulates the hypothalamic-pituitary gonadal axis <strong>in</strong>human males. J Cl<strong>in</strong> Endocr<strong>in</strong>ol Metab 90, 6609–6615.Dhillo WS, Chaudhri OB, Thompson EL, Murphy KG,Patterson M, Ramachandran R, Nijher GK, Amber V,Kokk<strong>in</strong>os A, Donaldson M, Ghatei MA, Bloom SR, 2007:Kisspept<strong>in</strong>-54 stimulates gonadotrop<strong>in</strong> release mostpotently dur<strong>in</strong>g the preovulatory phase of the menstrualcycle <strong>in</strong> women. J Cl<strong>in</strong> Endocr<strong>in</strong>ol Metab 92, 3958–3966.Dungan HM, Gottsch ML, Zeng H, Gragerov A, BergmannJE, Vassilatis DK, Clifton DK, Ste<strong>in</strong>er RA, 2007: The roleof kisspept<strong>in</strong>-GPR54 signal<strong>in</strong>g <strong>in</strong> the tonic regulation andsurge release of gonadotrop<strong>in</strong>-releas<strong>in</strong>g hormone ⁄ lute<strong>in</strong>iz<strong>in</strong>ghormone. J Neurosci 27, 12088–12095.Estrada KM, Clay CM, Pompolo S, Smith JT, Clarke IJ, 2006:Elevated Kiss-1 expression <strong>in</strong> the arcuate nucleus prior tothe cyclic preovulatory gonadotrop<strong>in</strong>-releas<strong>in</strong>g hormone ⁄lute<strong>in</strong>is<strong>in</strong>g hormone surge <strong>in</strong> the ewe suggests a stimulatoryrole for kisspept<strong>in</strong> <strong>in</strong> oestrogen-positive feedback. J Neuroendocr<strong>in</strong>ol18, 139–149.Francesch<strong>in</strong>i I, Lomet D, Cateau M, Delsol G, Tillet Y,Caraty A, 2006: Kisspept<strong>in</strong> immunoreactive cells of theov<strong>in</strong>e preoptic area and arcuate nucleus co-express estrogenreceptor alpha. Neurosci Lett 401, 225–230.Gottsch ML, Cunn<strong>in</strong>gham MJ, Smith JT, Popa SM, AcohidoBV, Crowley WF, Sem<strong>in</strong>ara S, Clifton DK, Ste<strong>in</strong>er RA,2004: A role for kisspept<strong>in</strong>s <strong>in</strong> the regulation of gonadotrop<strong>in</strong>secretion <strong>in</strong> the mouse. Endocr<strong>in</strong>ology 145, 4073–4077.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Control of Fertility <strong>in</strong> Females by Kisspept<strong>in</strong> 177Gottsch ML, Clifton DK, Ste<strong>in</strong>er RA, 2006: Kisspepept<strong>in</strong>-GPR54 signal<strong>in</strong>g <strong>in</strong> the neuroendocr<strong>in</strong>e reproductive axis.Mol Cell Endocr<strong>in</strong>ol 254 255, 91–96.Gu GB, Simerly RB, 1997: Projections of the sexuallydimorphic anteroventral periventricular nucleus <strong>in</strong> thefemale rat. J Comp Neurol 384, 142–164.Gutierrez-Pascual E, Mart<strong>in</strong>ez-Fuentes AJ, P<strong>in</strong>illa L, Tena-Sempere M, Malagon MM, Castano JP, 2007: Directpituitary effects of kisspept<strong>in</strong>: activation of gonadotrophsand somatotrophs and stimulation of lute<strong>in</strong>is<strong>in</strong>g hormoneand growth hormone secretion. J Neuroendocr<strong>in</strong>ol 19, 521–530.Han SK, Gottsch ML, Lee KJ, Popa SM, Smith JT, JakawichSK, Clifton DK, Ste<strong>in</strong>er RA, Herbison AE, 2005: Activationof gonadotrop<strong>in</strong>-releas<strong>in</strong>g hormone neurons by kisspept<strong>in</strong>as a neuroendocr<strong>in</strong>e switch for the onset of puberty. JNeurosci 25, 11349–11356.Herbison AE, 2007: Estrogen positive feedback to gonadotrop<strong>in</strong>-releas<strong>in</strong>ghormone (GnRH) neurons <strong>in</strong> the rodent: Thecase for the rostral periventricular area of the third ventricle(RP3V). Bra<strong>in</strong> Res Rev 57, 277–287.Irwig MS, Fraley GS, Smith JT, Acohido BV, Popa SM,Cunn<strong>in</strong>gham MJ, Gottsch ML, Clifton DK, Ste<strong>in</strong>er RA,2004: Kisspept<strong>in</strong> activation of gonadotrop<strong>in</strong> releas<strong>in</strong>g hormoneneurons and regulation of KiSS-1 mRNA <strong>in</strong> the malerat. Neuroendocr<strong>in</strong>ology 80, 264–272.Kadokawa H, Matsui M, Hayashi K, Matsunaga N, KawashimaC, Shimizu T, Kida K, Miyamoto A, 2008a:Peripheral adm<strong>in</strong>istration of kisspept<strong>in</strong>-10 <strong>in</strong>creases plasmaconcentrations of GH as well as LH <strong>in</strong> prepubertal Holste<strong>in</strong>heifers. J Endocr<strong>in</strong>ol 196, 331–334.Kadokawa H, Suzuki S, Hashizume T, 2008b: Kisspept<strong>in</strong>-10stimulates the secretion of growth hormone and prolact<strong>in</strong>directly from cultured bov<strong>in</strong>e anterior pituitary cells. AnimReprod Sci 105, 404–408.Kauffman AS, Gottsch ML, Roa J, Byquist AC, Crown A,Clifton DK, Hoffman GE, Ste<strong>in</strong>er RA, Tena-Sempere M,2007: Sexual differentiation of Kiss1 gene expression <strong>in</strong> thebra<strong>in</strong> of the rat. Endocr<strong>in</strong>ology 148, 1774–1783.K<strong>in</strong>oshita M, Tsukamura H, Adachi S, Matsui H, UenoyamaY, Iwata K, Yamada S, Inoue K, Ohtaki T, Matsumoto H,Maeda K, 2005: Involvement of central metast<strong>in</strong> <strong>in</strong> theregulation of preovulatory lute<strong>in</strong>iz<strong>in</strong>g hormone surge andestrous cyclicity <strong>in</strong> female rats. Endocr<strong>in</strong>ology 146, 4431–4436.Knobil E, 2005: Discovery of the hypothalamic gonadotrop<strong>in</strong>releas<strong>in</strong>ghormone pulse generator and of its physiologicsignificance. 1992. Am J Obstet Gynecol 193, 1765–1766.Kotani M, Detheux M, Vandenbogaerde A, Communi D,Vanderw<strong>in</strong>den JM, Le Poul E, Brezillon S, Tyldesley R,Suarez-Huerta N, Vandeput F, Blanpa<strong>in</strong> C, Schiffmann SN,Vassart G, Parmentier M, 2001: The metastasis suppressorgene KiSS-1 encodes kisspept<strong>in</strong>s, the natural ligands of theorphan G prote<strong>in</strong>-coupled receptor GPR54. J Biol Chem276, 34631–34636.Lee DK, Nguyen T, O’Neill GP, Cheng R, Liu Y, HowardAD, Coulombe N, Tan CP, Tang-Nguyen AT, George SR,O’Dowd BF, 1999: Discovery of a receptor related to thegalan<strong>in</strong> receptors. FEBS Lett 446, 103–107.Lents C, Barb C, 2007: Stimulatory actions of kisspept<strong>in</strong> ongonadotrop<strong>in</strong> secretion <strong>in</strong> prepubertal sw<strong>in</strong>e. Biol ReprodSpecial Issue 105 (Abstract No. 125).Maeda K, Adachi S, Inoue K, Ohkura S, Tsukamura H, 2007:Metast<strong>in</strong> ⁄ kisspept<strong>in</strong> and the control of estrous cycle <strong>in</strong> rats.Rev Endocr Metab Disord 8, 21–29.Matsui H, Takatsu Y, Kumano S, Matsumoto H, Ohtaki T,2004: Peripheral adm<strong>in</strong>istration of metast<strong>in</strong> <strong>in</strong>duces markedgonadotrop<strong>in</strong> release and ovulation <strong>in</strong> the rat. BiochemBiophys Res Commun 320, 383–388.Messager S, Chatzidaki EE, Ma D, Hendrick AG, Zahn D,Dixon J, Thresher RR, Mal<strong>in</strong>ge I, Lomet D, Carlton MB,Colledge WH, Caraty A, Aparicio SA, 2005: Kisspept<strong>in</strong>directly stimulates gonadotrop<strong>in</strong>-releas<strong>in</strong>g hormone releasevia G prote<strong>in</strong>-coupled receptor 54. Proc Natl Acad Sci USA102, 1761–1766.Moenter SM, Caraty A, Locatelli A, Karsch FJ, 1991: Patternof gonadotrop<strong>in</strong>-releas<strong>in</strong>g hormone (GnRH) secretion lead<strong>in</strong>gup to ovulation <strong>in</strong> the ewe: existence of a preovulatoryGnRH surge. Endocr<strong>in</strong>ology 129, 1175–1182.Muir AI, Chamberla<strong>in</strong> L, Elshourbagy NA, Michalovich D,Moore DJ, Calamari A, Szekeres PG, Sarau HM, ChambersJK, Murdock P, Steplewski K, Shabon U, Miller JE,Middleton SE, Darker JG, Larm<strong>in</strong>ie CG, Wilson S, BergsmaDJ, Emson P, Faull R, Philpott KL, Harrison DC, 2001:AXOR12, a novel human G prote<strong>in</strong>-coupled receptor,activated by the peptide KiSS-1. J Biol Chem 276, 28969–28975.Navarro VM, Castellano JM, Fernandez-Fernandez R,Barreiro ML, Roa J, Sanchez-Criado JE, Aguilar E,Dieguez C, P<strong>in</strong>illa L, Tena-Sempere M, 2004: Developmentaland hormonally regulated messenger ribonucleicacid expression of KiSS-1 and its putative receptor,GPR54, <strong>in</strong> rat hypothalamus and potent lute<strong>in</strong>iz<strong>in</strong>g hormone-releas<strong>in</strong>gactivity of KiSS-1 peptide. Endocr<strong>in</strong>ology145, 4565–4574.Navarro VM, Castellano JM, Fernandez-Fernandez R, TovarS, Roa J, Mayen A, Barreiro ML, Casanueva FF, Aguilar E,Dieguez C, P<strong>in</strong>illa L, Tena-Sempere M, 2005: Effects ofKiSS-1 peptide, the natural ligand of GPR54, on folliclestimulat<strong>in</strong>ghormone secretion <strong>in</strong> the rat. Endocr<strong>in</strong>ology146, 1689–1697.Ohtaki T, Sh<strong>in</strong>tani Y, Honda S, Matsumoto H, Hori A,Kanehashi K, Terao Y, Kumano S, Takatsu Y, Masuda Y,Ishibashi Y, Watanabe T, Asada M, Yamada T, SuenagaM, Kitada C, Usuki S, Kurokawa T, Onda H, Nishimura O,Fuj<strong>in</strong>o M, 2001: Metastasis suppressor gene KiSS-1 encodespeptide ligand of a G-prote<strong>in</strong>-coupled receptor. Nature 411,613–617.Ors<strong>in</strong>i MJ, Kle<strong>in</strong> MA, Beavers MP, Connolly PJ, MiddletonSA, Mayo KH, 2007: Metast<strong>in</strong> (KiSS-1) mimetics identifiedfrom peptide structure-activity relationship-derived pharmacophoresand directed small molecule database screen<strong>in</strong>g.J Med Chem 50, 462–471.Patterson M, Murphy KG, Thompson EL, Patel S, GhateiMA, Bloom SR, 2006: Adm<strong>in</strong>istration of kisspept<strong>in</strong>-54 <strong>in</strong>todiscrete regions of the hypothalamus potently <strong>in</strong>creasesplasma lute<strong>in</strong>is<strong>in</strong>g hormone and testosterone <strong>in</strong> male adultrats. J Neuroendocr<strong>in</strong>ol 18, 349–354.Pompolo S, Pereira A, Estrada KM, Clarke IJ, 2006: Colocalizationof kisspept<strong>in</strong> and gonadotrop<strong>in</strong>-releas<strong>in</strong>g hormone<strong>in</strong> the ov<strong>in</strong>e bra<strong>in</strong>. Endocr<strong>in</strong>ology 147, 804–810.Ritar AJ, Maxwell WM, Salamon S, 1984: Ovulation and LHsecretion <strong>in</strong> the goat after <strong>in</strong>travag<strong>in</strong>al progestagen sponge-PMSG treatment. J Reprod Fertil 72, 559–563.de Roux N, Gen<strong>in</strong> E, Carel JC, Matsuda F, Chaussa<strong>in</strong> JL,Milgrom E, 2003: Hypogonadotropic hypogonadism due toloss of function of the Kiss-1-derived peptide receptorGPR54. Proc Natl Acad Sci USA 100, 10972–10976.Sarkar DK, Chiappa SA, F<strong>in</strong>k G, Sherwood NM, 1976:Gonadotrop<strong>in</strong>-releas<strong>in</strong>g hormone surge <strong>in</strong> pro-oestrous rats.Nature 264, 461–463.Sem<strong>in</strong>ara SB, Messager S, Chatzidaki EE, Thresher RR,Acierno JS Jr, Shagoury JK, Bo-Abbas Y, Kuohung W,Schw<strong>in</strong>of KM, Hendrick AG, Zahn D, Dixon J, Kaiser UB,Slaugenhaupt SA, Gusella JF, O’Rahilly S, Carlton MB,Crowley WF Jr, Aparicio SA, Colledge WH, 2003: TheGPR54 gene as a regulator of puberty. N Engl J Med 349,1614–1627.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


178 A Caraty and I Francesch<strong>in</strong>iShahab M, Mastronardi C, Sem<strong>in</strong>ara SB, Crowley WF, OjedaSR, Plant TM, 2005: Increased hypothalamic GPR54signal<strong>in</strong>g: a potential mechanism for <strong>in</strong>itiation of puberty<strong>in</strong> primates. Proc Natl Acad Sci USA 102, 2129–2134.Smith JT, Cunn<strong>in</strong>gham MJ, Rissman EF, Clifton DK,Ste<strong>in</strong>er RA, 2005: Regulation of Kiss1 gene expression <strong>in</strong>the bra<strong>in</strong> of the female mouse. Endocr<strong>in</strong>ology 146, 3686–3692.Smith JT, Popa SM, Clifton DK, Hoffman GE, Ste<strong>in</strong>er RA,2006: Kiss1 neurons <strong>in</strong> the forebra<strong>in</strong> as central processorsfor generat<strong>in</strong>g the preovulatory lute<strong>in</strong>iz<strong>in</strong>g hormone surge. JNeurosci 26, 6687–6694.Smith JT, Rao A, Pereira A, Caraty A, Millar RP, Clarke IJ,2007a: Kisspept<strong>in</strong> is present <strong>in</strong> ov<strong>in</strong>e hypophysial portalblood, but does not <strong>in</strong>crease dur<strong>in</strong>g the preovulatorylute<strong>in</strong>iz<strong>in</strong>g hormone surge: Evidence that gonadotropes arenot direct targets of kisspept<strong>in</strong> <strong>in</strong> vivo. Endocr<strong>in</strong>ology 149,1979–1986.Smith JT, Clay CM, Caraty A, Clarke IJ, 2007b: KiSS-1messenger ribonucleic acid expression <strong>in</strong> the hypothalamusof the ewe is regulated by sex steroids and season.Endocr<strong>in</strong>ology 148, 1150–1157.Suzuki S, Kadokawa H, Hashizume T, 2007: Directkisspept<strong>in</strong>-10 stimulation on lute<strong>in</strong>iz<strong>in</strong>g hormone secretionfrom bov<strong>in</strong>e and porc<strong>in</strong>e anterior pituitary cells. AnimReprod Sci 103, 404–408.Thompson EL, Patterson M, Murphy KG, Smith KL, DhilloWS, Todd JF, Ghatei MA, Bloom SR, 2004: Central andperipheral adm<strong>in</strong>istration of kisspept<strong>in</strong>-10 stimulates thehypothalamic-pituitary-gonadal axis. J Neuroendocr<strong>in</strong>ol 16,850–858.Xia L, Van Vugt D, Alston EJ, Luckhaus J, Fer<strong>in</strong> M, 1992: Asurge of gonadotrop<strong>in</strong>-releas<strong>in</strong>g hormone accompanies theestradiol-<strong>in</strong>duced gonadotrop<strong>in</strong> surge <strong>in</strong> the rhesus monkey.Endocr<strong>in</strong>ology 131, 2812–2820.Author’s address (for correspondence): A. Caraty, UMR 6175,Physiologie de la <strong>Reproduction</strong> et des Comportements,INRA ⁄ CNRS ⁄ Université Tours ⁄ Haras Nationaux, 37380 Nouzilly,France. E-mail: caraty@tours.<strong>in</strong>ra.frConflict of <strong>in</strong>terest: This work was supported by an ‘‘Agence Nationalede la Recherche’’ (ANR) Grant. The authors have no furtherdeclarations.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 179–185 (2008); doi: 10.1111/j.1439-0531.2008.01159.xISSN 0936-6768Controll<strong>in</strong>g Animal Populations Us<strong>in</strong>g Anti-Fertility Vacc<strong>in</strong>esR Fayrer-HoskenDepartment of Large Animal Medic<strong>in</strong>e, College of Veter<strong>in</strong>ary Medic<strong>in</strong>e, University of Georgia, Athens, GA, USAContentsThe goal for fertility control of animal populations is thedevelopment of a safe, economical and effective contraceptive.One offshoot of the development of this technology is theacquisition of multiple therapeutic strategies for diseases, suchas immunotherapy probes for cancer. In the long run,successful population control requires multifactorial strategies.One component of population control is immunocontraception.Development of effective antigens for immunocontraceptivevacc<strong>in</strong>es has been remarkable and has greatly advancedour understand<strong>in</strong>g of the molecular mechanisms of fertilization.The chasm between the discovery of an antigen <strong>in</strong> thelaboratory, to the implementation of an effective field program,is immense. The zona pellucida (ZP) immunocontraceptivethat has been most extensively evaluated as a fertilityvacc<strong>in</strong>e antigen and the porc<strong>in</strong>e ZP has received particularattention. The long-term goal of population control would bethe use of a synthetic vacc<strong>in</strong>e, e.g. the ZP, tailored to a targetspecies. In the future, if populations’ levels are to be controlledby fertility vacc<strong>in</strong>es, we should consider that the vacc<strong>in</strong>atedanimals could receive other health protective agents at the sametime. For example, if a species were immunocontracepted, thenthey could be simultaneously vacc<strong>in</strong>ated aga<strong>in</strong>st habitatdiseases such as rabies (Plumb et al., Rev Sci Tech, 26, 2007,229).IntroductionThe ultimate goal of fertility control of a population isan agent that it is non-toxic, completely reversible, hasmulti-year efficacy, is easily adm<strong>in</strong>istered, is devoid ofbehavioural side-affects, cost-effective and is efficacious<strong>in</strong> multiple species and both sexes. This is an almostimpossible goal. The primary impetus for the developmentof a vacc<strong>in</strong>e for fertility regulation is safety, so thatit has no adverse hormonal or toxic side-effects. Thesesame issues are of particular <strong>in</strong>terest to researchers <strong>in</strong>companies concerned with human immunocontraception.We must remember that evolution has assured thatthe survival of a species is protected and centres on themechanisms of fertilization and early embryonic development.For survival of the species, the reproductiveprocesses of fertilization are probably protected bynumerous alternative (redundant) mechanisms. Therefore,it stands to reason that nature will attempt tothwart or circumvent reproductive control strategies.There are multiple antigens that might be targets forfertility control with vacc<strong>in</strong>es <strong>in</strong> animals. These targets<strong>in</strong>clude hormones, oocyte prote<strong>in</strong>s ⁄ glycoprote<strong>in</strong>s, spermprote<strong>in</strong>s and other molecules associated with fertilizationand early embryonic development. In addition, wemust consider that the <strong>in</strong>dividual animal may have aunique and variable immunological response to theseantigens. The presentation of these antigens to theanimals’ immune system will predicate the degree andseverity (Litscher and Wassarman 2007) of the animal’sfertility control. As part of the discussion, the developmentof an antigen from a complete product of purifiednative prote<strong>in</strong>s, to portions of a native prote<strong>in</strong>, to asynthetic prote<strong>in</strong> with a limited or altered compliment ofcarbohydrates, to a synthetic polypeptide ⁄ carbohydrateantigen, to a DNA-vectored or virally vectored vacc<strong>in</strong>eis <strong>in</strong>cluded.Our experiences <strong>in</strong> immunocontraceptives, from laboratoryto field studies, have revealed a number ofsurpris<strong>in</strong>g and unexpected paradoxes. For vacc<strong>in</strong>edevelopment, the use of contraceptive data from amodel species should be very circumspectly extrapolatedto a different target species <strong>in</strong> the field. From laboratorydevelopment to field delivery of a vacc<strong>in</strong>e, there areprofound practical problems. Vacc<strong>in</strong>e adm<strong>in</strong>istration <strong>in</strong>the laboratory environment often has nom<strong>in</strong>al relationshipto the <strong>in</strong>tricacies of field adm<strong>in</strong>istration. Policies forpracticality and techniques of field adm<strong>in</strong>istration mustbe considered for both efficacy and safety. Nevertheless,we must remember that cost-effective adm<strong>in</strong>istrationmight not be the best or safest strategy for adm<strong>in</strong>istrationof an immunocontraceptive vacc<strong>in</strong>e.Zona Pellucida (ZP) Vacc<strong>in</strong>esThe conceptualization of the ZP as an antigen forfertility control was considered at the beg<strong>in</strong>n<strong>in</strong>g of the20th century with reports from Metchnikoff (Shiverset al. 1972). In the early-1960s (Shivers and Metz 1962)to the mid-1970s (Yanagimachi et al. 1976), <strong>in</strong>vestigatorsconsidered the role of ZP-based immunocontraceptives.The most conspicuous advantage is thatimmunocontraceptive vacc<strong>in</strong>es, by their mechanism,appear to have no hormonal effects or deleterious sideeffects(Barber and Fayrer-Hosken 2000a; b). As reproductivecyclicity generally rema<strong>in</strong>s normal <strong>in</strong> the shortterm, the immunocontracepted animals have m<strong>in</strong>imalbehavioural or herd affects, as the <strong>in</strong>ter-oestrous <strong>in</strong>tervalwould be unaffected. Thus, herds of animals whosebehaviour and herd <strong>in</strong>tegrity is based on <strong>in</strong>tact reproductivecyclicity are unaffected by porc<strong>in</strong>e ZP (pZP)immunocontraceptives. Because the ZP glycoprote<strong>in</strong>sare a structurally unique and have very little homologywith other somatic prote<strong>in</strong>s, they provide ideal targetsfor immunocontraception (Barber and Fayrer-Hosken2000a). The ZP prote<strong>in</strong> structures are highly conservedacross several phyla and this facilitates <strong>in</strong>terspecies use(Barber and Fayrer-Hosken 2000b; Litscher and Wassarman2007) of ZP antigens. This conserved homology(Hedrick 1996) of the ZP glycoprote<strong>in</strong>s has allowedthe use of antigens derived from heterologous species(Barber and Fayrer-Hosken 2000b; Fayrer-HoskenÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


180 R Fayrer-Hoskenet al. 2000a,b, 2002; Barber et al. 2001). The use ofheterologous ZP antigens allows for standardization ofa vacc<strong>in</strong>e’s dose and adjuvant as well as its deliverymethod. In addition, the use of heterologous ZPobviates the necessity for a homologous source ofantigens, which for most exotic species would beimpossible. The successful use of heterologous ZPglycoprote<strong>in</strong>s suggests that the mechanism of sperm-ZP <strong>in</strong>teraction might have some commonality betweenspecies. For the same reason, it works as an immunocontraceptivethat affects multiple species, it also questionsthe precise mechanism of the species-specificity ofsperm-ZP <strong>in</strong>teraction. For much of the publishedliterature, the pZP has been the heterologous antigenof choice. To harvest enough native ZP for laboratoryor field studies has been labour-<strong>in</strong>tensive and <strong>in</strong>vestigatorshave considered alternative sources of ZP. Thehorse is one of the best-studied mammals as a model forimmunocontraception. The prototypical studies <strong>in</strong> thehorse were completed by Dr Irw<strong>in</strong> Liu (Liu and Shivers1982; Liu et al. 1989), who demonstrated the efficacyand reversibility of the pZP vacc<strong>in</strong>e. The horse has beenused as both a laboratory model (Liu et al. 1989; Williset al. 1994) and an ecological model (Kirkpatrick andTurner 2003). These immunocontraceptive studies <strong>in</strong> thehorse have shown that the pZP vacc<strong>in</strong>e is both safe andeffective. Long-term vacc<strong>in</strong>ations appear to have effectson the functionality of the ovary (Turner and Kirkpatrick2002), but these effects appear to be reversible withtime.Structure and orig<strong>in</strong> of the ZPThe ZP matrix consists of 3–4 glycoprote<strong>in</strong>s (ZP1, ZP2,ZP3 and ZP4) depend<strong>in</strong>g on the species. At a ratio of1 : 1 (Green 1997), ZP2 and ZP3 are <strong>in</strong>ter-tw<strong>in</strong>ed toform glycoprote<strong>in</strong> filaments that are cross-l<strong>in</strong>ked by ZP1(Wassarman and Mortillo 1991; Jov<strong>in</strong>e et al. 2002). TheZP glycoprote<strong>in</strong>s are comb<strong>in</strong>ed <strong>in</strong>to a microfibrillarmatrix. The ZP matrix is secreted around the oocyte andhas significant elastic properties. In some species, the<strong>in</strong>dividual glycoprote<strong>in</strong>s are produced <strong>in</strong> the ooplasmwith<strong>in</strong> the Golgi apparatus (Wassarman et al. 2005) andsubsequently moved through the cytoplasm and releasedoutside the oolemma <strong>in</strong>to the perivitell<strong>in</strong>e space.For the mare, the target antigens (eqZP) andvacc<strong>in</strong>ational antigens (pZP) have been well characterized.The equ<strong>in</strong>e research shows that there are threeequ<strong>in</strong>e ZP (eqZP) glycoprote<strong>in</strong>s us<strong>in</strong>g silver-sta<strong>in</strong>ed2D-PAGE. The glycoprote<strong>in</strong> families have molecularweights of 93–120 K, 73–90 K and 45–80 K (Milleret al. 1992). Common epitopes on the eqZP (Milleret al. 1992) were detected by rabbit antiserum aga<strong>in</strong>stheat-solubilized pZP (RaHSPZ) (Dunbar and Raynor1980), and gu<strong>in</strong>ea pig antiserum aga<strong>in</strong>st rabbit ZP(R55K) (Lee and Dunbar 1993; Lee et al. 1993).RaHSPZ recognized all three eqZP glycoprote<strong>in</strong> familiesand R55K only recognized the lowest molecularweight eqZP glycoprote<strong>in</strong> family eqZP3 (Miller et al.1992). This molecular data is complimented by thesuccess of mare immunocontraception <strong>in</strong> the field (Liuet al. 1989; Kirkpatrick et al. 1992). In addition, asanti-total-pZP antibodies and anti-R55K recognizeeqZP3, they could serve as a specific immunocontraceptiveantigen. This is important <strong>in</strong> target<strong>in</strong>g the ZP3molecule or portion of the molecule and would be thebest immunological approach for immunocontraceptionus<strong>in</strong>g a synthetic ZP.The formation of the eqZP is a collaborative processbetween the oocyte and cumulus cells (Kolle et al. 2007),and the f<strong>in</strong>al external layer of eqZP is a function of thecumulus cells. The collaborative secretion of ZP glycoprote<strong>in</strong>sis also seen <strong>in</strong> pig, cow, dog, rabbit, marmosetand rhesus monkey (Kolle et al. 2007). In other species,e.g. the cat and mouse, only the oocyte secretes ZP. Thetemporal secretion of ZP glycoprote<strong>in</strong>s is variablebetween species relative to oogenesis. In mouse, ZP2 isdeposited <strong>in</strong> the primordial follicle’s perivitell<strong>in</strong>e space,but ZP3 and ZP4 only appear with the formation of theprimary oocyte and the <strong>in</strong>itiation of its growth (Millaret al. 1993; Epifano et al. 1995). Therefore, there aredifferences <strong>in</strong> the cellular orig<strong>in</strong> of the secretion of ZPglycoprote<strong>in</strong>s, the temporal secretion of the ZP glycoprote<strong>in</strong>sand the manifestation of the antigenic determ<strong>in</strong>antson the ZP glycoprote<strong>in</strong>s.Although the precise molecular mechanism for theimmunocontraceptive effect of pZP rema<strong>in</strong>s to bedeterm<strong>in</strong>ed, the carbohydrate portion of the ZP glycoprote<strong>in</strong>has been implicated as an important aspect(Paterson et al. 1992). If immunocontraception is <strong>in</strong> partmediated by role of term<strong>in</strong>al carbohydrates, theirdistribution and configuration might hold the answer.Possible molecular explanations for the biochemicalmechanism of immunocontraception could <strong>in</strong>cludeblock<strong>in</strong>g sperm b<strong>in</strong>d<strong>in</strong>g sites (Barber and Fayrer-Hosken2000b) or structural changes <strong>in</strong> the ZP. When amare is successfully immunocontracepted, there are highcirculat<strong>in</strong>g levels of both IgG and IgM. The follicularIgG levels are dependent on the peripheral circulat<strong>in</strong>glevels (Husse<strong>in</strong> and Bourne 1984; Widders et al. 1984).IgG is probably the primary antibody to mediateimmunocontraception (Barber and Fayrer-Hosken2000b) as decl<strong>in</strong><strong>in</strong>g serum IgG levels are associated withreturn to fertility of the mare (Liu et al. 1989; Barberand Fayrer-Hosken 2000b). Another possible explanationfor the immunocontraceptive effects of pZP immunization<strong>in</strong> the mare is that dur<strong>in</strong>g the deposition offibrils of eqZP, anti-pZP IgG molecules might be<strong>in</strong>tercalated <strong>in</strong>to the matrix. The comb<strong>in</strong>ation of theIgG and eqZP matrix could cause the anti-pZP antibodiesto act like cross-l<strong>in</strong>kers (e.g. autoimmune reactions)and this could mimic the zona block. Theseimmunocontracepted zona-blocked oocytes would preventfertilization and this effect would rema<strong>in</strong> untilsystemic levels of IgG fall low enough to allow conception.This mechanism would be most relevant <strong>in</strong>animals, where cumulus cells participate <strong>in</strong> secret<strong>in</strong>gZP. This would provide a reasonable explanation as towhy animals, e.g. cats whose ZP is derived completelyfrom the oocyte, are unaffected by heterologous pZPvacc<strong>in</strong>ation (Jewgenow et al. 2000).Another possible mechanism for the immunocontraceptiveeffect of pZP (Barber and Fayrer-Hosken 2000b)is an affect on the sperm-zona b<strong>in</strong>d<strong>in</strong>g through therecognition of carbohydrate epitopes on the glycoprote<strong>in</strong>sof the ZP. It is clear that the ZP prote<strong>in</strong>s areÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Controll<strong>in</strong>g Animal Populations Us<strong>in</strong>g Anti-Fertility Vacc<strong>in</strong>es 181strongly conserved among species (Benoff 1997) and it ispossible that species-specificity of fertilization may bemoderated by differences <strong>in</strong> carbohydrates on the zonasurface. Current data demonstrate that vacc<strong>in</strong>ation withthe three pZP glycoprote<strong>in</strong>s <strong>in</strong>duces immunocontraception<strong>in</strong> multiple species of mammals. To expla<strong>in</strong>species-specificity of the carbohydrates and the species<strong>in</strong>dependenteffect of pZP, the author suggests that thehypothesis still has some merit. B<strong>in</strong>d<strong>in</strong>g of anti-pZP IgGto surface carbohydrate epitopes probably preventssperm b<strong>in</strong>d<strong>in</strong>g by direct and <strong>in</strong>direct <strong>in</strong>terference. Thisis supported by the observation that 63% glycosidicrecognition capability of our anti-pZP antibodies(Barber 2001). The rema<strong>in</strong>der of the polyclonal IgGantibodies recognizes the exposed prote<strong>in</strong> matrix. B<strong>in</strong>d<strong>in</strong>gof the ZP prote<strong>in</strong> matrix by these IgGs probablycompletely blocks the <strong>in</strong>duction of the acrosomereaction. Hence polyclonal IgGs block fertilization bypartial h<strong>in</strong>drance of b<strong>in</strong>d<strong>in</strong>g and possible blockade ofacrosome reaction <strong>in</strong>duction (Mahi-Brown et al. 1985;Henderson et al. 1988).Vacc<strong>in</strong>e preparationThe ZP glycoprote<strong>in</strong>s are secreted to comb<strong>in</strong>e with eachother <strong>in</strong> the perivitell<strong>in</strong>e space <strong>in</strong> a coord<strong>in</strong>ated process.However, <strong>in</strong> vitro, the prote<strong>in</strong> harvest<strong>in</strong>g of pZP forimmunocontraceptive vacc<strong>in</strong>es varies from laboratoryto laboratory. We also know that the purification andstorage of the ZP glycoprote<strong>in</strong>s varies from laboratoryto laboratory. The production and storage differencesmight also alter the site of presentation of the ZPglycoprote<strong>in</strong>s. The purification and storage of theprote<strong>in</strong>s might be extremely critical to antigen bioavailability.How have the glycoprote<strong>in</strong>s recomb<strong>in</strong>ed andwhat antigenic sites have been exposed? More importantly,does this represent the <strong>in</strong> vivo structure? Formouse prote<strong>in</strong>s, we know that the <strong>in</strong>dividual glycoprote<strong>in</strong>srecomb<strong>in</strong>e or repolymerize <strong>in</strong> an homomeric orheteromeric manner (Litscher et al. 2008). Repolymerizationoccurs under non-denatur<strong>in</strong>g conditions evenafter the treatment of the glycoprote<strong>in</strong>s with SDS, heat,urea or lyophilization. In the <strong>in</strong> vivo environment, thepolymerization of ZPs probably occurs <strong>in</strong> a coord<strong>in</strong>atedmolar <strong>in</strong>tegration. However, under <strong>in</strong> vitro conditions,especially with homomeric or heteromeric repolymerization,there could be novel aggregate presentations.The novel presentation could be <strong>in</strong> part an explanationof the effect on multiple species. The same rules ofantigen presentation also apply to synthetic vacc<strong>in</strong>es.For the synthetic or recomb<strong>in</strong>ant vacc<strong>in</strong>es, the antigenicityis also associated with the site of the source DNAsequence, the amount and quality of glycosylation, andthe tertiary assembly.The use of a complete vacc<strong>in</strong>e conta<strong>in</strong><strong>in</strong>g all threepZP glycoprote<strong>in</strong>s clearly works (Liu et al. 1989; Fayrer-Hoskenet al. 2000a,b, 2002; Dels<strong>in</strong>k et al. 2002;Stoops et al. 2006) as an immunocontraceptive agent <strong>in</strong>different species. A more def<strong>in</strong>ed synthetic or recomb<strong>in</strong>antvacc<strong>in</strong>e might be safer and more effective. Thenecessity to standardize a vacc<strong>in</strong>e product and have aready source has lead to the research <strong>in</strong>to synthetic ZPvacc<strong>in</strong>es (Choudhury et al. 2007; Duckworth et al.2007), but to date no synthetic vacc<strong>in</strong>e has been usedextensively.Another consideration for vacc<strong>in</strong>e usage is the role ofadjuvants. When ZP glycoprote<strong>in</strong>s are adjuvented byconjugation, the ZP prote<strong>in</strong>s are conjo<strong>in</strong>ed through thesulfhydryl l<strong>in</strong>kages, and are presented to the immunesystem <strong>in</strong> a specific conformation. This might expla<strong>in</strong>more profound and unique effects of an adjuvant. Wehave used pZP with an adjuvant ImjectÒ (ThermoFisher, IL, USA), a maliamide-activated keyhole limpethaemocyan<strong>in</strong>. The conjugated product produced immunocontraception<strong>in</strong> gilts and showed significant degenerativepathology of the follicles and was probably due topresentation of B-cell epitopes. The other aspect of thevacc<strong>in</strong>ation is the role of the adjuvant. The role of addedadjuvants to the pZP appears to be vitally important. Todate, complete Freund’s (Dels<strong>in</strong>k et al. 2007), <strong>in</strong>completeFreund’s (Hannesdottir et al. 2004; Duckworth et al.2007), modified Freund’s (Lyda et al. 2005) and synthetictrehalose dicorynomycolate (STDCM) (Fayrer-Hoskenet al. 2002) have been used successfully. The adjuvantand pZP comb<strong>in</strong>ation lead to stimulation of the immunemechanism. How much of it is due to the immunostimulantrole of the adjuvant and how much of it is dueto the pZP glycoprote<strong>in</strong> assembly after harvest<strong>in</strong>g?Vacc<strong>in</strong>ation method and route of deliveryFor free-roam<strong>in</strong>g populations, the antigen and adjuvantsgenerally will have to be delivered remotely. Thevacc<strong>in</strong>e might be delivered with darts or <strong>in</strong> baits. Incase of dart delivery, the measure of success ispredicated by field conditions as this might <strong>in</strong>cludestalk<strong>in</strong>g on foot to helicopter delivery and the degree ofsuccess varies. From captive situations, e.g. elephants <strong>in</strong>zoos where there are controlled conditions, vacc<strong>in</strong>ationsgenerally result <strong>in</strong> 100% efficacy (Fayrer-Hoskenet al. 1999). This is when compared with 80–100%vacc<strong>in</strong>ation titre efficacy <strong>in</strong> field when dart<strong>in</strong>g from ahelicopter (Fayrer-Hosken et al. 2000b). The practicalreality is that <strong>in</strong> the field, dart<strong>in</strong>g does not alwaysdeliver an <strong>in</strong>tramuscular <strong>in</strong>jection. Therefore, this is thepractical reality of field vacc<strong>in</strong>ation. The vacc<strong>in</strong>eformulation also plays a role <strong>in</strong> the success of adm<strong>in</strong>istration.The use of vacc<strong>in</strong>e pellets (Turner et al. 2002;Lane et al. 2007) or lactide-glycolide (Kirkpatrick et al.1996; Liu et al. 2005) preparations for longer-act<strong>in</strong>gvacc<strong>in</strong>es makes the adm<strong>in</strong>istration difficult. Oral bait<strong>in</strong>gof a species-specific fertility vacc<strong>in</strong>ation would be veryexcit<strong>in</strong>g as a strategy, as seen with oral vacc<strong>in</strong>ationaga<strong>in</strong>st rabies (Tordo et al. 2006). The possibility oforal vacc<strong>in</strong>es that employ plants (Tacket 2005) that areeng<strong>in</strong>eered to produce a glycosylated ZP product is anexcit<strong>in</strong>g possibility (Polk<strong>in</strong>ghorne et al. 2005). Oralvacc<strong>in</strong>es may provide a viable adm<strong>in</strong>istration route,especially for populations that are difficult to reach.Species-specificity or bait-specificity to deliver theimmunocontraceptive to only the target species presentsthe most worrisome contra<strong>in</strong>dication. While theremight be a possibility that baited or species-specificsites for <strong>in</strong>tra-nasal (Corner et al. 2001; Costant<strong>in</strong>oet al. 2007) delivery of immunocontraceptives, this canonly be evaluated <strong>in</strong> situ. The most important facet ofÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


182 R Fayrer-Hoskenvacc<strong>in</strong>e duration and efficacy is the magnitude andduration of serum antibody levels. For the nativeantigen, to all <strong>in</strong>tents and purposes, its antigenicstructure is similar for preparations from the samelaboratory. This is not the case <strong>in</strong> synthetic preparationswhere the selected DNA sequence and expressionsystem affect the product.Duration of immunocontraceptionMost of the above-mentioned adjuvants provide at least12 months of immunocontraception with one or twoboosters. Some reports <strong>in</strong>dicate longer effects with otherpreparations (Liu et al. 2005) with no booster<strong>in</strong>g or onebooster. The 1-year duration vacc<strong>in</strong>es can be adm<strong>in</strong>isteredfor several years without any side-effect. However,after multiple seasons of adm<strong>in</strong>istration, there areovarian effects as seen with the horses on AssateagueIslands. These cases that have an oestrogen phase ofoestrus, but did not appear to <strong>in</strong>itiate a luteal orprogesterone phase. The <strong>in</strong>itial (>5 years) effect oncyclicity of mares is reversed after several years when novacc<strong>in</strong>es have been re-adm<strong>in</strong>istered. Pregnant animals(Fayrer-Hosken et al. 2000b; Kirkpatrick and Turner2002) can be vacc<strong>in</strong>ated with no deleterious effects onthe foetus and growth of the offspr<strong>in</strong>g. These offspr<strong>in</strong>ghave been shown to grow normally and are fertile asadults.Current vacc<strong>in</strong>esCurrent vacc<strong>in</strong>es use synthetic or recomb<strong>in</strong>ant ZPfragments with or without glycosylation, DNA vacc<strong>in</strong>esor virally vectored vacc<strong>in</strong>es. How does the structure ofthese antigens affect fertility and by what mechanism dothey function? The selection of the specific portions ofthe DNA for the production of ZP glycoprote<strong>in</strong>s is thecurrent optimal technology. The selection of the ZPsequence is very important when an expression systemis selected. Immunocontraceptive antigens can beexpressed <strong>in</strong> bacterial systems (Hardy et al. 2008), yeastsystems (Tang et al. 2003), Ch<strong>in</strong>ese hamster ovary(CHO) systems (Zhao et al. 2004), plant systems (Tacket2005), viral systems (Hardy et al. 2003), <strong>in</strong>sect systems(Hardy et al. 2003; Choudhury et al. 2007) and mammaliancell l<strong>in</strong>es. The recomb<strong>in</strong>ant products of thesesystems have several excit<strong>in</strong>g properties. The antigenpurity and structure are optimized. The purity (Tanget al. 2003) of purified native prote<strong>in</strong>s has been aconcern and the effect of contam<strong>in</strong>at<strong>in</strong>g antigens has notbeen clearly documented. Once the appropriate antigenand expression system has been discovered for a specificspecies, abundant amounts of the antigen could beproduced <strong>in</strong> a cost-effective and repeatable process.These antigens and their production systems will be thefuture zenith of fertility control vacc<strong>in</strong>es. Anotherstrategy is to use a species-specific antigen with aspecies-specific virus to produce a virally vectoredimmunocontraceptive (Redwood et al. 2007). Thevirally vectored immunocontraceptive would solvemany of the practical problems of fertility control ofpopulations. Nevertheless, there is a concern that viralor antigen mutation could lead to the doomsday virus.SummaryThe ZP vacc<strong>in</strong>e is the most tested antigen fromlaboratory to field application. The vacc<strong>in</strong>e appearssafe and effective. The fertility control vacc<strong>in</strong>e does notappear to adversely affect the sociobiology of the targetspecies.Role of hormonal targets for vacc<strong>in</strong>es to control fertilityFertility control can also be achieved by the vacc<strong>in</strong>ationof male or female animals with antigens that arehormones. Several successful studies have been reportedus<strong>in</strong>g gonadotrop<strong>in</strong>-releas<strong>in</strong>g hormone (GnRH) orlute<strong>in</strong>iz<strong>in</strong>g hormone releas<strong>in</strong>g hormone (LHRH),human chorionic gonadotrop<strong>in</strong> (hCG) and folliclestimulat<strong>in</strong>ghormone (FSH).GnRH ⁄ LHRHThe hormonal vacc<strong>in</strong>es can be used as fertility controls,and also as therapeutic agents. Treatment of endocr<strong>in</strong>edisorders and neoplasms is an important facet ofhormonal vacc<strong>in</strong>es. A GnRH vacc<strong>in</strong>e <strong>in</strong> humans as atherapeutic agent is <strong>in</strong>creas<strong>in</strong>gly important. The classicalGnRH or GnRH-I is the ma<strong>in</strong> hormone and alterationsto its secretions will affect ovaries, testis, prostate andplacenta. The GnRH vacc<strong>in</strong>e has been used successfully<strong>in</strong> multiple species. The GnRH vacc<strong>in</strong>e must be conjugatedto a hapten or produced as a recomb<strong>in</strong>ant vacc<strong>in</strong>econta<strong>in</strong><strong>in</strong>g an immunogenic moiety <strong>in</strong> order to amplifythe antigenicity of the decapeptide. Conjugation toKLH, ovalbum<strong>in</strong>, more potent adjuvants or otheramplify<strong>in</strong>g agents is the essential for efficacy. It hasworked for variable periods of time <strong>in</strong> both sexes <strong>in</strong> pigs(Killian et al. 2006), sheep (Earl et al. 2006), horses(Turkstra et al. 2005; Imboden et al. 2006; Elhay et al.2007), deer (Curtis et al. 2002) and bison (Miller et al.2004). The primary problem associated with GnRHvacc<strong>in</strong>es is to produce durable titres.Over the years, several commercial preparations havebeen developed and marketed with greater or lesserfunctional product. Current preparations <strong>in</strong>clude GonaConÔ,ImprovacÔ (Dunshea et al. 2001), Equity andRepro-BlocÔ. The GnRH-analogue, GnRH-d6-Lys,was conjugated to recomb<strong>in</strong>ant Mycobacterium tuberculosishsp70 and adjuvanted with RIBI (STDCM) orIncomplete Freund’s was used <strong>in</strong> pre-pubertal mice.There was a statistically significant effect on the size ofthe urogenital complex and testosterone levels (RIBI).One of the most important animal uses of the GnRH-D6-Lys vacc<strong>in</strong>e is <strong>in</strong> beef production. This product is animportant alternative to hormonal growth promotersand their side-effects. The vacc<strong>in</strong>es have the desiredeffects through multiple treatments on both sexes withm<strong>in</strong>imal side-effects. This is a quality product for thecaptive environment, but for wild populations, themultiple adm<strong>in</strong>istrations pose the same problems aspZP vacc<strong>in</strong>es. Depend<strong>in</strong>g on the presentation andadjuvantation, GnRH vacc<strong>in</strong>es last from 1 to 2 years(Miller et al. 2000). The return to fertility with GnRHvacc<strong>in</strong>es is more rapid than pZP vacc<strong>in</strong>es. In the shortterm with GnRH vacc<strong>in</strong>es, there is a cessation ofÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Controll<strong>in</strong>g Animal Populations Us<strong>in</strong>g Anti-Fertility Vacc<strong>in</strong>es 183oestrous behaviour and cyclicity. This is when comparedwith pZP, where there is cessation of cyclicity after longtermexposure. While cessation of cyclicity might beacceptable <strong>in</strong> some species, it is more controversial ormight be contra-<strong>in</strong>dicated <strong>in</strong> herd-dependant species.Human chorionic gonadotrop<strong>in</strong>Human chorionic gonadotrop<strong>in</strong> is another target forfertility vacc<strong>in</strong>es primarily <strong>in</strong> humans. As the vacc<strong>in</strong>ehas little effect on wild populations, the hCG ismentioned for completeness (Cui et al. 2007).Role of sperm antigens for vacc<strong>in</strong>es to control fertilityThe presence of anti-sperm antigens is one of the mostpervasive causes of human <strong>in</strong>fertility. So, there has beensignificant research on the antigenic causes of <strong>in</strong>fertility.A series of excellent <strong>in</strong>vestigations has provided a seriesof candidate antigens that could be used for fertilitycontrol vacc<strong>in</strong>es. Unfortunately, a limited number oftrials outside the mouse model have been reported forthe practical use of ASA for immunocontraception.Epp<strong>in</strong> (O’Rand et al. 2007) was used <strong>in</strong> monkeys and78% became <strong>in</strong>fertile. The <strong>in</strong>fertility was reversible, butas a practical field agent, the Epp<strong>in</strong> ⁄ Freund’s comb<strong>in</strong>ationwould not work as the monkeys had to bevacc<strong>in</strong>ated every 3 weeks for the duration of the study.While many of the anti-sperm antigens have beendescribed, their use as fertility control vacc<strong>in</strong>es islimited.ConclusionsThe need for fertility control <strong>in</strong> the various ecosystems isobvious. For fertility modulation of populations, thereis no ‘magic agent’ for comprehensive control. For theshear size of the problem, nature will constantly attemptto circumvent control strategies. Scientists have providedmany solutions over the last decade, but there isstill a gulf between dissection of the molecule <strong>in</strong> thelaboratory to delivery of the antigen to the target species<strong>in</strong> the field. Certa<strong>in</strong>ly, the most used and def<strong>in</strong>ed fertilityvacc<strong>in</strong>e is the pZP glycoprote<strong>in</strong>-based immunocontraceptive.The vacc<strong>in</strong>e <strong>in</strong> its native and recomb<strong>in</strong>ant formsis effective, safe and relatively easily delivered. Anti-GnRH and anti-sperm fertility control vacc<strong>in</strong>es alsohold significant promise. In the future, the armoury oftarget antigens, adjuvants and delivery modalities usedwith critical evaluation of ecological and biodiversityissues, will allow safe and economical fertility control ofpopulations. Most importantly, the <strong>in</strong>tegrated use ofcontrol strategies with<strong>in</strong> the realm of a damagedplanetary ecosystem will assist with conservation andpreservation of the beauties of nature.AcknowledgementsThe author would like to dedicate this manuscript to JJ van Altenawho is recover<strong>in</strong>g from <strong>in</strong>juries obta<strong>in</strong>ed while dart<strong>in</strong>g wildlife forimmunocontraception. JJ van Altena has made the sacrifice for thetransition from laboratory science to field application. The authorwould also like to thank Irw<strong>in</strong> Liu and Barry Ball for their assistance<strong>in</strong> review<strong>in</strong>g the manuscript.ReferencesBarber MR, 2001: Comparison of the Mechanism of Action ofthe pZP Contraceptive <strong>in</strong> the Pig, Horse, Elephant and Dog.PhD Thesis, University of Georgia, Athens.Barber MR, Fayrer-Hosken RA, 2000a: Evaluation of somaticand reproductive immunotoxic effects of the porc<strong>in</strong>e zonapellucida vacc<strong>in</strong>ation. J Exp Zool 286, 641–646.Barber MR, Fayrer-Hosken RA, 2000b: Possible mechanismsof mammalian immunocontraception. J Reprod Immunol46, 103–124.Barber MR, Lee SM, Steffens WL, Ard M, Fayrer-HoskenRA, 2001: Immunolocalization of zona pellucida antigens <strong>in</strong>the ovarian follicle of dogs, cats, horses and elephants.Theriogenology 55, 1705–1717.Benoff S, 1997: Carbohydrates and fertilization: an overview.Mol Hum Reprod 3, 599–637.Choudhury S, Srivastava N, Narwal PS, Rath A, Jaiswal S,Gupta SK, 2007: Feasibility and challenges <strong>in</strong> the developmentof immunocontraceptive vacc<strong>in</strong>e based on zonapellucida glycoprote<strong>in</strong>s. Soc Reprod Fertil Suppl 63, 479–493.Corner LA, Buddle BM, Pfeiffer DU, Morris RS, 2001:Aerosol vacc<strong>in</strong>ation of the brushtail possum (Trichosurusvulpecula) with bacille Calmette-Guer<strong>in</strong>: the duration ofprotection. Vet Microbiol 81, 181–191.Costant<strong>in</strong>o HR, Illum L, Brandt G, Johnson PH, Quay SC,2007: Intranasal delivery: physicochemical and therapeuticaspects. Int J Pharm 337, 1–24.Cui C, Stevens VC, Schwendeman SP, 2007: Injectablepolymer microspheres enhance immunogenicity of a contraceptivepeptide vacc<strong>in</strong>e. Vacc<strong>in</strong>e 25, 500–509.Curtis PD, Pooler RL, Richmond ME, Miller LA, MattfeldGF, Quimby FW, 2002: Comparative effects of GnRH andporc<strong>in</strong>e zona pellucida (PZP) immunocontraceptive vacc<strong>in</strong>esfor controll<strong>in</strong>g reproduction <strong>in</strong> white-tailed deer (Odocoileusvirg<strong>in</strong>ianus). Reprod Suppl 60, 131–141.Dels<strong>in</strong>k AK, van Altena JJ, Kirkpatrick J, Grobler D, Fayrer-Hosken RA, 2002: Field applications of immunocontraception<strong>in</strong> African elephants (Loxodonta africana). ReprodSuppl 60, 117–124.Dels<strong>in</strong>k AK, van Altena JJ, Grobler D, Bertsch<strong>in</strong>ger HJ,Kirkpatrick JF, Slotow R, 2007: Implement<strong>in</strong>g immunocontraception<strong>in</strong> free-rang<strong>in</strong>g African elephants at Makalaliconservancy. J. S. Afr. Vet. Assoc. 78, 25–30.Duckworth JA, Wilson K, Cui X, Mol<strong>in</strong>ia FC, Cowan PE,2007: Immunogenicity and contraceptive potential of three<strong>in</strong>fertility-relevant zona pellucida 2 epitopes <strong>in</strong> the marsupialbrushtail possum (Trichosurus vulpecula). <strong>Reproduction</strong>133, 177–186.Dunbar BS, Raynor BD, 1980: Characterization of porc<strong>in</strong>ezona pellucida antigens. Biol Reprod 22, 941–954.Dunshea FR, Colantoni C, Howard K, McCauley I, JacksonP, Long KA, Lopaticki S, Nugent EA, Simons JA, Walker J,Hennessy DP, 2001: Vacc<strong>in</strong>ation of boars with a GnRHvacc<strong>in</strong>e (Improvac) elim<strong>in</strong>ates boar ta<strong>in</strong>t and <strong>in</strong>creasesgrowth performance. J. Anim. Sci. 79, 2524–2535.Earl ER, Waterston MM, Aughey E, Harvey MJ, Matschke C,Colston A, Ferro VA, 2006: Evaluation of two GnRH-Ibased vacc<strong>in</strong>e formulations on the testes function of entireSuffolk cross ram lambs. Vacc<strong>in</strong>e 24, 3172–3183.Elhay M, Newbold A, Britton A, Turley P, Dowsett K, WalkerJ, 2007: Suppression of behavioural and physiologicaloestrus <strong>in</strong> the mare by vacc<strong>in</strong>ation aga<strong>in</strong>st GnRH. AustVet J 85, 39–45.Epifano O, Liang LF, Familari M, Moos MC, Jr, Dean J,1995: Coord<strong>in</strong>ate expression of the three zona pellucidagenes dur<strong>in</strong>g mouse oogenesis. Development 121, 1947–1956.Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


184 R Fayrer-HoskenFayrer-Hosken RA, Bertsch<strong>in</strong>ger HJ, Kirkpatrick JF, GroblerD, Lamberski N, Honneyman G, Ulrich T, 1999: Contraceptivepotential of the porc<strong>in</strong>e zona pellucida vacc<strong>in</strong>e <strong>in</strong> theAfrican elephant (Loxodonta africana). Theriogenology 52,835–846.Fayrer-Hosken RA, Dookwah HD, Brandon CI, 2000a:Immunocontrol <strong>in</strong> dogs. Anim Reprod Sci 60–61, 365–373.Fayrer-Hosken RA, Grobler D, Van Altena JJ, Bertsch<strong>in</strong>gerHJ, Kirkpatrick JF, 2000b: Immunocontraception of Africanelephants. Nature 407, 149.Fayrer-Hosken RA, Barber MR, Crane M, Coll<strong>in</strong>s T, HodgdenR, 2002: Differences <strong>in</strong> immunocontraceptive responses<strong>in</strong> white-tailed deer (Odocoileus virg<strong>in</strong>ianus) and goats(Capra hircus). Reprod Suppl 60, 125–129.Green DP, 1997: Three-dimensional structure of the zonapellucida. Rev Reprod 2, 147–156.Hannesdottir SG, Han X, Lund T, S<strong>in</strong>gh M, Van Der Zee R,Roitt IM, Delves PJ, 2004: Changes <strong>in</strong> the reproductivesystem of male mice immunized with a GnRH-analogue conjugatedto mycobacterial hsp70. <strong>Reproduction</strong> 128, 365–371.Hardy CM, ten Have JF, Pek<strong>in</strong> J, Beaton S, Jackson RJ,Clydesdale G, 2003: Contraceptive responses of miceimmunized with purified recomb<strong>in</strong>ant mouse zona pellucidasubunit 3 (mZP3) prote<strong>in</strong>s. <strong>Reproduction</strong> 126, 49–59.Hardy CM, Beaton S, H<strong>in</strong>ds LA, 2008: Immunocontraception<strong>in</strong> mice us<strong>in</strong>g repeated, multi-antigen peptides: immunizationwith purified recomb<strong>in</strong>ant antigens. Mol Reprod Dev75, 126–135.Henderson CJ, Hulme MJ, Aitken RJ, 1988: Contraceptivepotential of antibodies to the zona pellucida. J Reprod Fertil83, 325–343.Hedrick JL, 1996: Comparative structural and antigenicproperties of zona pellucida glycoprote<strong>in</strong>s. J. Reprod. Fertil.Suppl. 50, 9–17.Husse<strong>in</strong> AM, Bourne FJ, 1984: Immunoglobul<strong>in</strong> concentrations<strong>in</strong> pig follicular fluid. Int J Fertil 29, 54–57.Imboden I, Janett F, Burger D, Crowe MA, Hassig M, ThunR, 2006: Influence of immunization aga<strong>in</strong>st GnRH onreproductive cyclicity and estrous behavior <strong>in</strong> the mare.Theriogenology 66, 1866–1875.Jewgenow K, Rohleder M, Wegner I, 2000: Differencesbetween antigenic determ<strong>in</strong>ants of pig and cat zonapellucida prote<strong>in</strong>s. J. Reprod. Fertil. 119, 15–23.Jov<strong>in</strong>e L, Qi H, Williams Z, Litscher E, Wassarman PM, 2002:The ZP doma<strong>in</strong> is a conserved module for polymerization ofextracellular prote<strong>in</strong>s. Nat Cell Biol 4, 457–461.Killian G, Miller L, Rhyan J, Doten H, 2006: Immunocontraceptionof Florida feral sw<strong>in</strong>e with a s<strong>in</strong>gle-dose GnRHvacc<strong>in</strong>e. Am J Reprod Immunol 55, 378–384.Kirkpatrick JF, Turner A, 2002: Reversibility of action andsafety dur<strong>in</strong>g pregnancy of immunization aga<strong>in</strong>st porc<strong>in</strong>ezona pellucida <strong>in</strong> wild mares (Equus caballus). Reprod Suppl60, 197–202.Kirkpatrick JF, Turner A, 2003: Absence of effects fromimmunocontraception on seasonal birth patterns and foalsurvival among barrier island wild horses. J Appl Anim WelfSci 6, 301–308.Kirkpatrick JF, Liu IM, Turner JW, Jr, Naugle R, Keiper R,1992: Long-term effects of porc<strong>in</strong>e zonae pellucidae immunocontraceptionon ovarian function <strong>in</strong> feral horses (Equuscaballus). J Reprod Fertil 94, 437–444.Kirkpatrick JF, Turner JW, Jr, Liu IK, Fayrer-Hosken R,1996: Applications of pig zona pellucida immunocontraceptionto wildlife fertility control. J Reprod Fertil Suppl 50,183–189.Kolle S, Dubois CS, Caillaud M, Lahuec C, S<strong>in</strong>owatz F,Goudet G, 2007: Equ<strong>in</strong>e zona prote<strong>in</strong> synthesis and ZPstructure dur<strong>in</strong>g folliculogenesis, oocyte maturation, andembryogenesis. Mol Reprod Dev 74, 851–859.Lane VM, Liu IK, Casey K, van Leeuwen EM, Flanagan DR,Murata K, Munro C, 2007: Inoculation of female Americanblack bears (Ursus americanus) with partially purifiedporc<strong>in</strong>e zona pellucidae limits cub production. ReprodFertil Dev 19, 617–625.Lee VH, Dunbar BS, 1993: Developmental expression of therabbit 55-kDa zona pellucida prote<strong>in</strong> and messenger RNA<strong>in</strong> ovarian follicles. Dev Biol 155, 371–382.Lee VH, Schwoebel E, Prasad S, Cheung P, Timmons TM,Cook R, Dunbar BS, 1993: Identification and structuralcharacterization of the 75-kDa rabbit zona pellucidaprote<strong>in</strong>. J Biol Chem 268, 12412–12417.Litscher ES, Wassarman PM, 2007: Egg extracellular coatprote<strong>in</strong>s: from fish to mammals. Histol Histopathol 22, 337–347.Litscher ES, Janssen WG, Darie CC, Wassarman PM, 2008:Purified mouse egg zona pellucida glycoprote<strong>in</strong>s polymerize<strong>in</strong>to homomeric fibrils under non-denatur<strong>in</strong>g conditions. JCell Physiol 214, 153–157.Liu IK, Shivers CA, 1982: Antibodies to the zona pellucida <strong>in</strong>mares. J Reprod Fertil Suppl 32, 309–313.Liu IK, Bernoco M, Feldman M, 1989: Contraception <strong>in</strong>mares heteroimmunized with pig zonae pellucidae. J ReprodFertil 85, 19–29.Liu IK, Turner JW, Jr, van Leeuwen EM, Flanagan DR,Hedrick JL, Murata K, Lane VM, Morales-Levy MP, 2005:Persistence of anti-zonae pellucidae antibodies follow<strong>in</strong>g as<strong>in</strong>gle <strong>in</strong>oculation of porc<strong>in</strong>e zonae pellucidae <strong>in</strong> thedomestic equ<strong>in</strong>e. <strong>Reproduction</strong> 129, 181–190.Lyda RO, Hall JR, Kirkpatrick JF, 2005: A comparison ofFreund’s Complete and Freund’s Modified Adjuvants usedwith a contraceptive vacc<strong>in</strong>e <strong>in</strong> wild horses (Equus caballus).J Zoo Wildl Med 36, 610–616.Mahi-Brown CA, Yanagimachi R, Hoffman JC, Huang TT,Jr, 1985: Fertility control <strong>in</strong> the bitch by active immunizationwith porc<strong>in</strong>e zonae pellucidae: use of different adjuvantsand patterns of estradiol and progesterone levels <strong>in</strong> estrouscycles. Biol Reprod 32, 761–772.Millar SE, Lader ES, Dean J, 1993: ZAP-1 DNA b<strong>in</strong>d<strong>in</strong>gactivity is first detected at the onset of zona pellucida geneexpression <strong>in</strong> embryonic mouse oocytes. Dev Biol 158, 410–413.Miller CC, Fayrer-Hosken RA, Timmons TM, Lee VH,Caudle AB, Dunbar BS, 1992: Characterization of equ<strong>in</strong>ezona pellucida glycoprote<strong>in</strong>s by polyacrylamide gel electrophoresisand immunological techniques. J Reprod Fertil 96,815–825.Miller LA, Johns BE, Killian GJ, 2000: Immunocontraceptionof white-tailed deer with GnRH vacc<strong>in</strong>e. Am J ReprodImmunol 44, 266–274.Miller LA, Rhyan JC, Drew M, 2004: Contraception ofbison by GnRH vacc<strong>in</strong>e: a possible means of decreas<strong>in</strong>gtransmission of brucellosis <strong>in</strong> bison. J Wildl Dis 40, 725–730.O’Rand MG, Widgren EE, Wang Z, Richardson RT, 2007:Epp<strong>in</strong>: an epididymal protease <strong>in</strong>hibitor and a target formale contraception. Soc Reprod Fertil Suppl 63, 445–453.Paterson M, Koothan PT, Morris KD, O’Byrne KT, BraudeP, Williams A, Aitken RJ, 1992: Analysis of the contraceptivepotential of antibodies aga<strong>in</strong>st native and deglycosylatedporc<strong>in</strong>e ZP3 <strong>in</strong> vivo and <strong>in</strong> vitro. Biol Reprod 46,523–534.Plumb G, Babiuk L, Mazet J, Olsen S, Rupprecht C, PastoretPP, Slate D, 2007: Vacc<strong>in</strong>ation <strong>in</strong> conservation medic<strong>in</strong>e.Rev Sci Tech 26, 229–241.Polk<strong>in</strong>ghorne I, Hamerli D, Cowan P, Duckworth J, 2005:Plant-based immunocontraceptive control of wildlife –‘‘potentials, limitations, and possums’’. Vacc<strong>in</strong>e 23, 1847–1850.Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Controll<strong>in</strong>g Animal Populations Us<strong>in</strong>g Anti-Fertility Vacc<strong>in</strong>es 185Redwood AJ, Harvey NL, Lloyd M, Lawson MA, Hardy CM,Shellam GR, 2007: Viral vectored immunocontraception:screen<strong>in</strong>g of multiple fertility antigens us<strong>in</strong>g mur<strong>in</strong>e cytomegalovirusas a vacc<strong>in</strong>e vector. Vacc<strong>in</strong>e 25, 698–708.Shivers CA, Metz CB, 1962: Inhibition of fertilization <strong>in</strong> frogeggs by univalent fragments of rabbit antibody. Proc. Soc.Exp. Biol. Med. 110, 385–387.Shivers CA, Dudkiewicz AB, Frankl<strong>in</strong> LE, Fussell EN, 1972:Inhibition of sperm-egg <strong>in</strong>teraction by specific antibody.Science 178, 1211–1213.Stoops MA, Liu IK, Shideler SE, Lasley BL, Fayrer-HoskenRA, Benirschke K, Murata K, van Leeuwen EM, AndersonGB, 2006: Effect of porc<strong>in</strong>e zonae pellucidae immunisationon ovarian follicular development and endocr<strong>in</strong>efunction <strong>in</strong> domestic ewes (Ovis aries). Reprod Fertil Dev18, 667–676.Tacket CO, 2005: Plant-derived vacc<strong>in</strong>es aga<strong>in</strong>st diarrhealdiseases. Vacc<strong>in</strong>e 23, 1866–1869.Tang J, Xie QX, Pan SP, Xiao LJ, Dong L, Zhang CX, SunCJ, 2003: [Expression of recomb<strong>in</strong>ant human zonapellucida-3 prote<strong>in</strong> (rhZP3) <strong>in</strong> Pichia pastoris]. Sheng WuGong Cheng Xue Bao 19, 758–762.Tordo N, Bahloul C, Jacob Y, Jallet C, Perr<strong>in</strong> P, Badrane H,2006: Rabies: epidemiological tendencies and control tools.Dev Biol (Basel) 125, 3–13.Turkstra JA, van der Meer FJ, Knaap J, Rottier PJ, TeerdsKJ, Colenbrander B, Meloen RH, 2005: Effects of GnRHimmunization <strong>in</strong> sexually mature pony stallions. AnimReprod Sci 86, 247–259.Turner A, Kirkpatrick JF, 2002: Effects of immunocontraceptionon population, longevity and body condition <strong>in</strong> wildmares (Equus caballus). Reprod Suppl 60, 187–195.Turner JW, Jr, Liu IK, Flanagan DR, Bynum KS, RutbergAT, 2002: Porc<strong>in</strong>e zona pellucida (PZP) immunocontraceptionof wild horses (Equus caballus) <strong>in</strong> Nevada: a 10 yearstudy. Reprod Suppl 60, 177–186.Wassarman PM, Mortillo S, 1991: Structure of the mouse eggextracellular coat, the zona pellucida. Int Rev Cytol 130,85–110.Wassarman PM, Jov<strong>in</strong>e L, Qi H, Williams Z, Darie C, LitscherES, 2005: Recent aspects of mammalian fertilizationresearch. Mol Cell Endocr<strong>in</strong>ol 234, 95–103.Widders PR, Stokes CR, David JS, Bourne FJ, 1984:Quantitation of the immunoglobul<strong>in</strong>s <strong>in</strong> reproductive tractsecretions of the mare. Res Vet Sci 37, 324–330.Willis L, Heusner G, Warren R, Kessler D, Fayrer-Hosken R,1994: Equ<strong>in</strong>e immunocontraception us<strong>in</strong>g porc<strong>in</strong>e zonapellucida: a new method for the remote delivery andcharacterization of the immune response. J Eq Vet Sci 14,364–370.Yanagimachi R, W<strong>in</strong>kelhake J, Nicolson GL, 1976:Immunological block to mammalian fertilization: survivaland organ distribution of immunoglobul<strong>in</strong> which <strong>in</strong>hibitsfertilization <strong>in</strong> vivo. Proc. Natl. Acad. Sci. USA 73, 2405–2408.Zhao M, Boja ES, Hoodbhoy T, Nawrocki J, Kaufman JB,Kresge N, Ghirlando R, Shiloach J, Pannell L, Lev<strong>in</strong>e RL,Fales HM, Dean J, 2004: Mass spectrometry analysisof recomb<strong>in</strong>ant human ZP3 expressed <strong>in</strong> glycosylationdeficientCHO cells. Biochemistry 43, 12090–12104.Author’s address (for correspondence): Richard Fayrer-Hosken,Department of Large Animal Medic<strong>in</strong>e, College of Veter<strong>in</strong>ary Medic<strong>in</strong>e,University of Georgia, Athens, GA 30602-7385, USA. E-mail:rfh@vet.uga.eduConflict of <strong>in</strong>terest: The author has not declared any conflict of<strong>in</strong>terests.Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 186–192 (2008); doi: 10.1111/j.1439-0531.2008.01160.xISSN 0936-6768The Expand<strong>in</strong>g Role of Recomb<strong>in</strong>ant Gonadotrop<strong>in</strong>s <strong>in</strong> Assisted <strong>Reproduction</strong>TE Adams 1 and I Boime 21 Department of Animal Science, University of California, Davis, CA, USA; 2 Department of Molecular Biology and Pharmacology, Wash<strong>in</strong>gtonUniversity School of Medic<strong>in</strong>e, St Louis, MO, USAContentsUs<strong>in</strong>g recomb<strong>in</strong>ant gonadotrop<strong>in</strong>s for assisted reproduction ofdomestic species is still <strong>in</strong> its <strong>in</strong>fancy. Yet, the purity, potencyand pathogen-free nature of recomb<strong>in</strong>ant gonadotrop<strong>in</strong>s makethem attractive alternatives to tissue-derived gonadotropicagents. In this study, the authors summarize the work to dateus<strong>in</strong>g recomb<strong>in</strong>ant gonadotrop<strong>in</strong>s to enhance the - fertility ofdomestic animals and they discussed their recent studiesexam<strong>in</strong><strong>in</strong>g the biopotency of s<strong>in</strong>gle cha<strong>in</strong> analogues of humangonadotrop<strong>in</strong>s. In these studies, s<strong>in</strong>gle cha<strong>in</strong> analogues offollicle stimulat<strong>in</strong>g hormone (Fca), chorionic gonadotrop<strong>in</strong>(CGba) or a gonadotrop<strong>in</strong> construct with dual activity(FcCGba) were adm<strong>in</strong>istered to sheep pre-treated with antiseradirected aga<strong>in</strong>st GnRH. Ovulation was <strong>in</strong>duced 3 days afteranalogue adm<strong>in</strong>istration us<strong>in</strong>g hCG (1000 IU, iv). AlthoughFca or CGba alone <strong>in</strong>duced only modest oestradiol productiondur<strong>in</strong>g the pre-hCG period, serum concentrations of oestradiolwere markedly <strong>in</strong>creased (p < 0.05) 3 days after adm<strong>in</strong>istrationof FcCGba or the Fca +CGba comb<strong>in</strong>ation. F<strong>in</strong>alovarian weight was significantly <strong>in</strong>creased (p < 0.05) <strong>in</strong>animals receiv<strong>in</strong>g Fca, Fca +CGba or FcCGba. Collectively,these observations demonstrate that the s<strong>in</strong>gle cha<strong>in</strong> analoguesof the human gonadotrop<strong>in</strong>s are active <strong>in</strong> sheep.IntroductionThe multiple ovulation-embryo transfer (MOET) technologyis one of the most important management toolsthat producers can use to accelerate the pace of geneticimprovement <strong>in</strong> commercial herds and flocks (Galliet al. 2003; Hasler 2003). Yet, the use of the MOETtechnology <strong>in</strong> the animal <strong>in</strong>dustries <strong>in</strong> North America isapproach<strong>in</strong>g a plateau (Hasler 2003). One critical factorlimit<strong>in</strong>g the more general acceptance of this technologyis its reliance on hormone-<strong>in</strong>duced superovulation andthe variation and <strong>in</strong>consistency <strong>in</strong>herent <strong>in</strong> that process(Kanitz et al. 2002). Until recently, the exogenoushormones used to <strong>in</strong>duce the superovulatory responsewere gonadotrop<strong>in</strong>s derived from pituitary or placentaltissue. The heterogeneous nature of tissue-derivedgonadotrop<strong>in</strong>s and <strong>in</strong>tr<strong>in</strong>sic difference between animals<strong>in</strong> follicular development result <strong>in</strong> marked animal toanimal variation <strong>in</strong> the magnitude of the superovulatoryresponse (Monniaux et al. 1983). The purity andpotency of recomb<strong>in</strong>ant gonadotrop<strong>in</strong>s may reduce thevariation associated with hormone-<strong>in</strong>duced superovulationand, thereby, facilitate more general acceptance ofthis valuable tool for genetic improvement.Non-Recomb<strong>in</strong>ant Gonadotrop<strong>in</strong>sConventional, tissue-derived gonadotrop<strong>in</strong>s used <strong>in</strong> theanimal <strong>in</strong>dustries (Boland et al. 1991; Kanitz et al. 2002)<strong>in</strong>clude ov<strong>in</strong>e and porc<strong>in</strong>e follicle stimulat<strong>in</strong>g hormone(FSH) and equ<strong>in</strong>e and human chorionic gonadotrop<strong>in</strong>(eCG and hCG, respectively). Pituitary and placentalgonadotrop<strong>in</strong>s are composed of two subunits, commonlydesignated as the a and b subunits (Pierce andParsons 1981; Hearn and Gomme 2000). The a subunitis shared by all gonadotrop<strong>in</strong>s produced by a givenspecies, while the b subunit of each gonadotrop<strong>in</strong> isunique. For example, the dimeric (ab) composition ofhuman LH, FSH and CG consists of a hormone specificb subunit and a common a subunit (Fig. 1). Althoughthe a and b subunits are not covalently l<strong>in</strong>ked, theconfiguration of each subunit is stabilized by several<strong>in</strong>tra-cha<strong>in</strong> disulphide bridges.A feature of the gonadotrop<strong>in</strong>s that markedly <strong>in</strong>fluencespotency and duration of response is the degree ofglycosylation. Indeed, a significant portion of the massof gonadotrop<strong>in</strong>s resides <strong>in</strong> the oligosaccharide cha<strong>in</strong>sadded dur<strong>in</strong>g co- and post-translational process<strong>in</strong>g.Pituitary-derived gonadotrop<strong>in</strong>s conta<strong>in</strong> three or fourasparag<strong>in</strong>e-associated (N-l<strong>in</strong>ked) oligosaccharidegroups. The a subunits of human, bov<strong>in</strong>e and ov<strong>in</strong>egonadotrop<strong>in</strong>s carry oligosaccharide cha<strong>in</strong>s attached totwo asparag<strong>in</strong>e residues. Similarly, the b subunits carryone (hLHb) or two (hFSHb) N-l<strong>in</strong>ked carbohydratecha<strong>in</strong>s. Human chorionic gonadotrop<strong>in</strong>, like FSH, hastwo N-l<strong>in</strong>ked cha<strong>in</strong>s associated with the b subunit. Inaddition, the b subunit of hCG conta<strong>in</strong>s four oligosaccharidecha<strong>in</strong>s l<strong>in</strong>ked to ser<strong>in</strong>e residues (O-l<strong>in</strong>kedcha<strong>in</strong>s) located <strong>in</strong> the carboxy-term<strong>in</strong>al portion of CGb.The N-l<strong>in</strong>ked glycans of LH and FSH are a heterogeneousmix of di- and tri-branched oligosaccharidecha<strong>in</strong>s (Green and Baenziger 1988a; b). Heterogeneity isalso evident at the branch term<strong>in</strong>i, with term<strong>in</strong>alsulphate residues common <strong>in</strong> LH while sialylatedterm<strong>in</strong>i predom<strong>in</strong>ate <strong>in</strong> FSH (Green and Baenziger1988b). The degree of sialylation also varies acrossspecies. For example, 88% of the oligosaccharide cha<strong>in</strong>sof human FSH end with sialic acid, while only 38% ofterm<strong>in</strong>i <strong>in</strong> ov<strong>in</strong>e FSH are sialylated. In contrast, theN-l<strong>in</strong>ked oligosaccharide cha<strong>in</strong>s <strong>in</strong> hCG are much moreuniform and generally carry term<strong>in</strong>al sialic acid residues(Kessler et al. 1979b). The O-l<strong>in</strong>ked glycans of hCG arealso highly sialylated (Kessler et al. 1979a).The carbohydrate portion of the gonadotrop<strong>in</strong>s<strong>in</strong>fluences the fold<strong>in</strong>g, assembly, secretion, clearanceand biological activity of the gonadotrop<strong>in</strong>s (Thotakuraand Blithe 1995). The liver plays an active role <strong>in</strong> theclearance and degradation of the gonadotrop<strong>in</strong>s (Fieteet al. 1991). Indeed, hepatocytes conta<strong>in</strong> high concentrationsof a receptor that specifically recognizes thesulphated, but not the sialylated, oligosaccharide cha<strong>in</strong>sof the gonadotrop<strong>in</strong>s (Roseman and Baenziger 2000).Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Recomb<strong>in</strong>ant Gonadotrop<strong>in</strong>s <strong>in</strong> Assisted <strong>Reproduction</strong> 187Fig. 1. Tissue-derived gonadotrop<strong>in</strong>s (LH, FSH and hCG) are composedof non-covalently l<strong>in</strong>ked a and b subunits each of which carryone (LHb) or two (a, FSHb and CGb) N-l<strong>in</strong>ked oligosaccharide cha<strong>in</strong>s(•). The carboxy-term<strong>in</strong>al peptide (CTP) of hCG (dashed l<strong>in</strong>e) alsocarries four O-l<strong>in</strong>ked glycans (*). In s<strong>in</strong>gle cha<strong>in</strong> gonadotrop<strong>in</strong>s, the aand b doma<strong>in</strong>s are l<strong>in</strong>ked by CTP. The chimeric s<strong>in</strong>gle cha<strong>in</strong>gonadotrop<strong>in</strong> with dual activity (FCCGa) conta<strong>in</strong>s FSHb, CGb anda doma<strong>in</strong>s l<strong>in</strong>ked by CTPAs a consequence, LH, a sulphated gonadotrop<strong>in</strong>, has ashort half-life. Conversely, the richly sialylated placentalcounterpart, hCG, has a much longer functional life.Deficiencies of Non-Recomb<strong>in</strong>antGonadotrop<strong>in</strong>sThe heterogeneous nature of tissue-derived gonadotrop<strong>in</strong>preparations results <strong>in</strong> significant variation amongbatches with respect to purity and biopotency (Murphyet al. 1984; Phillips et al. 1993; Kanitz et al. 2002). Inaddition, exogenous gonadotrop<strong>in</strong>s of equ<strong>in</strong>e or porc<strong>in</strong>eorig<strong>in</strong> can <strong>in</strong>duce an immune response that may limit therepetitive use of these preparations <strong>in</strong> sheep and cattle(Bod<strong>in</strong> et al. 1997; Drion et al. 2001). Furthermore, theshort half-life of ov<strong>in</strong>e, porc<strong>in</strong>e and human FSHpreparations necessitates cont<strong>in</strong>uous or repetitiveadm<strong>in</strong>istration, <strong>in</strong>creas<strong>in</strong>g the time and expense associatedwith animal sort<strong>in</strong>g and handl<strong>in</strong>g (Alcivar et al.1992; Kanitz et al. 2002). Conversely, the long life ofeCG may compromise embryo viability (Mart<strong>in</strong>uk et al.1991) and often requires the adm<strong>in</strong>istration of anti-eCGsera dur<strong>in</strong>g the post-ovulatory period (Gonzalez et al.1994). In addition, tissue-derived gonadotrop<strong>in</strong>s oftenconta<strong>in</strong> differ<strong>in</strong>g proportions of LH and FSH activity(Kanitz et al. 2002). This heterogeneity likely contributesto variation <strong>in</strong> the physiological response (Murphyet al. 1984). A grow<strong>in</strong>g concern is the possibility thattissue-derived gonadotrop<strong>in</strong>s may harbour viruses, bacteriaor prions that may result <strong>in</strong> the <strong>in</strong>advertentdevelopment of disease <strong>in</strong> treated animals (Reichl et al.2002; Galli et al. 2003; Matorras and Rodriguez-Escudero2003). Indeed, certa<strong>in</strong> European countries havebanned the use of pituitary-derived gonadotrop<strong>in</strong>s <strong>in</strong>superovulation protocols (Galli et al. 2003).Recomb<strong>in</strong>ant Gonadotrop<strong>in</strong>sRecomb<strong>in</strong>ant forms of LH and FSH are generated byimmortalized cells transfected with gene constructsencod<strong>in</strong>g the a and b subunits (Kaetzel et al. 1985;Keene et al. 1989; Lunenfeld 2004). The transfected cellsare ma<strong>in</strong>ta<strong>in</strong>ed <strong>in</strong> culture and represent a renewablesource of dimeric gonadotrop<strong>in</strong> (Howles 1996). Olijveand co-workers (Olijve et al. 1996) transfected cells witha construct that <strong>in</strong>corporated the DNA encod<strong>in</strong>g the aand b subunits of human FSH <strong>in</strong>to a s<strong>in</strong>gle vector. Inaddition, multiple copies of the composite constructwere stably <strong>in</strong>serted <strong>in</strong> the transfected cells (Olijve et al.1996), <strong>in</strong>creas<strong>in</strong>g the efficiency of dimeric FSH production<strong>in</strong> the cell culture system. In addition, recentadvances <strong>in</strong> manufactur<strong>in</strong>g and purification generaterecomb<strong>in</strong>ant gonadotrop<strong>in</strong> preparations that are uniform<strong>in</strong> isoform profile and glycan composition (Driebergenand Baer 2003; Bassett and Driebergen 2005).These advances <strong>in</strong> recomb<strong>in</strong>ant gonadotrop<strong>in</strong> technologyaddress many of the concerns that have developedregard<strong>in</strong>g the use of tissue-derived gonadotrop<strong>in</strong>s.Current recomb<strong>in</strong>ant technology produces pharmaceuticalgrade LH and FSH of def<strong>in</strong>ed am<strong>in</strong>o acid andcarbohydrate composition (Olijve et al. 1996). Currentpurification procedures ensure that the purity, biopotencyand batch-to-batch consistency are very high(Daya 2004). Furthermore, the generation of therecomb<strong>in</strong>ants <strong>in</strong> a serum-free system by immortalizeddisease-free cells ensures that the recomb<strong>in</strong>ant gonadotrop<strong>in</strong>sare not vectors of disease. An additional benefitof the recomb<strong>in</strong>ant gonadotrop<strong>in</strong>s is that the high purityand consistent biopotency across batches permits dos<strong>in</strong>gbased on mass (Driebergen and Baer 2003).The gene constructs encod<strong>in</strong>g the gonadotrop<strong>in</strong>s aregenerally <strong>in</strong>serted <strong>in</strong>to a cell l<strong>in</strong>e derived from ovariantissue of a Ch<strong>in</strong>ese hamster. The Ch<strong>in</strong>ese hamster ovary(CHO) cells are used from this purpose because theyaccept transfection easily (Olijve et al. 1996). In addition,CHO cells conta<strong>in</strong> the <strong>in</strong>tracellular mach<strong>in</strong>eryrequired to form the <strong>in</strong>tra-cha<strong>in</strong> disulphide bridges andcorrectly complete the fold<strong>in</strong>g and assembly of the dimer(Howles 1996). Furthermore, CHO cells constitutivelysecrete the recomb<strong>in</strong>ant prote<strong>in</strong> <strong>in</strong>to the culturemedium, a characteristic that facilitates gonadotrop<strong>in</strong>recovery and purification. Most importantly, the patternof glycosylation <strong>in</strong> CHO cells mirrors that found <strong>in</strong>pituitary-derived gonadotrop<strong>in</strong>s. Yet, CHO cells lackthe sulphotransferase conta<strong>in</strong>ed <strong>in</strong> gonadotrope cells(Smith et al. 1992). As a consequence, the oligosaccharidecha<strong>in</strong>s of recomb<strong>in</strong>ant gonadotrop<strong>in</strong>s carry term<strong>in</strong>alsialic acid groups, rather than the term<strong>in</strong>al sulphateresidues that predom<strong>in</strong>ate <strong>in</strong> pituitary-derived gonadotrop<strong>in</strong>s(Smith et al. 1990; Baenziger et al. 1992). Thepractical consequence of sialic acid term<strong>in</strong>i is that therecomb<strong>in</strong>ant gonadotrop<strong>in</strong>s are afforded protectionfrom the hepatic prote<strong>in</strong>s that <strong>in</strong>duce the rapid degradationof gonadotrop<strong>in</strong>s with sulphated term<strong>in</strong>i (Baenzigeret al. 1992; Roseman and Baenziger 2000).Use of Recomb<strong>in</strong>ant Dimeric Gonadotrop<strong>in</strong>sThe efficacy of recomb<strong>in</strong>ant FSH <strong>in</strong> the treatment of<strong>in</strong>fertility <strong>in</strong> humans was exam<strong>in</strong>ed <strong>in</strong> a series of studiesconducted <strong>in</strong> the 1990s (Devroey et al. 1992; Germondet al. 1992) and the use of these recomb<strong>in</strong>ant gonadotrop<strong>in</strong>preparations has become common <strong>in</strong> <strong>in</strong> vitroÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


188 TE Adams and I Boimefertilization programs (Daya 2004). In the animal<strong>in</strong>dustries, recomb<strong>in</strong>ant forms of bov<strong>in</strong>e (Wilson et al.1993; Wehrman et al. 1996) and human (Takagi et al.2001) FSH (rbFSH and rhFSH, respectively) have beenused to <strong>in</strong>duce superovulation <strong>in</strong> beef cattle. Recomb<strong>in</strong>antporc<strong>in</strong>e FSH (rpFSH) has also been produced andthe biological activity has been verified us<strong>in</strong>g an <strong>in</strong> vitroSertoli cell bioassay and an <strong>in</strong> vivo assay based on follicledevelopment <strong>in</strong> hypophysectomized mice treated withrpFSH (Inaba et al. 1997).The comprehensive studies of Wilson and co-workers(Wilson et al. 1993) demonstrated that rbFSH was aseffective as pituitary-derived FSH <strong>in</strong> promot<strong>in</strong>g thesuperovulatory response <strong>in</strong> mature cows. The treatmentprotocol required rbFSH adm<strong>in</strong>istration (im) at 12 h<strong>in</strong>tervals for 3–5 days. The optimal response was notedat a total dose of 24 mg rbFSH. This level of rbFSH iscomparable to the total dose of purified pituitary FSHrequired to <strong>in</strong>duce superovulation <strong>in</strong> beef and dairycows (Alcivar et al. 1992; Kanitz et al. 2002). Thedevelopment of multiple pre-ovulatory follicles was also<strong>in</strong>duced <strong>in</strong> heifers receiv<strong>in</strong>g twice daily <strong>in</strong>jections ofrhFSH over a 4-day-treatment period (Takagi et al.2001). The magnitude of the rhFSH-<strong>in</strong>duced responsewas comparable to the follicular response <strong>in</strong>duced byeCG. Yet, when compared with eCG-treated heifers,serum concentrations of oestradiol dur<strong>in</strong>g the preovulatoryperiod were significantly reduced <strong>in</strong> heifersreceiv<strong>in</strong>g rhFSH. As a consequence, the <strong>in</strong>duction of thepre-ovulatory surge of LH was delayed. Takagi and coworkerssuggest that endogenous production of LH isnot sufficient to support oestradiol synthesis dur<strong>in</strong>grhFSH-<strong>in</strong>duced follicle development. Therefore, concurrentadm<strong>in</strong>istration of LH and rhFSH may berequired to achieve an ovulatory response that iscomparable to that <strong>in</strong>duced by eCG.Dimeric CTP-Modified Gonadotrop<strong>in</strong>sIn contrast to pituitary-derived gonadotrop<strong>in</strong>s, placentalgonadotrop<strong>in</strong>s are cleared slowly and, as a consequence,the duration of response <strong>in</strong>duced by eCG orhCG is much longer than the response <strong>in</strong>duced by LH orFSH (Mart<strong>in</strong>uk et al. 1991; Baenziger et al. 1992). Thelong half-life of the placental gonadotrop<strong>in</strong>s is due, <strong>in</strong>part, to a carboxyl term<strong>in</strong>al peptide (CTP) extension ofthe b subunit that is subject to extensive O-l<strong>in</strong>kedglycosylation (Kessler et al. 1979a; Bousfield et al.2001). The critical impact of the CTP on gonadotrop<strong>in</strong>clearance was demonstrated by Matzuk and co-workers(Matzuk et al. 1990), who produced a truncated form ofhCGb lack<strong>in</strong>g the CTP doma<strong>in</strong>. Although the truncatedvariant of hCG was recognized with high aff<strong>in</strong>ity by theLH receptor, the <strong>in</strong> vivo potency was dramaticallyreduced by deletion of the CTP doma<strong>in</strong>. Similarly,<strong>in</strong>corporat<strong>in</strong>g a CTP doma<strong>in</strong> <strong>in</strong>to the b subunits of FSHor TSH markedly <strong>in</strong>creases the half-life of the CTPmodifiedforms of FSH and TSH (Fares et al. 1992;Joshi et al. 1995).Although the bioactivity of CTP-modified dimericFSH has not been exam<strong>in</strong>ed <strong>in</strong> domestic species, theefficacy of an hFSH analogue <strong>in</strong>corporat<strong>in</strong>g the CTPdoma<strong>in</strong> (hFSH-CTP) has been exam<strong>in</strong>ed <strong>in</strong> humans.The hFSH analogue is effective <strong>in</strong> promot<strong>in</strong>g spermatogenesisand follicle development (Bouloux et al.2001; Balen et al. 2004). Importantly, <strong>in</strong> comparisonwith rhFSH, the rate of clearance of hFSH-CTP ismarkedly reduced and development of pre-ovulatoryfollicles is <strong>in</strong>duced by a s<strong>in</strong>gle <strong>in</strong>jection of the FSHanalogue. This is <strong>in</strong> contrast to the repetitive <strong>in</strong>jectionschedule that is required to promoted follicle maturationus<strong>in</strong>g pituitary-derived FSH. Collectively, thesedata demonstrate that the CTP doma<strong>in</strong> extends thefunctional life of FSH and simplifies the ovulation<strong>in</strong>duction protocol by reduc<strong>in</strong>g the frequency ofadm<strong>in</strong>istration.S<strong>in</strong>gle Cha<strong>in</strong> Analogues of LH and FSHA recent advance <strong>in</strong> the production of recomb<strong>in</strong>antgonadotrop<strong>in</strong>s is the generation of s<strong>in</strong>gle cha<strong>in</strong> analoguesof the normally dimeric prote<strong>in</strong>s (Garcia-Campayoand Boime 2001). The s<strong>in</strong>gle cha<strong>in</strong> technology<strong>in</strong>volves transfection of CHO cells with a plasmid thatcarries DNA encod<strong>in</strong>g both subunits comb<strong>in</strong>ed <strong>in</strong> as<strong>in</strong>gle gene construct. The l<strong>in</strong>ker sequence that jo<strong>in</strong>s thea and b doma<strong>in</strong>s of the construct encodes the CTPportion of hCGb. The prote<strong>in</strong> product secreted bytransfected CHO cells is a s<strong>in</strong>gle peptide cha<strong>in</strong> thatconta<strong>in</strong>s a and b doma<strong>in</strong>s l<strong>in</strong>ked through the CTPsegment. To date, s<strong>in</strong>gle cha<strong>in</strong> analogues of hFSH(FSHb–CTP–a; Sugahara et al. 1996), hLH (LHb–CTP–a; Garcia-Campayo et al. 1997), equ<strong>in</strong>e LH(eLHb–CTP–a; Jablonka-Shariff et al. 2007) and hCG(CGb–a; Sugahara et al. 1995) have been developed.The s<strong>in</strong>gle cha<strong>in</strong> analogues of the gonadotrop<strong>in</strong>s arerecognized by the appropriate receptor (LH or FSHreceptor) with high aff<strong>in</strong>ity and the s<strong>in</strong>gle cha<strong>in</strong> analogues<strong>in</strong>duce the normal <strong>in</strong>tracellular second messengerresponse. The bioactivity of the s<strong>in</strong>gle cha<strong>in</strong> analoguesdemonstrates that the fold<strong>in</strong>g of the a and b components<strong>in</strong>to the configuration required to associate with thegonadotrop<strong>in</strong> receptor is not impaired by the s<strong>in</strong>glecha<strong>in</strong> organization of the a and b doma<strong>in</strong>s. The correctfold<strong>in</strong>g is likely facilitated by the <strong>in</strong>terven<strong>in</strong>g CTPsequence. Importantly, the high proportion of prol<strong>in</strong>eand ser<strong>in</strong>e residues <strong>in</strong> the CTP sequence makes thissegment highly flexible and allows the a and b doma<strong>in</strong>sof the s<strong>in</strong>gle cha<strong>in</strong> to fold <strong>in</strong>to the active configuration.An additional benefit of <strong>in</strong>corporat<strong>in</strong>g the CTPsequence <strong>in</strong>to the s<strong>in</strong>gle cha<strong>in</strong> is that, much like thedimeric CTP-modified gonadotrop<strong>in</strong>s discussed above,the CTP portion of the s<strong>in</strong>gle cha<strong>in</strong> reduces the rate ofclearance. An additional practical consequence of produc<strong>in</strong>ggonadotrop<strong>in</strong>s as s<strong>in</strong>gle cha<strong>in</strong> prote<strong>in</strong>s is thatdimerization, the rate limit<strong>in</strong>g step <strong>in</strong> the synthesis ofconventional gonadotrop<strong>in</strong>s, is bypassed. This uniquefeature of the s<strong>in</strong>gle cha<strong>in</strong> prote<strong>in</strong>s enhances theefficiency of synthesis and secretion by transfectedCHO cells (Sugahara et al. 1996).The recent studies of Jablonka-Shariff and co-workersexam<strong>in</strong>ed the activity of a s<strong>in</strong>gle cha<strong>in</strong> analogue ofequ<strong>in</strong>e LH (Jablonka-Shariff et al. 2007). The analoguewas as potent as pituitary-derived eLH <strong>in</strong> promot<strong>in</strong>gtestosterone production by equ<strong>in</strong>e Leydig cells <strong>in</strong>culture. The s<strong>in</strong>gle cha<strong>in</strong> eLH analogue also <strong>in</strong>creasedÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Recomb<strong>in</strong>ant Gonadotrop<strong>in</strong>s <strong>in</strong> Assisted <strong>Reproduction</strong> 189the serum concentration of testosterone after adm<strong>in</strong>istrationto stallions. The s<strong>in</strong>gle cha<strong>in</strong> analogue was alsoas effective as hCG <strong>in</strong> <strong>in</strong>duc<strong>in</strong>g ovulation <strong>in</strong> mares(Yoon et al. 2007). Indeed, ovulation was evident with<strong>in</strong>48 h of analogue adm<strong>in</strong>istration <strong>in</strong> mares with a maturefollicle (>35 mm) at the time of adm<strong>in</strong>istration. Ovulationwas noted with<strong>in</strong> the same temporal w<strong>in</strong>dow <strong>in</strong>only 15% of control mares receiv<strong>in</strong>g vehicle alone. Yoonand colleagues suggest that the s<strong>in</strong>gle cha<strong>in</strong> analogue ofeLH may be useful <strong>in</strong> accelerat<strong>in</strong>g ovulation and,thereby, facilitat<strong>in</strong>g timed mat<strong>in</strong>g and <strong>in</strong>sem<strong>in</strong>ationus<strong>in</strong>g frozen ⁄ thawed semen.The bioactivity of s<strong>in</strong>gle cha<strong>in</strong> analogues of hFSH hasbeen assessed <strong>in</strong> rodent (Weenen et al. 2004) andprimate (Kle<strong>in</strong> et al. 2002) models. Adm<strong>in</strong>istration ofa s<strong>in</strong>gle dose of the FSH analogue to prepuberal rats<strong>in</strong>duced a significant <strong>in</strong>crease <strong>in</strong> ovarian weight and<strong>in</strong>hib<strong>in</strong> secretion (Weenen et al. 2004) and promoted thedevelopment of multiple antral follicles (Trousdale et al.2007). In contrast, ovarian weight and <strong>in</strong>hib<strong>in</strong> productiondid not differ from sal<strong>in</strong>e-treated control animals <strong>in</strong>rats receiv<strong>in</strong>g a s<strong>in</strong>gle <strong>in</strong>jection of dimeric rhFSH. This<strong>in</strong>dicates that the functional life of the s<strong>in</strong>gle cha<strong>in</strong>analogue is greater than that of dimeric FSH. Indeed,direct assessment of the half-life <strong>in</strong>dicated that theclearance of the s<strong>in</strong>gle cha<strong>in</strong> analogue was three timesslower than the clearance of dimeric FSH <strong>in</strong> the rodentmodel. Follicle development and oestradiol productionwere also <strong>in</strong>creased <strong>in</strong> monkeys receiv<strong>in</strong>g a s<strong>in</strong>gle<strong>in</strong>jection of the s<strong>in</strong>gle cha<strong>in</strong> FSH analogue (Kle<strong>in</strong> et al.2002). As <strong>in</strong> the rodent model, the half-life of the FSHanalogue was markedly <strong>in</strong>creased relative to the half-lifeof recomb<strong>in</strong>ant dimeric FSH. Collectively, these studiesdemonstrate that the s<strong>in</strong>gle cha<strong>in</strong> form of FSH is activeand long-lived.Gonadotrop<strong>in</strong>s with Dual ActivityA novel variation of the s<strong>in</strong>gle cha<strong>in</strong> technology <strong>in</strong>volvesthe <strong>in</strong>corporation of two or more different b subunitdoma<strong>in</strong>s <strong>in</strong>to the s<strong>in</strong>gle cha<strong>in</strong> prote<strong>in</strong> (Kanda et al.1999; Garcia-Campayo and Boime 2001). When exam<strong>in</strong>edus<strong>in</strong>g <strong>in</strong> vitro assay systems a s<strong>in</strong>gle cha<strong>in</strong>gonadotrop<strong>in</strong> with a, hFSHb and hCGb doma<strong>in</strong>s(FSHb–CTP–CGb–a) <strong>in</strong>teracted with LH and FSHreceptors and <strong>in</strong>duced cAMP production that wascomparable to the level <strong>in</strong>duced by dimeric LH andFSH (Kanda et al. 1999). The dual activity of thechimeric gonadotrop<strong>in</strong> suggests that the s<strong>in</strong>gle cha<strong>in</strong>prote<strong>in</strong> can assume a secondary structure that isrecognized by both gonadotrop<strong>in</strong> receptors. Yet, recentstudies <strong>in</strong>dicate that the chimeric prote<strong>in</strong>s assume twodifferent conformations, one that is recognized by theFSH receptor and a second configuration that permitsrecognition by the LH receptor (Garcia-Campayo et al.2004). In other words, the s<strong>in</strong>gle cha<strong>in</strong> prote<strong>in</strong>s secretedfrom transfected CHO cells represent a heterogeneouspopulation of conformations, some with the capacity toactivate the LH receptor and others capable of activat<strong>in</strong>gthe FSH receptor. The dual activity of the chimerichuman gonadotrop<strong>in</strong>s is similar to the bifunctionalpotential of eCG. Indeed, the dually active chimericgonadotrop<strong>in</strong> was as effective as eCG <strong>in</strong> promot<strong>in</strong>gfollicle development, enhanc<strong>in</strong>g of ovarian weight andaugment<strong>in</strong>g aromatase activity <strong>in</strong> immature mice (Jablonka-Shariffet al. 2006). The ovulation <strong>in</strong>ductionaction of hCG was also mimicked by the dually activechimeric gonadotrop<strong>in</strong> <strong>in</strong> eCG-primed immature mice.In our recent studies, the authors have compared thebiopotency of the dually active human gonadotrop<strong>in</strong>with s<strong>in</strong>gle cha<strong>in</strong> analogues of hFSH and hCG (Lemkeet al. 2008). The research model we used employed ewestreated with progesterone-conta<strong>in</strong><strong>in</strong>g vag<strong>in</strong>al <strong>in</strong>serts(CIDR’s) and anti-GnRH sera to suppress secretion ofendogenous gonadotrop<strong>in</strong>s (Fig. 2). Ewes received as<strong>in</strong>gle <strong>in</strong>jection of the dually active gonadotrop<strong>in</strong>(FSHb–CTP–CGb–a; 5 IU⁄ kg, iv), s<strong>in</strong>gle cha<strong>in</strong> analoguesof hFSH (FSHb–CTP–a; Fca; 5IU⁄ kg, iv) orCG (CGb–a; CGba; 5IU⁄ kg, iv), or the FSH and CGanalogues <strong>in</strong> comb<strong>in</strong>ation (Fca +CGba; both at5IU⁄ kg, iv) at the time of CIDR removal. Controlanimals received vehicle alone. Ovulation was <strong>in</strong>ducedby hCG (1000 IU, iv) <strong>in</strong>jection 3 days after adm<strong>in</strong>istrationof the s<strong>in</strong>gle cha<strong>in</strong> gonadotrop<strong>in</strong>s.As oestradiol production requires the concertedaction of both LH and FSH, the authors used oestradiolsecretion as a measure of the bifunctional capacity of thes<strong>in</strong>gle cha<strong>in</strong> gonadotrop<strong>in</strong>s. Significant oestradiol secretionwas not evident <strong>in</strong> ewes receiv<strong>in</strong>g Fca or CGbaalone. Yet, serum levels of oestradiol were markedly<strong>in</strong>creased <strong>in</strong> ewes receiv<strong>in</strong>g Fca and CGba <strong>in</strong> comb<strong>in</strong>ation.The results of this study demonstrate that thes<strong>in</strong>gle cha<strong>in</strong> analogues of hFSH and hCG are active <strong>in</strong>sheep. These data also clearly illustrate that the s<strong>in</strong>glecha<strong>in</strong> construct that <strong>in</strong>corporates FSHb and CGbdoma<strong>in</strong>s has both FSH and LH activity (Table 1).Similarly, follicle development was promoted by thedually active chimeric gonadotrop<strong>in</strong>s and the comb<strong>in</strong>edtreatment with Fca and CGba. This augmented follicularresponse is <strong>in</strong>dicated by the <strong>in</strong>crease <strong>in</strong> ovarianweight and corpora lutea number that were noted <strong>in</strong>ovarian tissue collected 11 days after adm<strong>in</strong>istration ofFig. 2. The oestrous activity of yearl<strong>in</strong>g ewes was synchronized us<strong>in</strong>gprogesterone-conta<strong>in</strong><strong>in</strong>g vag<strong>in</strong>al <strong>in</strong>serts (CIDRs). Secretion of endogenousgonadotrop<strong>in</strong>s was suppressed by passive immunization withantisera directed aga<strong>in</strong>st GnRH (Anti-GnRH) 1 day prior to CIDRremoval. <strong>Animals</strong> (n = 6 ewes ⁄ group) received recomb<strong>in</strong>ant gonadotrop<strong>in</strong>(CGba, Fca, FcCGba or CGba +Fca <strong>in</strong> comb<strong>in</strong>ation) atCIDR removal. Chimeric gonadotrop<strong>in</strong>s were adm<strong>in</strong>istered at a doseof 5 IU ⁄ kg (iv). Control animals received conditioned media of nontransfectedCHO-cells. Ovulation was <strong>in</strong>duced by hCG (1000 IU, iv)on day 3 and ovarian tissue was collected on day 11Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


190 TE Adams and I BoimeTable 1. Effect of s<strong>in</strong>gle cha<strong>in</strong> human gonadotrop<strong>in</strong>s on pre-ovulatoryserum concentrations of oestradiol (concentration 3 days after adm<strong>in</strong>istrationof s<strong>in</strong>gle cha<strong>in</strong> gonadotrop<strong>in</strong>) and f<strong>in</strong>al ovarian weight andcorpora lutea per animalTreatment(5 IU ⁄ kg, iv) nPre-ovulatoryoestradiol (pg ⁄ ml)Ovarianweight (g)Corpora lutea(CLs ⁄ animal)Control 6 1.1 ± .6 a 1.3 ± .1 a 0 aCGba 6 3.2 ± 1.3 a 2.8 ± .2 b 1.0 ± .4 bFCa 6 3.7 ± 1.1 a 14.0 ± 1.4 c 9.5 ± 2.4 cFCa +CGba 6 34.4 ± 9.8 b 11.1 ± 1.0 c 11.3 ± 2.7 cFcCGba 6 44.0 ± 8.2 b 10.5 ± 1.0 c 6.7 ± 2.7 cOvarian parameters were assessed 11 days after adm<strong>in</strong>istration of s<strong>in</strong>gle cha<strong>in</strong>gonadotrop<strong>in</strong>s. Values with different superscripts differed significantly(p < 0.05).the s<strong>in</strong>gle cha<strong>in</strong> gonadotrop<strong>in</strong>s. Although Fca alone wasunable to <strong>in</strong>duce a significant <strong>in</strong>crease <strong>in</strong> oestradiolproduction, this s<strong>in</strong>gle cha<strong>in</strong> analogue of hFSH didpromote the development of multiple follicles that were<strong>in</strong>duced to ovulate by the hCG challenge adm<strong>in</strong>istered3 days after s<strong>in</strong>gle cha<strong>in</strong> gonadotrop<strong>in</strong>. These observations<strong>in</strong>dicate that follicle development and oestradiolbiosynthesis are separable phenomena. In addition, thisobservation illustrates that a s<strong>in</strong>gle <strong>in</strong>jection of the s<strong>in</strong>glecha<strong>in</strong> gonadotrop<strong>in</strong>s is sufficient to <strong>in</strong>duce the superovulatoryresponse. This is <strong>in</strong> marked contrast to themultiple <strong>in</strong>jection regimen required to <strong>in</strong>duce the superovulatoryresponse us<strong>in</strong>g pituitary-derived or dimericrecomb<strong>in</strong>ant FSH.The Next Generation of Recomb<strong>in</strong>antGonadotrop<strong>in</strong>sThe s<strong>in</strong>gle cha<strong>in</strong> technology opens the possibility ofl<strong>in</strong>k<strong>in</strong>g multiple bioactive doma<strong>in</strong>s <strong>in</strong> a s<strong>in</strong>gle recomb<strong>in</strong>antprote<strong>in</strong>. The development of recomb<strong>in</strong>ant prote<strong>in</strong>s<strong>in</strong>corporat<strong>in</strong>g portions of LH and FSH to generateprote<strong>in</strong>s with dual activity is a clear example of thepotential of the s<strong>in</strong>gle cha<strong>in</strong> technology. Modificationsof this technology may result <strong>in</strong> the generation of s<strong>in</strong>glecha<strong>in</strong> gonadotrop<strong>in</strong>s specifically tailored to meet thefunctional requirements of a targeted physiologicresponse. For example, a s<strong>in</strong>gle cha<strong>in</strong> gonadotrop<strong>in</strong>with dual activity may be required to <strong>in</strong>duce follicledevelopment and ovulation <strong>in</strong> the prepuberal period ornon-breed<strong>in</strong>g season while a long-lived s<strong>in</strong>gle cha<strong>in</strong>gonadotrop<strong>in</strong> with s<strong>in</strong>gular FSH activity may be optimalto <strong>in</strong>duce a superovulatory response dur<strong>in</strong>g thebreed<strong>in</strong>g season. Similarly, a very long-lived gonadotrop<strong>in</strong>may be useful <strong>in</strong> promot<strong>in</strong>g spermatogenesis <strong>in</strong>elite breed<strong>in</strong>g males while a s<strong>in</strong>gle cha<strong>in</strong> with a moremoderate functional life may be optimal for follicledevelopment. Comb<strong>in</strong><strong>in</strong>g the potential of molecularbiology with the utility of the s<strong>in</strong>gle cha<strong>in</strong> technologywill allow scientists and production eng<strong>in</strong>eers to developselective analogues of the gonadotrop<strong>in</strong>s that aredesigned to address a specific application.The s<strong>in</strong>gle cha<strong>in</strong> technology may also be used togenerate functional prote<strong>in</strong>s that <strong>in</strong>clude non-gonadotrop<strong>in</strong>doma<strong>in</strong>s. For example, Trousdale and coworkers(Trousdale et al. 2007) recently generated a s<strong>in</strong>gle cha<strong>in</strong>construct that comb<strong>in</strong>ed the functional doma<strong>in</strong> ofvascular endothelial growth factor (VEGF) with adoma<strong>in</strong> encod<strong>in</strong>g the s<strong>in</strong>gle cha<strong>in</strong> variant of FSH. Theresult<strong>in</strong>g bifunctional prote<strong>in</strong> displayed angiogenic andFSH activity <strong>in</strong> <strong>in</strong> vitro assay systems and <strong>in</strong>creasedfollicle development and perifollicular angiogenesis<strong>in</strong> vivo. Similarly, multi-functional prote<strong>in</strong>s with LH,FSH and TSH activity have been generated us<strong>in</strong>g thes<strong>in</strong>gle cha<strong>in</strong> technology (Garcia-Campayo et al. 2002).Collectively, these observations demonstrate that thes<strong>in</strong>gle cha<strong>in</strong> technology is not limited to gonadotrop<strong>in</strong>sbut may also be expanded to <strong>in</strong>clude other functionaldoma<strong>in</strong>s that may synergize with the gonadotrop<strong>in</strong>component to enhance the fertility of domestic species.Another recent advance (Low et al. 2005) is thel<strong>in</strong>kage of the s<strong>in</strong>gle cha<strong>in</strong> form of FSH to the Fcdoma<strong>in</strong> of immunoglob<strong>in</strong> G 1 (IgG 1 ). The s<strong>in</strong>gle cha<strong>in</strong>FSH-Fc prote<strong>in</strong> reta<strong>in</strong>ed FSH activity and the <strong>in</strong>clusionof the Fc doma<strong>in</strong> markedly prolonged the functional lifeof the chimeric prote<strong>in</strong>. In addition, the <strong>in</strong>clusion of theFc doma<strong>in</strong> permitted Fc receptor mediated absorptionacross epithelial barriers <strong>in</strong> the <strong>in</strong>test<strong>in</strong>e and lungs. Thisopens the possibility of adm<strong>in</strong>ister<strong>in</strong>g multifunctionalgonadotrop<strong>in</strong> constructs orally or by <strong>in</strong>halation. IndeedLow and coworkers (Low et al. 2005) demonstrated thatthe physiological response to FSH (<strong>in</strong>creased testisweight and <strong>in</strong>creased <strong>in</strong>hib<strong>in</strong> production) was evident <strong>in</strong>rats and monkeys after oral or pulmonary adm<strong>in</strong>istration.ConclusionTissue-derived gonadotrop<strong>in</strong>s have facilitated the developmentand application of reproductive technologiesthat <strong>in</strong>crease the productivity of herds and flocks byreduc<strong>in</strong>g management and labour costs and <strong>in</strong>creas<strong>in</strong>gthe pace of genetic improvement. Yet, variation <strong>in</strong> thepotency, purity and availability of natural gonadotrop<strong>in</strong>smay limit further advances. The high purity,potency and longevity of recomb<strong>in</strong>ant dimeric gonadotrop<strong>in</strong>sand novel s<strong>in</strong>gle cha<strong>in</strong> gonadotrop<strong>in</strong> analoguesmake these gonadotrop<strong>in</strong>s practical and effective alternativesto tissue-derived preparations <strong>in</strong> superovulatoryprotocols and out-of-season breed<strong>in</strong>g programs.AcknowledgementsThis project was supported by National Research Initiative CompetitiveGrant 5-35203-16274 from the USDA Cooperative StateResearch, Education, and Extension Service Animal <strong>Reproduction</strong>Program and the California Agricultural Experiment Station.ReferencesAlcivar AA, Maurer RR, Anderson LL, 1992: Endocr<strong>in</strong>echanges <strong>in</strong> beef heifers superovulated with follicle-stimulat<strong>in</strong>ghormone (FSH-P) or human menopausal gonadotrop<strong>in</strong>.J Anim Sci 70, 224–231.Baenziger JU, Kumar S, Brodbeck RM, Smith PL, BeranekMC, 1992: Circulatory half-life but not <strong>in</strong>teraction with thelutrop<strong>in</strong> ⁄ chorionic gonadotrop<strong>in</strong> receptor is modulated bysulfation of bov<strong>in</strong>e lutrop<strong>in</strong> oligosaccharides. Proc NatlAcad Sci USA 89, 334–338.Balen AH, Mulders AG, Fauser BC, Schoot BC, RenierMA, Devroey P, Struijs MJ, Mannaerts BM, 2004:Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Recomb<strong>in</strong>ant Gonadotrop<strong>in</strong>s <strong>in</strong> Assisted <strong>Reproduction</strong> 191Pharmacodynamics of a s<strong>in</strong>gle low dose of long-act<strong>in</strong>grecomb<strong>in</strong>ant follicle-stimulat<strong>in</strong>g hormone (FSH-carboxyterm<strong>in</strong>al peptide, corifollitrop<strong>in</strong> alfa) <strong>in</strong> women with WorldHealth Organization group II anovulatory <strong>in</strong>fertility. J Cl<strong>in</strong>Endocr<strong>in</strong>ol Metab 89, 6297–6304.Bassett RM, Driebergen R, 2005: Cont<strong>in</strong>ued improvements <strong>in</strong>the quality and consistency of follitrop<strong>in</strong> alfa, recomb<strong>in</strong>anthuman FSH. Reprod Biomed Onl<strong>in</strong>e 10, 169–177.Bod<strong>in</strong> L, Drion PV, Remy B, Brice G, Cognie Y, Beckers JF,1997: Anti-PMSG antibody levels <strong>in</strong> sheep subjected annuallyto oestrus synchronisation. Reprod Nutr Dev 37, 651–660.Boland MP, Gould<strong>in</strong>g D, Roche JF, 1991: Alternativegonadotroph<strong>in</strong>s for superovulation of cattle. Theriogenology35, 5–17.Bouloux PM, Handelsman DJ, Jockenhovel F, Nieschlag E,Rab<strong>in</strong>ovici J, Frasa WL, de Bie JJ, Voortman G, Itskovitz-Eldor J, 2001: First human exposure to FSH-CTP <strong>in</strong>hypogonadotrophic hypogonadal males. Hum Reprod 16,1592–1597.Bousfield GR, Butnev VY, Butnev VY, 2001: Identification oftwelve O-glycosylation sites <strong>in</strong> equ<strong>in</strong>e chorionic gonadotrop<strong>in</strong>beta and equ<strong>in</strong>e lute<strong>in</strong>iz<strong>in</strong>g hormone ss by solid-phaseEdman degradation. Biol Reprod 64, 136–147.Daya S, 2004: Follicle-stimulat<strong>in</strong>g hormone <strong>in</strong> cl<strong>in</strong>ical practice:an update. Treat Endocr<strong>in</strong>ol 3, 161–171.Devroey P, Van Steirteghem A, Mannaerts B, Coel<strong>in</strong>ghBenn<strong>in</strong>k H, 1992: First s<strong>in</strong>gleton term birth after ovariansuperovulation with rhFSH. Lancet 340, 1108–1109.Driebergen R, Baer G, 2003: Quantification of follicle stimulat<strong>in</strong>ghormone (follitrop<strong>in</strong> alfa): is <strong>in</strong> vivo bioassay stillrelevant <strong>in</strong> the recomb<strong>in</strong>ant age? Curr Med Res Op<strong>in</strong> 19, 41–46.Drion PV, DeRoover R, Houta<strong>in</strong> JY, McNamara EM, RemyB, Sulon J, Beckers JF, 2001: Increase of plasma eCGb<strong>in</strong>d<strong>in</strong>g rate after adm<strong>in</strong>istration of repeated high dose ofeCG to cows. Reprod Nutr Dev 41, 207–215.Fares FA, Suganuma N, Nishimori K, LaPolt PS, Hsueh AJ,Boime I, 1992: Design of a long-act<strong>in</strong>g follitrop<strong>in</strong> agonist byfus<strong>in</strong>g the C-term<strong>in</strong>al sequence of the chorionic gonadotrop<strong>in</strong>beta subunit to the follitrop<strong>in</strong> beta subunit. Proc NatlAcad Sci USA 89, 4304–4308.Fiete D, Srivastava V, H<strong>in</strong>dsgaul O, Baenziger JU, 1991: Ahepatic reticuloendothelial cell receptor specific for SO4-4GalNAc beta 1,4GlcNAc beta 1,2Man alpha that mediatesrapid clearance of lutrop<strong>in</strong>. Cell 67, 1103–1110.Galli C, Duchi R, Crotti G, Tur<strong>in</strong>i P, Ponderato N, Colleoni S,Lagut<strong>in</strong>a I, Lazzari G, 2003: Bov<strong>in</strong>e embryo technologies.Theriogenology 59, 599–616.Garcia-Campayo V, Boime I, 2001: Novel recomb<strong>in</strong>antgonadotrop<strong>in</strong>s. Trends Endocr<strong>in</strong>ol Metab 12, 72–77.Garcia-Campayo V, Sato A, Hirsch B, Sugahara T, Muyan M,Hsueh AJ, Boime I, 1997: Design of stable biologicallyactive recomb<strong>in</strong>ant lutrop<strong>in</strong> analogs. Nat Biotechnol 15,663–667.Garcia-Campayo V, Kumar TR, Boime I, 2002: Thyrotrop<strong>in</strong>,follitrop<strong>in</strong>, and chorionic gonadotrop<strong>in</strong> expressed as a s<strong>in</strong>glemultifunctional unit reveal remarkable permissiveness <strong>in</strong>receptor–ligand <strong>in</strong>teractions. Endocr<strong>in</strong>ology 143, 3773–3778.Garcia-Campayo V, Jablonka-Shariff A, Boime I, 2004: As<strong>in</strong>gle-cha<strong>in</strong> bifunctional gonadotrop<strong>in</strong> analog is secretedfrom Ch<strong>in</strong>ese hamster ovary cells as two dist<strong>in</strong>ct bioactivespecies. J Biol Chem 279, 44286–44293.Germond M, Dessole S, Senn A, Loumaye E, Howles C,Beltrami V, 1992: Successful <strong>in</strong>-vitro fertilisation andembryo transfer after treatment with recomb<strong>in</strong>ant humanFSH. Lancet 339, 1170.Gonzalez A, Wang H, Carruthers TD, Murphy BD, MapletoftRJ, 1994: Superovulation <strong>in</strong> the cow with pregnant mareserum gonadotroph<strong>in</strong>: effects of dose and antipregnant mareserum gonadotroph<strong>in</strong> serum. Can Vet J 35, 158–162.Green ED, Baenziger JU, 1988a: Asparag<strong>in</strong>e-l<strong>in</strong>ked oligosaccharideson lutrop<strong>in</strong>, follitrop<strong>in</strong>, and thyrotrop<strong>in</strong>. I. Structuralelucidation of the sulfated and sialylatedoligosaccharides on bov<strong>in</strong>e, ov<strong>in</strong>e, and human pituitaryglycoprote<strong>in</strong> hormones. J Biol Chem 263, 25–35.Green ED, Baenziger JU, 1988b: Asparag<strong>in</strong>e-l<strong>in</strong>ked oligosaccharideson lutrop<strong>in</strong>, follitrop<strong>in</strong>, and thyrotrop<strong>in</strong>. II. Distributionsof sulfated and sialylated oligosaccharides onbov<strong>in</strong>e, ov<strong>in</strong>e, and human pituitary glycoprote<strong>in</strong> hormones.J Biol Chem 263, 36–44.Hasler JF, 2003: The current status and future of commercialembryo transfer <strong>in</strong> cattle. Anim Reprod Sci 79, 245–264.Hearn MT, Gomme PT, 2000: Molecular architecture andbiorecognition processes of the cyst<strong>in</strong>e knot prote<strong>in</strong> superfamily:part I. The glycoprote<strong>in</strong> hormones. J Mol Recognit13, 223–278.Howles CM, 1996: Genetic eng<strong>in</strong>eer<strong>in</strong>g of human FSH(Gonal-F). Hum Reprod Update 2, 172–191.Inaba T, Mori J, Ohmura M, Kato Y, Tomizawa K, Kato T,Ihara T, Sato I, Ueda S, 1997: Baculovirus-<strong>in</strong>sect cellproduction of bioactive porc<strong>in</strong>e FSH. Theriogenology 47,491–499.Jablonka-Shariff A, Kumar TR, Eklund J, Comstock A,Boime I, 2006: S<strong>in</strong>gle-cha<strong>in</strong>, triple-doma<strong>in</strong> gonadotrop<strong>in</strong>analogs with disulfide bond mutations <strong>in</strong> the alpha-subunitelicit dual follitrop<strong>in</strong> and lutrop<strong>in</strong> activities <strong>in</strong> vivo. MolEndocr<strong>in</strong>ol 20, 1437–1446.Jablonka-Shariff A, Roser JF, Bousfield GR, Wolfe MW,Sibley LE, Colg<strong>in</strong> M, Boime I, 2007: Expression andbioactivity of a s<strong>in</strong>gle cha<strong>in</strong> recomb<strong>in</strong>ant equ<strong>in</strong>e lute<strong>in</strong>iz<strong>in</strong>ghormone (reLH). Theriogenology 67, 311–320.Joshi L, Murata Y, Wondisford FE, Szkudl<strong>in</strong>ski MW, DesaiR, We<strong>in</strong>traub BD, 1995: Recomb<strong>in</strong>ant thyrotrop<strong>in</strong> conta<strong>in</strong><strong>in</strong>ga beta-subunit chimera with the human chorionicgonadotrop<strong>in</strong>-beta carboxy-term<strong>in</strong>us is biologically active,with a prolonged plasma half-life: role of carbohydrate <strong>in</strong>bioactivity and metabolic clearance. Endocr<strong>in</strong>ology 136,3839–3848.Kaetzel DM, Browne JK, Wondisford F, Nett TM, ThomasonAR, Nilson JH, 1985: Expression of biologically activebov<strong>in</strong>e lute<strong>in</strong>iz<strong>in</strong>g hormone <strong>in</strong> Ch<strong>in</strong>ese hamster ovary cells.Proc Natl Acad Sci USA 82, 7280–7283.Kanda M, Jablonka-Shariff A, Sato A, Pixley MR, Bos E,Hiro’oka T, Ben-Menahem D, Boime I, 1999: Geneticfusion of an alpha-subunit gene to the follicle-stimulat<strong>in</strong>ghormone and chorionic gonadotrop<strong>in</strong>-beta subunit genes:production of a bifunctional prote<strong>in</strong>. Mol Endocr<strong>in</strong>ol 13,1873–1881.Kanitz W, Becker F, Schneider F, Kanitz E, Leid<strong>in</strong>g C,Nohner HP, Pohland R, 2002: Superovulation <strong>in</strong> cattle:practical aspects of gonadotrop<strong>in</strong> treatment and <strong>in</strong>sem<strong>in</strong>ation.Reprod Nutr Dev 42, 587–599.Keene JL, Matzuk MM, Otani T, Fauser BC, Galway AB,Hsueh AJ, Boime I, 1989: Expression of biologically activehuman follitrop<strong>in</strong> <strong>in</strong> Ch<strong>in</strong>ese hamster ovary cells. J BiolChem 264, 4769–4775.Kessler MJ, Mise T, Ghai RD, Bahl OP, 1979a: Structureand location of the O-glycosidic carbohydrate units ofhuman chorionic gonadotrop<strong>in</strong>. J Biol Chem 254, 7909–7914.Kessler MJ, Reddy MS, Shah RH, Bahl OP, 1979b: Structuresof N-glycosidic carbohydrate units of human chorionicgonadotrop<strong>in</strong>. J Biol Chem 254, 7901–7908.Kle<strong>in</strong> J, Lobel L, Pollak S, Fer<strong>in</strong> M, Xiao E, Sauer M,Lustbader JW, 2002: Pharmacok<strong>in</strong>etics and pharmacodynamicsof s<strong>in</strong>gle-cha<strong>in</strong> recomb<strong>in</strong>ant human follicle-stimulat<strong>in</strong>ghormone conta<strong>in</strong><strong>in</strong>g the human chorionicÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


192 TE Adams and I Boimegonadotrop<strong>in</strong> carboxyterm<strong>in</strong>al peptide <strong>in</strong> the rhesus monkey.Fertil Steril 77, 1248–1255.Lemke EP, Adams BM, Jablonka-Shariff A, Boime I, AdamsTE, 2008: Us<strong>in</strong>g novel monomeric gonadotrop<strong>in</strong>s withs<strong>in</strong>gular (FSH-like) or dual (LH- and FSH-like) activity to<strong>in</strong>duce follicle development <strong>in</strong> sheep. J Endocr<strong>in</strong>ol 196, 593–600.Low SC, Nunes SL, Bitonti AJ, Dumont JA, 2005: Oral andpulmonary delivery of FSH-Fc fusion prote<strong>in</strong>s via neonatalFc receptor-mediated transcytosis. Hum Reprod 20, 1805–1813.Lunenfeld B, 2004: Historical perspectives <strong>in</strong> gonadotroph<strong>in</strong>therapy. Hum Reprod Update 10, 453–467.Mart<strong>in</strong>uk SD, Mann<strong>in</strong>g AW, Black WD, Murphy BD, 1991:Effects of carbohydrates on the pharmacok<strong>in</strong>etics andbiological activity of equ<strong>in</strong>e chorionic gonadotrop<strong>in</strong> <strong>in</strong> vivo.Biol Reprod 45, 598–604.Matorras R, Rodriguez-Escudero FJ, 2003: Prions, ur<strong>in</strong>arygonadotroph<strong>in</strong>s and recomb<strong>in</strong>ant gonadotroph<strong>in</strong>s. HumReprod 18, 1129–1130.Matzuk MM, Hsueh AJ, Lapolt P, Tsafriri A, Keene JL,Boime I, 1990: The biological role of the carboxyl-term<strong>in</strong>alextension of human chorionic gonadotrop<strong>in</strong> beta-subunit.Endocr<strong>in</strong>ology 126, 376–383.Monniaux D, Chup<strong>in</strong> D, Saumande J, 1983: Superovulatoryresponses of cattle. Theriogenology 19, 55–81.Murphy BD, Mapletoft RJ, Manns J, Humphrey WD, 1984:Variability <strong>in</strong> gonadotroph<strong>in</strong> preparations as a factor <strong>in</strong> thesuperovulatory response. Theriogenology 21, 117–125.Olijve W, de Boer W, Mulders JW, van Wezenbeek PM, 1996:Molecular biology and biochemistry of human recomb<strong>in</strong>antfollicle stimulat<strong>in</strong>g hormone (Puregon). Mol Hum Reprod2, 371–382.Phillips DJ, Hudson NL, Lun S, Condell LA, McNatty KP,1993: Biopotency <strong>in</strong> vitro and metabolic clearance rates offive pituitary preparations of follicle stimulat<strong>in</strong>g hormone.Reprod Fertil Dev 5, 181–190.Pierce JG, Parsons TF, 1981: Glycoprote<strong>in</strong> hormones: structureand function. Annu Rev Biochem 50, 465–495.Reichl H, Balen A, Jansen CA, 2002: Prion transmission <strong>in</strong>blood and ur<strong>in</strong>e: what are the implications for recomb<strong>in</strong>antand ur<strong>in</strong>ary-derived gonadotroph<strong>in</strong>s? Hum Reprod 17,2501–2508.Roseman DS, Baenziger JU, 2000: Molecular basis of lutrop<strong>in</strong>recognition by the mannose ⁄ GalNAc-4-SO4 receptor. ProcNatl Acad Sci USA 97, 9949–9954.Smith PL, Kaetzel D, Nilson J, Baenziger JU, 1990: Thesialylated oligosaccharides of recomb<strong>in</strong>ant bov<strong>in</strong>e lutrop<strong>in</strong>modulate hormone bioactivity. J Biol Chem 265, 874–881.Smith PL, Skelton TP, Fiete D, Dharmesh SM, BeranekMC, MacPhail L, Broze GJ Jr, Baenziger JU, 1992: Theasparag<strong>in</strong>e-l<strong>in</strong>ked oligosaccharides on tissue factor pathway<strong>in</strong>hibitor term<strong>in</strong>ate with SO4-4GalNAc beta 1,4GlcNAc beta 1,2 Mana alpha. J Biol Chem 267,19140–19146.Sugahara T, Pixley MR, M<strong>in</strong>ami S, Perlas E, Ben-MenahemD, Hsueh AJ, Boime I, 1995: Biosynthesis of a biologicallyactive s<strong>in</strong>gle peptide cha<strong>in</strong> conta<strong>in</strong><strong>in</strong>g the human commonalpha and chorionic gonadotrop<strong>in</strong> beta subunits <strong>in</strong> tandem.Proc Natl Acad Sci USA 92, 2041–2045.Sugahara T, Sato A, Kudo M, Ben-Menahem D, Pixley MR,Hsueh AJ, Boime I, 1996: Expression of biologically activefusion genes encod<strong>in</strong>g the common alpha subunit and thefollicle-stimulat<strong>in</strong>g hormone beta subunit. Role of a l<strong>in</strong>kersequence. J Biol Chem 271, 10445–10448.Takagi M, Kim IH, Izadyar F, Hyttel P, Bevers MM,Dieleman SJ, Hendriksen PJ, Vos PL, 2001: Impaired f<strong>in</strong>alfollicular maturation <strong>in</strong> heifers after superovulation withrecomb<strong>in</strong>ant human FSH. <strong>Reproduction</strong> 121, 941–951.Thotakura NR, Blithe DL, 1995: Glycoprote<strong>in</strong> hormones:glycobiology of gonadotroph<strong>in</strong>s, thyrotroph<strong>in</strong> and freealpha subunit. Glycobiology 5, 3–10.Trousdale RK, Pollak SV, Kle<strong>in</strong> J, Lobel L, Funahashi Y,Feirt N, Lustbader JW, 2007: S<strong>in</strong>gle-cha<strong>in</strong> bifunctionalvascular endothelial growth factor (VEGF)-follicle-stimulat<strong>in</strong>ghormone (FSH)-C-term<strong>in</strong>al peptide (CTP) is superiorto the comb<strong>in</strong>ation therapy of recomb<strong>in</strong>ant VEGF plusFSH-CTP <strong>in</strong> stimulat<strong>in</strong>g angiogenesis dur<strong>in</strong>g ovarian folliculogenesis.Endocr<strong>in</strong>ology 148, 1296–1305.Weenen C, Pena JE, Pollak SV, Kle<strong>in</strong> J, Lobel L, TrousdaleRK, Palmer S, Lustbader EG, Ogden RT, Lustbader JW,2004: Long-act<strong>in</strong>g follicle-stimulat<strong>in</strong>g hormone analogsconta<strong>in</strong><strong>in</strong>g N-l<strong>in</strong>ked glycosylation exhibited <strong>in</strong>creased bioactivitycompared with o-l<strong>in</strong>ked analogs <strong>in</strong> female rats. JCl<strong>in</strong> Endocr<strong>in</strong>ol Metab 89, 5204–5212.Wehrman ME, Fike KE, Kojima FN, Bergfeld EG, Cupp AS,Mariscal V, Sanchez T, K<strong>in</strong>der JE, 1996: Development ofpersistent ovarian follicles dur<strong>in</strong>g synchronization of estrus<strong>in</strong>fluences the superovulatory response to FSH treatment <strong>in</strong>cattle. Theriogenology 45, 593–610.Wilson JW, Jones AL, Moore K, Looney CR, Bondioli KR,1993: Superovulation of cattle with a recomb<strong>in</strong>ant-DNAbov<strong>in</strong>e follicle stimulat<strong>in</strong>g hormone. Anim Reprod Sci 33,71–82.Yoon MJ, Boime I, Colg<strong>in</strong> M, Niswender KD, K<strong>in</strong>g SS,Alvarenga M, Jablonka-Shariff A, Pearl CA, Roser JF,2007: The efficacy of a s<strong>in</strong>gle cha<strong>in</strong> recomb<strong>in</strong>ant equ<strong>in</strong>elute<strong>in</strong>iz<strong>in</strong>g hormone (reLH) <strong>in</strong> mares: <strong>in</strong>duction of ovulation,hormone profiles, and <strong>in</strong>ter-ovulatory <strong>in</strong>tervals. DomestAnim Endocr<strong>in</strong>ol 33, 470–479.Author’s address (for correspondence): TE Adams, Department ofAnimal Science, University of California, Davis, CA, USA. E-mail:teadams@ucdavis.eduConflict of <strong>in</strong>terest: TE Adams declares no conflict of <strong>in</strong>terest; B Irv<strong>in</strong>gis a paid consultant for AspenBio Pharma Inc.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 193–199 (2008); doi: 10.1111/j.1439-0531.2008.01161.xISSN 0936-6768Recent Progress <strong>in</strong> Embryonic Stem Cell Research and Its Application <strong>in</strong> <strong>Domestic</strong>SpeciesTAL Brev<strong>in</strong>i, S Anton<strong>in</strong>i, G Pennarossa and F GandolfiDepartment of Animal Science, Centre for Embryonic Stem Cell Research, Laboratory of Biomedical Embryology, University of Milan, ItalyContentsMany reports described cell l<strong>in</strong>es derived <strong>in</strong> domestic species,which presented several important features typical of embryonicstem cells (ESCs). Such features unfortunately did not<strong>in</strong>clude the capacity to generate germ-l<strong>in</strong>e chimeras, thereforelimit<strong>in</strong>g the possibility to use these cells as tools for the geneticmanipulation. However, farm animal ESCs may still be usefulfor the generation of transgenic animals as usually have a selfrenewalcapacity more prolonged than normal primarycultures thus <strong>in</strong>creas<strong>in</strong>g the possibility to transform and selectcells to be used as nucleus donors <strong>in</strong> clon<strong>in</strong>g procedures. Farmanimal ESCs may also be an excellent experimental model <strong>in</strong>pre-cl<strong>in</strong>ical trials, assess<strong>in</strong>g the feasibility of cell therapybecause of the close morphological and physiological resemblanceto humans of species like the pig. However, thepersistent lack of standard methods for the derivation,ma<strong>in</strong>tenance and characterization of ESCs <strong>in</strong> domestic speciesstimulated the search for alternatives. Embryonic germ cellsmay represent such an alternative. Indeed, these cells showed ahigher plasticity than ESCs as contributed to embryonicdevelopment form<strong>in</strong>g chimeric newborns but, as for ESCs,standardization is still far away and efficiency is very low.Recent results <strong>in</strong>dicated spermatogonial stem cells as possibletools for germ-l<strong>in</strong>e genetic modifications with some proof ofpr<strong>in</strong>ciple results already achieved. But, a real break throughcould arrive from the multipotent germ-l<strong>in</strong>e stem cells,virtually equivalent to ESC, derived from newborn and adultmouse testis.IntroductionThe important achievements already reached by embryonicstem cells (ESCs) research have been recentlycelebrated with the Nobel price awarded to the scientistswho derived these cells for the first time and used themto identify the function of genes (Evans and Kaufman1981; Doetschman et al. 1987; Thomas and Capecchi1987). However, this aspect of stem cell research hashardly reached the general public that, on the contrary,is eagerly await<strong>in</strong>g for ESCs to fulfil the next bigachievement: to provide an unlimited source of replacementmaterial for the therapy of <strong>in</strong>jured, degenerated orotherwise damaged tissues. It is difficult to tell, atpresent, if this ambitious goal will ever be reached byESCs but the simple hope has fuelled a great deal ofresearch efforts as well as of hot debate.Such efforts have <strong>in</strong>volved domestic species from theearly days with the first reports on putative ESCs <strong>in</strong>sheep, pig and cow be<strong>in</strong>g published <strong>in</strong> 1990 (Evans et al.1990; Piedrahita et al. 1990a,b).As for the other species, also <strong>in</strong> domestic animals, thefocus of the research has shifted dur<strong>in</strong>g the years.Initially, the ma<strong>in</strong> aim of stem cell research was toimprove transgenesis efficiency and control of transgeneexpression (Seamark 1994). The vision was to obta<strong>in</strong> anabundance of totipotent ESC to be transformed accord<strong>in</strong>gto plan and to take advantage of the variety ofprocedures available to ensure that those cells could beidentified, cloned and ma<strong>in</strong>ta<strong>in</strong>ed as isogenetic cell l<strong>in</strong>es.The publication, of evidence that pluripotent pig ESCl<strong>in</strong>es were able to form chimeras, a few years from thefirst reports <strong>in</strong> mice, (Wheeler 1994; Gerfen and Wheeler1995; Chen et al. 1999) susta<strong>in</strong>ed for a while this visionbut, to date, it has not been possible to demonstrate the<strong>in</strong>tegration of embryo-derived cell l<strong>in</strong>es <strong>in</strong>to the germl<strong>in</strong>e either <strong>in</strong> pig or <strong>in</strong> any other domestic species (Keeferet al. 2007). Indeed, it has not been possible even toobta<strong>in</strong> cell l<strong>in</strong>es able to consistently and significantlycontribute to embryo development upon <strong>in</strong>jection <strong>in</strong>toblastocyst <strong>in</strong> farm animals <strong>in</strong>clud<strong>in</strong>g pig, with theexception of one report <strong>in</strong> rabbit (Schoonjans et al.1996).In the absence of cell l<strong>in</strong>es capable to generate germl<strong>in</strong>echimeras, the hope to use embryo-derived cell l<strong>in</strong>esas a tool to improve transgenesis <strong>in</strong> domestic animalsshifted towards the use of these cells as nuclear donors<strong>in</strong> nuclear transfer procedures. This pathway immediatelyproved to be much more effective with the birth ofthe first lamb obta<strong>in</strong>ed from an embryonic cell l<strong>in</strong>e(Campbell et al. 1996). However, the follow<strong>in</strong>g birth ofDolly (Wilmut et al. 1997) suggested that <strong>in</strong> order tomanipulate farm animal germ l<strong>in</strong>e, ESCs were notnecessary. In fact, the formal demonstration of transgenicsheep generated through the transfer of transformedsomatic cells followed <strong>in</strong> a short <strong>in</strong>terval of time(Schnieke et al. 1997) and was soon applied to otherspecies (Cibelli et al. 1998).This diverted the <strong>in</strong>terest from farm animal ESCs fora while. However, recent data <strong>in</strong> the mouse <strong>in</strong>dicat<strong>in</strong>gthat, <strong>in</strong> this species, ESCs are more amenable toreprogramm<strong>in</strong>g through nuclear transfer than theirsomatic counterparts (Hochedl<strong>in</strong>ger and Jaenisch2006), may <strong>in</strong>duce a renew <strong>in</strong>terest <strong>in</strong> deriv<strong>in</strong>g stableESCs as nuclear donors. Even if this phenomenon maynot be true for other species or for all mouse stra<strong>in</strong>s(Oback and Wells 2007), the ability of putative ESCs toproliferate <strong>in</strong> vitro without undergo<strong>in</strong>g through senescence,may represent per se an important potentialadvantage <strong>in</strong> comparison to primary cultures of somaticcells. A prolonged <strong>in</strong> vitro life span, if fact, represents adist<strong>in</strong>ct advantage when genetic modifications need tobe <strong>in</strong>serted <strong>in</strong>to cell l<strong>in</strong>es as these procedure are usuallyfollowed by extensive selective processes that, <strong>in</strong> turn,require several cell replications. Therefore, it is importantthat the transformed cell l<strong>in</strong>e rema<strong>in</strong>s viable andÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


194 TAL Brev<strong>in</strong>i, S Anton<strong>in</strong>i, G Pennarossa and F Gandolfikaryotypically stable for as long as possible <strong>in</strong> order toallow its use as a source of transgenic nuclei.The derivation of ESCs from human embryos(Thomson et al. 1998) is another major reason for therecent renewed <strong>in</strong>terest <strong>in</strong> farm animal ESCs. Thisimportant result brought the full realization of thetherapeutic potential of stem cells and made it clear thatthese cells are perfect candidates for regenerative medic<strong>in</strong>e,tissue repair and gene therapy. However, it wassoon clear also that, before rout<strong>in</strong>ely apply<strong>in</strong>g ESCs as atherapeutic tool several issues need to be addressed.Reliable ESCs differentiation protocols are needed,enabl<strong>in</strong>g the isolation of highly purified cell l<strong>in</strong>eages tobe used <strong>in</strong> transplantation and <strong>in</strong> comb<strong>in</strong>ation withways to circumvent the immunological rejection oftransplanted cells. Ultimately, the safety of us<strong>in</strong>g thesecells <strong>in</strong> human patients has to be ascerta<strong>in</strong>ed. To achievethis, <strong>in</strong> vitro tests are be<strong>in</strong>g performed <strong>in</strong> both mouseand human cell l<strong>in</strong>es, while <strong>in</strong> vivo experiments are,understandably, limited to the mouse. However, markedmorphological and physiological differences, a reducedgenetic variability and a short life-span make the mousemodel <strong>in</strong> many respect rather unsatisfactory whenexperimental outcomes need to be extrapolated tohuman cl<strong>in</strong>ical applications. To this purpose, outbredlarge animal models will be <strong>in</strong>creas<strong>in</strong>gly <strong>in</strong>strumental <strong>in</strong>safety and efficiency assessment of ESC-based therapeuticmethodologies.What Is an Embryonic Stem Cell?At this po<strong>in</strong>t we must clarify that a formally correctdef<strong>in</strong>ition of embryonic stem cells implies the satisfactionof several criteria <strong>in</strong>clud<strong>in</strong>g: derivation withouttransformation or immortalization; stable diploidkaryotype, to be clonogenic, unlimited self-renewalcapacity, ability to generate all foetal and adult celltypes <strong>in</strong> vitro and <strong>in</strong> teratomas, <strong>in</strong>corporation <strong>in</strong>toembryonic development and contribution to all germlayers <strong>in</strong> chimera, germ-l<strong>in</strong>e colonization and transmission(Smith 2001). Any cell l<strong>in</strong>e which fails to satisfy allthese requirements should be def<strong>in</strong>ed as stem cell-like orother dubitative descriptions.This cautionary nomenclature has been followed formany years when deal<strong>in</strong>g with domestic animals celll<strong>in</strong>es of embryonic orig<strong>in</strong> (Galli et al. 1994; Prelle et al.1999). However, the def<strong>in</strong>ition of ESC has changedupon the isolation of human embryonic cell l<strong>in</strong>es(Thomson et al. 1998) and it has recently been proposedas cells that can extensively self-renew <strong>in</strong> vitro, ma<strong>in</strong>ta<strong>in</strong>a normal karyotype, can differentiate <strong>in</strong>to derivatives ofall three germ layers, express Oct-4 and a panel of otherpluripotency genes, show telomerase activity (Hoffmanand Carpenter 2005).If this or similar def<strong>in</strong>itions are taken <strong>in</strong>to consideration,it is clear that many of the embryonic cell l<strong>in</strong>esderived from mammalian embryos, different frommouse, can qualify as ESCs. This is not only a matterof appropriate nomenclature, but also support theconcept that large animal ESCs may be a useful,mean<strong>in</strong>gful experimental and pre-cl<strong>in</strong>ical model forhuman ESCs (hESCs) and for their therapeutic applications.Are ‘True’ ESCs Unique to the Mouse?The genetic backgroundEmbryonic stem cells are most commonly derived fromthe epiblast, a specific cell subpopulation of the <strong>in</strong>ner cellmass of the blastocyst (Ralston and Rossant 2005). It iswell known that, at least <strong>in</strong> the mouse, ESCs can be re<strong>in</strong>troduced<strong>in</strong>to the Inner Cell Mass (ICM) re-enter<strong>in</strong>gthe process of embryonic development (Bradley et al.1984). However, the two cell types, ESCs and epiblast,are not equivalent, because the epiblast exists onlytransiently <strong>in</strong> the embryo and does not act as a stem cellcompartment <strong>in</strong> vivo whereas ESCs form a stable cell l<strong>in</strong>e<strong>in</strong> vitro. Therefore, ESCs are the result of a selection andadaptation process to the culture environment and, assuch, could be considered more an artefact than aphysiological cell type (Chambers and Smith 2004). Ifthis is the case, it has been suggested that the significantdifferences observed between species <strong>in</strong> their capacity toorig<strong>in</strong>ate pluripotent cell l<strong>in</strong>es from early embryos maydepend ma<strong>in</strong>ly on their ICM ability to adapt to anarbitrary set of artificial conditions (Smith 2001). Indeed,the standard protocols for deriv<strong>in</strong>g mouse ESCs(mESCs) work efficiently only with 129 and, to a lowerextent, C57BL ⁄ 6 stra<strong>in</strong>s (Robertson 1987) whereas thesame procedures are not equally successful with blastocystof different genetic background (Kawase et al.1994), confirm<strong>in</strong>g that there is a strong genetic componentto ESC derivation (Smith 2001). S<strong>in</strong>ce geneticbackground has such a strong <strong>in</strong>fluence on the <strong>in</strong>itialfrequency of establish<strong>in</strong>g ESCs and on their subsequentstability <strong>in</strong> culture, the differences observed betweenmESC and those derived <strong>in</strong> large animal species,<strong>in</strong>clud<strong>in</strong>g primates, may be caused by the impact ofgenetic heterogeneity typical of these species and which isvirtually absent <strong>in</strong> <strong>in</strong>bred mouse stra<strong>in</strong>s.Pre-implantation developmentDetailed studies <strong>in</strong> mouse embryos determ<strong>in</strong>ed that thefirst differentiation process occurs at the late morulastage when the outer cells adopt an epithelial structure.This event is followed by the first appearance of theblastocoel that marks the divergence of the first twol<strong>in</strong>eages: trophectoderm and <strong>in</strong>ner cell mass. Uponblastocyst expansion differentiation cont<strong>in</strong>ues with theICM form<strong>in</strong>g two further cell l<strong>in</strong>eages: the epiblast andthe primitive endoderm or hypoblast. Between days 3.5and 4.5, the epiblast will give rise to the embryo itselfand the hypoblast will evolve <strong>in</strong> the extra embryonicendoderm and later will contribute to the yolk sac. Inother species, these events take place over a longerperiod of time. Although the three early embryonicl<strong>in</strong>eages are present <strong>in</strong> all eutherian mammals, the timebetween their formation and fertilization is substantiallyshorter <strong>in</strong> mouse and human than <strong>in</strong> domestic ungulates.Detailed studies <strong>in</strong> mice have established thatESCs are derivatives of the epiblast (Brook and Gardner1997). In humans, blastocyst formation of the threeearly l<strong>in</strong>eages takes approximately 6 days (Dvash andBenvenisty 2004), whereas, for <strong>in</strong>stance, <strong>in</strong> pig andbov<strong>in</strong>e embryos epiblast formation beg<strong>in</strong>s at hatch<strong>in</strong>gand is complete towards day 12 (Vejlsted et al. 2005,Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Farm <strong>Animals</strong> Embryonic Stem Cells 1952006). In practical terms, this translates <strong>in</strong> no def<strong>in</strong>edepiblast be<strong>in</strong>g present <strong>in</strong> these species before hatch<strong>in</strong>gwhich <strong>in</strong> vivo occurs on late day 6 or on day 7 ofdevelopment <strong>in</strong> pig and days 8–9 <strong>in</strong> cow (Hunter 1974;Renard and Heyman 1979; Betteridge and Flechon1988). At this stage the ICM is present, def<strong>in</strong>ed as thecells comprised between the Rauber’s layer, separat<strong>in</strong>g itfrom the uter<strong>in</strong>e lumen, and the visceral endoderm.Which is to say that, <strong>in</strong> order to obta<strong>in</strong> <strong>in</strong> vivo producedpig and cow blastocysts equivalent to their mur<strong>in</strong>ecounterparts, the uterus must be flushed on days 7–8and 8–9, respectively, when embryos are completelyhatched and expanded (Betteridge and Flechon 1988;Vejlsted et al. 2006). Upon hatch<strong>in</strong>g, pig blastocystsreta<strong>in</strong> a round shape and dur<strong>in</strong>g days 8 and 9, thehypoblast is formed from the ICM and graduallyproliferates as a confluent cell layer surround<strong>in</strong>g theblastocoel cavity. At the same time and through day 10<strong>in</strong> pig and day 12 <strong>in</strong> cow, the polar trophectoderm thatcovers the epiblast, referred to as the Rauber’s layer,beg<strong>in</strong>s to degenerate until is completely lost leav<strong>in</strong>g theepiblast directly exposed to the uter<strong>in</strong>e lumen (Maddox-Hyttel et al. 2003; Flechon et al. 2004). The area wherethe epiblast has surfaced is spherical, has a whitishcolour under the stereomicroscope and is referred to asthe embryonic disc whose <strong>in</strong>ternal surface is covered bycuboidal hypoblast cells. Between days 11 and 13,development cont<strong>in</strong>ues with the embryonic disk and thewhole conceptus beg<strong>in</strong>s to assume an ovoid shape. Atthis stage, the first signs of polarity become visible alsoat the stereo microscope <strong>in</strong> the form of a crescentshapedthicken<strong>in</strong>g on the posterior third (Vejlsted et al.2006). This thicken<strong>in</strong>g will orig<strong>in</strong>ate, with<strong>in</strong> a couple ofdays, the primitive streak which accompanies theappearance of def<strong>in</strong>ed mesoderm and endoderm layers.This differs from the mouse where mesoderm andendoderm differentiation follows rather than precedesthe primitive streak formation (Tam and Behr<strong>in</strong>ger1997). Around days 13 and 14 while the primitive streakis still clearly visible, the major part of the epiblasttransforms <strong>in</strong>to neural ectoderm and forms the neuralplate. This corresponds to a gradual down regulation oftypical pluripotency maker OCT4 which is substitutedby b-tubul<strong>in</strong> III expression, a marker of neural differentiation.Therefore, it can be assumed that embryos atthis stage are no longer suitable for ESC derivation. Insummary, the extended pre-implantation period togetherwith the formation of an embryonic disk makesungulate embryo epiblast available for a much longertime than <strong>in</strong> rodents and primates. As a consequence,the ICM available <strong>in</strong> the pre-hatch<strong>in</strong>g blastocyst maynot be exactly equivalent to the mouse epiblast usuallyrecovered for ESC derivation.However, it must be noted that, <strong>in</strong> pig and cow, a bigvariation <strong>in</strong> size is observed <strong>in</strong> embryos collected at thesame time and embryos of the same size showsubstantial differences <strong>in</strong> the development of theembryo proper. This means that, when post-hatch<strong>in</strong>gembryos are used for ESC derivation, the simple<strong>in</strong>dication of the day of collection may comprise awide range of development. This wide variation mayexpla<strong>in</strong> why a recent survey of the literature hasshowed no obvious effect of embryo age on theefficiency of ESC l<strong>in</strong>e derivation, at least <strong>in</strong> pig (Brev<strong>in</strong>iet al. 2007b).Although derivation efficiency was not affected by theage of the embryo, the quality of the cell l<strong>in</strong>es mighthave been different as suggested by some recent f<strong>in</strong>d<strong>in</strong>gs<strong>in</strong> mouse. Two <strong>in</strong>dependent groups, <strong>in</strong> fact, showed thatmESC derived from early post-implantation embryoscorrespond more closely to hESC rather than mESC, <strong>in</strong>all aspects tested so far (Brons et al. 2007; Tesar et al.2007) <strong>in</strong>clud<strong>in</strong>g be<strong>in</strong>g leukemia <strong>in</strong>hibitory factor (LIF)and bone morphogenetic prote<strong>in</strong>-4 (BMP4) <strong>in</strong>dependentbut requir<strong>in</strong>g activ<strong>in</strong> and FGF for efficient self-renewal.Interest<strong>in</strong>gly, these cell l<strong>in</strong>es, named epiblast stem cells(EpiSCs) were unable to form chimeras (Tesar et al.2007) or did so at a very low efficiency (Brons et al.2007) closely resembl<strong>in</strong>g, <strong>in</strong> this respect, to domesticanimal cell l<strong>in</strong>es. Moreover, EpiSCs did not showdependency from a specific genetic background similarlyto human and farm animal cell l<strong>in</strong>es.Therefore, differences <strong>in</strong> pre-attachment developmentand tim<strong>in</strong>g of isolation may be another factor added togenetic variation expla<strong>in</strong><strong>in</strong>g species-specific differences<strong>in</strong> ESC derivation efficiency and pluripotency.It has been speculated that cells able to give rise to‘true’ ESCs are overgrown by more rapidly divid<strong>in</strong>gslightly later cells (Lovell-Badge 2007) and this wouldexpla<strong>in</strong> the differences between those few mouse stra<strong>in</strong>swere ESC derivation is possible and all other species<strong>in</strong>clud<strong>in</strong>g human.How Different Are ESCs from DifferentSpecies?Recent evidence, based on the comparison between thetrascriptomes of mESC and hESC, leads to the conclusionthat mESCs are substantially different from humancell l<strong>in</strong>es. In fact, comb<strong>in</strong><strong>in</strong>g together the results ofdifferent studies, it appears that only 13–55% oftranscripts enriched <strong>in</strong> mESCs are also enriched <strong>in</strong>human l<strong>in</strong>es but when comparison is performed amongdifferent hESC l<strong>in</strong>es the overlap raise to 85–99% (seeEckfeldt et al. 2005; for detailed references). Thesubstantial differences determ<strong>in</strong>ed between hESC andmESC expression profiles are followed by some morphologicaland functional differences between the celll<strong>in</strong>es of the two species (Laslett et al. 2003; G<strong>in</strong>is et al.2004; Rao and Zandstra 2005). For <strong>in</strong>stance, hESCstypically grow <strong>in</strong> tightly adherent, flattened groupsrather than <strong>in</strong> rounded clumps and tolerate physicalseparation very poorly. ESCs of both typically grow well<strong>in</strong> presence of mouse embryonic fibroblasts; hESCs donot require the presence of LIF to activate JAK-STAT3transcription factors <strong>in</strong> order to ma<strong>in</strong>ta<strong>in</strong> their pluripotency<strong>in</strong> culture. ESCs of both the species can bema<strong>in</strong>ta<strong>in</strong>ed <strong>in</strong> culture without a feeder layer, but <strong>in</strong> thiscondition hESCs require the presence of fibroblastgrowth factor-2 and activ<strong>in</strong>, whereas mESCs need LIFand bone morphogenetic prote<strong>in</strong>-4 (BMP4). In contrast,BMP4 <strong>in</strong>duces trophoblast differentiation <strong>in</strong> hESCs(Ludwig et al. 2006; Brons et al. 2007; Tesar et al.2007). Furthermore, although they express the same setof factors known to be required for pluripotency <strong>in</strong>mouse ESCs (such as OCT4, SOX2 and NANOG),Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


196 TAL Brev<strong>in</strong>i, S Anton<strong>in</strong>i, G Pennarossa and F Gandolfisome other markers are not shared (Brivanlou et al.2003).Data currently available on ungulate ESC characteristics,morphology and properties are def<strong>in</strong>itively morelimited than those available for mESC or hESC, but it isalready possible to del<strong>in</strong>eate some common tracts aswell as some differences.For <strong>in</strong>stance, classic hESC and mESC markers suchas OCT4, SSEA1, SSEA4 and alkal<strong>in</strong>e phosphatase are<strong>in</strong>deed expressed by ungulates ICM and embryo-derivedcell l<strong>in</strong>es; however, the same genes are also expressed <strong>in</strong>the trophectoderm and endoderm (van Eijk et al. 1999;Kirchhof et al. 2000; Talbot et al. 2002; He et al. 2006).NANOG, another well-characterized ESC marker <strong>in</strong>human and mouse cell l<strong>in</strong>es looks more reliable s<strong>in</strong>ce ithas been shown to be expressed <strong>in</strong> pig ESC-like cell l<strong>in</strong>es(Brev<strong>in</strong>i et al. 2007a; b) and to be strongly downregulated<strong>in</strong> capr<strong>in</strong>e trophectoderm while be<strong>in</strong>g stronglyexpressed <strong>in</strong> the ICM (He et al. 2006).Spontaneous differentiation of embryo-derived celll<strong>in</strong>es is a common problem when work<strong>in</strong>g with ungulatecells even <strong>in</strong> the presence of a STO feeder layer or ofother cell types (Brev<strong>in</strong>i et al. 2007a; b; Keefer et al.2007). The addition of cytok<strong>in</strong>es and growth factors,such as EGF, LIF, SCF and FGF, that <strong>in</strong>hibit spontaneousdifferentiation of hESC and mESC have beenreported to be <strong>in</strong>effective <strong>in</strong> other species (Talbot et al.1993, 1995; Moore and Piedrahita 1997). These additionsto the culture medium are done without evidenceof the presence on ungulate ESCs of the relatedreceptors and are ma<strong>in</strong>ly based on previous experience<strong>in</strong> the mur<strong>in</strong>e species. In prelim<strong>in</strong>ary studies, we havefound that pig cell l<strong>in</strong>es do not express LIF receptor<strong>in</strong>dicat<strong>in</strong>g that the addition of this cytok<strong>in</strong>e to theculture medium is not essential for the ma<strong>in</strong>tenance ofpluripotency. However, <strong>in</strong> our experience, the presenceof LIF <strong>in</strong> the culture medium seems to <strong>in</strong>hibit thedifferentiation process because it prevented embryoidbodies formation (Brev<strong>in</strong>i et al. 2007a). On the contrary,prelim<strong>in</strong>ary <strong>in</strong>formation suggest that LIF receptor andgp130 are both expressed <strong>in</strong> bov<strong>in</strong>e ICM and prelim<strong>in</strong>aryoutgrowth (Pant and Keefer 2006).Although the fundamental properties of ESC arecommon to the different species, overall, these observationssuggest that several details are species-specific.Alternatives to ESCsEmbryonic germ cellsGiven the current limitations affect<strong>in</strong>g the derivationand ma<strong>in</strong>tenance of large animal ESCs, it is worth<strong>in</strong>vestigat<strong>in</strong>g the availability of alternative sources ofcells with similar properties.The most obvious candidates are embryonic germcells (EGCs), orig<strong>in</strong>ally derived from the primordialgerm cells isolated from 8.5 dpc mouse embryos(Matsui et al. 1992), these cells are <strong>in</strong> most aspects<strong>in</strong>dist<strong>in</strong>guishable from ESCs <strong>in</strong>clud<strong>in</strong>g the capability toform chimeras and to give germ-l<strong>in</strong>e transmission(Smith 2001), although at a lower rate than ESCs.The capacity of EGCs to erase impr<strong>in</strong>ts is the ma<strong>in</strong>difference so far described between EGCs and ESCs(Tada et al. 1997). The derivation of EGCs fromhuman embryos (Shamblott et al. 1998) has ignited aconsiderable <strong>in</strong>terest <strong>in</strong> this type of cells as it happenedwith ESCs.Primordial germ cell-derived cell l<strong>in</strong>es have beenderived from rabbit, pig, cow and sheep, although withdifferent plasticity depend<strong>in</strong>g on the species (reviewed byPrelle et al. 1999). The best results, so far, have beenobta<strong>in</strong>ed with pig EGCs which were able to contributeto liveborn chimeric offspr<strong>in</strong>gs both normal (Shim et al.1997) and after transgenic transformation (Piedrahitaet al. 1998; Mueller et al. 1999). However, these <strong>in</strong>itialencourag<strong>in</strong>g results have not been susta<strong>in</strong>ed by furtherprogress and, at present, the efficiency of chimerageneration rema<strong>in</strong>s low with a weak contribution ofthe EGC to the newborn piglets (Rui et al. 2004). Inaddition, as for ESC, no germ-l<strong>in</strong>e transmission has everbeen described.The use of EGCs as nuclear donors has been tested <strong>in</strong>cattle with moderate success (Zakhartchenko et al.1999); however, current data available <strong>in</strong> the literaturedo not allow to evaluate whether domestic animalEGCs have the characteristics of <strong>in</strong>def<strong>in</strong>ite self-renewal<strong>in</strong> vitro that would be useful for us<strong>in</strong>g them as avaluable source of karyoplasts as mentioned previously.Pig EGC l<strong>in</strong>es are capable of differentiat<strong>in</strong>g <strong>in</strong>to a widerange of cell types <strong>in</strong> culture, <strong>in</strong>clud<strong>in</strong>g endodermal,trophoblast-like, epithelial-like, fibroblast-like and neuron-likecells. Moreover, when cultured <strong>in</strong> suspension,these cells formed embryoid bodies (Shim and Anderson1998). Benign teratomas were obta<strong>in</strong>ed after transplantationof days 34 and 37, bov<strong>in</strong>e genital ridges under thekidney capsule of athymic mice (Choi and Anderson1998).The characterization of farm animal EGCs rema<strong>in</strong>smore superficial compared with that of ESCs, but theavailable data suggest that EGCs show a better plasticityand a comparable ability of self-renewal, thereforeencourag<strong>in</strong>g further research with these cells that mayrepresent a useful tool both for genetic manipulationand as a model for cell therapies.Spermatogonial stem cellsAnother alternative is represented by the spermatogonialstem cells (SSCs) found <strong>in</strong> the testis which have theunique potential for both self-renewal and production ofdifferentiated daughter cells which will ultimately formspermatozoa (for an excellent review, see Dobr<strong>in</strong>ski2005).It has been demonstrated that it is possible to fullyrestore male fertility by the transplantation of spermatogonialcells from fertile donor mice to the testes of<strong>in</strong>fertile recipient mice and this worked also betweenspecies when rat SSCs were transplanted <strong>in</strong>to mice testis(reviewed by McLaren 1998). Unfortunately, this onlyworks with<strong>in</strong> limited phylogenetic distance betweendonor and recipient species, s<strong>in</strong>ce transplantation <strong>in</strong>the mouse testis of germ cells from non-rodent donorsrang<strong>in</strong>g from rabbits, dogs, pigs, cattle, horses and<strong>in</strong>clud<strong>in</strong>g non-human primates and humans, resulted <strong>in</strong>colonization of the mouse testis but spermatogenesisbecame arrested at the stage of spermatogonial expansion(Dobr<strong>in</strong>ski 2005). However, recent data suggestÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Farm <strong>Animals</strong> Embryonic Stem Cells 197that it may be possible to circumvent this limitation byco-transplant<strong>in</strong>g SSCs together with the support<strong>in</strong>gSertoli cells (Sh<strong>in</strong>ohara et al. 2003).Although the ambitious goal to obta<strong>in</strong> mature spermof one species <strong>in</strong> the testis of a different one has not beenreached <strong>in</strong> domestic species, transplants of SSCsbetween different <strong>in</strong>dividuals of the same species hasbeen successful <strong>in</strong> pigs and goats (Honaramooz et al.2002, 2003a; b). Through this <strong>in</strong>trigu<strong>in</strong>g technique, ithas even been possible to transplant SSCs obta<strong>in</strong>ed froma transgenic buck and to transmit to the progeny thetransgene through the sperm ejaculated by a normalbuck who received the transgenic SSCs (Honaramoozet al. 2003b). These data support the vision that SSCscan represent the ideal way to propagate transgenesthrough the sperm, mak<strong>in</strong>g the ideal substitute of ESCsfor this purpose.Multipotent germ stem cellsIndeed, this could be just the beg<strong>in</strong>n<strong>in</strong>g of a moreextensive use of SSCs as suggested by a recent f<strong>in</strong>d<strong>in</strong>gobta<strong>in</strong>ed dur<strong>in</strong>g the prolonged culture of these cells<strong>in</strong> vitro which was orig<strong>in</strong>ally aimed at their transformation.It has been noticed that among mouse post-natalSSCs cultured <strong>in</strong> vitro for an extended period of time,ESC-like cells appeared upon treat<strong>in</strong>g the culture with amixture of cytok<strong>in</strong>es and growth factors (Kanatsu-Sh<strong>in</strong>ohara et al. 2004). These cells have been namedmultipotent germ stem cells (mGSCs) as they arecompletely different from SSCs. In fact, whereas SSCscan give rise only to spermatozoa, mGSCs show all theproperties typical of mESCs <strong>in</strong>clud<strong>in</strong>g pluripotency andthe ability to generate germ-l<strong>in</strong>e chimeras (Sh<strong>in</strong>oharaand Kanatsu-Sh<strong>in</strong>ohara 2007).Recently, the isolation of mGSCs has been describedalso from adult mouse testis (Guan et al. 2006) furtherenhanc<strong>in</strong>g the potential utility of these cells as a viablealternative to ESCs both as a way to genetically modifythe germ l<strong>in</strong>e and a source of undifferentiated cells fortherapeutic purposes.Extensive efforts have been spent <strong>in</strong> the attempt toobta<strong>in</strong> similar cell l<strong>in</strong>es <strong>in</strong> other species and <strong>in</strong> human <strong>in</strong>particular, but, at present, no positive results have beenannounced. Work on farm animals has lead to theidentification of germ cell-specific markers <strong>in</strong> sheep(Rodriguez-Sosa et al. 2006) and pig (Luo et al. 2006;Goel et al. 2007) which should be <strong>in</strong>strumental <strong>in</strong>reach<strong>in</strong>g an highly purified cell population.ConclusionsThe long quest for farm animal ESCs has not lead yet tothe derivation of a bona fide ESC l<strong>in</strong>e, conv<strong>in</strong>c<strong>in</strong>glydisplay<strong>in</strong>g all the properties of analogous cells <strong>in</strong> mouse.Many reports have described cell l<strong>in</strong>es <strong>in</strong> domesticspecies, which presented several important featurestypical of ESCs. Such features may be sufficient <strong>in</strong> orderto use these cells as tools for the genetic manipulation astheir self-renewal may be more prolonged than that ofnormal primary cultures, thus enabl<strong>in</strong>g the possibility totransform and select the cells to be used as nucleusdonors <strong>in</strong> cell transfer experiments. An alternative use offarm animal ESCs is as an excellent experimental model<strong>in</strong> pre-cl<strong>in</strong>ical trial assess<strong>in</strong>g the feasibility of cell therapybecause of the closer morphological and physiologicalresemblance to humans of species like the pig whencompared with the mouse.However, the persistent lack of standard methods forthe derivation, ma<strong>in</strong>tenance and characterization ofESCs <strong>in</strong> domestic species stimulates the search foralternatives.Embryonic germ cells may represent such an alternative.Indeed, these cells showed a higher plasticity thanESCs as they were able to contribute to embryonicdevelopment form<strong>in</strong>g chimeric newborns. However, asfor ESCs, standardization is still far away and efficiencyis very low.Recent results <strong>in</strong>dicated SSCs as possible tools forgerm-l<strong>in</strong>e genetic modifications with some proof ofpr<strong>in</strong>ciple results already achieved. But, a real breakthough could arise from the isolation of mGSCs,virtually equivalent to ESCs, from newborn and adultmouse testis. Unfortunately, at present, we do not knowwhether such cells are, once aga<strong>in</strong>, only typical of themouse with the other species only hav<strong>in</strong>g a pale copy ofthe orig<strong>in</strong>al.AcknowledgementsThis work is supported by MUSRT PRIN 2005, 2006, FIRST 2005,2006, 2007.ReferencesBetteridge KJ, Flechon J-E, 1988: The anatomy and physiologyof pre-attachement bov<strong>in</strong>e embryos. Theriogenology 29,155–187.Bradley A, Evans M, Kaufman MH, Robertson E, 1984:Formation of germ-l<strong>in</strong>e chimaeras from embryo-derivedteratocarc<strong>in</strong>oma cell l<strong>in</strong>es. Nature 309, 255–256.Brev<strong>in</strong>i T, Anton<strong>in</strong>i S, Cillo F, Crestan M, Gandolfi F, 2007a:Porc<strong>in</strong>e embryonic stem cells: facts, challenges and hopes.Theriogenology 68S, S206–S213.Brev<strong>in</strong>i TAL, Tosetti V, Crestan M, Anton<strong>in</strong>i S, Gandolfi F,2007b: Derivation and characterization of pluripotent celll<strong>in</strong>es from pig embryos of different orig<strong>in</strong>s. Theriogenology67, 54–63.Brivanlou AH, Gage FH, Jaenisch R, Jessell T, Melton D,Rossant J, 2003: Stem cells. Sett<strong>in</strong>g standards for humanembryonic stem cells. Science 300, 913–916.Brons IG, Smithers LE, Trotter MW, Rugg-Gunn P, Sun B,Chuva de Sousa Lopes SM, Howlett SK, Clarkson A,Ahrlund-Richter L, Pedersen RA, Vallier L, 2007: Derivationof pluripotent epiblast stem cells from mammalianembryos. Nature 448, 191–195.Brook FA, Gardner RL, 1997: The orig<strong>in</strong> and efficientderivation of embryonic stem cells <strong>in</strong> the mouse. Proc NatlAcad Sci U S A 94, 5709–5712.Campbell KH, McWhir J, Ritchie WA, Wilmut I, 1996: Sheepcloned by nuclear transfer from a cultured cell l<strong>in</strong>e. Nature380, 64–66.Chambers I, Smith A, 2004: Self-renewal of teratocarc<strong>in</strong>omaand embryonic stem cells. Oncogene 23, 7150–7160.Chen LR, Shiue YL, Bertol<strong>in</strong>i L, Medrano JF, BonDurant RH,Anderson GB, 1999: Establishment of pluripotent cell l<strong>in</strong>esfrom porc<strong>in</strong>e preimplantation embryos. Theriogenology 52,195–212.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


198 TAL Brev<strong>in</strong>i, S Anton<strong>in</strong>i, G Pennarossa and F GandolfiChoi SJ, Anderson GB, 1998: Development of tumors frombov<strong>in</strong>e primordial germ cells transplanted to athymic mice.Anim Reprod Sci 52, 17–25.Cibelli JB, Stice SL, Golueke PJ, Kane JJ, Jerry J, Blackwell C,Ponce de Leon FA, Robl JM, 1998: Cloned transgeniccalves produced from nonquiescent fetal fibroblasts. Science280, 1256–1258.Dobr<strong>in</strong>ski I, 2005: Germ cell transplantation and testis tissuexenograft<strong>in</strong>g <strong>in</strong> domestic animals. Anim Reprod Sci 89, 137–145.Doetschman T, Gregg RG, Maeda N, Hooper ML, MeltonDW, Thompson S, Smithies O, 1987: Targeted correction ofa mutant HPRT gene <strong>in</strong> mouse embryonic stem cells. Nature330, 576–578.Dvash T, Benvenisty N, 2004: Human embryonic stem cells asa model for early human development. Best Pract Res Cl<strong>in</strong>Obstet Gynaecol 18, 929–940.Eckfeldt CE, Mendenhall EM, Verfaillie CM, 2005: Themolecular repertoire of the ‘almighty’ stem cell. Nat RevMol Cell Biol 6, 726–737.van Eijk MJ, van Rooijen MA, Mod<strong>in</strong>a S, Scesi L, Folkers G,van Tol HT, Bevers MM, Fisher SR, Lew<strong>in</strong> HA, RakacolliD, Galli C, de Vaureix C, Trounson AO, Mummery CL,Gandolfi F, 1999: Molecular clon<strong>in</strong>g, genetic mapp<strong>in</strong>g, anddevelopmental expression of bov<strong>in</strong>e POU5F1. Biol Reprod60, 1093–1103.Evans MJ, Kaufman MH, 1981: Establishment <strong>in</strong> culture ofpluripotential cells from mouse embryos. Nature 292, 154–156.Evans MJ, Notarianni E, Laurie S, Moor RM, 1990: Derivationand prelim<strong>in</strong>ary characterization of pluripotent celll<strong>in</strong>es from porc<strong>in</strong>e and bov<strong>in</strong>e blastocysts. Theriogenology33, 125–128.Flechon JE, Degrouard J, Flechon B, 2004: Gastrulationevents <strong>in</strong> the prestreak pig embryo: ultrastructure and cellmarkers. Genesis 38, 13–25.Galli C, Lazzari G, Flechon JE, Moor RM, 1994: Embryonicstem cells <strong>in</strong> farm animals. Zygote 2, 385–389.Gerfen RW, Wheeler DA, 1995: Isolation of embryonic celll<strong>in</strong>esfrom porc<strong>in</strong>e blastocysts. Anim Biotechnol 6, 1–14.G<strong>in</strong>is I, Luo Y, Miura T, Thies S, Brandenberger R, Gerecht-Nir S, Amit M, Hoke A, Carpenter MK, Itskovitz-Eldor J,Rao MS, 2004: Differences between human and mouseembryonic stem cells. Dev Biol 269, 360–380.Goel S, Sugimoto M, M<strong>in</strong>ami N, Yamada M, Kume S, ImaiH, 2007: Identification, isolation, and <strong>in</strong> vitro culture ofporc<strong>in</strong>e gonocytes. Biol Reprod 77, 127–137.Guan K, Nayernia K, Maier LS, Wagner S, Dressel R, Lee JH,Nolte J, Wolf F, Li M, Engel W, Hasenfuss G, 2006:Pluripotency of spermatogonial stem cells from adult mousetestis. Nature 440, 1199–1203.He S, Pant D, Schiffmacher A, Bischoff S, Melican D, Gav<strong>in</strong>W, Keefer C2006: Developmental expression of pluripotencydeterm<strong>in</strong><strong>in</strong>g factors <strong>in</strong> capr<strong>in</strong>e embryos: novel pattern ofNANOG prote<strong>in</strong> localization <strong>in</strong> the nucleolus. Mol ReprodDev 73, 1512–1522.Hochedl<strong>in</strong>ger K, Jaenisch R, 2006: Nuclear reprogramm<strong>in</strong>gand pluripotency. Nature 441, 1061–1067.Hoffman LM, Carpenter MK, 2005: Characterization andculture of human embryonic stem cells. Nat Biotechnol 23,699–708.Honaramooz A, Megee SO, Dobr<strong>in</strong>ski I, 2002: Germ celltransplantation <strong>in</strong> pigs. Biol Reprod 66, 21–28.Honaramooz A, Behboodi E, Blash S, Megee SO, Dobr<strong>in</strong>ski I,2003a: Germ cell transplantation <strong>in</strong> goats. Mol Reprod Dev64, 422–428.Honaramooz A, Behboodi E, Megee SO, Overton SA,Galant<strong>in</strong>o-Homer H, Echelard Y, Dobr<strong>in</strong>ski I, 2003b:Fertility and germl<strong>in</strong>e transmission of donor haplotypefollow<strong>in</strong>g germ cell transplantation <strong>in</strong> immunocompetentgoats. Biol Reprod 69, 1260–1264.Hunter RH, 1974: Chronological and cytological details offertilization and early embryonic development <strong>in</strong> thedomestic pig, Sus scrofa. Anat Rec 178, 169–185.Kanatsu-Sh<strong>in</strong>ohara M, Inoue K, Lee J, Yoshimoto M,Ogonuki N, Miki H, Baba S, Kato T, Kazuki Y, ToyokuniS, Toyoshima M, Niwa O, Oshimura M, Heike T, NakahataT, Ish<strong>in</strong>o F, Ogura A, Sh<strong>in</strong>ohara T, 2004: Generation ofpluripotent stem cells from neonatal mouse testis. Cell 119,1001–1012.Kawase E, Suemori H, Takahashi N, Okazaki K, HashimotoK, Nakatsuji N, 1994: Stra<strong>in</strong> difference <strong>in</strong> establishment ofmouse embryonic stem (ES) cell l<strong>in</strong>es. Int J Dev Biol 38,385–390.Keefer CL, Pant D, Blomberg L, Talbot NC, 2007: Challengesand prospects for the establishment of embryonic stem celll<strong>in</strong>es of domesticated ungulates. Anim Reprod Sci 98, 147–168.Kirchhof N, Carnwath JW, Lemme E, Anastassiadis K,Scholer H, Niemann H, 2000: Expression pattern of Oct-4<strong>in</strong> preimplantation embryos of different species. Biol Reprod63, 1698–1705.Laslett AL, Filipczyk AA, Pera MF, 2003: Characterizationand culture of human embryonic stem cells. TrendsCardiovasc Med 13, 295–301.Lovell-Badge R, 2007: Many ways to pluripotency. NatBiotechnol 25, 1114.Ludwig TE, Levenste<strong>in</strong> ME, Jones JM, Berggren WT, MitchenER, Frane JL, Crandall LJ, Daigh CA, Conard KR,Piekarczyk MS, Llanas RA, Thomson JA, 2006: Derivationof human embryonic stem cells <strong>in</strong> def<strong>in</strong>ed conditions. NatBiotechnol 24, 185–187.Luo J, Megee S, Rathi R, Dobr<strong>in</strong>ski I, 2006: Prote<strong>in</strong> geneproduct 9.5 is a spermatogonia-specific marker <strong>in</strong> the pigtestis: application to enrichment and culture of porc<strong>in</strong>espermatogonia. Mol Reprod Dev 73, 1531–1540.Maddox-Hyttel P, Alexopoulos NI, Vajta G, Lewis I, RogersP, Cann L, Callesen H, Tveden-Nyborg P, Trounson A,2003: Immunohistochemical and ultrastructural characterizationof the <strong>in</strong>itial post-hatch<strong>in</strong>g development of bov<strong>in</strong>eembryos. <strong>Reproduction</strong> 125, 607–623.Matsui Y, Zsebo K, Hogan BL, 1992: Derivation of pluripotentialembryonic stem cells from mur<strong>in</strong>e primordial germcells <strong>in</strong> culture. Cell 70, 841–847.McLaren A, 1998: Germ cells and germ cell transplantation.Int J Dev Biol 42, 855–860.Moore K, Piedrahita JA, 1997: The effects of human leukemia<strong>in</strong>hibitory factor (hLIF) and culture medium on <strong>in</strong> vitrodifferentiation of cultured porc<strong>in</strong>e <strong>in</strong>ner cell mass (pICM).In Vitro Cell Dev Biol Anim 33, 62–71.Mueller S, Prelle K, Rieger N, Petznek H, Lassnig C, LukschU, Aigner B, Baetscher M, Wolf E, Mueller M, Brem G,1999: Chimeric pigs follow<strong>in</strong>g blastocyst <strong>in</strong>jection of transgenicporc<strong>in</strong>e primordial germ cells. Mol Reprod Dev 54,244–254.Oback B, Wells DN, 2007: Donor cell differentiation, reprogramm<strong>in</strong>g,and clon<strong>in</strong>g efficiency: elusive or illusive correlation?Mol Reprod Dev 74, 646–654.Pant D, Keefer CL, 2006: Gene expression <strong>in</strong> cultures of<strong>in</strong>ner cell masses isolated from <strong>in</strong> vitro produced and <strong>in</strong>vivo derived bov<strong>in</strong>e blastocysts. Reprod Fertil Dev 18,110.Piedrahita JA, Anderson GB, BonDurant RH, 1990a: Influenceof feeder layer type on the efficiency of isolation ofporc<strong>in</strong>e embryo-derived cell l<strong>in</strong>es. Theriogenology 34, 865–877.Piedrahita JA, Anderson GB, Bondurant RH, 1990b: On theisolation of embryonic stem cells: Comparative behavior ofÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Farm <strong>Animals</strong> Embryonic Stem Cells 199mur<strong>in</strong>e, porc<strong>in</strong>e and ov<strong>in</strong>e embryos. Theriogenology 34,879–901.Piedrahita JA, Moore K, Oetama B, Lee CK, Scales N,Ramsoondar J, Bazer FW, Ott T, 1998: Generation oftransgenic porc<strong>in</strong>e chimeras us<strong>in</strong>g primordial germ cellderivedcolonies. Biol Reprod 58, 1321–1329.Prelle K, Vassiliev IM, Vassilieva SG, Wolf E, Wobus AM,1999: Establishment of pluripotent cell l<strong>in</strong>es from vertebratespecies – present status and future prospects. Cells TissuesOrgans 165, 220–236.Ralston A, Rossant J, 2005: Genetic regulation of stem cellorig<strong>in</strong>s <strong>in</strong> the mouse embryo. Cl<strong>in</strong> Genet 68, 106–112.Rao BM, Zandstra PW, 2005: Culture development for humanembryonic stem cell propagation: molecular aspects andchallenges. Curr Op<strong>in</strong> Biotechnol 16, 568.Renard JP, Heyman Y, 1979: Variable development ofsuperovulated bov<strong>in</strong>e embryos between day 6 and day 12.Ann Biol Anim Biochim Biophys 19, 1589–1598.Robertson EJ, 1987: Teratocarc<strong>in</strong>omas and Embryonic StemCells, a Practical Approach. IRL Press, Oxford, Wash<strong>in</strong>gtonDC.Rodriguez-Sosa JR, Dobson H, Hahnel A, 2006: Isolation andtransplantation of spermatogonia <strong>in</strong> sheep. Theriogenology66, 2091–2103.Rui R, Shim H, Moyer AL, Anderson DL, Penedo CT, RoweJD, BonDurant RH, Anderson GB, 2004: Attempts toenhance production of porc<strong>in</strong>e chimeras from embryonicgerm cells and preimplantation embryos. Theriogenology61, 1225–1235.Schnieke AE, K<strong>in</strong>d AJ, Ritchie WA, Mycock K, Scott AR,Ritchie M, Wilmut I, Colman A, Campbell KH, 1997:Human factor IX transgenic sheep produced by transfer ofnuclei from transfected fetal fibroblasts. Science 278, 2130–2133.Schoonjans L, Albright GM, Li JL, Collen D, MoreadithRW, 1996: Pluripotential rabbit embryonic stem (ES) cellsare capable of form<strong>in</strong>g overt coat color chimeras follow<strong>in</strong>g<strong>in</strong>jection <strong>in</strong>to blastocysts. Mol Reprod Dev 45, 439–443.Seamark RF, 1994: Progress and emerg<strong>in</strong>g problems <strong>in</strong>livestock transgenesis: a summary perspective. Reprod FertilDev 6, 653–657.Shamblott MJ, Axelman J, Wang S, Bugg EM, Littlefield JW,Donovan PJ, Blumenthal PD, Hugg<strong>in</strong>s GR, Gearhart JD,1998: Derivation of pluripotent stem cells from culturedhuman primordial germ cells. Proc Natl Acad Sci U S A 95,13726–13731.Shim H, Anderson GB, 1998: In vitro survival and proliferationof porc<strong>in</strong>e primordial germ cells. Theriogenology 49,521–528.Shim H, Gutierrez-Adan A, Chen LR, BonDurant RH,Behboodi E, Anderson GB, 1997: Isolation of pluripotentstem cells from cultured porc<strong>in</strong>e primordial germ cells. BiolReprod 57, 1089–1095.Sh<strong>in</strong>ohara T, Kanatsu-Sh<strong>in</strong>ohara M2007: Culture and geneticmodification of mouse germl<strong>in</strong>e stem cells. Ann NY AcadSci, Annals. 1120, 58–71.Sh<strong>in</strong>ohara T, Orwig KE, Avarbock MR, Br<strong>in</strong>ster RL, 2003:Restoration of spermatogenesis <strong>in</strong> <strong>in</strong>fertile mice by Sertolicell transplantation. Biol Reprod 68, 1064–1071.Smith AG, 2001: Embryo-derived stem cells: of mice and men.Annu Rev Cell Dev Biol 17, 435–462.Tada M, Tada T, Lefebvre L, Barton SC, Surani MA, 1997:Embryonic germ cells <strong>in</strong>duce epigenetic reprogramm<strong>in</strong>g ofsomatic nucleus <strong>in</strong> hybrid cells. EMBO J 16, 6510–6520.Talbot NC, Rexroad CE Jr, Pursel VG, Powell AM, Nel ND,1993: Cultur<strong>in</strong>g the epiblast cells of the pig blastocyst. InVitro Cell Dev Biol Anim 29A, 543–554.Talbot NC, Powell AM, Rexroad CE Jr, 1995: In vitropluripotency of epiblasts derived from bov<strong>in</strong>e blastocysts.Mol Reprod Dev 42, 35–52.Talbot NC, Caperna TJ, Wells KD, 2002: The PICM-19 celll<strong>in</strong>e as an <strong>in</strong> vitro model of liver bile ductules: effects ofcAMP <strong>in</strong>ducers, biopeptides and pH. Cells Tissues Organs171, 99–116.Tam PP, Behr<strong>in</strong>ger RR, 1997: Mouse gastrulation: theformation of a mammalian body plan. Mech Dev 68, 3–25.Tesar PJ, Chenoweth JG, Brook FA, Davies TJ, Evans EP,Mack DL, Gardner RL, McKay RD, 2007: New cell l<strong>in</strong>esfrom mouse epiblast share def<strong>in</strong><strong>in</strong>g features with humanembryonic stem cells. Nature 448, 196–199.Thomas KR, Capecchi MR, 1987: Site-directed mutagenesisby gene target<strong>in</strong>g <strong>in</strong> mouse embryo-derived stem cells. Cell51, 503–512.Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA,Swiergiel JJ, Marshall VS, Jones JM, 1998: Embryonic stemcell l<strong>in</strong>es derived from human blastocysts. Science 282,1145–1147.Vejlsted M, Avery B, Schmidt M, Greve T, Alexopoulos N,Maddox-Hyttel P, 2005: Ultrastructural and immunohistochemicalcharacterization of the bov<strong>in</strong>e epiblast. BiolReprod 72, 678–686.Vejlsted M, Du Y, Vajta G, Maddox-Hyttel P, 2006: Posthatch<strong>in</strong>gdevelopment of the porc<strong>in</strong>e and bov<strong>in</strong>e embryodef<strong>in</strong><strong>in</strong>gcriteria for expected development <strong>in</strong> vivo and <strong>in</strong>vitro. Theriogenology 65, 153–165.Wheeler MB, 1994: Development and validation of sw<strong>in</strong>eembryonic stem cells: a review. Reprod Fertil Dev 6, 563–568.Wilmut I, Schnieke AE, McWhir J, K<strong>in</strong>d AJ, Campbell KH,1997: Viable offspr<strong>in</strong>g derived from fetal and adult mammaliancells. Nature 385, 810–813.Zakhartchenko V, Durcova-Hills G, Schernthaner W, StojkovicM, Reichenbach HD, Mueller S, Ste<strong>in</strong>born R, MuellerM, Wenigerk<strong>in</strong>d H, Prelle K, Wolf E, Brem G, 1999:Potential of fetal germ cells for nuclear transfer <strong>in</strong> cattle.Mol Reprod Dev 52, 421–426.Author’s address (for correspondence): Fulvio Gandolfi, Departmentof Animal Science, Centre for Embryonic Stem Cell Research,Laboratory of Biomedical Embryology, University of Milan, ViaCecoria 10, 20133 Milano, Italy. E-mail: fulvio.gandolfi@unimi.itConflict of <strong>in</strong>terest: The authors declare no conflict of <strong>in</strong>terests.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 200–206 (2008); doi: 10.1111/j.1439-0531.2008.01162.xISSN 0936-6768<strong>Reproduction</strong> <strong>in</strong> <strong>Domestic</strong> BuffaloBMAO PereraFaculty of Veter<strong>in</strong>ary Medic<strong>in</strong>e and Animal Science, University of Peradeniya, Peradeniya, Sri LankaContentsThe domestic buffalo is an <strong>in</strong>dispensable livestock resource tomillions of smallholder farmers <strong>in</strong> develop<strong>in</strong>g countries,particularly <strong>in</strong> Asia. Although its reproductive biology isbasically similar to that of cattle, there are importantdifferences and unique characteristics that need to be considered<strong>in</strong> order to apply modern reproductive technologies toimprove its productivity. Under most smallholder productionsystems, the reproductive efficiency of buffalo is compromisedby factors related to climate, management, nutrition anddiseases. However, when managed and fed properly, buffalocan have good fertility and provide milk, calves and draughtpower over a long productive life. The basic technicalproblems associated with artificial <strong>in</strong>sem<strong>in</strong>ation <strong>in</strong> buffalowere largely overcome two decades ago, but the technologyhas not had the expected impact <strong>in</strong> some develop<strong>in</strong>g countries,because largely of <strong>in</strong>frastructural and logistic problems.Approaches <strong>in</strong>volv<strong>in</strong>g the use of hormones for treat<strong>in</strong>ganoestrus and for synchroniz<strong>in</strong>g oestrus have had vary<strong>in</strong>grates of success, depend<strong>in</strong>g on the protocols used and the<strong>in</strong>cidence of underly<strong>in</strong>g problems that cause <strong>in</strong>fertility.Embryo technologies such as multiple ovulation embryotransfer, <strong>in</strong> vitro embryo production, cryopreservation andclon<strong>in</strong>g are be<strong>in</strong>g <strong>in</strong>tensively studied but have had far lowersuccess rates than <strong>in</strong> cattle. Improv<strong>in</strong>g the productivity ofbuffalo requires an understand<strong>in</strong>g of their potential andlimitations under each farm<strong>in</strong>g system, development of simple<strong>in</strong>tervention strategies to ameliorate deficiencies <strong>in</strong> management,nutrition and healthcare, followed by judicious applicationof reproductive technologies that are susta<strong>in</strong>able withthe resources available to buffalo farmers.IntroductionThe world population of domestic water buffalo (Bubalusbubalis) is estimated to be approximately 172million, of which 166 million (96%) are <strong>in</strong> Asia, and therema<strong>in</strong>der ma<strong>in</strong>ly <strong>in</strong> the Mediterranean and Lat<strong>in</strong>American regions. Buffalo have been classified <strong>in</strong> totwo ma<strong>in</strong> ‘types’: the river type located <strong>in</strong> South Asiaand the swamp type spread across the South-East Asianregion. Systems of buffalo production vary widelythrough the different regions of the world (Perera et al.2005) and are determ<strong>in</strong>ed by a matrix of several<strong>in</strong>teract<strong>in</strong>g factors that <strong>in</strong>clude climate (tropical ortemperate, humid or arid), location (rural, peri-urbanor urban), cropp<strong>in</strong>g systems (ra<strong>in</strong>-fed or irrigated,annual or perennial crops), type of operation (small orlarge farm, subsistence or commercial) and the primarypurpose (milk, meat, draught, capital or mixed). Thebuffalo has been traditionally regarded as a poorbreeder with low reproductive efficiency, characterizedby late atta<strong>in</strong>ment of puberty and maturity, seasonalityof calv<strong>in</strong>g, long postpartum anoestrus, poor expressionof oestrous signs, low conception rates and long calv<strong>in</strong>g<strong>in</strong>tervals (Perera 1999; Barile 2005). This can be attributedto factors such as harsh environments, lack of yearroundfeed supply and m<strong>in</strong>imal managerial <strong>in</strong>puts, <strong>in</strong> themajority of farm<strong>in</strong>g systems under which buffalo areraised. Studies <strong>in</strong> Pakistan (Usmani et al. 1990), SriLanka (Perera et al. 1987) and Brazil (Vale 1997) haveshown that they can have good fertility if they aremanaged and fed properly so as to overcome suchstresses.The reproductive physiology and endocr<strong>in</strong>ology ofdomestic buffalo and their comparative aspects withcattle were comprehensively reviewed two decades agoby Dobson and Kamonpatana (1986) and a decadelater, subsequent advances <strong>in</strong> knowledge on buffaloreproduction were reviewed by Madan et al. (1996) andPerera (1999). A recent publication of the Food andAgriculture Organization edited by Borghese (2005)conta<strong>in</strong>s a series of comprehensive review papers on thebuffalo, <strong>in</strong>clud<strong>in</strong>g the production systems <strong>in</strong> the worldand their reproductive and productive characteristics.This paper will therefore limit its objectives to: (a)highlight<strong>in</strong>g some of the reproductive characteristics ofbuffalo that have a bear<strong>in</strong>g on their fertility andproductivity, (b) review<strong>in</strong>g recent advances <strong>in</strong> modernreproductive technologies and (c) discuss<strong>in</strong>g the potentialapplications and limitations of these technologies forimprov<strong>in</strong>g buffalo production.Genetics and Breed<strong>in</strong>gThe phylogeny of water buffalo is still a matter ofdebate. Based on studies of mitochondrial DNA(mtDNA) of swamp and river buffalo, together withanalysis of data published from South-East Asian andAustralian water buffalo, Kierste<strong>in</strong> et al. (2004) concludedthat both types of buffalo descend from onedomestication event, probably <strong>in</strong> the Indian subcont<strong>in</strong>ent.They also found evidence for <strong>in</strong>trogression of wildAsian buffalo (Bubalus arnee) mtDNA <strong>in</strong>to domesticswamp buffalo. However, studies by Kumar et al. (2007)showed that river and swamp buffalo are dist<strong>in</strong>guished<strong>in</strong>to two dist<strong>in</strong>ct clades, <strong>in</strong>dicat<strong>in</strong>g that the two typeswere domesticated <strong>in</strong>dependently. This was supportedby studies <strong>in</strong> Ch<strong>in</strong>a (Lei et al. 2007) that showed twomtDNA l<strong>in</strong>eages with divergence estimated at18 000 years ago, <strong>in</strong>dicat<strong>in</strong>g <strong>in</strong>dependent domesticationevents for the swamp buffalo from Ch<strong>in</strong>a and the riverbuffalo from the Indian subcont<strong>in</strong>ent.From the practical aspects of buffalo breed<strong>in</strong>g, thedisparity <strong>in</strong> the number of chromosomes <strong>in</strong> swamp(2n = 48) and river (2n = 50) buffalo has relevance(Huang et al. 2003). The F1 hybrids have 49 chromosomes,while the F2 hybrids have 48, 49 or 50 chromosomes.The backcrosses have two different karyotypeÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


<strong>Reproduction</strong> <strong>in</strong> <strong>Domestic</strong> Buffalo 201categories each, with 2n = 48 and 2n = 49 <strong>in</strong> the threequarters swamp types and 2n = 49 and 2n = 50 <strong>in</strong> thethree quarters river types (Harisah et al. 1989). Thedistribution of chromosome categories among the F2hybrids and backcrosses suggests that only geneticallybalanced gametes of the F1 hybrids are capable ofproduc<strong>in</strong>g viable F2 and backcross generations, andthat crossbreds with 2n = 49 had lower fertility thancrossbreds with 2n = 50 (Huang et al. 2003).<strong>Reproduction</strong> <strong>in</strong> the FemaleBuffaloes are polyoestrus and are capable of breed<strong>in</strong>gthroughout the year. Yet, <strong>in</strong> many countries, a seasonalpattern of calv<strong>in</strong>g has been observed. In some locations,where annual changes <strong>in</strong> ra<strong>in</strong>fall determ<strong>in</strong>e the availabilityand quality of feed, ovarian activity commencessome 2–3 months after the onset of ra<strong>in</strong>s, followed byconceptions that result <strong>in</strong> a peak calv<strong>in</strong>g season10 months later (Perera et al. 1987). In Italy, however,where buffalo are fed with a constant balanced diet, adist<strong>in</strong>ct seasonal reproductive pattern is also found, andthe <strong>in</strong>ference from a series of studies is that seasonality is<strong>in</strong>fluenced by photoperiod, mediated by melaton<strong>in</strong>secretion (Zicarelli 1997). In India, where summer heatstress can be severe, high prolact<strong>in</strong> secretion has beenidentified as a factor contribut<strong>in</strong>g to acyclicity and poorfertility by lower<strong>in</strong>g progesterone secretion dur<strong>in</strong>g thesummer months (Roy and Prakash 2007). The ma<strong>in</strong>reproductive characteristics of buffalo, based on reportsfrom various countries on river and swamp buffalounder a wide range of management systems, aresummarized <strong>in</strong> Table 1.Puberty, ovarian characteristics and oestrous cyclesBuffalo heifers usually atta<strong>in</strong> puberty when they reachapproximately 55–60% of adult body weight, but theage at which this occurs can be highly variable (Table 1),be<strong>in</strong>g <strong>in</strong>fluenced by genotype, nutrition, management,social environment, climate and year or season of birth.Although buffalo atta<strong>in</strong> puberty later than cattle, theyhave a longer reproductive life, which compensates forthe early disadvantage. The ovaries of cyclic buffaloheifers have a reservoir of only 10 000 to 20 000primordial follicles (Danell 1987) compared withTable 1. Summary of reproductive characteristics of buffalo (adaptedfrom Perera et al. 2005)Parameter Mean RangeAge at puberty (months) 30 16–46Weight at puberty (kg) 275 200–350Length of oestrous cycle (days) 21 17–26Length of oestrus (h) 10 5–27Time of ovulationAfter onset of oestrus (h) 34 24–48After end of oestrus (h) 14 6–21Length of gestation (days)River type 310 300–320Swamp type 330 320–340Birth weight of calves (kg) 26 22–36Involution of uterus (days) 30 25–35approximately 150 000 <strong>in</strong> cattle. This could be one ofthe reasons for the lower yield of embryos when multipleovulation embryo transfer (MOET) is attempted <strong>in</strong>buffalo (see section ‘Multiple ovulation embryo transfer’).Various treatments have been attempted for earlier<strong>in</strong>duction of puberty <strong>in</strong> buffalo heifers and one regimethat has been successful is the use of ProgesteroneReleas<strong>in</strong>g Intravag<strong>in</strong>al Devices (PRID) left <strong>in</strong> place for12 days followed by treatment with Equ<strong>in</strong>e ChorionicGonadotroph<strong>in</strong> (eCG) at the time of withdrawal (Barileet al. 2001). This resulted <strong>in</strong> conception rates of 50–60%at 60 days after treatment, compared with 22% <strong>in</strong>controls.Ovarian follicular growth dur<strong>in</strong>g the oestrous cycle <strong>in</strong>buffalo is similar to that observed <strong>in</strong> cattle and ischaracterized by waves of follicular recruitment, growthand regression. Studies <strong>in</strong> Brazil found that 63% ofanimals had two follicular waves dur<strong>in</strong>g a cycle while33% had three follicular waves (Baruselli et al. 1997),and that the latter group had a longer luteal phase,result<strong>in</strong>g <strong>in</strong> a longer oestrous cycle, than the former(mean 24.0 vs 21.8 days). However, a recent study onsuckled Indian buffalo showed that five out of eightanimals showed a s<strong>in</strong>gle wave of follicular growth, whilethe others had two waves (Awasthi et al. 2006). Thetemporal changes of progesterone <strong>in</strong> blood and milkdur<strong>in</strong>g the oestrous cycle are similar to those <strong>in</strong> cattle,but the concentration is relatively lower (Dobson andKamonpatana 1986; Perera et al. 1987).A major difference between buffalo and cattle is that,external signs of oestrus are less obvious <strong>in</strong> the former,with homosexual behaviour between females be<strong>in</strong>g rare(Perera 1987). The ma<strong>in</strong> behavioural signs are restlessness,bellow<strong>in</strong>g and frequent void<strong>in</strong>g of small quantitiesof ur<strong>in</strong>e. Externally detectable physical changes <strong>in</strong>cludeswell<strong>in</strong>g of the vulva, result<strong>in</strong>g <strong>in</strong> effacement of thehorizontal wr<strong>in</strong>kles that are present on its externalsurface and this, together with vestibular redden<strong>in</strong>g, canbe detected by regular exam<strong>in</strong>ation of <strong>in</strong>dividualanimals under conf<strong>in</strong>ed systems. Mucus, secreted fromthe cervix dur<strong>in</strong>g oestrus, is less copious than <strong>in</strong> cattleand does not usually hang as strands from the vulva buttends to accumulate on the floor of the vag<strong>in</strong>a and bedischarged either when the animal is ly<strong>in</strong>g down or withthe ur<strong>in</strong>e. Thus, <strong>in</strong> females that are housed or tethered, agood practice is to exam<strong>in</strong>e the floor near the rear of theanimal each morn<strong>in</strong>g and even<strong>in</strong>g for signs of mucus.These factors have contributed to the observation thatsilent ovulation (also termed as silent oestrus) is morecommon <strong>in</strong> buffalo than <strong>in</strong> cattle. Awasthi et al. (2007)found that buffalo with silent ovulation follow<strong>in</strong>gtreatment with prostagland<strong>in</strong> had slower growth rateand smaller diameter of the ovulatory follicle, and alonger <strong>in</strong>terval from treatment to ovulation, than thosewith overt oestrous signs. Studies are warranted todeterm<strong>in</strong>e whether such differences also occur <strong>in</strong> spontaneouslyovulat<strong>in</strong>g animals.The duration of oestrus and the <strong>in</strong>tervals from theonset and end of oestrus to ovulation are shown <strong>in</strong>Table 1. In hot climates, the duration of oestrus tends tobe short and the signs may be exhibited only dur<strong>in</strong>g thenight or early morn<strong>in</strong>g. In contrast, studies on Italianbuffalo show that they have longer duration of oestrousÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


202 BMAO Pererabehaviour and a longer <strong>in</strong>terval from the beg<strong>in</strong>n<strong>in</strong>g ofoestrus to ovulation, with the <strong>in</strong>terval from peak LHconcentration to ovulation be<strong>in</strong>g 25 ± 13 h <strong>in</strong> animalsthat conceived to artificial <strong>in</strong>sem<strong>in</strong>ation (AI) and46 ± 18 h <strong>in</strong> those that did not (Moioli et al. 1998;Borghese 2005). The results from most studies <strong>in</strong>dicatethat the appropriate time for breed<strong>in</strong>g, whether bynatural service or AI, is dur<strong>in</strong>g the latter part of theoestrous period. Thus, the AM ⁄ PM rule, which isapplied <strong>in</strong> cattle, where females detected <strong>in</strong> oestrus <strong>in</strong>the morn<strong>in</strong>g are bred <strong>in</strong> the afternoon of the same dayand those detected <strong>in</strong> the afternoon or even<strong>in</strong>g are bredthe next morn<strong>in</strong>g, also applies <strong>in</strong> buffalo.Pregnancy, parturition and the postpartum periodThe duration of gestation <strong>in</strong> the buffalo ranges from 300to 330 days with a mean of approximately 310 days forriver types and 320 days for swamp types (Perera and deSilva 1985). Early diagnosis of pregnancy can be carriedout by ultrasound scann<strong>in</strong>g from approximately 20 daysonwards (Presicce et al. 2001) and by rectal palpationfrom approximately 45 days onwards. The techniquesare basically similar to those rout<strong>in</strong>ely used <strong>in</strong> cattle, butthe longer gestation period has to be taken account of <strong>in</strong>assess<strong>in</strong>g the stage of pregnancy. The ma<strong>in</strong> laboratorymethod of diagnosis is based on measurement ofprogesterone 20–23 days after breed<strong>in</strong>g (Perera et al.1980). The process of parturition and the changesoccurr<strong>in</strong>g <strong>in</strong> hormones such as progesterone, oestroneand the ma<strong>in</strong> metabolite of prostagland<strong>in</strong> F 2a aroundthe period of parturition <strong>in</strong> buffalo are similar to those<strong>in</strong> cattle (Perera et al. 1981). Involution of the uterus isusually completed <strong>in</strong> 25–35 days after calv<strong>in</strong>g (Lohanet al. 2004), and the stimulus of suckl<strong>in</strong>g shortens the<strong>in</strong>volution time (Usmani et al. 1990).The period of postpartum anoestrus or acyclicity ishighly variable <strong>in</strong> the buffalo and is usually longer than<strong>in</strong> cattle under comparative management conditions.Under good conditions, buffalo can resume cyclicity by30–90 days, but factors such as poor nutrition and bodycondition (Baruselli et al. 2001), suckl<strong>in</strong>g management(Usmani et al. 1990) and climate (which also <strong>in</strong>fluencesnutrition through feed quality and availability) candelay this considerably. For example, <strong>in</strong>digenous buffalo<strong>in</strong> Sri Lanka raised under free-graz<strong>in</strong>g conditions withabundant natural feed and suckl<strong>in</strong>g restricted to onceper day resumed cyclicity by 30–60 days after calv<strong>in</strong>g,but those under harsh conditions with free suckl<strong>in</strong>g bythe calves rema<strong>in</strong>ed acyclic for 150–200 days (Pereraet al. 1987). A review of literature from Egypt, India andPakistan (El-Wishy 2007) showed that only 34–49% ofbuffalo showed oestrus dur<strong>in</strong>g the first 90 days aftercalv<strong>in</strong>g and 31–42% rema<strong>in</strong>ed anoestrus for more than150 days. Both postpartum ovulation and oestrusoccurred later <strong>in</strong> swamp than <strong>in</strong> river buffalo. The firstpostpartum ovulation was frequently followed by one ormore short oestrous cycles (


<strong>Reproduction</strong> <strong>in</strong> <strong>Domestic</strong> Buffalo 203Reproductive TechnologiesArtificial <strong>in</strong>sem<strong>in</strong>ationThe procedures for process<strong>in</strong>g and us<strong>in</strong>g buffalo semenfor AI are based ma<strong>in</strong>ly on techniques developed forcattle with some modifications (Vale 1997; IAEA 2005).The ma<strong>in</strong> differences are <strong>in</strong> the semen diluents, with Trisbuffers, egg yolk, skim milk and coconut water be<strong>in</strong>gcommonly used as <strong>in</strong>gredients for preserv<strong>in</strong>g semen <strong>in</strong>both chilled (+4°C) and deep-frozen forms. The cryoprotectiveagent used for freez<strong>in</strong>g buffalo semen isglycerol, at a f<strong>in</strong>al concentration of 6.5–7.0%. Variousadditives have been <strong>in</strong>vestigated for possible beneficialeffects on buffalo spermatozoa dur<strong>in</strong>g freez<strong>in</strong>g andthaw<strong>in</strong>g, and the two prote<strong>in</strong>s that help to ma<strong>in</strong>ta<strong>in</strong>higher post-thaw motility are oviductal prote<strong>in</strong>s (Kumaresanet al. 2005) and Bradyk<strong>in</strong><strong>in</strong> (Shukla and Misra2007). Buffalo spermatozoa, subjected to freez<strong>in</strong>g andthaw<strong>in</strong>g, appear to have a shorter fertile lifespan <strong>in</strong>sidethe female tract than those <strong>in</strong> fresh semen (Moioli et al.1998). Thus, proper detection of heat and tim<strong>in</strong>g of AIbecome critical when frozen semen is used and may beone reason for the lower conception rates, while anothercould be the narrow cervix of the buffalo, which makesAI more difficult.Apart from the technical aspects, there are manyconstra<strong>in</strong>ts which <strong>in</strong>fluence the success of AI <strong>in</strong> buffalo,especially <strong>in</strong> develop<strong>in</strong>g countries. These <strong>in</strong>clude factorsthat impede the effective and timely delivery of services,such as poor communication, lack of transport and<strong>in</strong>adequate remuneration for AI technicians, and limitationsof smallholder farm<strong>in</strong>g systems, such as poornutrition and reproductive management of cows.Separation of X and Y spermatozoaIdentification of X- and Y-bear<strong>in</strong>g spermatozoa <strong>in</strong>buffalo can be performed by fluorescence <strong>in</strong> situhybridization, us<strong>in</strong>g the cattle Y-chromosome-specificBC1.2 probe or the X- and Y-specific probes from theyak (Re´vay et al. 2003). Use of high-speed flowcytometric cell sort<strong>in</strong>g, followed by deep AI carriedout near the utero-tubal junction (UTJ) with 2.5 millionlive frozen-thawed sperm, resulted <strong>in</strong> 43% conceptionrate with eight out of n<strong>in</strong>e fetuses correspond<strong>in</strong>g to thepredicted sex (Presicce et al. 2005). Semen depositionnear the UTJ of buffalo can be performed us<strong>in</strong>g theGhent device that was developed for cattle (Presicceet al. 2004). Sex-sorted sperm has also been usedsuccessfully for <strong>in</strong> vitro fertilization (IVF) and embryotransfer (Lu et al. 2007).Oestrous synchronizationThe procedures used <strong>in</strong> the past for oestrous synchronization<strong>in</strong> buffalo were empirically based on thosedeveloped for cattle, aimed at either <strong>in</strong>duc<strong>in</strong>g prematureluteolysis us<strong>in</strong>g prostagland<strong>in</strong>s or prolong<strong>in</strong>g the lutealphase us<strong>in</strong>g progestagens (Perera 1987). However, theefficacy of prostagland<strong>in</strong>s for synchroniz<strong>in</strong>g ovulation isnow known to be dependent upon progesterone concentrationand ovarian follicular status at the time oftreatment (Brito et al. 2002). Use of the two-dose regimeof prostagland<strong>in</strong> overcomes some of the above limitations,but manipulation of follicular development isnecessary to achieve better synchrony and improvedfertility (De Rensis and Lo´pez-Gatius 2007). Therefore,most current protocols <strong>in</strong>clude GnRH or gonadotroph<strong>in</strong>s<strong>in</strong> comb<strong>in</strong>ation with prostagland<strong>in</strong>s, progesteroneor oestradiol. The ‘Ovsynch’ protocol (GnRH, prostagland<strong>in</strong>7 days later and second GnRH 2 days later) hasbeen successful <strong>in</strong> synchroniz<strong>in</strong>g ovulation <strong>in</strong> 70–90% ofbuffalo with conception rates rang<strong>in</strong>g from 33% to 60%(Baruselli et al. 1999; Paul and Prakash 2005). Inaddition to the type of protocol selected, the follow<strong>in</strong>gfactors must also be addressed to achieve success <strong>in</strong>buffalo: (a) selection of animals that are <strong>in</strong> goodnutritional condition and free from disease; (b) preventionof stress dur<strong>in</strong>g the procedures for treatment andAI, especially under tropical conditions, where animalsmay be herded together or moved to other locations;and (c) where seasonal differences exist, schedul<strong>in</strong>gtreatment for the more favourable periods when themajority of animals are cycl<strong>in</strong>g.Multiple ovulation embryo transferThe current status of MOET and other advancedreproductive technologies <strong>in</strong> buffalo has been recentlyreviewed (Drost 2007). Studies <strong>in</strong> many countries haveconfirmed that buffalo have a lower superovulatoryresponse than cattle, attributed ma<strong>in</strong>ly to the smallerpopulation of recruitable follicles <strong>in</strong> the ovary (Madanet al. 1996; Manik et al. 2002). A further limitationappears to be the relatively low rate of transfer ofoocytes to the oviduct and ⁄ or impaired transport of ovaand embryos <strong>in</strong> the reproductive tract (Baruselli et al.2000). In a large scale MOET operation <strong>in</strong> India (Misraet al. 1994), the total embryo yield per treatment<strong>in</strong>creased from 1.77 to 3.83 over 5 years, and thenumber of viable embryos from 0.92 to 2.13; thetransfer of 469 embryos resulted <strong>in</strong> a pregnancy rateof 17% and a calv<strong>in</strong>g rate of 9.8%. In Italy, Campanileet al. (1995) transferred 76 buffalo embryos andobta<strong>in</strong>ed a pregnancy rate of 30%, with no significantdifference between fresh or frozen-thawed embryos andbetween morulae or blastocysts. Yet, the overall rate ofsuccess <strong>in</strong> terms of calves born per superovulation is stilltoo low for MOET to be widely applicable under fieldfarm<strong>in</strong>gconditions. Two situations <strong>in</strong> which it mighthave a role are for establish<strong>in</strong>g nucleus breed<strong>in</strong>g stocksor for conservation of genetic resources.In vitro embryo production and cryopreservationThe low efficiency of MOET <strong>in</strong> buffalo has led to an<strong>in</strong>creased <strong>in</strong>terest <strong>in</strong> <strong>in</strong> vitro embryo production (IVEP)technologies for achiev<strong>in</strong>g rapid genetic improvement.The sequence of procedures <strong>in</strong>volv<strong>in</strong>g recovery ofoocytes from ovaries of slaughtered animals by directaspiration or from live animals by ultrasound-guidedtransvag<strong>in</strong>al ovum pick-up (OPU), followed by <strong>in</strong> vitromaturation, IVF and <strong>in</strong> vitro culture have been studied<strong>in</strong> buffalo over the past decade (Boni et al. 1996; Madanet al. 1996; Hufana-Duran et al. 2004), but the successÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


204 BMAO Pererarates are lower than those achieved <strong>in</strong> cattle. Thecollection of immature oocytes from slaughtered buffaloby aspiration of antral follicles can yield one to threeoocytes per ovary that are suitable for IVEP. The OPUtechnique can be used for repeatedly collect<strong>in</strong>g oocytes<strong>in</strong> cycl<strong>in</strong>g buffalo once or twice every week (Boni et al.1996; Manik et al. 2006) or <strong>in</strong> prepubertal heifers andcyclic cows after treatment with follicle-stimulat<strong>in</strong>ghormone (FSH) (Techakumphu et al. 2004). The yieldof cumulus–oocyte complexes us<strong>in</strong>g OPU <strong>in</strong> non-superovulatedbuffalo is approximately 1–2 per ovary percollection, whereas after FSH treatment it is approximately2–3 per ovary (Boni et al. 1997; Promdireg et al.2005). The cleavage rate of oocytes selected as suitablefor IVEP is approximately 50%, but only approximately10% develop <strong>in</strong>to morulae and blastocysts (Manik et al.2006). Maturation of buffalo oocytes <strong>in</strong> vitro occursearlier than <strong>in</strong> cattle (Neglia et al. 2003).For the cryopreservation of buffalo oocytes, the slowfreez<strong>in</strong>g procedure has been less successful than themethod of vitrification, and solid surface vitrificationappears to be superior to <strong>in</strong>-straw vitrification, us<strong>in</strong>gethylene glycol as the cryoprotectant (Boonkusol et al.2007). Recent studies <strong>in</strong> Philipp<strong>in</strong>es on the transfer ofvitrified embryos derived from oocytes collected fromslaughtered river type animals resulted <strong>in</strong> a pregnancyrate of 16% and a calv<strong>in</strong>g rate of 11% <strong>in</strong> river typerecipients (Hufana-Duran et al. 2004), and a calv<strong>in</strong>grate of 10% <strong>in</strong> swamp type recipients (Hufana-Duranet al. 2007). In Ch<strong>in</strong>a, transfer of fresh embryos derivedfrom repeated OPU to recipients after natural oestrusresulted <strong>in</strong> 35% calv<strong>in</strong>gs, while transfer of embryosderived from abattoir ovaries to synchronized recipientsresulted <strong>in</strong> 15% calv<strong>in</strong>gs (Liang et al. 2007).Clon<strong>in</strong>gResearch on clon<strong>in</strong>g by somatic cell nuclear transfer <strong>in</strong>buffalo is still <strong>in</strong> its early stages, but some basicunderstand<strong>in</strong>g has been achieved regard<strong>in</strong>g parthenogenetic,<strong>in</strong> vitro and <strong>in</strong> vivo development of embryosreconstructed by transferr<strong>in</strong>g donor nuclei from foetaland adult fibroblast cells to enucleated buffalo oocytes.Embryos reconstructed us<strong>in</strong>g foetal fibroblasts werecapable of develop<strong>in</strong>g to blastocyst stage (Meena andDas 2006) and some pregnancies were detected after thetransfer of cloned blastocysts <strong>in</strong>to recipients, but nonewere carried to term (Saikhun et al. 2004). Clonedembryos that are capable of develop<strong>in</strong>g to the morulaand blastocyst stages have also been constructed us<strong>in</strong>genucleated rabbit oocytes as recipient cytoplasm andcow, swamp buffalo, pig and elephant fibroblasts asdonor nuclei (Numchaisrika et al. 2007). When clon<strong>in</strong>gtechnology does become established <strong>in</strong> the buffalo,problems that have been encountered <strong>in</strong> other species,such as high <strong>in</strong>cidence of developmental abnormalities,will need to be addressed before it can have widepractical applications.Conclusions<strong>Domestic</strong> buffalo are generally regarded as hav<strong>in</strong>g lowreproductive efficiency. This is largely because of theconditions under which the majority of them are raised,be<strong>in</strong>g smallholder farm<strong>in</strong>g systems with harsh environments,poor nutrition and m<strong>in</strong>imal managerial <strong>in</strong>puts.However, they can have good fertility when managedand fed properly. Modern methods <strong>in</strong> molecular geneticsare help<strong>in</strong>g to unravel the evolutionary and geneticstatus of the river and swamp types of buffalo, but froma practical viewpo<strong>in</strong>t, the disparity <strong>in</strong> the number ofchromosomes <strong>in</strong> the two types needs to be considered <strong>in</strong>cross-breed<strong>in</strong>g programmes <strong>in</strong> order to avoid decl<strong>in</strong>e <strong>in</strong>fertility. Buffalo, cows and bulls are capable of breed<strong>in</strong>gthroughout the year but often show seasonal fluctuations<strong>in</strong> fertility because of climatic and nutritionalfactors that modulate ovarian and testicular functions.The physiology and endocr<strong>in</strong>ology of reproduction <strong>in</strong>buffalo are basically similar to those <strong>in</strong> cattle, but someimportant differences exist that must be considered <strong>in</strong>attempts to improve reproductive efficiency through theuse of modern reproductive technologies. A majorfactor limit<strong>in</strong>g wider uptake of AI by buffalo farmersis the difficulty <strong>in</strong> detect<strong>in</strong>g oestrus. Although improvedprotocols of oestrous synchronization can overcome thisproblem, there are many other factors such as nutritionalstatus, seasonality and reproductive managementthat need to be addressed to achieve success. Embryotechnologies that <strong>in</strong>clude MOET and IVEP have beenvigorously studied over the past two decades, but thesuccess rates rema<strong>in</strong> below that achieved <strong>in</strong> cattlebecause of many <strong>in</strong>herent biological features that areunique to the buffalo. Once the technological problemsare overcome, the successful practical application ofthese methods will need to be preceded by measure toovercome the managerial and nutritional causes of<strong>in</strong>fertility that are common <strong>in</strong> the majority of currentbuffalo farm<strong>in</strong>g systems.ReferencesAwasthi MK, Khare A, Kavani FS, Siddiquee GM, PanchalMT, Shah RR, 2006: Is one-wave follicular growth dur<strong>in</strong>gthe estrous cycle a usual phenomenon <strong>in</strong> water buffaloes(Bubalus bubalis)? Anim Reprod Sci 92, 241–253.Awasthi MK, Kavani FS, Siddiquee GM, Sarvaiya NP,Derashri HJ, 2007: Is slow follicular growth the cause ofsilent estrus <strong>in</strong> water buffaloes? Anim Reprod Sci 99, 258–268.Bahga CS, Khokar BS, 1991: Effect of different seasons onconcentration of plasma lute<strong>in</strong>iz<strong>in</strong>g hormone and sem<strong>in</strong>alquality vis-a` -vis freezability of buffalo bulls (Bubalus bubalis).Int J Biometeorol 35, 222–224.Barile VL, 2005: Review article: improv<strong>in</strong>g reproductiveefficiency <strong>in</strong> female buffaloes. Livest Prod Sci 92, 183–194.Barile VL, Galasso A, Marchiori E, Pacelli C, Montemurro N,Borghese A, 2001: Effect of PRID treatment on conceptionrate <strong>in</strong> Mediterranean buffalo heifers. Livest Prod Sci 68,283–287.Baruselli PS, Mucciolo R, Vis<strong>in</strong>t<strong>in</strong> GA, Viana VC, Arruda RP,Madureira EH, 1997: Ovarian follicular dynamics dur<strong>in</strong>gthe estrous cycle <strong>in</strong> buffalo (Bubalus bubalis). Theriogenology47, 1531–1547.Baruselli PS, Madureira EH, Vis<strong>in</strong>t<strong>in</strong> JA, Barnabe RC,Amaral R, 1999: Timed <strong>in</strong>sem<strong>in</strong>ation us<strong>in</strong>g synchronizationof ovulation <strong>in</strong> buffalo. Rev Bras Reprod Anim 23, 360–362.Baruselli PS, Madureira EH, Vis<strong>in</strong>t<strong>in</strong> JA, Porto-Filho R,Carvalho NAT, Campanile G, Zicarelli L, 2000: Failure ofÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


<strong>Reproduction</strong> <strong>in</strong> <strong>Domestic</strong> Buffalo 205oocytes entry <strong>in</strong>to oviduct <strong>in</strong> superovulated buffalo. Theriogenology53, 491.Baruselli PS, Barnabe VH, Barnabe RC, Vis<strong>in</strong>t<strong>in</strong> JA, Molero-Filho JR, Porto R, 2001: Effect of body condition score atcalv<strong>in</strong>g on postpartum reproductive performance <strong>in</strong> buffalo.Buffalo J 17, 53–65.Boni R, Roviello S, Zicarelli L, 1996: Repeated ovum-pick-up<strong>in</strong> Italian Mediterranean buffalo cows. Theriogenology 46,899–909.Boni R, Rovello S, Gasparr<strong>in</strong>i B, Zicarelli L, 1997: Pregnanciesestablished after transferr<strong>in</strong>g embryos yielded by ovumpick-up and <strong>in</strong> vitro embryo production <strong>in</strong> Italian buffalocows. In: Proceed<strong>in</strong>gs of the Fifth World Buffalo Congress,Caserta, Italy, pp. 787–792.Boonkusol D, Faisaikarm T, D<strong>in</strong>nyes A, Kitiyanant Y, 2007:Effects of vitrification procedures on subsequent developmentand ultrastructure of <strong>in</strong> vitro-matured swamp buffalo(Bubalus bubalis) oocytes. Reprod Fertil Dev 19, 383–391.Borghese A(ed.), 2005: Buffalo Production and Research.FAO Regional Office for Europe, Technical Series 67, Foodand Agriculture Organization, Rome.Brito LF, Satrapa R, Marson EP, Kastelic JP, 2002: Efficacyof PGF(2alpha) to synchronize estrus <strong>in</strong> water buffalo cows(Bubalus bubalis) is dependent upon plasma progesteroneconcentration, corpus luteum size and ovarian follicularstatus before treatment. Anim Reprod Sci 73, 23–35.Campanile G, Boni R, Esposito L, Spadetta M, Zicarelli L,1995: Embryo transfer <strong>in</strong> mediterranean buffalo cow bred <strong>in</strong>Italy. Bubalus bubalis 3, 63.Danell B, 1987: Oestrus Behaviour, Ovarian Morphology andCyclical Variation <strong>in</strong> Follicular System and Endocr<strong>in</strong>ePattern <strong>in</strong> Water Buffalo Heifers. PhD Thesis, SwedishUniversity of Agricultural Sciences, Uppsala.De Rensis F, Lo´pez-Gatius F, 2007: Protocols for synchroniz<strong>in</strong>gestrus and ovulation <strong>in</strong> buffalo (Bubalus bubalis): areview. Theriogenology 67, 209–216.Dobson H, Kamonpatana M, 1986: A review of female cattlereproduction with special reference to a comparison betweenbuffaloes, cows and zebu. J Reprod Fertil 77, 1–36.Drost M, 2007: Advanced reproductive technology <strong>in</strong> thewater buffalo. Theriogenology 68, 450–453.El-Wishy AB, 2007: The postpartum buffalo. II. Acyclicity andanestrus. Anim Reprod Sci 97, 216–36.Harisah M, Azmi TI, Hilmi M, Vidyadaran MK, Bongso TA,Nava ZM, Momongan V, Basrur PK, 1989: Identification ofcrossbred buffalo genotypes and their chromosome segregationpatterns. Genome 32, 999–1002.Huang YJ, Shang JH, Liang MM, Zhang XF, Huang FX,2003: Studies of chromosomal heredity and fertility ofprogenies (2n = 49) crossed between river and swampbuffalo [<strong>in</strong> Ch<strong>in</strong>ese]. Yi Chuan 25, 155–159.Hufana-Duran D, Pedro PB, Ventur<strong>in</strong>a HV, Hufana RD,Salazar AL, Duran PG, Cruz LC, 2004: Post-warm<strong>in</strong>ghatch<strong>in</strong>g and birth of live calves follow<strong>in</strong>g transfer of <strong>in</strong>vitro-derived vitrified water buffalo (Bubalus bubalis) embryos.Theriogenology 61, 1429–1439.Hufana-Duran D, Pedro PB, Ventur<strong>in</strong>a HV, Duran PG, CruzLC, 2007: Full-term delivery of river buffalo calves(2n = 50) from <strong>in</strong> vitro-derived vitrified embryos by swampbuffalo recipients (2n = 48). Livest Sci 107, 213–219.IAEA, 2005: Improv<strong>in</strong>g Artificial Breed<strong>in</strong>g of Cattle andBuffalo <strong>in</strong> Asia: Guidel<strong>in</strong>es and Recommendations. IAEA-TECDOC 1480, International Atomic Energy Agency,Vienna. (http://www-pub.iaea.org/MTCD/publications/PDF/TE_1480_web.pdf)Kierste<strong>in</strong> G, Vall<strong>in</strong>oto M, Silva A, Schneider MP, Iannuzzi L,Brenig B, 2004: Analysis of mitochondrial D-loop regioncasts new light on domestic water buffalo (Bubalus bubalis)phylogeny. Mol Phylogenet Evol 30, 308–324.Kumar S, Nagarajan M, Sandhu JS, Kumar N, Behl V,Nishanth G, 2007: Mitochondrial DNA analyses of Indianwater buffalo support a dist<strong>in</strong>ct genetic orig<strong>in</strong> of river andswamp buffalo. Anim Genet 38, 227–232.Kumaresan A, Ansari MR, Garg A, 2005: Modulation ofpost-thaw sperm functions with oviductal prote<strong>in</strong>s <strong>in</strong>buffaloes. Anim Reprod Sci 90, 73–84.Lei CZ, Zhang W, Chen H, Lu F, Liu RY, Yang XY, ZhangHC, Liu ZG, Yao LB, Lu ZF, Zhao ZL, 2007: Independentmaternal orig<strong>in</strong> of Ch<strong>in</strong>ese swamp buffalo (Bubalus bubalis).Anim Genet 38, 97–102.Liang X, Zhang X, Yang B, Cheng M, Huang F, Pang C, Q<strong>in</strong>gG, Liao C, Wei S, Senatore EM, Bella A, Presicce GA, 2007:Pregnancy and calv<strong>in</strong>g rates follow<strong>in</strong>g transfer of <strong>in</strong>-vitroproducedriver and F1 (river x swamp) buffalo (Bubalusbubalis) embryos <strong>in</strong> recipients on natural oestrus or synchronisedfor ovulation. Reprod Fertil Dev 19, 670–676.Lohan IS, Malik RK, Kaker ML, 2004: Uter<strong>in</strong>e <strong>in</strong>volutionand ovarian follicular growth dur<strong>in</strong>g early postpartumperiod of Murrah buffaloes (Bubalus bubalis). Asian-australasJ Anim Sci 17, 313–316.Lu YQ, Liang XW, Zhang M, Wang WL, Kitiyanant Y, LuSS, Meng B, Lu KH, 2007: Birth of tw<strong>in</strong>s after <strong>in</strong> vitrofertilization with flow-cytometric sorted buffalo (Bubalusbubalis) sperm. Anim Reprod Sci 100, 192–196.Madan ML, Das SK, Palta P, 1996: Application of reproductivetechnology to buffaloes. Anim Reprod Sci 42,299–306.Malfatti A, Barbato O, Tod<strong>in</strong>i L, Terzano GM, DebenedettiA, Borghese A, 2006: Blood testosterone levels <strong>in</strong> ItalianMediterranean buffalo bulls managed <strong>in</strong> two differentbreed<strong>in</strong>g conditions. Theriogenology 65, 1137–1144.Manik RS, Palta P, S<strong>in</strong>gla SK, Sharma V, 2002: Folliculogenesis<strong>in</strong> buffalo (Bubalus bubalis): a review. Reprod FertilDev 14, 315–325.Manik RS, Chauhan MS, Gupta V, S<strong>in</strong>gla SK, Palta P, 2006:In vitro fertilization of oocytes obta<strong>in</strong>ed through transvag<strong>in</strong>aloocyte retrieval from cyclic Murrah buffaloes (Bubalusbubalis). Reprod Fertil Dev 18, 219–219.Meena CR, Das SK, 2006: Development of water buffalo(Bubalus bubalis) embryos from <strong>in</strong> vitro matured oocytesreconstructed with fetal sk<strong>in</strong> fibroblast cells as donor nuclei.Anim Reprod Sci 93, 258–267.Misra AK, Kasiraj R, Rao MM, Reddy NSR, Joshi BV1994:Embryo transfer <strong>in</strong> buffalo <strong>in</strong> India: Progress <strong>in</strong> the last fiveyears. In: Proceed<strong>in</strong>gs of the IVth World Buffalo Congress,Sao Paulo, Brazil, 3, 501–504.Moioli BM, Napolitano F, Puppo S, Barile VL, Terzano GM,Borghese A, Malfatti A, Catalano A, Pilla AM, 1998:Patterns of oestrus, time of LH release and ovulation andeffects of time of artificial <strong>in</strong>sem<strong>in</strong>ation <strong>in</strong> Mediterraneanbuffalo cows. Anim Sci 66, 87–91.Nanda AS, Brar PS, Prabhakar S, 2003: Enhanc<strong>in</strong>g reproductiveperformance <strong>in</strong> dairy buffalo: major constra<strong>in</strong>ts andachievements. Reprod Suppl 61, 27–36.Neglia G, Gasparr<strong>in</strong>i B, Di Palo R, De Rosa C, Zicarelli L,Campanile G, 2003: Comparison of pregnancy rates withtwo estrus synchronization protocols <strong>in</strong> Italian Mediterraneanbuffalo cows. Theriogenology 60, 125–133.Numchaisrika P, Thongphakdee A, Rungsiwiwut R, PruksananondaK, Virutamasen P, Techakumphu M, 2007: Thedevelopment of <strong>in</strong>tra- and <strong>in</strong>ter-species cloned embryosderived from rabbit oocytes: the effect of donor cell sources.Reprod Fertil Dev 19, 153–153.Paul V, Prakash BS, 2005: Efficacy of the ovsynch protocol forsynchronization of ovulation and fixed-time artificial <strong>in</strong>sem<strong>in</strong>ation<strong>in</strong> Murrah buffaloes (Bubalus bubalis). Theriogenology64, 1049–1060.Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


206 BMAO PereraPerera BMAO, 1987: A review of experiences with oestroussynchronisation <strong>in</strong> buffaloes <strong>in</strong> Sri Lanka. Buffalo J Suppl 1,105–114.Perera BMAO, 1999: <strong>Reproduction</strong> <strong>in</strong> water buffalo: comparativeaspects and implications for management. J ReprodFertil Suppl 54, 157–168.Perera BMAO, de Silva LNA, 1985: Gestation length <strong>in</strong><strong>in</strong>digenous (Lanka) and exotic (Murrah) buffaloes <strong>in</strong> SriLanka. Buffalo J 1, 83–87.Perera BMAO, Pathiraja N, Motha MXJ, Weerasekera DA,1979: Seasonal differences <strong>in</strong> plasma testosterone profiles <strong>in</strong>buffalo bulls. Theriogenology 12, 33–38.Perera BMAO, Pathiraja N, Abeywardena SA, Motha MXJ,Abeygunawardena H, 1980: Early pregnancy diagnosis <strong>in</strong>buffaloes from plasma progesterone concentration. Vet Rec106, 104–106.Perera BMAO, Abeygunawardena H, Thamotharam A, K<strong>in</strong>dahlH, Edqvist LE, 1981: Peripartal changes of oestrone,progesterone and prostagland<strong>in</strong> <strong>in</strong> the water buffalo. Theriogenology15, 463–467.Perera BMAO, de Silva LNA, Kuruwita VY, KarunaratneAM, 1987: Postpartum ovarian activity, uter<strong>in</strong>e <strong>in</strong>volutionand fertility <strong>in</strong> <strong>in</strong>digenous buffaloes at a selected villagelocation <strong>in</strong> Sri Lanka. Anim Reprod Sci 14, 115–127.Perera BMAO, Abeygunawardena H, Vale WG, ChantalakhanaC, 2005: Buffalo. In: Livestock and Wealth Creation –Improv<strong>in</strong>g the Husbandry of <strong>Animals</strong> Kept by Poor People<strong>in</strong> Develop<strong>in</strong>g Countries. Owen E, Kitayi A, Jayasuriya N,Smith T (Eds). Livestock Production Programme, NaturalResources International Limited, Nott<strong>in</strong>gham, UK, pp.451–471.Presicce GA, De Santis G, Stecco R, Senatore E, De MauroGJ, Terzano GM, 2001: Foetal gender determ<strong>in</strong>ation byultrasound <strong>in</strong> the Mediterranean Italian buffalo (Bubalusbubalis). Theriogenology 55, 532.Presicce GA, Verberckmoes SB, Senatore EM, Pascale MD,Jeroen Dewulf JB, Van Soom AB, 2004: Assessment of anew utero-tubal junction <strong>in</strong>sem<strong>in</strong>ation device <strong>in</strong> the MediterraneanItalian water buffalo (Bubalus bubalis) under fieldconditions. Bubalus Bubalis 1, 58–64.Presicce GA, Verberckmoes SB, Senatore EM, Kl<strong>in</strong>c P, RathD, 2005: First established pregnancies <strong>in</strong> MediterraneanItalian buffaloes (Bubalus bubalis) follow<strong>in</strong>g deposition ofsexed spermatozoa near the utero tubal junction. ReprodDomest Anim 40, 73–75.Promdireg A, Adulyanubap W, S<strong>in</strong>glor J, Na-Chiengmai A,Techakumphu M, 2005: Ovum pick-up <strong>in</strong> cycl<strong>in</strong>g andlactat<strong>in</strong>g postpartum swamp buffaloes (Bubalis bubalis).Reprod Domest Anim 40, 145–149.Re´vay T, Kova´cs A, Presicce GA, Rens W, Gustavsson I,2003: Detection of water buffalo sex chromosomes <strong>in</strong>spermatozoa by fluorescence <strong>in</strong> situ hybridization. ReprodDomest Anim 38, 377–379.Roy KS, Prakash BS, 2007: Seasonal variation and circadianrhythmicity of the prolact<strong>in</strong> profile dur<strong>in</strong>g the summermonths <strong>in</strong> repeat-breed<strong>in</strong>g Murrah buffalo heifers. ReprodFertil Dev 19, 569–575.Saikhun J, Kitiyanant N, Songtavees<strong>in</strong> C, Pavasuthipaisit K,Kitiyanant Y, 2004: Development of swamp buffalo (Bubalusbubalis) embryos after parthenogenetic activation andnuclear transfer us<strong>in</strong>g serum fed or starved fetal fibroblasts.Reprod Nutr Dev 44, 65–78.Shukla MK, Misra AK, 2007: Effect of bradyk<strong>in</strong><strong>in</strong> on Murrahbuffalo (Bubalus bubalis) semen cryopreservation. AnimReprod Sci 97, 175–179.S<strong>in</strong>g C, 2003: Response of anestrus rural buffaloes (Bubalusbubalis) to <strong>in</strong>travag<strong>in</strong>al progesterone implants and PGF2alpha<strong>in</strong>jection <strong>in</strong> summer. J Vet Sci 4, 137–141.Techakumphu M, Promdireg A, Na-Chiengmai A, PhutikanitN, 2004: Repeated oocyte pick up <strong>in</strong> prepubertal swampbuffalo (Bubalus bubalis) calves after FSH superstimulation.Theriogenology 61, 1705–1711.Usmani RH, Dailey RA, Inskeep EK, 1990: Effects of limitedsuckl<strong>in</strong>g and vary<strong>in</strong>g prepartum nutrition on postpartumreproductive traits of milked buffaloes. J Dairy Sci 73, 1564–1570.Vale WG, 1997: News on reproduction biotechnology <strong>in</strong>males. In: Proceed<strong>in</strong>gs of the Vth World Buffalo Congress,Caserta, Italy, pp. 103–123.Zicarelli L, 1997: Reproductive seasonality <strong>in</strong> buffalo. BubalusBubalis Suppl 4, 29–52.Author’s address (for correspondence): BMA Osw<strong>in</strong> Perera, Faculty ofVeter<strong>in</strong>ary Medic<strong>in</strong>e and Animal Science, University of Peradeniya,Peradeniya 20400, Sri Lanka. E-mail: osw<strong>in</strong>p@pdn.ac.lkConflict of <strong>in</strong>terest: The author declares no conflict of <strong>in</strong>terest.Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 207–212 (2008); doi: 10.1111/j.1439-0531.2008.01163.xISSN 0936-6768Postpartum Ovarian Activity <strong>in</strong> South Asian Zebu CattlePS Brar and AS NandaDepartment of Animal <strong>Reproduction</strong>, Gynaecology and Obstetrics, Guru Angad Dev Veter<strong>in</strong>ary and Animal Sciences University, Ludhiana, Punjab,IndiaContentsTimely onset of postpartum ovarian activity is vital foroptimal reproductive performance of dairy cows. Muchdepends upon genetic constitution of an animal althoughseveral factors <strong>in</strong>terplay to govern the onset of postpartumovarian activity. South Asian zebu cattle have much longerservice period when compared with other exotic or crossbredcattle reared <strong>in</strong> the same Asian environment, which suggestsdifferences <strong>in</strong> their genetic makeup. However, the cows withsame genetic configuration expressed better reproductivepotential when reared under different environment, such as<strong>in</strong> Brazil and Mexico, which suggests the role of extr<strong>in</strong>sicfactors such as management, nutrition, environment anddisease conditions. Better management of animals (provisionof proper shade, water and hous<strong>in</strong>g, efficient oestrous detectionand timely <strong>in</strong>sem<strong>in</strong>ation), good quality nutrition supplementedwith appropriate m<strong>in</strong>erals and vitam<strong>in</strong>s, prevention ofdiseases (vacc<strong>in</strong>ation, deworm<strong>in</strong>g, suitable therapeutic <strong>in</strong>terventions)and application of biotechnology have helped <strong>in</strong>improv<strong>in</strong>g postpartum ovarian activity and, therefore, reproductiveperformance of zebu cattle <strong>in</strong> Asia. No comprehensivestudy appears to have been carried out on the various aspectsof reproduction <strong>in</strong> zebu cattle reared under South Asian socioagro-climaticconditions. This paper is a modest effort tocollect what ever <strong>in</strong>formation available and to critically reviewthe postpartum ovarian activity <strong>in</strong> zebu cattle with specialreference to the effect of the various managemental practicesand pharmacological <strong>in</strong>terventions.IntroductionZebu cattle (Bos <strong>in</strong>dicus), world’s oldest domesticatedcattle, orig<strong>in</strong>ated <strong>in</strong> Western Asia and subsequentlymigrated to larger areas <strong>in</strong> Asia, Africa and South andCentral America. There are approximately 75 recognizedbreeds, of which 30 exist <strong>in</strong> India and Pakistanalone (Chenoweth 1994). Sahiwal, Gir, Amritmahal,Haryana, Rathi, Tharparkar and Nelore are some of themost prom<strong>in</strong>ent breeds found <strong>in</strong> South Asia. Orig<strong>in</strong>at<strong>in</strong>gfrom the wild, most of these are dual purpose breeds;males be<strong>in</strong>g used as draft animals and females for milkproduction. Lately, however, with mechanization ofagricultural operations and commercialization of dairy<strong>in</strong>g,high produc<strong>in</strong>g milk breeds became preferred. So<strong>in</strong>tense has been the need and desire for enhanced milkproduction that crossbreed<strong>in</strong>g of zebu with exotic milkbreeds became fashionable <strong>in</strong> the early sixties andonwards. With this, zebu cattle however faced neglectand their numbers were drastically reduced. Hav<strong>in</strong>gevolved under tropical conditions, zebu were betteradapted to hot and dry climatic conditions (Barcelos etal. 1989), can utilize low-quality roughages and havebetter disease resistance when compared with Bostaurus. Their quality of milk is comparable or evenbetter than <strong>in</strong> crossbred cattle (Joshi et al. 2001). Zebucontributes significantly to draft power and meatproduction <strong>in</strong> many South Asian countries.The reproductive efficiency of Asian zebu (Table 1),however, is much lower than <strong>in</strong> their counterparts <strong>in</strong> thewestern world, the Bos taurus. They have delayed onsetof puberty, poorly expressed oestrus, long <strong>in</strong>ter-calv<strong>in</strong>g<strong>in</strong>tervals (ICI) and dist<strong>in</strong>ct seasonality of reproductivetraits. In particular, they suffer from prolonged postpartumanoestrous periods (Abeygunawardena andDematawewa 2004) which make their rear<strong>in</strong>g uneconomical.Hence, it is important to understand their basicreproductive physiology, identify major impedimentsand devise proper <strong>in</strong>terventions to improve postpartumovarian activity.Physiology of Ovarian ActivityThe developmental and the structural properties of thepre-antral follicles are similar <strong>in</strong> B. <strong>in</strong>dicus and B. taurus.The follicular wave pattern studied <strong>in</strong> some zebu breeds(e.g. <strong>in</strong> Rathi cattle; Gaur and Purohit 2007) revealedtwo (78.57%) and three follicular waves (21.42%) withcharacteristics similar to Bos taurus. Two (6.67%), three(60.00%), four (26.67%) and five (6.67%) follicularwaves have also been reported <strong>in</strong> Gir cows (Viana et al.2000). Certa<strong>in</strong> characteristics of the follicular wave suchas rate of growth and atresia <strong>in</strong> dom<strong>in</strong>ant and subord<strong>in</strong>atefollicles were similar <strong>in</strong> cows with differentnumbers of follicular waves (Table 2). There was nodifference of cycle length (21.11 ± 1.76 and22.25 ± 1.71 days) and circulatory progesterone levelsdur<strong>in</strong>g dioestrus (14.24 ± 4.61 and 16.15 ± 4.45 nM)<strong>in</strong>cows with one or more follicular waves.Some scientists have considered postpartum pituitaryfunctions <strong>in</strong> zebu apparently equivalent to Europeancattle (Chenoweth 1994). However, their reproductiveendocr<strong>in</strong>e profiles are generally lower. The basal (Bo etal. 2003) and GnRH-<strong>in</strong>duced LH release is lower <strong>in</strong> zebuthan <strong>in</strong> exotic breeds of cattle and their crosses (Portilloet al. 2008), although the follicular size and ovulationrates between Angus cows and Angus-Brahman crossesdid not differ.Circulatory progesterone profiles are also normallylower <strong>in</strong> zebu (Baruselli et al. 2004), althoughthe pattern of changes <strong>in</strong> its concentrations dur<strong>in</strong>gdifferent stages of reproduction is similar to that <strong>in</strong>B. taurus. Average serum progesterone concentrationson days 0 (1.30 ± 0.57 nM), 7 (3.87 ± 0.47 nM) and 15(9.41 ± 1.93 nM) of oestrous cycle and on 22 ofpregnancy (11.16 ± 2.13 nM) have been reported(Chakurkar et al. 2004). Ovariectomized zebu cattlerelease progesterone follow<strong>in</strong>g exogenous challengesÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


208 PS Brar and AS NandaZebu breed; country of observationsPostpartumanoestrousperiod (days)Services perconception ICI (days) ReferenceTable 1. Postpartum fertility traits<strong>in</strong> some breeds of zebu cows <strong>in</strong>AsiaPunganur; India 152 ± 14 1.57 ± 0.1 473 ± 18 Bramhaiah et al. (2003)Ongole; India 113 ± 2 1.53 ± 0.1 407 ± 2 Baburao and Rao (1999)Sahiwal; India 292 ± 15 – 582 ± 15 Katiyar et al. (1993)Non-descript <strong>in</strong>digenous; Indonesia 151 ± 56 3.0 – Tjiptosumirat et al. (2007)Non-descript <strong>in</strong>digenous; Malaysia 114 ± 63 2.8 – Nord<strong>in</strong> et al. (2007)Non-descript <strong>in</strong>digenous; Myammar 125 ± 32 1.6 – W<strong>in</strong> et al. (2007)Table 2. Characteristics of follicular waves <strong>in</strong> Rathi cowsParameterFollicular wavesWave onset (days) 2.10 ± 0.4 10.55 ± 0.6Wave duration (days) 13.35 ± 1.7 10.45 ± 1.0Max. diameter (mm) of dom<strong>in</strong>ant follicle 11.75 ± 1.6 14.65 ± 1.2Day of max. diameter (days) 5.00 ± 0.6 17.5 ± 0.5Growth rate (mm ⁄ day) 1.81 ± 0.3 1.16 ± 0.2Growth period (days) 3.10 ± 0.5 8.90 ± 0.8Onset of atresia (days) 10.50 ± 1.1 –Length of atresia (days) 6.50 ± 0.8 –Atresia rate (mm ⁄ day) 0.55 ± 0.2 –Largest subord<strong>in</strong>ate follicle diameter (mm) 7.42 ± 0.6 7.60 ± 0.9with ACTH, <strong>in</strong>dicat<strong>in</strong>g that adrenal glands can be anextra ovarian source of progesterone (Bolanos et al.1997).The overall average oestradiol concentrations on days–1, 0, 5, 10, 14, 18 and 22 of oestrus <strong>in</strong> Rathi cows were29.36 ± 6.90, 37.25 ± 8.00, 18.24 ± 1.58,24.77 ± 4.22, 21.76 ± 5.50, 22.35 ± 3.81 and34.97 ± 7.74 pM, respectively (Purohit et al. 2000).These values are much lower than <strong>in</strong> western breeds,especially dur<strong>in</strong>g oestrus (Dobson 1978). This may bethe cause for a very high <strong>in</strong>cidence of silent oestrus <strong>in</strong>Rathi cows under Indian conditions.Postpartum <strong>Reproduction</strong>Asian zebu cattle have high <strong>in</strong>cidence of postpartumanoestrus (48.7%, Abeygunawardena and Dematawewa2004) lead<strong>in</strong>g to very long ‘open days’ (113–291 days;Table 1). Anoestrous zebu crosses cont<strong>in</strong>ued to havebasal concentrations of FSH (0.75 ± 0.10 nM) andoestradiol-17 b (6.31 ± 0.62 pM; Mondhe et al. 1989)for very long-period postpartum and exhibited the firstprogesterone rise above 2.0 nM by 66.1 ± 6.8 dayspostpartum (Lyimo et al. 2004). In approximately 45%of zebu crosses, the first postpartum oestrous cycles wereof short duration (3.18 nM, which <strong>in</strong>dicated ovulation followed by activeluteal functions, were noticed as early as 25 dayspostpartum <strong>in</strong> Ongole cattle and the first stand<strong>in</strong>goestrus was observed between 40 and 45 dayspostpartum (Venkantanaidu et al. 2007). This suggeststhat under favourable conditions, the Asian zebu cattlehave the potential of breed<strong>in</strong>g as early <strong>in</strong> postpartumperiod as their western counterparts. This has furtherbeen v<strong>in</strong>dicated by the performance of zebu cattle rearedunder relatively better environmental and nutritionalconditions <strong>in</strong> North and South Americas (Chenoweth1994; Bo et al. 2003).Crossbreed<strong>in</strong>g zebu with exotic dairy breedssubstantially improved their milk production but hadvariable effects on reproduction. While Akhtar et al.(2007) reported elongation of ICI <strong>in</strong> crosses of pure cowswith Pakistani <strong>in</strong>digenous breeds, others claimedsignificant improvement <strong>in</strong> their various reproductiontraits, at least <strong>in</strong> F1 generation. The F2 generation with>75% exotic blood, however, started hav<strong>in</strong>g multipletype of reproductive problems. The average ICI<strong>in</strong>creased from 437 ± 28.9 days <strong>in</strong> F1 generation to451 ± 34.3 days <strong>in</strong> F2 onwards with >80% HF<strong>in</strong>heritance (S<strong>in</strong>gh 2005). The gross uter<strong>in</strong>e <strong>in</strong>volution<strong>in</strong> zebu cows was completed by approximately 30 days ofcalv<strong>in</strong>g but was delayed to 35.14 ± 1.21 days <strong>in</strong> cowswith >75% exotic blood (Deshmukh and Kaik<strong>in</strong>i 1990).Endocr<strong>in</strong>e Interventions to Hasten Onset ofPostpartum Ovarian ActivityAssessment of circulatory progesterone profiles has beensuccessfully used for early diagnosis of pregnancy <strong>in</strong>certa<strong>in</strong> South Asian zebu cattle (Boettcher 2007). Thiswould allow early <strong>in</strong>terventions <strong>in</strong> non-pregnant cows tohave overall beneficiary effects on cattle production.Progesterone radioimmunoassay (RIA) was 100accurate <strong>in</strong> diagnos<strong>in</strong>g non-pregnancy and 84–90%pregnancy on 20–24 days after artificial <strong>in</strong>sem<strong>in</strong>ation(AI) <strong>in</strong> Bangladesh, Malaysia, Indonesia and Myanmarzebu cattle (Nord<strong>in</strong> et al. 2007; Shamsudd<strong>in</strong> et al. 2007;Tjiptosumirat et al. 2007).Certa<strong>in</strong> hormonal <strong>in</strong>terventions that have commonlybeen used to improve reproductive efficiency <strong>in</strong> Bostaurus have also been <strong>in</strong>vestigated <strong>in</strong> certa<strong>in</strong> Bos <strong>in</strong>dicusbreeds. Exogenous adm<strong>in</strong>istration of GnRH,progestogens and ⁄ or PGF 2µ hastened the onset ofovarian activity and <strong>in</strong>duced ovulation <strong>in</strong> postpartumanoestrous zebu cows. S<strong>in</strong>gle <strong>in</strong>jection of 250 lg GnRH(ReceptalÒ, Intervet, Holland) <strong>in</strong>duced ovulation <strong>in</strong>Asian zebu cows hav<strong>in</strong>g good body condition score(BCS) and well-grown follicles on the ovariesÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Postpartum Ovarian Activity <strong>in</strong> South Asian Zebu Cattle 209(Abeygunawardena et al. 1992). The results were evenbetter (75% ovulation) when GnRH was <strong>in</strong>jected on theday of lowest vag<strong>in</strong>al impedance (Sahu et al. 2005a)which normally happens under oestrogen <strong>in</strong>fluence,aga<strong>in</strong> suggest<strong>in</strong>g the presence of active follicles <strong>in</strong> suchcases. Adm<strong>in</strong>istration of GnRH <strong>in</strong> Gir cows just beforeAI reduced early embryonic mortality and enhancedpregnancy rates by 20%. The concentration of circulat<strong>in</strong>gprogesterone on days 0, 14 and 22 post-treatment,however, was similar to the untreated controls (0.73 vs0.86; 9.92 vs 10.78 and 16.28 vs 14.72 nM, respectively;Shelar et al. 2002)Application of progestogens, more often <strong>in</strong> the formof norgestomet ear implants (CrestarÒ, Intervet, Holland)embedded subcutaneously for 9–12 days <strong>in</strong> zebucattle yielded variable results. Approximately 35% and60% cows manifested synergistic sexual behaviourbetween 31 and 57 days after norgestomet implantremoval <strong>in</strong> dry and wet seasons, respectively (Medranoet al. 1996). S<strong>in</strong>gh et al. (2002) reported oestrousresponses <strong>in</strong> approximately 71% zebu cows with overallconception rate of up to 83%. Ovulatory oestrus hasalso been reported <strong>in</strong> 90% Deoni cows with conceptionrate of 70% with 1.85 services per conception (Markendeyaand Bharkad 2004). Solano et al. (2001),however, could achieve ovulations <strong>in</strong> only 16% Bos<strong>in</strong>dicus cows follow<strong>in</strong>g norgestomet implants alone.Oestrogen adm<strong>in</strong>istered along with norgestomet implants<strong>in</strong>creased the expression of behavioural oestrusbut had no bear<strong>in</strong>g on ovulation rate. Incorporation ofequ<strong>in</strong>e chorionic gonadotrop<strong>in</strong> (eCG) to progestogentherapy <strong>in</strong>duced oestrus <strong>in</strong> 100% anoestrous zebu cattlewith good BCS (Abeygunawardena et al. 1992).Kathiresan et al. (2001) advocated two simultaneousnorgestomet implants <strong>in</strong>stead of one <strong>in</strong> Kangeyam cows.While one implant <strong>in</strong>creased LH pulsatile release andma<strong>in</strong>ta<strong>in</strong>ed the dom<strong>in</strong>ant follicle, two implants suppressedLH release with pattern ak<strong>in</strong> to a mid-lutealphase and restored dom<strong>in</strong>ant follicle turn over whichwas useful for oestrous synchronization. Oestrus couldalso be synchronized <strong>in</strong> cycl<strong>in</strong>g zebu with two PGF 2µ<strong>in</strong>jections 11 days apart. Serum progesterone profileshowed that 67% and 100% of the animals responded tothe first and the second <strong>in</strong>jection, respectively (Obasi etal. 1999). Ovsynch oestrous synchronization protocol,which proved meritorious <strong>in</strong> European breeds, performedpoorly <strong>in</strong> zebu (Baruselli et al. 2004) but proveduseful <strong>in</strong> <strong>in</strong>duc<strong>in</strong>g ovarian cyclicity <strong>in</strong> some anoestrousBos taurus · Bos <strong>in</strong>dicus crosses under tropical conditions(Ahuja and Montiel 2004).Zebu cows respond well to superovulatory protocolsbe<strong>in</strong>g used <strong>in</strong> European cows. Treatment with FSH as acomponent of the superovulatory protocols generated11.62 ± 1.28 ovulations and recovered 6.25 ± 2.63embryos of which, 2.35 ± 0.96 were viable and transferable.F<strong>in</strong>ally, the subsequent calv<strong>in</strong>g rate <strong>in</strong> the implantedrecipient cows was 27.28% (Chakurkar et al. 2004).Although the results from the use of the aforesaidhormonal regimes were encourag<strong>in</strong>g, yet their adoptionrema<strong>in</strong>ed m<strong>in</strong>imal. Under field conditions <strong>in</strong> Asia, zebucattle are generally reared at zero <strong>in</strong>put system. Poor basaldiet, BCS and general management normally prevalentunder these circumstances are bound to affect overallreproductive performance of the cattle. Besides, highcosts of hormonal <strong>in</strong>terventions is a deterrent. This alongwith lack of <strong>in</strong>terest <strong>in</strong> promotion of <strong>in</strong>digenous zebucows could be the cause of lack of any proved superovulationand embryo transfer programme <strong>in</strong> zebu cattle.Managemental Aspects of the Onset ofPostpartum Ovarian ActivityQuality of management of animals affects postpartumreproduction <strong>in</strong> all species of animals, and so is <strong>in</strong> zebu.Oestrous detection was missed once or several times <strong>in</strong>34.7% of the cows, most likely because of poor oestrousdetection system or high <strong>in</strong>cidence of silent oestrus(Lyimo et al. 2004). Oestrous detection is even worst <strong>in</strong>zebu cows reared by nomads and kept on free graz<strong>in</strong>gpastures. Prevalence of a strong phenomenon of hierarchy<strong>in</strong> zebu cattle may also affect oestrous expressionand mat<strong>in</strong>g <strong>in</strong> subord<strong>in</strong>ate cows (Solano et al. 2004).Body weight, heart girth and height at withers play asignificant role <strong>in</strong> determ<strong>in</strong><strong>in</strong>g social ranks among Rathicows. Dom<strong>in</strong>ance ranks have also been positivelycorrelated (p < 0.05) to lactation yield (Prasad et al.1996). Environmental extremes, especially the summer,are stressful to zebu cows. Scorch<strong>in</strong>g sun restrictsgraz<strong>in</strong>g, thereby affect<strong>in</strong>g body condition and ultimatelyovarian activity (Mitloehner and Laube 2003), cowslower <strong>in</strong> social rank<strong>in</strong>g be<strong>in</strong>g more vulnerable.Long-term exposure of Bos <strong>in</strong>dicus to heat stress haddeleterious effects on follicular dynamics and oocytecompetence (de S Torres-Ju´nior et al. 2008). Provisionof appropriate shade and cool<strong>in</strong>g practices can beemployed to decrease effects of heat stress <strong>in</strong> cattle(Nienaber and Hahn 2007).Dynamics of body weight at calv<strong>in</strong>g, and thereforenutritional status, have great bear<strong>in</strong>g on postpartumovarian activity <strong>in</strong> zebu. Early ga<strong>in</strong> <strong>in</strong> body weightpostpartum was associated with early resumption ofovarian activity <strong>in</strong> Gir cows (Patil and Deshpande1981). Nutritional deficiency, especially dur<strong>in</strong>g latepregnancy and early postpartum period, results <strong>in</strong> majorloss of body weight and prolonged postpartum anoestrus<strong>in</strong> zebu cattle (Montiel and Ahuja 2005). Supplementaryfeed<strong>in</strong>g of UMMB to small zebu cows ofBangladesh <strong>in</strong>itiated ovarian cyclicity by25.50 ± 3.39 days postpartum when compared with>60 days <strong>in</strong> the control group (Ghosh et al. 1993).Similar observations have been reported follow<strong>in</strong>gUMMB supplementation <strong>in</strong> several other South Asiancountries (Boettcher 2007). Supplementary feed<strong>in</strong>g oftrace m<strong>in</strong>erals and vitam<strong>in</strong> E for a period of 2 weeks<strong>in</strong>duced oestrus with<strong>in</strong> 10 days <strong>in</strong> 65% anoestrousDeoni cows and all conceived to 1.9 services perconception (Markandeya et al. 2002).There is a strong bull effect on postpartum fertility,especially <strong>in</strong> feed-supplemented zebu cows. Exposure tobulls shortened the average length of postpartumanoestrus <strong>in</strong> zebu cattle from 119 to 95 days. Nutritionalsupplementation to these further helped their fertility.All such cows exhibited oestrus with<strong>in</strong> 150 days postpartumaga<strong>in</strong>st only 69% controls (Rekwot et al. 2004).Suckl<strong>in</strong>g is known to suppress postpartum fertility <strong>in</strong>most species of mammals and is commonly practiced <strong>in</strong>Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


210 PS Brar and AS Nandalactat<strong>in</strong>g animals throughout Indian subcont<strong>in</strong>ent, especiallyunder small hold<strong>in</strong>g farm<strong>in</strong>g system (S<strong>in</strong>gh et al.2006). The phenomenon of suckl<strong>in</strong>g appears to be more<strong>in</strong>tense <strong>in</strong> zebu cows than <strong>in</strong> crossbred cows. A study onrestricted suckl<strong>in</strong>g revealed that zebu suckled for 36% ofthe graz<strong>in</strong>g time which was significantly longer than <strong>in</strong>crossbred cows. In addition, the duration of eachsuckl<strong>in</strong>g bout and number of suckl<strong>in</strong>g bouts weresignificantly higher <strong>in</strong> zebu cows than <strong>in</strong> crossbred.The duration and frequency of cross-suckl<strong>in</strong>g and <strong>in</strong>tersuckl<strong>in</strong>gdecreased with <strong>in</strong>crease <strong>in</strong> age of calves, andboth behaviour variables were significantly higher forcrossbred calves than for zebu calves. Although zebucows had lower milk yield, the total duration of suckl<strong>in</strong>gand preferential bond<strong>in</strong>g towards their own offspr<strong>in</strong>gswere higher than <strong>in</strong> crossbred cows (Das 1999). Therewas no significant difference <strong>in</strong> the growth rate ofdivergent follicles between suckl<strong>in</strong>g restricted and suckl<strong>in</strong>gnon-restricted cows (0.87 vs 0.93 mm ⁄ day, respectively).However, with 72-h suckl<strong>in</strong>g restriction, 94.7%postpartum cows showed signs of oestrus when comparedwith 46.6% <strong>in</strong> the control group (p < 0.05;Mahecha et al. 2003). Temporary suspension of suckl<strong>in</strong>genhanced the oestrous synchronization success <strong>in</strong> Nelorecows (Moraes et al. 2003).Studies on improv<strong>in</strong>g success of AI <strong>in</strong> Ongole cowsrevealed better conception rates with semen depositeddeep <strong>in</strong>trauter<strong>in</strong>e (73.63%) than <strong>in</strong> body of uterus(63.79%) or mid-cervix (53.68%) (Rao and Naidu2001). Wrong time <strong>in</strong>sem<strong>in</strong>ation (9–15%), a majorcause <strong>in</strong>fertility <strong>in</strong> zebu cows, has been reported <strong>in</strong> themost develop<strong>in</strong>g countries <strong>in</strong>clud<strong>in</strong>g Bangladesh, Ch<strong>in</strong>aand Myanmar. Further, microbial endometritis <strong>in</strong> morethan 11% zebu cows forms other factor limit<strong>in</strong>g thesuccess of AI (Shamsudd<strong>in</strong> et al. 2007).Zebu cattle <strong>in</strong> tropical Asia are normally kept bysmall farmers and often under poor managementalconditions. They are prone to parasitic <strong>in</strong>festations, animportant pathological variable caus<strong>in</strong>g prolongedpostpartum anoestrus <strong>in</strong> zebu cattle. In a study <strong>in</strong>central parts of India, S<strong>in</strong>gh and Saxena (2006) revealedbursate worms <strong>in</strong>festation <strong>in</strong> 100% anoestrous Haryanacows. Substantially, higher proportion of these came<strong>in</strong>to oestrus with<strong>in</strong> 20 days after deworm<strong>in</strong>g (26% vs14% untreated cows). Ecto- and endoparasitism ishighly prevalent <strong>in</strong> zebu cows <strong>in</strong> tropical Asia. Luckily,however, they have better <strong>in</strong>nate immunity aga<strong>in</strong>st theseand other tropical diseases compared with the crossbredcattle (Chenoweth 1994). Their reproduction is, therefore,less likely to be affected by such diseases than theircrossbred counterparts.It is thus evident that the reproductive efficiency ofzebu cows could be improved through provision ofproper nutrition, management, especially dur<strong>in</strong>g periparum,calf wean<strong>in</strong>g and improved oestrous detection(Mugerwa et al. 1991).Conservation of Zebu CattleConservation of zebu cattle is very important ow<strong>in</strong>gto their merits of better disease resistance, thermotoleranceand crude fibre utilization than <strong>in</strong> crossbredand exotic cows. Overall <strong>in</strong>cidence of cl<strong>in</strong>ical reproductivehealth problems was lesser <strong>in</strong> zebu than <strong>in</strong>their crosses with exotic blood (22% vs 36%). Studiesby Mandal et al. (2005) revealed that <strong>in</strong>cidence ofabortion, still birth, premature birth, retention ofplacenta, dystocia and total parturient disorders were3.16%, 2.70%, 0.45%, 7.00%, 0.90% and 14.21%,respectively. More cows suffered from parturientdisorders dur<strong>in</strong>g w<strong>in</strong>ter, followed by summer, andra<strong>in</strong>y seasons. Seasons, however, appeared to have nomajor effect on mortality rate <strong>in</strong> adult zebu cattle(Prasad et al. 2004), ow<strong>in</strong>g perhaps to their <strong>in</strong>natetolerance to extremes of tropical temperature andhumidity. Pre-implantation zebu embryos are lessvulnerable to elevated temperature than are embryosfrom European breeds (Hansen 2004). In response tothreats of global warm<strong>in</strong>g, further development andpropagation of zebu cattle are vital.From the forego<strong>in</strong>g, it is evident that the zebu cattlehas been and is an <strong>in</strong>tegral and vital part of socioeconomiclife, especially <strong>in</strong> rural tropical Asia. However,these animals did not attract the attention of thescientists and the policy makers alike, and postpartumreproductive physiology rema<strong>in</strong>ed unexplored and thezebu unexploited. The fact that several breeds of Asianzebu have performed better <strong>in</strong> American and Australiancont<strong>in</strong>ents, emphasis is needed on studies to developstrategies to exploit the full production potential ofthese breeds with<strong>in</strong> Asia too. As the postpartum periodis the key w<strong>in</strong>dow <strong>in</strong> the entire spectrum of reproductiveprocess, much more work needs to be carried out tounderstand the physiology and the limit<strong>in</strong>g factors sothat appropriate <strong>in</strong>terventions to enhance fertility <strong>in</strong>zebu cattle are developed.ReferencesAbeygunawardena H, Dematawewa CM, 2004: Pre-pubertaland postpartum anoestrus <strong>in</strong> tropical zebu cattle. AnimReprod Sci 82–83, 373–387.Abeygunawardena H, Abayawansa WD, Kaduwela SC, 1992:Postpartum anostrus <strong>in</strong> cattle and buffaloes;experiences withhormonal therapy. In: Proceed<strong>in</strong>gs of 45th Annual Conventionof Sri Lanka Veter<strong>in</strong>ary Association, Colombo, SriLanka 11–12 December 1992, pp 21.Ahuja C, Montiel F, 2004: Induction of ovulation with GnRHand PGF2 alpha <strong>in</strong> lactat<strong>in</strong>g Bos taurus · Bos <strong>in</strong>dicus cows.Acta Vet Hung 52, 501–508.Akhtar T, Lodhi LA, Khanum SA, Rashad MA, Hussa<strong>in</strong> M,2007: Improv<strong>in</strong>g reproductive efficiency <strong>in</strong> an artificial<strong>in</strong>sem<strong>in</strong>ation programme through early non-pregnancydiagnosis, management and tra<strong>in</strong><strong>in</strong>g. In: Boettcher P (ed.),Improv<strong>in</strong>g the Reproductive Management of Dairy CattleSubjected to Artificial Insem<strong>in</strong>ation, Vienna, Austria.IAEA-TECDOC-1533, pp. 103–112.Baburao K, Rao KS, 1999: Reproductive performance ofOngole cattle. Indian J Anim Reprod 20, 120–122.Barcelos AF, Garcia JA, Cardoso RM, Torres CAA, FerreiraBA, Americo GJ, Maciel CR, Alves CA, 1989: Effects ofclimate and diet on the physiological reactions of buffaloes,zebu cattle, Bos taurus and their crosses. 1. Rectal temperatureand respiration rate. Rev Soc Bras Zoot 18, 32–41.Baruselli PS, Reis EL, Marques MO, Nasser LF, Bo GA,2004: The use of hormonal treatments to improve reproductiveperformance of anoestrus beef cattle <strong>in</strong> tropicalclimates. Anim Reprod Sci 82–83, 479–486.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Postpartum Ovarian Activity <strong>in</strong> South Asian Zebu Cattle 211Bo GA, Baruselli PS, Mart<strong>in</strong>ez MF, 2003: Pattern andmanipulation of follicular development <strong>in</strong> Bos <strong>in</strong>dicus cattle.Anim Reprod Sci 78, 307–326.Boettcher P, 2007: Improv<strong>in</strong>g the Reproductive Managementof Dairy Cattle Subjected to Artificial Insem<strong>in</strong>ation, Vienna,Austria. IAEA-TECDOC-1533.Bolanos JM, Mol<strong>in</strong>a JR, Forsberg M, 1997: Effect of bloodsampl<strong>in</strong>g and adm<strong>in</strong>istration of ACTH on cortisol andprogesterone levels <strong>in</strong> ovariectomized zebu cows (Bos<strong>in</strong>dicus).Acta Vet Scand 38, 1–7.Bramhaiah KV, Rao ST, Reddy YR, 2003: <strong>Reproduction</strong> <strong>in</strong>Punjanur cattle. Indian J Anim Reprod 24, 51.Chakurkar EB, Deopurkar VL, Bakshi SA, Ch<strong>in</strong>chkar SR,2004: Success rate of embryo transfer and serum progesteronelevels <strong>in</strong> recipients of gir and its crosses under farmconditions. Indian Vet J 81, 250–254.Chenoweth PJ, 1994: Aspects of reproduction <strong>in</strong> female Bos<strong>in</strong>dicus cattle: a review. Aust Vet J 71, 422–426.Das SM, 1999: Performance and behaviour of the cow and calf<strong>in</strong> restricted suckl<strong>in</strong>g and artificial rear<strong>in</strong>g systems. Acta-Universitatis-Agri-Sueciae-Agraria 145, 36.de S Torres-Ju´nior JR, de FA Pires M, de Sa´ WF, de MFerreira A, Viana JH, Camargo LS, Ramos AA, FolhadellaIM, Polisseni J, de Freitas C, Clemente CA, de Sa´ Filho MF,Paula-Lopes FF, Baruselli PS, 2008: Effect of maternal heatstresson follicular growth and oocyte competence <strong>in</strong> Bos<strong>in</strong>dicus cattle. Theriogenology 69, 155–166.Deshmukh AW, Kaik<strong>in</strong>i AS, 1990: Studies on <strong>in</strong>volution ofuterus <strong>in</strong> relation to postpartum oestrus <strong>in</strong> Sahiwal · Jerseycrossbreds with three levels of <strong>in</strong>heritance. Indian J AnimReprod 11, 122–124.Dobson H, 1978: Plasma gonadotrop<strong>in</strong>s and estradiol dur<strong>in</strong>gestrus <strong>in</strong> the cow. J Reprod Fert 52, 51–53.Gaur M, Purohit GN, 2007: Follicular dynamics <strong>in</strong> Rathi (Bos<strong>in</strong>dicus) cattle. Vetr<strong>in</strong>arski Arhiv 77, 177–186.Ghosh A, Alam MGS, Akbar MA, 1993: Effect of ureamolasses-m<strong>in</strong>eralblock supplementation on postpartumovarian activity <strong>in</strong> Zebu cows. Anim Reprod Sci 31, 61–67.Hansen PJ, 2004: Physiological and cellular adaptations ofzebu cattle to thermal stress. Anim Reprod Sci 82–83, 349–360.Joshi BK, S<strong>in</strong>gh A, Gandhi RS, 2001: Performance evaluation,conservation and improvement of Sahiwal cattle <strong>in</strong> India.Anim Genet Res Inf 31, 43–54.Kathiresan D, Arosh AJ, Devanathan TG, Subramanian A,Na<strong>in</strong>ar MA, Nachimuthu K, 2001: Dose of Norgestometand its effect on the secretary pattern of hormones <strong>in</strong>Kangeyam cows. Indian J Anim Reprod 22, 1–5.Katiyar SS, S<strong>in</strong>gh C, Katiyar MP, 1993: Effect of parity onsome productive and reproductive traits <strong>in</strong> sahiwal cattle(Bos <strong>in</strong>dicus). Indian J Anim Reprod 14, 30–31.Lyimo ZC, Nkya R, Schoonman L, Eerdenburg FJCM, 2004:Postpartum reproductive performance of crossbred dairycattle on smallholder farms <strong>in</strong> sub-humid coastal Tanzania.Trop Anim Health Prod 36, 269–279.Mahecha LL, Henao D, Cardona M, Olivera AM, 2003: Effectof suckl<strong>in</strong>g restriction on heat presentation <strong>in</strong> postpartumzebu cows. Revista Colombiana de Ciencias Pecuarias 16,215–219.Mandal DK, Tayagi S, Mukherjee S, 2005: Seasonal variation<strong>in</strong> the <strong>in</strong>cidence of parturient disorders of Sahiwal cattle.Indian J Anim Reprod 26, 58–59.Markendeya NM, Bharkad GP, 2004: Controlled breed<strong>in</strong>gwith norgestomet ear implant for <strong>in</strong>duction of postpartum oestrus <strong>in</strong> Deoni cows. Indian J Anim Reprod 25,53–54.Markandeya NM, Bhikane AU, Bharkad GP, 2002: Cl<strong>in</strong>icalresponse to microm<strong>in</strong>erals with vitam<strong>in</strong> E supplementation<strong>in</strong> anoestrus Deoni cows. Indian J Anim Reprod 23, 77–78.Medrano EA, Hernandez O, Lamothe C, Gal<strong>in</strong>a CS, 1996:Evidence of asynchrony <strong>in</strong> the onset of signs of oestrus <strong>in</strong>zebu cattle treated with a progestogen ear implant. Res VetSci 60, 51–54.Mitloehner FM, Laube RB, 2003: Chronobiological <strong>in</strong>dicatorsof heat stress <strong>in</strong> Bos <strong>in</strong>dicus cattle <strong>in</strong> the tropics. J Anim VetAdv 2, 654–659.Mondhe DM, Dopurkar VL, Hukeri VB, Mantri AM,H<strong>in</strong>gane VR, Kaik<strong>in</strong>i AS, 1989: Serum FSH and oestradiol-17bconcentrations <strong>in</strong> postpartum anoestrus crossbredcattle. Indian J Anim Reprod 10, 6–9.Montiel F, Ahuja C, 2005: Body condition and suckl<strong>in</strong>g asfactors <strong>in</strong>fluenc<strong>in</strong>g the duration of postpartum anestrus <strong>in</strong>cattle: a review. Anim Reprod Sci 85, 1–26.Moraes TT, Okuda HT, Bianch<strong>in</strong>ni SE, 2003: The calv<strong>in</strong>gafter the first estrous period observed after oestrus synchronizationprotocol associated with the temporary <strong>in</strong>terruptionof suckl<strong>in</strong>g. Revista Brasileira de Reproducao Animal27, 666–673.Mugerwa EM, Tegegne A, Ketema H, 1991: Patterns ofpostpartum oestrus onset and associated plasma progesteroneprofiles <strong>in</strong> Bos <strong>in</strong>dicus cows <strong>in</strong> Ethiopia. Anim ReprodSci 24, 273–284.Nienaber JA, Hahn GL, 2007: Livestock production systemmanagement responses to thermal challenges. Int J Biometeorol52, 149–157.Nord<strong>in</strong> Y, Za<strong>in</strong>i N, Wan Zahari WM (2007) Reproductivestatus follow<strong>in</strong>g artificial <strong>in</strong>sem<strong>in</strong>ation and factors affect<strong>in</strong>gconception rate <strong>in</strong> dairy cows <strong>in</strong> smallholder productionsystems. In: Boettcher P (ed.) Improv<strong>in</strong>g the ReproductiveManagement of Dairy Cattle Subjected to ArtificialInsem<strong>in</strong>ation, Vienna, Austria. IAEA-TECDOC-1533, pp.79–92.Obasi OL, Ogwu D, Mai H, 1999: Efficiency of prostagland<strong>in</strong>F2 alpha <strong>in</strong> the <strong>in</strong>duction of physiological oestrus <strong>in</strong> Zebucattle. J Sus Agri Env 1, 114–118.Patil JS, Deshpande BR, 1981: Study of body weight changesdur<strong>in</strong>g antepartum, parturition and postpartum period <strong>in</strong>Gir cows with special reference to exhibition of postpartumactions. Indian Vet J 58, 376–379.Portillo GE, Bridges GA, de Araujo JW, Shaw MKV, SchrickFN, Thatcher WW, Yelich JV, 2008: Response to GnRH onday 6 of the estrous cycle is dim<strong>in</strong>ished as the percentage ofBos <strong>in</strong>dicus breed<strong>in</strong>g <strong>in</strong>creases <strong>in</strong> Angus, Brangus, andBrahman · Angus heifers. Anim Reprod Sci 103, 38–51.Prasad S, Mittal JP, Kaushish SK, Prasad S, 1996: Dom<strong>in</strong>ancepattern <strong>in</strong> free graz<strong>in</strong>g zebu cattle. Indian J Anim ProdManag 12, 99–103.Prasad S, Ramachandran N, Raju S, 2004: Mortality patterns<strong>in</strong> dairy animals under organized herd managementconditions at Karnal, India. Trop Anim Health Prod 36,645–654.Purohit GN, Datt M, Shama SS, Upadhyaya RC, 2000:Estradol profile of Rathi cattle dur<strong>in</strong>g estrus cycle. Indian JAnim Reprod 21, 6–7.Rao BK, Naidu VG, 2001: Effect of site of semen depositionon conception rate <strong>in</strong> Ongole cows. Indian J Anim Sci 22,60–62.Rekwot PI, Ak<strong>in</strong>pelumi OP, Sekoni VO, Eduvie LO, OyedipeEO, 2004: Effects of nutritional supplementation andexposure to bulls on resumption of post-partum ovarianactivity <strong>in</strong> Bunaji (Bos <strong>in</strong>dicus) cattle. Vet J 167, 67–71.Sahu SK, Tiwari RP, Mishra UK, 2005a: Treatment ofpostpartum anoestrus Sahiwal cows on the basis of VMIand ultrasonography us<strong>in</strong>g s<strong>in</strong>gle <strong>in</strong>jection of GnRH. IndianJ Anim Reprod 26, 99–101.Sahu SK, Tiwari RP, Mishra UK, Mukherejee K, 2005b:Ovarian activity and vag<strong>in</strong>al resistance <strong>in</strong> postpartumanoestrus Sahiwal cows. Indian J Anim Reprod 26, 109–112.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


212 PS Brar and AS NandaShamsudd<strong>in</strong> M, Hosse<strong>in</strong> MS, Siddiqui MAR, Khan AHMSI,Bari FY, Alam MF, Rahman M, Sayem ASM, Momont H,2007: Use of milk progesterone radioimmunoassay andcomputer applications for community based reproductivehealth services <strong>in</strong> smallholder dairy farms of Bangladesh. <strong>in</strong>:Boettcher P (ed.) Improv<strong>in</strong>g the Reproductive Managementof Dairy Cattle Subjected to Artificial Insem<strong>in</strong>ation, Vienna,Austria. IAEA-TECDOC-1533, pp. 9–22.Shelar RR, Deopurkar VL, Bakshi SA, Gulavane SU, 2002:Efficacy of pre-<strong>in</strong>sem<strong>in</strong>ation treatment with GnRH forimprov<strong>in</strong>g conception rate <strong>in</strong> repeat breeder cows. Indian JAnim Reprod 23, 69–70.S<strong>in</strong>gh A, 2005: Crossbreed<strong>in</strong>g of cattle for <strong>in</strong>creas<strong>in</strong>g milkproduction <strong>in</strong> India – A review. Indian J Anim Sci 75,383–390.S<strong>in</strong>gh CP, Saxena A, 2006: Study of effect of anthelm<strong>in</strong>tictreatment on metabolic profile of anoestrus Haryana cows.Indian J Anim Reprod 27, 91–93.S<strong>in</strong>gh U, S<strong>in</strong>gh I, Khar SK, S<strong>in</strong>gh U, S<strong>in</strong>gh I, 2002: Influenceof season, age and postpartum <strong>in</strong>terval on reproductiveparameters of norgestomet treated anestrus zebu cattle.Indian J Anim Reprod 23, 113–116.S<strong>in</strong>gh AK, Brar PS, Nanda AS, Parkash BS, 2006: Effect ofsuckl<strong>in</strong>g on basal and GnRH <strong>in</strong>duced LH release <strong>in</strong>postpartum dairy buffaloes. Anim Reprod Sci 95,244–250.Solano J, Orihuela A, Gal<strong>in</strong>a CS, Montiel F, 2001: Sexualbehavior of Zebu cattle (Bos <strong>in</strong>dicus) follow<strong>in</strong>g estrous<strong>in</strong>duction by Syncro-Mate B, with or without estrogen<strong>in</strong>jection. Physiol Behav 71, 503–508.Solano J, Gal<strong>in</strong>do F, Orihuela A, Gal<strong>in</strong>a CS, 2004: The effectof social rank on the physiological response dur<strong>in</strong>g repeatedstressful handl<strong>in</strong>g <strong>in</strong> Zebu cattle (Bos <strong>in</strong>dicus). Physiol Behav82, 679–683.Tjiptosumirat T, Tuasikal BJ, Murni AP, Lelanan<strong>in</strong>gtyas N,Darwati S, Ariyanto A, Yunita F, Mondrida G, Tr<strong>in</strong><strong>in</strong>gsih ,Setyowati S, Sutari , Toleng AL, Arman C, Rizal Y, 2007:Improvement of the efficiency of artificial <strong>in</strong>sem<strong>in</strong>ationservices through the use of radioimmunoassay and acomputer database application. In: Boettcher P (ed.)Improv<strong>in</strong>g the Reproductive Management of Dairy CattleSubjected to Artificial Insem<strong>in</strong>ation. IAEA-TECDOC-1533,pp. 57–78.Venkantanaidu G, Rao SA, Rao BK, 2007: Progesteroneprofile <strong>in</strong> postpartum lactat<strong>in</strong>g Ongole (zebu) cows. Indian JAnim Reprod 28, 12–14.Viana JHM, Ferreira A de M, Sa WF de, Camargo LS de, AFerreira A de, Sa WF, de A Camargo LS, 2000: Folliculardynamics <strong>in</strong> zebu cattle. Pesqui Agropecu Bras 35, 2501–2509.W<strong>in</strong> N, W<strong>in</strong> YT, Kyi SS, Myatt A, 2007: Evaluation ofreproductive performance of cattle bred by artificial <strong>in</strong>sem<strong>in</strong>ation<strong>in</strong> Myanmar through the use of progesteroneradioimmunoassay. In: Boettcher P (ed.) Improv<strong>in</strong>g theReproductive Management of Dairy Cattle Subjected toArtificial Insem<strong>in</strong>ation, Vienna, Austria. IAEA-TECDOC-1533, pp. 93–102.Author’s address (for correspondence): PS Brar, Department ofAnimal <strong>Reproduction</strong>, Gynaecology and Obstetrics, Guru AngadDev Veter<strong>in</strong>ary and Animal Sciences University, Ludhiana, Punjab,India. E-mail: parkashs<strong>in</strong>gh@satyam.net.<strong>in</strong>Conflict of <strong>in</strong>terest: All authors declare no conflict of <strong>in</strong>terests.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 213–216 (2008); doi: 10.1111/j.1439-0531.2008.01164.xISSN 0936-6768Mother–Offspr<strong>in</strong>g Interactions <strong>in</strong> Zebu CattleMJR Paranhos da Costa 1,2 , A Schmidek 1,3 and LM Toledo 1,41 Grupo ETCO – Grupo de Estudos e Pesquisas em Etologia e Ecologia Animal; 2 Departamento de Zootecnia, FCAV-UNESP; 3 Programa de Po´sGraduaça˜o em Gene´tica e Melhoramento Animal, Faculdade de Cieˆncias Agra´rias e Veter<strong>in</strong>a´rias, UNESP, Jaboticabal; 4 APTA, Po´lo Regional doVale do Ribeira, UPD Registro, Sa˜o Paulo, BrazilContentsThe knowledge of the <strong>in</strong>teraction between mother andoffspr<strong>in</strong>g might contribute to enhance the welfare of theoffspr<strong>in</strong>g and to improve the reproductive efficiency of thecow. However, there is still little <strong>in</strong>formation available aboutsuch <strong>in</strong>teraction <strong>in</strong> some cattle breeds. A series of observationalstudies were set up, address<strong>in</strong>g the mother–offspr<strong>in</strong>grelationships of Nelore, Guzerat and Gyr cattle breeds. Firstly,the behaviour of cows and calves around the time ofparturition was described, and then, the underly<strong>in</strong>g factorsthat affect the calves’ survival and development were studied.Special attention was given to the failure or delay <strong>in</strong> the firstsuckl<strong>in</strong>g. The results together are <strong>in</strong>dicative of geneticvariability for some studied variables, <strong>in</strong>dicat<strong>in</strong>g the possibilityof selection for calf vigour (us<strong>in</strong>g latency to stand up andlatency to suckle as its <strong>in</strong>dicators) and maternal ability (us<strong>in</strong>gpercentage of time <strong>in</strong> contact with the calves), <strong>in</strong> spite of theestimates of heritability were low and presented high standarddeviation for all variables. The <strong>in</strong>dividual variability <strong>in</strong> theirsuckl<strong>in</strong>g behaviour and the efficiency <strong>in</strong> first suckl<strong>in</strong>g cannot beexpla<strong>in</strong>ed by a s<strong>in</strong>gle isolated underly<strong>in</strong>g factor. By now, thereare some results available, although there are many questionswithout answers. The field is still open for the development offuture research.IntroductionThe adaptation of a newborn calf to the extra-uter<strong>in</strong>eenvironment is clearly dependent to the expression ofappropriate behaviour, presented by itself and by itsmother. It must stand up and suckle as soon as possible,while its mother must take care of it, lick<strong>in</strong>g, protect<strong>in</strong>gaga<strong>in</strong>st predators and rema<strong>in</strong><strong>in</strong>g quiet, stand<strong>in</strong>g, whenthe calf tries to get the teat <strong>in</strong>to its mouth and suckle(Lidfors 1994; Fraser and Broom 1997; Paranhos daCosta and Cromberg 1998). The importance of thesebehaviours for the calves’ survival has been reported(Schmidek 2003; Ribeiro et al. 2007), and when amother licks its offspr<strong>in</strong>g it also stimulates the calf’srespiratory and circulatory systems, removes the foetalmembranes from its body and stimulates the defecationand ur<strong>in</strong>ation (Lidfors 1994; Fraser and Broom 1997).The first suckl<strong>in</strong>g must occur soon after calv<strong>in</strong>g, and thelack or delay of it <strong>in</strong>creases the <strong>in</strong>cidence of calfmortality (Paranhos da Costa and Cromberg 1998;Schmidek et al. 2008).Quality and <strong>in</strong>tensity of maternal behaviour dependson a complex <strong>in</strong>teraction among genetic, physiologicalfactors and maternal experience (Edwards and Broom1982; Lawrence and Fowler 1997; Paranhos da Costaand Cromberg 1998).After the recognition of its own offspr<strong>in</strong>g, a cowusually does not allow other calves to suckle. However,some previous studies described the occurrence ofallosuckl<strong>in</strong>g <strong>in</strong> cattle (when a cow allows other calf,than its own, to suckle), rang<strong>in</strong>g from 3.0% (Lewandrowskiand Hurnik 1983; Das et al. 2000) to 19.02% ofthe total suckl<strong>in</strong>g events (Víchova´ and Bartosˇ 2005), butit seems to be rare <strong>in</strong> Zebu cattle (Paranhos da Costaet al. 2006a).Theoretically, these variations could be resultant frommany causal factors, genetic and environmental. This isa vague statement and does not help to understand thebiological phenomenon neither to solve the practicalproblems result<strong>in</strong>g from it. There are some empiricalevidences that, under restricted milk and space conditions,the probability of allosuckl<strong>in</strong>g <strong>in</strong>creases <strong>in</strong> waterbuffalo (Murphey et al. 1991, 1995) and the fact thatallosuckl<strong>in</strong>g <strong>in</strong>creases the competition for milk amongwater buffalo calves (Paranhos da Costa et al. 2000).This statement is, <strong>in</strong> some extension, supported by theresults with cattle, for example, Paranhos da Costa et al.(2006a) studied Zebu cattle kept <strong>in</strong> free-range conditions,the cows were not milked and the calves had freeaccess to suckle, and under these conditions, the authorsdid not record any s<strong>in</strong>gle case of allosuckl<strong>in</strong>g. On thecontrary, the studies of Lewandrowski and Hurnik(1983), Das et al. (2000) and Vı´chova´ and Bartosˇ (2005)reported the occurrence of allosuckl<strong>in</strong>g <strong>in</strong> cattle, andunder their studies conditions, the cows and calves hadsome restriction <strong>in</strong> space and milk availability. In spiteof the research <strong>in</strong>terests on this subject, for theoreticaland practical reasons, the underly<strong>in</strong>g factors of allosuckl<strong>in</strong>gand its role on the rate of calf survival and calfperformance are still not well expla<strong>in</strong>ed.The mother–offspr<strong>in</strong>g relationships of cattle are stillnot well understood <strong>in</strong> many aspects, probably becauseof the complex scenario and the challeng<strong>in</strong>g situations<strong>in</strong>volved on their expression. The aim of this article is topresent some results achieved through the behaviouralstudies of Zebu cattle <strong>in</strong> Brazil and to <strong>in</strong>troduce somepractical recommendations about how to improve themanagement based on these studies.The Importance of the Behavioural Studies toIncrease Reproductive EfficiencyThe reproductive (and economic) efficiency of a cow isdirectly dependent of the survival of its offspr<strong>in</strong>g. Theknowledge of the mother and offspr<strong>in</strong>g behaviour isimportant to identify situations that could <strong>in</strong>crease therisk of calf weakness, abandon or death, giv<strong>in</strong>g theopportunity for one to develop appropriate strategies tom<strong>in</strong>imize these problems and the respective economicÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


214 MJR Paranhos da Costa, A Schmidek and LM Toledolosses (Cromberg and Paranhos da Costa 1997; Schmideket al. 2008).Cows usually isolate themselves from the herd whennear to calve. However, when they are <strong>in</strong> a smallpaddock, they are not able to do it, result<strong>in</strong>g <strong>in</strong> a higherrisk of fail or delay <strong>in</strong> the first suckl<strong>in</strong>g. This is because,under high density conditions, the social <strong>in</strong>teractionsamong parturient cows are more frequent and sometimescalves spend time try<strong>in</strong>g to suckle <strong>in</strong> other cowsthan <strong>in</strong> their own mothers (Ribeiro et al. 2004), justbecause these cows are close to the parturient cow andits respective calf, or because some cows show <strong>in</strong>teresttowards a calf, behav<strong>in</strong>g like its mother.Throughout the parturition process, cows usually lyedown and stand up just after deliver<strong>in</strong>g the calves(Paranhos da Costa and Cromberg 1998). The identificationof this simple behaviour has practical relevance, asthe risk of calf death is higher when the cow delivers <strong>in</strong> astand<strong>in</strong>g (16.1%) rather than <strong>in</strong> a recumbent position(4.2%) (Paranhos da Costa et al. 2006b). Better understand<strong>in</strong>gof this situation and its underly<strong>in</strong>g factors mayassist to solve this problem. Some factors that affect thecows’ postures when deliver<strong>in</strong>g their calves were alreadyidentified, among them: lack of calv<strong>in</strong>g experience,presence of potential predators near the parturient cows(Toledo 2005; Paranhos da Costa 2008).The episodes of aggressive behaviour towards anewborn calf are also important. Accord<strong>in</strong>g to Schmideket al. (2006), they were more frequent <strong>in</strong> primiparousthan <strong>in</strong> multiparous cows (55.7% and 22.0%,respectively), result<strong>in</strong>g on higher frequency of calvesthat did not succeed to suckle after 3 h after birth(15.7% and 5.7%, respectively) and higher latency forfirst suckl<strong>in</strong>g (102.6 and 76.0 m<strong>in</strong>, respectively). The riskof calf mortality also <strong>in</strong>crease with the delay <strong>in</strong> the firstsuckl<strong>in</strong>g latency above 3 h (Schmidek et al. 2006), andby this, we recommend the need of review<strong>in</strong>g the criticallimit for the cattle first suckl<strong>in</strong>g latency, usually reportedas 6 h after calv<strong>in</strong>g (Broom 1983).The presence of potential predators <strong>in</strong> the calv<strong>in</strong>gplace also affects the mother–young relationship. Underour environmental conditions, special attention wasgiven to the black vultures (Coragyps atratus) becausethis species is more and more present at the cattlecalv<strong>in</strong>g places, disturb<strong>in</strong>g the formation of the mother–offspr<strong>in</strong>g bond and attack<strong>in</strong>g directly the calves. Thema<strong>in</strong> effect of the presence of black vulture was todecrease the time that the cows spend <strong>in</strong> contact withtheir calves, as usually they spend more time <strong>in</strong>vigilance. This situation resulted <strong>in</strong> negative effects onthe latencies to stand up and first suckl<strong>in</strong>g (Toledo2005).Weather conditions (Toledo et al. 2007), calf weightat birth, udder shape and teat size (Ventrop andMichanek 1992) are other identified factors that wouldresult <strong>in</strong> fail or delay <strong>in</strong> the first suckl<strong>in</strong>g.Failure <strong>in</strong> the first suckl<strong>in</strong>g <strong>in</strong>creases the mortality rate ofcalvesIn animal husbandry, this subject has crucial importancebecause the death of one animal means an economicalloss (Cromberg and Paranhos da Costa 1997).It is expected that cow–calf behaviour dur<strong>in</strong>g the firsthours after calv<strong>in</strong>g has a direct effect on the calfmortality; by this, the knowledge of the mother–offspr<strong>in</strong>g behaviour has practical significance, becauseit can be used to evaluate the strategies of managementand selection <strong>in</strong> beef cattle. In spite of this, there are notmuch data about this subject, especially when consider<strong>in</strong>gcattle bred <strong>in</strong> Brazil.In one of our studies (Schmidek et al. 2008), the highpercentage of newborn Guzerat calves that fail <strong>in</strong> thefirst suckl<strong>in</strong>g stimulated us to study the genetic andenvironmental factors underly<strong>in</strong>g this situation, and itseffect on calf mortality. Data from 1527 calves (bornbetween 1992 and 2004) revealed that 279 (18.3%) ofthem failed <strong>in</strong> first suckl<strong>in</strong>g and were helped to suckle,more than 6 h after birth. Two major factors wereidentified for the occurrence of the failure <strong>in</strong> the firstsuckl<strong>in</strong>g: cows with big udders or teats and calvesweigh<strong>in</strong>g less than 25 kg at birth. The risk of death forcalves that failed <strong>in</strong> the first suckl<strong>in</strong>g was 28% timeshigher (p < 0.01) than those that were able to suckle ontime. The estimated maternal effect presented lowheritability (0.08) and the correspondent direct effectwas null. These low heritability estimates <strong>in</strong>dicate thatthe genetic progress through selection criteria aga<strong>in</strong>stfailure of the first suckl<strong>in</strong>g might be achieved only <strong>in</strong>long term. However, the conformation of the udder andthe birth weight of the calf are two possible <strong>in</strong>dicatorsfor immediate action to help the calf to suckle as soon aspossible.Rout<strong>in</strong>e <strong>in</strong>spections <strong>in</strong> the maternity area to detecteventual problems with parturient cows and neonates(ideally three times <strong>in</strong> the day: early morn<strong>in</strong>g, noon andlate afternoon) could m<strong>in</strong>imize the <strong>in</strong>cidence of thefailure <strong>in</strong> the first suckl<strong>in</strong>g of newborn calves (Paranhosda Costa 2008; Schmidek et al. 2008).Variability <strong>in</strong> the expression of cow and calf behavioursamong and with<strong>in</strong> breedsThe differences <strong>in</strong> suckl<strong>in</strong>g behaviour seem to beproduced by a complex comb<strong>in</strong>ation of genetic andenvironmental factors, which would result <strong>in</strong> a particularbehavioural relationship style of a mother–offspr<strong>in</strong>gpair (Paranhos da Costa et al. 2006a). There arescientific evidences of variability among Zebu breeds,among herds of the same breed and among <strong>in</strong>dividualswith<strong>in</strong> herd; for example, the latency to stand up afterparturition varied significantly between Nelore andGuzerat breed (Paranhos da Costa and Cromberg1998), between two herds of Nelore breed (Toledo et al.2007) and among several progenies of Nelore bulls(Schmidek 2003).It is usually accepted that the behavioural variationbetween species, breeds, populations and <strong>in</strong>dividuals hasa genetic bases, by this, the most common strategy toidentify genetic variability <strong>in</strong> behaviour is the comparisonamong breeds (Hohenboken 1986; Buchenauer1999). For example, the variation <strong>in</strong> maternal behaviourbetween beef and dairy cows was reported (Le Ne<strong>in</strong>dre1989; Buchenauer 1999), show<strong>in</strong>g that beef cowsdisplayed better maternal behaviour than dairy cows.Besides, beef calves are usually more agile to stand upÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Mother–Offspr<strong>in</strong>g Interactions 215and suckle than the dairy ones. The reason for suchdifference is probably because of selection, as we expectthat dairy cows give milk without the offspr<strong>in</strong>g stimulation.In such conditions, most of the calves’ needs aresupplied by human be<strong>in</strong>gs, which result <strong>in</strong> the relaxationof the selection pressure, turn<strong>in</strong>g the survival of calvesless dependent on its own and mother behaviours.There are few studies estimat<strong>in</strong>g the genetic varianceof mother–offspr<strong>in</strong>g behaviour, probably due ma<strong>in</strong>ly tothe difficulties for data record<strong>in</strong>g. Only three studiesaddress<strong>in</strong>g this specific subject were found. Two of themwith lambs (Cloete et al. 1998, 2002), showed lowcoefficients of heritability (h 2 , direct and maternal) forthe latencies to stand up (h 2 = 0.10 ± 0.05 and0.09 ± 0.04, respectively) and to suckle after stand<strong>in</strong>gup (h 2 = 0.07 ± 0.04 and 0.19 ± 0.04, respectively),and one study with Nelore cattle (Schmidek 2003),which also showed low coefficients of heritability for thelatencies for calves to stand up, to suckle after stand upand to suckle (h 2 = 0.16 ± 0.17, 0.09 ± 0.16 and0.13 ± 0.18, respectively) and the percentages of timespent by cows <strong>in</strong> contact with the calves and disturb<strong>in</strong>gthe calves (h 2 = 0.22 ± 0.14, 0.00 ± 0.12 and0.13 ± 0.18, respectively). These results are suggestive,<strong>in</strong>dicat<strong>in</strong>g the existence of additive genetic variabilityfor, at least, three variables: the latencies to stand upand to suckle, (which could be used as an <strong>in</strong>dicator ofcalf vigour) and the percentage of time that a cow was <strong>in</strong>contact with its calf (which is an <strong>in</strong>dicator of maternalability).ConclusionsThe rate of calf survival is an important <strong>in</strong>dicator ofreproductive efficiency <strong>in</strong> a cattle herd. As reported,there are many factors determ<strong>in</strong><strong>in</strong>g the risk of calfdeath, and the role of most of them is still not wellunderstood. It is necessary to keep on work<strong>in</strong>g to clarifythis subject and understand the relationship among cowand calf behaviour and their reproductive performance.The results presented <strong>in</strong> this paper br<strong>in</strong>g some light onthe subject, and re<strong>in</strong>force the idea that the appropriatedexpression of mother–offspr<strong>in</strong>g behaviour is essential toachieve a high level <strong>in</strong> the reproductive performance.Tak<strong>in</strong>g the results of the behavioural studies <strong>in</strong> account,one can adopt some practical handl<strong>in</strong>g procedures todecrease the risk of fail or delay <strong>in</strong> the first suckl<strong>in</strong>g. Thedef<strong>in</strong>ition of the space availability <strong>in</strong> the birth paddock,for example, allows a cow to move away from the herdjust before birth, and this is important to establish anappropriated attachment between mother and offspr<strong>in</strong>g.Special attention should be given to the first calv<strong>in</strong>gcows, <strong>in</strong>clud<strong>in</strong>g a special paddock for calv<strong>in</strong>g. Theyrequire better environment and more <strong>in</strong>tensive assistanceto achieve good reproductive performance; do<strong>in</strong>gthis, it is possible to decrease the risk of abandonedcalves and first suckl<strong>in</strong>g fail or delay.It is also recommended to organize periodic <strong>in</strong>spections<strong>in</strong> the calv<strong>in</strong>g paddock, ideally three times <strong>in</strong> theday (early morn<strong>in</strong>g, noon and late afternoon). Do<strong>in</strong>gthis, it will be possible to assess the risk for cows andneonates, identify<strong>in</strong>g situations that would result <strong>in</strong>problems for both, and giv<strong>in</strong>g the opportunity to actpreventively, reduc<strong>in</strong>g deaths and optimiz<strong>in</strong>g the reproductiveefficiency of the herd.The results also <strong>in</strong>dicate the possibility to improve thecalf survival through selective breed<strong>in</strong>g, consider<strong>in</strong>gbehavioural traits (percentage of time spent by a cow <strong>in</strong>contact with its offspr<strong>in</strong>g, and the latencies to stand upand to suckle) as <strong>in</strong>dicators of maternal ability and calfvigour, respectively.In conclusion, there is some <strong>in</strong>formation availableabout the mother–offspr<strong>in</strong>g <strong>in</strong>teractions that could beused to improve Zebu cattle reproductive performance.However, the picture is not complete; there are manyquestions without answers and problems to be solved.The field is still open for the development of futureresearch.AcknowledgementsF<strong>in</strong>ancial support was provided by Fapesp (Fundação de Ampaso doEstado de Sa˜ o Paulo) and CNPq (Conselho Nacional de DesenvolvimentoCientifico e Technologico).ReferencesBroom DM, 1983: Cow-calf and sow-piglet behaviour <strong>in</strong>relation to colostrum <strong>in</strong>gestion. Ann Rech Vet 14, 342–348.Buchenauer D, 1999: Genetics of behaviour <strong>in</strong> cattle. In: FriesR, Ruv<strong>in</strong>sky A(eds). The Genetics of the Cattle. CABInternational, Wall<strong>in</strong>gford, pp. 365–390.Cloete SWP, Olivier JJ, Scholtz AJ, 1998: Aspects of lamb<strong>in</strong>gand neonatal behaviour of dual-purpose sheep. In: Proceed<strong>in</strong>gsof the 6th World Congress on Genetic Applied toLivestock Production, Armidale, NSW, Australia, vol. 27,pp. 39–42.Cloete SWP, Scholtz AJ, Gilmour AR, Olivier JJ, 2002:Genetic and environmental effects on lamb<strong>in</strong>g and neonatalbehaviour of Dormer and AS Mutton Mer<strong>in</strong>o lambs. LivestProd Sci 78, 183–193.Cromberg VU, Paranhos da Costa MJR, 1997: Mamandologo, para crescer a receita. Anualpec 97, 215–217.Das SM, Redbo I, Wiktorsson H, 2000: Effect of age of calf onsuckl<strong>in</strong>g behaviour and other behavioural activities of Zebuand crossbred calves dur<strong>in</strong>g restricted suckl<strong>in</strong>g periods.Appl Anim Behav Sci 67, 47–57.Edwards SA, Broom D, 1982: Behavioural <strong>in</strong>teractions ofdairy cows with their newborn calves and the effects ofparity. Anim Behav 30, 525–535.Fraser AF, Broom DM, 1997: Parturient behaviour. In: FraserAF, Broom DM (eds), Farm Animal Behaviour andWelfare. CAB International, Wall<strong>in</strong>gford, UK, pp. 208–218.Hohenboken WD, 1986: Inheritance of behavioural characteristics<strong>in</strong> livestock: a review. Anim Breed Abstr 54, 623–639.Lawrence TLJ, Fowler VR, 1997: Growth of Farm <strong>Animals</strong>.CAB International, Wall<strong>in</strong>gford, 330 pp.Le Ne<strong>in</strong>dre P, 1989: Influence of rear<strong>in</strong>g conditions and breedon social relationships of mother and young. Appl AnimBehav Sci 23, 129–140.Lewandrowski NM, Hurnik JF, 1983: Nurs<strong>in</strong>g and crossnurs<strong>in</strong>gbehaviour of beef cattle <strong>in</strong> conf<strong>in</strong>ement. Can J AnimSci 63, 849–853.Lidfors L, 1994: Mother-young behaviour <strong>in</strong> cattle. Thesis(Doctoral). Swedish University of Agricultural Science,Skara, Sweden.Murphey RM, Paranhos da Costa MJR, Lima LOS, DuarteFAM, 1991: Communal suckl<strong>in</strong>g <strong>in</strong> water buffalo (Bubalusbubalis). Appl Anim Behav Sci 28, 341–352.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


216 MJR Paranhos da Costa, A Schmidek and LM ToledoMurphey RM, Paranhos da Costa MJR, Silva RG, SouzaRC, 1995: Allonurs<strong>in</strong>g <strong>in</strong> river buffalo, Bubalus bubalis:nepotism, <strong>in</strong>competence or thievery? Anim Behav 49,1611–1616.Paranhos da Costa MJR, 2008: Improv<strong>in</strong>g the welfare ofcattle: practical experience <strong>in</strong> Brazil. In: Dawk<strong>in</strong>s MS,Bonney R(eds), The Future of Animal Farm<strong>in</strong>g. BlackwellPublish<strong>in</strong>g, Oxford, pp 145–152.Paranhos da Costa MJR, Cromberg VU, 1998: Relac¸o˜ esmaterno-filiais em bov<strong>in</strong>os de corte nas primeiras horas apo´so parto. In: Paranhos da Costa MJR, Cromberg VU(eds),Comportamento materno em mamı´feros: bases teo´ricas eaplicac¸o˜ es aos rum<strong>in</strong>antes domésticos. Sociedade Brasileirade Etologia, Sa˜ o Paulo, pp. 215–236.Paranhos da Costa MJR, Andriolo A, Oliveira JFS, SchmidekWR, 2000: Suckl<strong>in</strong>g and allosuckl<strong>in</strong>g <strong>in</strong> river buffalo calvesand its relation with weight ga<strong>in</strong>. Appl Anim Behav Sci 66,1–10.Paranhos da Costa MJR, Albuquerque LG, Eler JP, Silva JAII, de V, 2006a: Suckl<strong>in</strong>g behaviour of Nelore, Gir andCaracu calves and their crosses. Appl Anim Behav Sci 101,276–287.Paranhos da Costa MJR, Schmidek A, Toledo LM, 2006b:Boas pra´ticas de manejo: bezerros ao nascimento. EditoraFunep, Jaboticabal-SP, 36 pp.Ribeiro L, Toledo LM, Paranhos da Costa MJR, 2004:Influeˆ ncia de locais do parto no comportamento de vacas ebezerros da raça Nelore. In: ZOOTEC, Brası´lia. Anais.Brası´lia. 1 CD Rom .Ribeiro ARB, Alencar MM, Paranhos da Costa MJR, Negra˜ oJA, 2007: Effects of sire breed-graz<strong>in</strong>g system and environmentalparameters on the behaviour of beef calves just afterbirth. Appl Anim Behav Sci 107, 198–205.Schmidek A, 2003: Ana´lise de fatores genéticos e ambientaisrelacionados a caracterı´sticas de vigor e qualidade materna,para as raças Nelore e Guzera´. Dissertac¸ão de Mestrado,Faculdade de Cieˆ ncias Agrárias e Veter<strong>in</strong>a´rias, UniversidadeEstadual Paulista, Jaboticabal-SP, Brasil.Schmidek A, Paranhos da Costa MJR, Mercadante MEZ,Toledo LM, 2006: The effect of newborn calves vigour <strong>in</strong>their mortality probability. In: Proceed<strong>in</strong>gs of the 40thInternational Congress of the International Society ofApplied Ethology, Bristol, 221 pp.Schmidek A, Mercadante MEZ, Paranhos da Costa MJR,Figueiredo LA, 2008: Falhas na primeira mamada em umrebanho da rac¸a Guzera´: fatores predisponentes, reflexos nasobreviveˆ ncia do bezerro e paraˆ metros genéticos. Rev Brasde Zootec 37, 998–1004.Toledo LM, 2005: Fatores <strong>in</strong>tervenientes no comportamentode vacas e bezerros do parto ate´ a primeira mamada. Tese deDoutorado, Faculdade de Cieˆ ncias Agrárias e Veter<strong>in</strong>a´rias,Universidade Estadual Paulista, Jaboticabal-SP, Brasil.Toledo LM, Paranhos da Costa MJR, Titto EAL, Figueiredo LA,Ablas DS, 2007: Impactos de varia´veis clima´ticas na agilidadede bezerros Nelore neonatos. Ciência Rural 37, 1399–1404.Ventrop M, Michanek P, 1992: The importance of udder andteat conformation for teat seek<strong>in</strong>g by the newborn calf.J Dairy Sci 75, 262–268.Vı´chová J, Bartosˇ L, 2005: Allosuckl<strong>in</strong>g <strong>in</strong> cattle: ga<strong>in</strong> orcompensation? Appl Anim Behav Sci 94, 223–235.Author’s address (for correspondence): Mateus JR Paranhos da Costa,Departamento de Zootecnia, FCAV-UNESP, 14884-900 Jaboticabal-Sa˜ o Paulo, Brazil. E-mail: mpcosta@fcav.unesp.brConflict of <strong>in</strong>terest: All authors declare no conflict of <strong>in</strong>terests.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 217–223 (2008); doi: 10.1111/j.1439-0531.2008.01165.xISSN 0936-6768An Update on <strong>Reproduction</strong> <strong>in</strong> Yak and MithunBS Prakash 1 , M Sarkar 2 and M Mondal 31 Dairy Cattle Physiology Division, NDRI, Karnal, Haryana; 2 National Research Centre on Yak, Dirang, Arunachal Pradesh; 3 National ResearchCentre on Mithun, Jharnapani, Nagaland, IndiaContentsYak and mithun are two domesticated herbivores of higheconomic importance to the farm<strong>in</strong>g community liv<strong>in</strong>g <strong>in</strong>highlands. Improved yak and mithun production couldsignificantly enhance the liv<strong>in</strong>g standards of these highlanders.Over the years, their dw<strong>in</strong>dl<strong>in</strong>g numbers have been a cause ofserious concern. In view of the lack of knowledge on thereproductive physiology of these rum<strong>in</strong>ants, studies have beenundertaken to <strong>in</strong>vestigate their reproductive endocr<strong>in</strong>ology <strong>in</strong>recent years. This paper attempts to present the latest<strong>in</strong>formation on the endocr<strong>in</strong>e changes associated with theirvarious reproductive processes viz. growth and puberty,oestrous cyclicity and oestrous behaviour, ovulation, and,pregnancy and parturition. The paper also provides the recentdevelopments on research done towards the enhancement ofyak and mithun reproductive efficiencies through endocr<strong>in</strong>eand embryo biotechniques.IntroductionThe yak is a unique bov<strong>in</strong>e species domesticated by thehighlanders <strong>in</strong>habit<strong>in</strong>g the high altitude <strong>in</strong>hospitableterra<strong>in</strong>s <strong>in</strong> the middle and <strong>in</strong>ner Himalayan region, andsurvives only at high altitudes between 2500 and6000 m. The world’s total yak population is estimatedto be approximately 14 million of which more than 90%is <strong>in</strong> Ch<strong>in</strong>a (Wiener et al. 2003). India possessesapproximately 70 000 yaks <strong>in</strong> the high reaches ofHimalayan region. The animal is a seasonal breederand the factors which are considered to be responsiblefor seasonality <strong>in</strong> breed<strong>in</strong>g <strong>in</strong>clude nutrition, climate andaltitude. Mithun (Bos frontalis), believed to be thedescendent of wild gaur (Bos gaurus gaurus) of India andMyanmar, is an animal of special significance for thetribals of North Eastern-Hills region of India and isbasically used for beef purpose besides its use <strong>in</strong> bartertrade, marriage gift, etc. Probably, mithun is the onlyrum<strong>in</strong>ant that can browse on biomass most efficiently ondifficult steep hill slopes under adverse climatic conditionprevail<strong>in</strong>g <strong>in</strong> the region. At present, India had atotal population of 0.27 million mithuns (M<strong>in</strong>istry ofAgriculture Department of Animal Husbandry, Dairy<strong>in</strong>g& Fisheries 2003).Survey reports reveal that majority of Indian yaks andmithuns suffer from various reproductive problems likelate maturity, long calv<strong>in</strong>g <strong>in</strong>terval, poor oestrousexpression and repeat breed<strong>in</strong>g; <strong>in</strong> addition yaks areseasonal breeders. The <strong>in</strong>formation related to basicendocr<strong>in</strong>ology is, therefore, paramount not only forbetter understand<strong>in</strong>g of these animals and improv<strong>in</strong>ggrowth, <strong>in</strong>duc<strong>in</strong>g early puberty and improv<strong>in</strong>g itsfertility but also for application of endocr<strong>in</strong>e biotechniquessuch as oestrus synchronization and set timeAI, superovulation and embryo biotechniques forimplementation of ex situ as well as <strong>in</strong> situ conservationof valuable germplasm. The present paper highlightssome of the recent research f<strong>in</strong>d<strong>in</strong>gs on reproductiveendocr<strong>in</strong>ology and subsequent use of various biotechnologicaltools for augment<strong>in</strong>g fertility <strong>in</strong> these twowonderful species.Development of Hormone AssaysWe have developed sensitive enzyme immuno-assay(EIA) methods us<strong>in</strong>g the second antibody coat<strong>in</strong>gtechnique on microtitre plates for the determ<strong>in</strong>ationof LH (Mondal et al. 2005a; Sarkar and Prakash2005a), oxytoc<strong>in</strong> (Mondal et al. 2006d; Sarkar andPrakash 2006), GH (Mondal et al. 2005b; Sarkar et al.2007d), prolact<strong>in</strong> (Sarkar and Prakash 2006; Mondalet al. 2007a), total oestrogen, oestradiol-17b (Sarkaret al. 2006b; Mondal et al. 2007b), cortisol (Sarkar et al.2007b) and PGFM (Mondal et al. 2006a; Sarkar et al.2007c).Growth and Puberty <strong>in</strong> Yak and MithunThe role of GH <strong>in</strong> postnatal somatic growth is wellestablished (Breier and Gluckman 1991). Age at puberty<strong>in</strong>fluences production efficiency <strong>in</strong> animals (Short andBellows 1971; Hoffman and Funk 1992). Factors modulat<strong>in</strong>gthe endocr<strong>in</strong>e regulation of onset of puberty arenot fully understood. Physiological mechanism thatcontrols puberty by chang<strong>in</strong>g <strong>in</strong>terplay between hormonessecreted from the hypothalamus, the anteriorpituitary and the gonads lead to a transition from sexualquiescence to sexual function (Hull and Harvey 2001).Among these complex endocr<strong>in</strong>e <strong>in</strong>teraction, GH acts asa common hormonal l<strong>in</strong>k (Hull and Harvey 2001).Plasma progesterone level which <strong>in</strong>creases with theadvancement of age and <strong>in</strong>creas<strong>in</strong>g body weight isconsidered to be an <strong>in</strong>dicator of puberty <strong>in</strong> domesticanimals (El-Nouty 1971). We found that with <strong>in</strong>creas<strong>in</strong>gage and body weight (BW), GH and GH per 100 kg BWboth decreased and the animal with higher plasma GHand GH per 100 kg BW showed higher growth rates <strong>in</strong>mithuns (Mondal et al. 2007c) and yaks (Sarkar et al.2008).In grow<strong>in</strong>g and adult mithuns, we found that GHpatterns consisted of frequent pulses of vary<strong>in</strong>gamplitude. Growth hormone pulses occurred at anaverage frequency of 0.69 and 0.48 ⁄ h <strong>in</strong> grow<strong>in</strong>g andadult mithuns, respectively (Mondal et al. 2004,2005d). Peak and mean GH levels were associatedpositively (r = 0.98, p < 0.001) with rates of weightga<strong>in</strong>. Results from these studies <strong>in</strong>dicated that the GHÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


218 BS Prakash, M Sarkar and M MondalProgesterone, LH (ng/ml)4.03.53.02.52.01.51.00.50.0Progesterone LH GHGroup-I Group-II Group-III Group-IV Group-V Group-VIGroupsFig. 1. Changes <strong>in</strong> plasma progesterone, LH and GH for consecutive6 weeks <strong>in</strong> mithuns of six different age groups from birth throughadulthood. Group I (0–3 months; n = 9), group II (>3–6 months;n = 7), group III (>6–12 months; n = 11), group IV (>12–18months; n = 10), group V (>18–24 months; n = 10) and group VI(>24–31 months; n = 12). Blood samples were collected from allanimals for six consecutive weekspulses occur at frequent <strong>in</strong>tervals throughout the dayand night and alterations <strong>in</strong> GH levels and patternsare elicited more by pulse amplitude than frequencymodulation. LH pulses occurred at an average rate of0.54 ⁄ h (5 pulses ⁄ 9 h). The mean plasma LH levelwas correlated with body weight (r = 0.82; p < 0.05)and pulse amplitude <strong>in</strong> grow<strong>in</strong>g mithuns (Mondalet al. 2005c). There was higher LH concentrationswith higher pulsatility and greater amplitude <strong>in</strong> prepubertalmithuns than exhibited <strong>in</strong> grow<strong>in</strong>g mithuns(Mondal et al. 2005e). Our results showed <strong>in</strong>creas<strong>in</strong>gtrend of plasma progesterone and LH concentrations<strong>in</strong> the process of onset of puberty <strong>in</strong> mithun (Fig. 1).Plasma GH levels were recorded to be doubled dur<strong>in</strong>gpubertal process (Fig. 1). The optimum LH pulsefrequency and amplitude required for onset of puberty<strong>in</strong> mithun were ‡9 pulses ⁄ 24 h and ‡1.6 ng ⁄ ml,respectively.Possible use of GH-releas<strong>in</strong>g factor as an <strong>in</strong>ducer ofpubertyKeep<strong>in</strong>g <strong>in</strong> view the promis<strong>in</strong>g results obta<strong>in</strong>ed recentlyof us<strong>in</strong>g exogenous GH-releas<strong>in</strong>g hormone (GHRH)for growth enhancement (Mondal and Prakash 2004)and the suggestion of Gelato and Merriam (1986) thatGHRH be<strong>in</strong>g active <strong>in</strong> a wide range of species andhence its treatment could potentially be used toaccelerate growth of animals of commercial importance,we first standardized the dosage of GHRH <strong>in</strong>mithun (Mondal et al. 2006c) and then went on to testthe effects of exogenous GHRH on secretion patternsof GH and LH as a prelim<strong>in</strong>ary study for the first timeever <strong>in</strong> this rum<strong>in</strong>ant species (Mondal et al. 2006e).Our results suggest that 10 lg GHRH per 100 kg BWis the dosage, which can be used for augmentation ofmithun production. Us<strong>in</strong>g this dosage, GHRH wasfound to <strong>in</strong>crease plasma GH and LH pulse frequencyand amplitude with higher mean LH levels <strong>in</strong> grow<strong>in</strong>gmithuns suggest<strong>in</strong>g the possibility of us<strong>in</strong>g such strategyfor enhancement of maturity and ⁄ or pubertyprocess <strong>in</strong> this species.160140120100806040200GH (ng/ml)Endocr<strong>in</strong>e Changes Associated withReproductive Processes <strong>in</strong> Yaks and MithunsRelationship of plasma oestradiol-17b, total oestrogenand progesterone to oestrous behaviour <strong>in</strong> mithun cowsWe carried out studies to (1) establish the characteristicsof oestrous behaviour <strong>in</strong> mithun cows and (2) determ<strong>in</strong>ethe relationships between this behaviour and theplasma concentrations of oestradiol-17b (E2), totaloestrogen and progesterone. Among the behaviouralsigns of oestrus, the cow to be mounted by bull (100%)was the best <strong>in</strong>dicator of oestrus followed by stand<strong>in</strong>g tobe mounted by other females (92% Mondal et al.2006b). Rectal exam<strong>in</strong>ation of the reproductive tractrevealed a relaxed cervix, turgid uterus and ovarieshav<strong>in</strong>g palpable follicles <strong>in</strong> all animals (Mondal et al.2006g). The length of oestrous cycle and duration ofoestrus were recorded to be 21.8 ± 0.69 days and12.6 ± 1.34 h, respectively. E2 and total oestrogenprofiles dur<strong>in</strong>g the peri-oestrous period (Mondal et al.2006f) showed that the mean highest peak concentrationsof E2 (27.29 ± 0.79 pg ⁄ ml) and total oestrogen(45.69 ± 2.32 pg ⁄ ml) occurred at 3.90 ± 2.27 and3.89 ± 2.26 h prior to the onset of oestrus, respectively(Fig. 2a). Plasma progesterone concentration was basal(0.14 ± 0.001 ng ⁄ ml) dur<strong>in</strong>g the peri-oestrous period.Plasma E2 and total oestrogen were found to <strong>in</strong>creasefrom 6 days before oestrus to reach a peak level on theday of oestrus and decl<strong>in</strong>e thereafter to basal level onday 3 of the cycle (Fig. 2b). The plasma progesteroneconcentration was the lowest on the day of oestrusshow<strong>in</strong>g gradual <strong>in</strong>crease to register a peak level on day15 of the cycle.Plasma prolact<strong>in</strong> and oxytoc<strong>in</strong> profiles dur<strong>in</strong>g cyclicity <strong>in</strong>mithun and yak cowsIn yaks the plasma prolact<strong>in</strong> concentration althoughhigh at oestrus, its level fluctuated throughout the cycle,without any def<strong>in</strong>ite trend (Sarkar 2004).In mithuns, the plasma prolact<strong>in</strong> concentration washigh at oestrus and showed fluctuation thereafter withno def<strong>in</strong>ite trend throughout the cycle (Mondal et al.2007a). Results on prolact<strong>in</strong> profile suggest an <strong>in</strong>volvementof prolact<strong>in</strong> <strong>in</strong> the ovulation process <strong>in</strong> this species.The mean plasma oxytoc<strong>in</strong> concentration dur<strong>in</strong>gdifferent days of the oestrous cycle was found to besignificantly different (p < 0.001; Mondal et al. 2006d).The plasma oxytoc<strong>in</strong> concentration was low at oestrus.In mithun cows, two peaks of oxytoc<strong>in</strong> were recorded ondays 6 and 18 of the oestrous cycle.Tim<strong>in</strong>g of ovulation <strong>in</strong> relation to onset of oestrus and LHpeak <strong>in</strong> mithun cowsMondal et al. (2006g) found that the preovulatory LHsurges <strong>in</strong> mithun cows consisted of several pulses(2.92 ± 0.26 pulses ⁄ animal). The peak level of LH for<strong>in</strong>dividual mithun varied from 6.99 ± 0.44 to 12.69 ±2.10 ng ⁄ ml. The concentration of LH dur<strong>in</strong>g surge was10.83 ± 0.76 ng ⁄ ml. The duration of LH surge was6.98 ± 0.22 h. LH surge started 1.23 ± 0.18 h afteronset of oestrus. Ovulation occurred at 26.92 ± 0.31Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


<strong>Reproduction</strong> Augmentation <strong>in</strong> Yak and Mithun 219(a)Estradiol-17b and totalestrogen (pg/ml)403020100E2TE(b)4.5 P4 TE4.03.53.02.52.01.51.00.50.0Progesterone (ng/ml)E2403020100Estradiol-17b, total estrogen(pg/ml)–24–18–12 –60 6 11 17 23 29Periestrous period (h)–6 –4 –2 0 2 4 6 8 10 12 14 16 18 20Days of estrous cycleFig. 2. Mean (±SEM) plasma oestradiol-17b (E2) and total oestrogen (TE) profiles of mithun cows dur<strong>in</strong>g peri-oestrous period. Blood sampleswere collected hourly <strong>in</strong>tervals for 24 h prior to onset of oestrus, at 15-m<strong>in</strong> <strong>in</strong>tervals for 9 h post-onset of oestrus and thereafter at 2 h <strong>in</strong>tervals till31 h post-onset of oestrus (a). Mean plasma E2, TE and progesterone (P4) profiles dur<strong>in</strong>g the different days of oestrous cycle <strong>in</strong> mithun cows(n = 12). Blood samples were collected daily for the entire cycle (b)LH (ng/ml)1086420LHProgesteroneOvulation0 2 4 6 8 10 12 14 16 18 20 22 24 26 28 30 32 34 36 38 40Hours after the onset of estrus1.000.750.500.250.00Fig. 3. Changes <strong>in</strong> the plasma LH and progesterone profile (mean ±SEM) <strong>in</strong> mithun cows (n = 12) after onset of oestrus. Blood sampleswere collected at 15 m<strong>in</strong> <strong>in</strong>tervals after the <strong>in</strong>itial expression of heatsymptoms by the mithuns for 9 h period and thereafter at an <strong>in</strong>tervalof 2 h till 4 h post-ovulation. Ovulation was confirmed by rectalpalpation at 2 h <strong>in</strong>tervalsafter the onset of oestrus and 18.63 ± 0.35 h after theend of LH surge (Fig. 3).Seasonal breed<strong>in</strong>g pattern <strong>in</strong> yaks and associatedendocr<strong>in</strong>e changesYaks are considered seasonal breeders. However, <strong>in</strong>formationabout the breed<strong>in</strong>g season is rather conflict<strong>in</strong>g.The onset and the end of the breed<strong>in</strong>g season areaffected by ecological factors such as climate, grassgrowth, latitude and altitude. Follow<strong>in</strong>g their longperiod of deprivation and weight loss over the w<strong>in</strong>ter,the female yaks come <strong>in</strong>to the breed<strong>in</strong>g season whentemperature and humidity start to rise, and grass beg<strong>in</strong>sto grow which also improves their body condition. InWest Kameng and Tawang districts of ArunachalPradesh <strong>in</strong> India, the breed<strong>in</strong>g season reaches its peak<strong>in</strong> July and August when temperature is at its highestand grass growth is at its best and lasts up to November.Sarkar et al. (2006b) estimated plasma progesterone,total oestrogen and oestradiol-17b profile dur<strong>in</strong>g breed<strong>in</strong>g(July to November) and non-breed<strong>in</strong>g season(February to March) <strong>in</strong> yak. Plasma progesterone wasvery low (£0.2 ng ⁄ ml) at oestrus, thereafter started toProgesterone (ng/ml)rise with a sharp <strong>in</strong>crease dur<strong>in</strong>g the late luteal phaseand reached a peak on day 15–16 of the cycle, decl<strong>in</strong><strong>in</strong>grapidly thereafter to basal levels at oestrus.Dur<strong>in</strong>g non-breed<strong>in</strong>g season, <strong>in</strong> 50% (4 ⁄ 8) of theanimals studied, plasma progesterone level was at basallevel as anticipated. However, cyclic changes <strong>in</strong> plasmaprogesterone were seen <strong>in</strong> three yaks while the plasmaprogesterone level stayed high throughout the sampl<strong>in</strong>gperiod <strong>in</strong> one animal. There were clear <strong>in</strong>dications ofcyclic luteal activity <strong>in</strong> a large proportion of animalseven dur<strong>in</strong>g the non-breed<strong>in</strong>g season, although oestrussymptoms were not exhibited (Sarkar et al. 2006a). Inthe same study, plasma total oestrogen and oestradiol-17b concentrations were high at oestrus and thendecl<strong>in</strong>ed to basal level on day 2 of the cycle and anothersmall elevation was found between days 8 and 12 of thecycle (Sarkar et al. 2006a). This elevation of thehormones suggests the possibility of additional follicularwaves <strong>in</strong> yaks.Circadian rhythmicity <strong>in</strong> circulatory melaton<strong>in</strong> concentrationswas exhibited with low concentrations dur<strong>in</strong>gdaytime and high dur<strong>in</strong>g night <strong>in</strong> both the periodsunder <strong>in</strong>vestigation. Similar observations have beenrecorded <strong>in</strong> cattle (Berthelot et al. 1990), buffalo(Borghese et al. 1994), sheep (Kennaway et al. 1977)and goats (Kloren and Norton 1995). A circadianrhythmicity of prolact<strong>in</strong> release was also seen with amaximum mean concentration at 04:00 h and a m<strong>in</strong>imumat 14:00 h <strong>in</strong> breed<strong>in</strong>g season and with a maximumvalue at 00:00 h and a m<strong>in</strong>imum at 12:00 h <strong>in</strong> nonbreed<strong>in</strong>gmonth. The observations on diurnal prolact<strong>in</strong>pattern <strong>in</strong> yaks seems to be <strong>in</strong> sharp contrast to thoserecorded <strong>in</strong> cattle and buffaloes where higher prolact<strong>in</strong>concentrations have been recorded dur<strong>in</strong>g daytime(S<strong>in</strong>gh and Madan 1993; Gustafson 1994). Diurnalrhythmicity <strong>in</strong> hormonal or biochemical constituents is<strong>in</strong>fluenced by many factors <strong>in</strong>clud<strong>in</strong>g metabolic rate,nutrition and environmental conditions. Positive relationshipof prolact<strong>in</strong> secretion to stress related situationsis also well known (Raud et al. 1971; Tucker 1971).Under extremely cold conditions at night, yaks areexposed to greater stress. The condition is reverseddur<strong>in</strong>g the daytime, under the <strong>in</strong>fluence of sunlight. Thismay be the reason for higher prolact<strong>in</strong> concentrationÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


220 BS Prakash, M Sarkar and M Mondaldur<strong>in</strong>g night times. Sarkar and Prakash (2005c) alsoobserved that the overall plasma prolact<strong>in</strong> levels dur<strong>in</strong>gday as well as night time were found to be more <strong>in</strong> nonbreed<strong>in</strong>gmonth than the correspond<strong>in</strong>g time <strong>in</strong> breed<strong>in</strong>gmonth. This marked <strong>in</strong>crease of prolact<strong>in</strong> level <strong>in</strong> nonbreed<strong>in</strong>gseason may be stress related and associatedwith harsh environmental condition particularly lowtemperature as well as nutritional stress dur<strong>in</strong>g the nonbreed<strong>in</strong>gseason. The relationship between prolact<strong>in</strong> andmelaton<strong>in</strong> secretion <strong>in</strong> blood plasma dur<strong>in</strong>g breed<strong>in</strong>g(November) and non-breed<strong>in</strong>g (February) months was<strong>in</strong>vestigated <strong>in</strong> yaks by Sarkar and Prakash (2005c). Thepositively correlated (0.8 <strong>in</strong> breed<strong>in</strong>g season and 0.7 <strong>in</strong>non-breed<strong>in</strong>g season) circadian rhythms of plasmamelaton<strong>in</strong> and prolact<strong>in</strong> is not clearly understood.T<strong>in</strong>dal (1974) reported that melaton<strong>in</strong> profoundlymodulates prolact<strong>in</strong> release from the adenohypophysis.However, different species <strong>in</strong>terpret the p<strong>in</strong>eal gland’shormonal signal <strong>in</strong> a fundamentally different manner.This prelim<strong>in</strong>ary <strong>in</strong>vestigation opens the gates forresearchers to unravel the <strong>in</strong>terrelationship of thetwo hormones for more comprehensive and detailed<strong>in</strong>vestigation.Tim<strong>in</strong>g of ovulation and plasma LH <strong>in</strong> yakYaks ovulated at 30.5 ± 0.8 h (28–34 h) after theonset of spontaneous oestrus. Preovulatory LH surgeoccurred 3.0 ± 0.7 h after the commencement ofoestrus signs with the surge last<strong>in</strong>g 7.3 ± 0.6 h. Ovulationtook place 20.3 ± 1.0 h after LH surge (18–26 h)(Sarkar and Prakash 2005a).Hormone concentrations dur<strong>in</strong>g pregnancy and periparturientperiod <strong>in</strong> yakProgesterone profiles were similar <strong>in</strong> pregnant and nonpregnantyaks up to 14 days after oestrus; however, <strong>in</strong>pregnant yaks concentrations were significantly higheron day 19 and tended to <strong>in</strong>crease gradually thereafter.Plasma progesterone concentrations decreased rapidlyon day 120, then <strong>in</strong>creased to reach a maximum level onday 210. Concentrations decreased aga<strong>in</strong> 20 days beforeparturition reach<strong>in</strong>g basal levels at parturition. Oestradiol-17blevels <strong>in</strong> plasma and milk <strong>in</strong>creased graduallyuntil day 23 after conception, decreased abruptly on day60, and <strong>in</strong>creased aga<strong>in</strong> to reach a maximum level atparturition. Oestradiol concentrations decreased aga<strong>in</strong>after parturition to reach similar levels as measureddur<strong>in</strong>g mat<strong>in</strong>g (Yu et al. 1993).Profiles of progesterone, cortisol and 13,14-dihydro-15-keto-PGF 2a (PGFM) <strong>in</strong> blood plasma of yaks dur<strong>in</strong>gperi-parturient period were determ<strong>in</strong>ed (Sarkar et al.2007b,c). Plasma PGFM level showed an <strong>in</strong>creas<strong>in</strong>gtrend beg<strong>in</strong>n<strong>in</strong>g day 4 prior to parturition (0.73 ± 0.16ng ⁄ ml) and <strong>in</strong>creased steeply thereafter to reach a peaklevel (17.16 ± 1.31 ng ⁄ ml) on the day of parturition.The levels, then, decl<strong>in</strong>ed sharply on day 1 post-partumto reach 1.20 ± 0.40 ng ⁄ ml and thereafter decl<strong>in</strong>edgradually over the days to reach 0.28 ± 0.09 ng ⁄ ml onday 20 post-partum and this level was ma<strong>in</strong>ta<strong>in</strong>ed withfluctuation with<strong>in</strong> narrow limits thereafter till conclusionof the blood sampl<strong>in</strong>g on day 90 post-calv<strong>in</strong>g.Plasma cortisol level showed an <strong>in</strong>creas<strong>in</strong>g trend beg<strong>in</strong>n<strong>in</strong>gday 2 prior to parturition (2.36 ± 0.65 ng ⁄ ml) and<strong>in</strong>creased steeply thereafter to reach a peak level(26.65 ± 5.28 ng ⁄ ml) on the day of parturition. Thelevels, then, decl<strong>in</strong>ed gradually over the days andreached 2.36 ± 0.25 ng ⁄ ml on day 3 post-partum, andthis level was ma<strong>in</strong>ta<strong>in</strong>ed with fluctuation with<strong>in</strong> narrowlimits thereafter till conclusion of the blood sampl<strong>in</strong>g onday 7 post-calv<strong>in</strong>g. The plasma progesterone concentrationon day 7 before parturition was high(2.10 ± 0.10 ng ⁄ ml). The hormone concentrationdecreased gradually followed by an abrupt fall on theday of parturition (0.24 ± 0.04 ng ⁄ ml).Endocr<strong>in</strong>e Biotechniques to Enhance FertilitySilent or non-detected oestrus is one of the majorcontributory factors <strong>in</strong> both yak and mithun reproductionlead<strong>in</strong>g to poor reproductive efficiency <strong>in</strong> theseanimals (Sarkar and Prakash 2005a; Mondal et al.2006g).Synchronization of oestrus us<strong>in</strong>g PGF 2a <strong>in</strong> mithunsWe tried synchronization of oestrus us<strong>in</strong>g double<strong>in</strong>jection of PGF 2a at 11 days apart <strong>in</strong> cyclic mithuncows. Plasma LH characteristics and time of ovulation<strong>in</strong> respect to LH peak and onset of oestrus <strong>in</strong> mithuncows synchronized for oestrus with PGF 2a are presented<strong>in</strong> Table 1. All the cyclic animals that were subjected tooestrus synchronization us<strong>in</strong>g PGF 2a responded to thetreatment. The <strong>in</strong>tensity of oestrus <strong>in</strong> the mithun cowssynchronized with double <strong>in</strong>jection of PGF 2a wascomparable to that of spontaneous oestrus (MondalM, Rajkhowa C, and Prakash BS., unpublished data).Synchronization of oestrus us<strong>in</strong>g CIDR <strong>in</strong> mithunsExperiments were conducted to synchronize oestrusus<strong>in</strong>g CIDR <strong>in</strong> cyclic and post-partum mithun cows. Inboth categories of animal, synchronized oestrus us<strong>in</strong>gCIDR showed more prom<strong>in</strong>ent behavioural signs thanspontaneous heat (unpublished data). More <strong>in</strong>terest<strong>in</strong>gly,application of CIDR on day 45–50 after parturition<strong>in</strong>duced first post-partum oestrus immediatelyafter uter<strong>in</strong>e <strong>in</strong>volution (day 53–58 post-parturition).Unlike other bov<strong>in</strong>es, mithun cows exhibit firstpost-partum oestrus at approximately day 102 ± 19.6post-partum.Table 1. Plasma LH characteristics and tim<strong>in</strong>g of ovulation <strong>in</strong>mithuns subjected to two <strong>in</strong>jection of PGF 2a at 11 days apart foroestrus synchronizationParameters Mean ± SEM RangeHighest LH peak concentration (ng ⁄ ml) 12.54 ± 1.37 7.34–22.65Duration of LH surge (h) 14.76 ± 1.47 10–19Time from:Second PGF 2a <strong>in</strong>jection to onset of oestrus 43.52 ± 5.93 36–58Onset of oestrus to onset of LH surge (h) 2.45 ± 0.43 1.5–3.25Onset of oestrus to ovulation (h) 33.95 ± 1.41 27–39After end of LH surge to ovulation (h) 20.45 ± 0.89 17–24After end of LH surge to ovulation (h) 20.45 ± 0.89 17 to 24Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


<strong>Reproduction</strong> Augmentation <strong>in</strong> Yak and Mithun 221Application of Ovsynch protocol <strong>in</strong> yaks and mithunsThe Ovsynch protocol (Pursley et al. 1995) was developedas a breed<strong>in</strong>g strategy to elim<strong>in</strong>ate the need foroestrus detection. This method of oestrus synchronizationwas based on control of both ovulation andfollicular growth (Pursley et al. 1995). The protocol iscomposed of an <strong>in</strong>jection of GnRH at random stages ofoestrous cycle to synchronize a new follicle waveemergence. Seven days later PGF 2a is given to regressthe orig<strong>in</strong>al and the newly formed corpora lutea (CL),followed by a second GnRH <strong>in</strong>jection 48 h later to<strong>in</strong>duce a synchronous ovulation 24–32 h later thatallows for fixed time artificial <strong>in</strong>sem<strong>in</strong>ation 12–16 hafter the second GnRH <strong>in</strong>jection.We tested the efficacy of ovulation synchronization <strong>in</strong>yaks (Sarkar and Prakash 2005b) and mithuns (unpublisheddata) us<strong>in</strong>g the Ovsynch protocol. Ovulation wasdetected by rectal palpation at 2 h <strong>in</strong>tervals from the<strong>in</strong>itial signs of oestrus till ovulation. Ovsynch protocolfor synchronization of ovulation produced a response of87.5% and 100% <strong>in</strong> yak and mithun, respectively.Ovulation time and LH characteristics follow<strong>in</strong>govsynch protocol <strong>in</strong> mithun is presented <strong>in</strong> Table 2.The LH peak values among <strong>in</strong>dividual yaks rangedfrom 10.2 to 40 ng ⁄ ml (n = 8) with a mean of22.8 ± 5.1 ng ⁄ ml. LH concentration (mean ± SEM)<strong>in</strong>creased steeply from 0.98 ± 0.66 ng ⁄ ml at the time ofsecond GnRH <strong>in</strong>jection to peak value of16.04 ± 4.30 ng ⁄ ml 2 h post-GnRH adm<strong>in</strong>istrationdropp<strong>in</strong>g sharply thereafter to basal levels of£0.31 ng ⁄ ml. The duration of LH peak (mean ±SEM) was 4.69 ± 0.36 h with a range of 2.75 to 5.75 h.In seven yaks, the circulatory levels of plasmaprogesterone were basal (£0.2 ng ⁄ ml) at the time ofsecond GnRH adm<strong>in</strong>istration, and rema<strong>in</strong>ed basal tillovulation. It was concluded that the Ovsynch protocolcould be applied successfully for fixed time AI <strong>in</strong> yakand mithun.Application of Heatsynch protocol <strong>in</strong> yaksHeatsynch is a newly developed synchronization protocolthat uses the less expensive hormone oestradiolcypionate (ECP) <strong>in</strong> place of the second GnRH <strong>in</strong>jection<strong>in</strong> Ovsynch protocol (Lopes et al. 2000).We tested the efficacy of <strong>in</strong>duction of oestrus,synchronization of ovulation and timed artificial <strong>in</strong>sem<strong>in</strong>ation<strong>in</strong> anoestrous yaks us<strong>in</strong>g the Heatsynch protocol(Sarkar et al. 2007e). All the animals responded toHeatsynch treatment for <strong>in</strong>duction of oestrus andsynchronization of ovulation. The high degree ofTable 2. Plasma LH characteristics and tim<strong>in</strong>g of ovulation <strong>in</strong>mithuns (n = 23) subjected to Ovsynch protocolParameters Mean ± SEM RangeHighest LH peak concentration (ng ⁄ ml) 12.23 ± 0.66 9.03–17.22Duration of LH surge (h) 8.25 ± 1.38 6–12Time from:Onset of LH surge after second1.90 ± 0.44 1.25–2.75GnRH <strong>in</strong>jection (h)Ovulation after second GnRH <strong>in</strong>jection (h) 26.75 ± 2.02 19–33Ovulation after end of LH surge (h) 18.62 ± 1.69 15–27ovulation synchronization could be attributed to thehighly synchronized LH peaks <strong>in</strong> the treated animals. Inanoestrous yaks treated with the Heatsynch protocoland subjected to TAI 40% pregnancy rates wererecorded. Heatsynch protocol could therefore be successfullyutilized for improv<strong>in</strong>g fertility <strong>in</strong> anoestrusyaks too.Superovulation and embryo transferSuperovulatory treatments are widely used <strong>in</strong> embryotransfer programs to <strong>in</strong>crease the supply of embryosfrom animals of superior genetic merit. In a trial byZagdsuren et al. (1997) three female yaks were superovulatedus<strong>in</strong>g progesterone and FSH. Oestrus wasdetected by a teaser male and the females were <strong>in</strong>sem<strong>in</strong>ated24–36 h after the last FSH <strong>in</strong>jection. Numbers ofCL and ovarian follicles detected by rectal palpationand laparoscopy averaged 5.0 ± 0.6 and 1.3 ± 0.9respectively, but embryo recovery was not reported.Davaa et al. (2000) used FSH and PMSG to <strong>in</strong>ducesuperovulation <strong>in</strong> yak cows. Oestrus was detected34.1 ± 0.52 h after the prostagland<strong>in</strong> treatment. Theaverage number of ovarian follicles was 5.4 ± 0.65, ofwhich 4.5 ± 0.43 ovulated.Sarkar et al. (2006a) used four yaks for superovulationfollow<strong>in</strong>g Ovsynch program. On day 20 after thefirst GnRH <strong>in</strong>jection, females received a total dose of200 mg Folltrop<strong>in</strong>, twice daily, equal doses, over 4 daysperiod <strong>in</strong> association with two <strong>in</strong>jections of prostagland<strong>in</strong>analogues on 48 and 60 h after <strong>in</strong>itiation ofsuperovulation. The females were mated and the numberof CL was recorded on day 7 after the last <strong>in</strong>jectionof FSH. Only one animal was flushed non-surgically forembryo recovery 7 days after mat<strong>in</strong>g. The averagenumber of palpable CL was 2 ± 0.71. A total of threeembryos were recovered on non-surgical flush<strong>in</strong>g from as<strong>in</strong>gle animal. One embryo was transferred to a recipientyak cow, which produced one female yak calf after258 days. This is the first yak calf reported throughembryo transfer technology. In another trial cyclic yaks(n = 10) were synchronized us<strong>in</strong>g Ovsynch protocol.On day 10 after expected oestrus, animals received atotal of 200 mg Folltrop<strong>in</strong>, twice daily over 4 days.From 9 yaks, 27 ovulations were detected and 16embryos were recovered (Sarkar et al. 2007a). Plasmaprogesterone profiles from <strong>in</strong>dividual yaks suggestedthat a poor superovulatory response <strong>in</strong> terms of embryorecovery <strong>in</strong> some animals was caused by the lysis of CLbefore flush<strong>in</strong>g which was carried out 7 days aftersuperovulatory ooestrus. It was suggested that flush<strong>in</strong>g5 days post-superovulatory oestrus could improve thesuperovulatory response <strong>in</strong> this species.ConclusionsWe have presented here our major research efforts onsome aspects of reproduction <strong>in</strong> mithuns and yaks. Our<strong>in</strong>itial efforts on develop<strong>in</strong>g hormone assay procedureshave helped us to successfully extend recent endocr<strong>in</strong>etechniques for fertility improvement <strong>in</strong> these species.The results of these studies hold promise for undertak<strong>in</strong>gthese techniques on a larger scale.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


222 BS Prakash, M Sarkar and M MondalHowever, extensive research work is also required onelucidation of hormone receptor <strong>in</strong>terplay at the targetsite to understand the precise molecular role of hormones<strong>in</strong> regulation of reproductive cyclicity <strong>in</strong> thesespecies. An understand<strong>in</strong>g of hormonal <strong>in</strong>terplay atreceptor level is essential for alleviat<strong>in</strong>g reproductiveproblems of endocr<strong>in</strong>e orig<strong>in</strong> which <strong>in</strong> turn will furtherimprove the efficiency of reproduction.ReferencesBerthelot XM, Lautentie JP, Ravault J, Ferney J, Touta<strong>in</strong> PL,1990: Circadian profile and production rate of melaton<strong>in</strong> <strong>in</strong>the cow. Domest Anim Endocr<strong>in</strong>ol 7, 315–319.Borghese A, Barile VL, Tergaro GM, Pilla AM, ParmegganiA, 1994: Melaton<strong>in</strong> trend dur<strong>in</strong>g seasons <strong>in</strong> heifers andbuffalo cows. Proceed<strong>in</strong>gs of the 4th Buffalo Congress, SaoPaulo, Brazil, 27–30 June, Vol. 3, pp. 528–530.Breier BH, Gluckman PD, 1991: The regulation of postnatalgrowth: nutritional <strong>in</strong>fluences on endocr<strong>in</strong>e pathways andfunction of the somatotrophic axis. Lives Prod Sci 27, 77–94.Davaa M, Altankhuag N, Zagdsuren Y, 2000: The prelim<strong>in</strong>aryexperiment to <strong>in</strong>duce superovulation <strong>in</strong> female yaks. Int YakNews Lett 5, 78.El-Nouty F, 1971: The effect of different systems before andafter wean<strong>in</strong>g on age at puberty and age at first conception<strong>in</strong> buffalo heifers. MSc Thesis, Faculty of Agriculture, A<strong>in</strong>Shams University, Egypt.Gelato MC, Merriam GR, 1986: Growth hormone releas<strong>in</strong>ghormone. Ann Rev Physiol 48, 569–591.Gustafson GM, 1994: Effect of changes <strong>in</strong> light on hormonalsecretion and milk production of dairy cows <strong>in</strong> earlylactation. Acta Agric Scand 44, 160–168.Hoffman PC, Funk DA, 1992: Applied dynamics of dairyreplacement growth and management. J Dairy Sci 75, 2504–2515.Hull KL, Harvey S, 2001: Growth hormone: roles <strong>in</strong> femalereproduction. J Endocr<strong>in</strong>ol 168, 1–23.Kennaway DJ, Frith RG, Phillipou G, Seamark RW, 1977:Patterns of progesterone, melaton<strong>in</strong>, and prolact<strong>in</strong> secretion<strong>in</strong> ewes ma<strong>in</strong>ta<strong>in</strong>ed <strong>in</strong> four different photoperiods. J Endocr<strong>in</strong>ol97, 229–242.Kloren WRL, Norton BW, 1995: Melaton<strong>in</strong> and fleece growth<strong>in</strong> Australian cashmere goats. Small Rum<strong>in</strong> Res 17, 179–185.Lopes FL, Arnold DR, Williams J, Pancarci SM, Thatcher MJ,Drost M, Thatcher WW, 2000: Use of estradiol cypionate fortimed <strong>in</strong>sem<strong>in</strong>ation. J Dairy Sci 83, 216 (Abstr).M<strong>in</strong>istry of Agriculture Department of Animal Husbandry,Dairy<strong>in</strong>g & Fisheries, 2003: Seventeenth Livestock Census,Government of India.Mondal M, Prakash BS, 2004: Effects of long-term growthhormone-releas<strong>in</strong>g factor (GRF) adm<strong>in</strong>istration on patternsof GH and LH secretion <strong>in</strong> grow<strong>in</strong>g female buffaloes(Bubalus bubalis). <strong>Reproduction</strong> 127, 45–55.Mondal M, Dhali A, Rajkhowa C, Prakash BS, 2004:Secretion patterns of growth hormone <strong>in</strong> grow<strong>in</strong>g captivemithuns (Bos frontalis). Zool Sci 21, 1125–1129.Mondal M, Prakash B, Rajkhowa C, Prakash BS, 2005a:Development and validation of a simple, sensitive, secondantibody format enzyme immunoassay (EIA) for LHdeterm<strong>in</strong>ation <strong>in</strong> mithun (Bos frontalis) plasma. J ImmunImmunochem 26, 157–167.Mondal M, Rajkhowa C, Prakash BS, 2005b: Standardizationand validation of a simple, sensitive, second antibody formatenzyme immunoassay (EIA) for GH determ<strong>in</strong>ation <strong>in</strong>mithun (Bos frontalis) plasma. Zoo Biol 24, 483–493.Mondal M, Rajkhowa C, Prakash BS, 2005c: Secretionpatterns of lute<strong>in</strong>iz<strong>in</strong>g hormone <strong>in</strong> grow<strong>in</strong>g captive mithuns(Bos frontalis). Reprod Biol 5, 227–235.Mondal M, Rajkhowa C, Prakash BS, 2005d: Twenty-fourhourrhythmicity of growth hormone <strong>in</strong> captive adultmithuns (Bos frontalis). Biol Rhythm Res 36, 255–262.Mondal M, Rajkhowa C, Prakash BS, 2005e: Twenty-fourhoursecretion patterns of lute<strong>in</strong>iz<strong>in</strong>g hormone <strong>in</strong> captiveprepubertal female mithuns (Bos frontalis). Gen ComEndocr<strong>in</strong>ol 144, 197–203.Mondal M, Meyer HHD, Rajkhowa C, Prakash BS, 2006a:Highly sensitive second antibody format enzymeimmunoassayfor determ<strong>in</strong>ation of 13,14-dihydro-15-keto-PGF 2a <strong>in</strong>blood plasma of mithun (Bos frontalis). Prost Other LipidMed 80, 100–109.Mondal M, Rajkhowa C, Prakash BS, 2006b: Behavioralestrous signs can predict the time of ovulation <strong>in</strong> mithun(Bos frontalis). Theriogenology 66, 1391–1396.Mondal M, Rajkhowa C, Prakash BS, 2006c: Determ<strong>in</strong>ationof effective dosage of GH-releas<strong>in</strong>g factor for blood GHresponses <strong>in</strong> Mithun (Bos frontalis). J Anim Physiol AnimNutr 90, 453–458.Mondal M, Rajkhowa C, Prakash BS, 2006d: Development ofa biot<strong>in</strong>–streptavid<strong>in</strong> amplified enzyme immunoassay foroxytoc<strong>in</strong> and its application dur<strong>in</strong>g milk ejection andreproductive cycle <strong>in</strong> the mithun (Bos frontalis). Zool Sci23, 633–639.Mondal M, Rajkhowa C, Prakash BS, 2006e: Exogenous GHreleas<strong>in</strong>ghormone <strong>in</strong>creases GH and LH secretion <strong>in</strong>grow<strong>in</strong>g mithuns (Bos frontalis). Gen Com Endocr<strong>in</strong>ol149, 197–204.Mondal M, Rajkhowa C, Prakash BS, 2006f: Relationship ofplasma estradiol-17b, total estrogen and progesterone toestrous behavior <strong>in</strong> mithun (Bos frontalis) cows. HormBehav 49, 626–633.Mondal M, Rajkhowa C, Prakash BS, 2006g: Oestrousbehavior and tim<strong>in</strong>g of ovulation <strong>in</strong> relation to onset ofoestrus and LH peak <strong>in</strong> mithun (Bos frontalis) cows. ReprodDomest Anim 41, 479–484.Mondal M, Rajkhowa C, Prakash BS, 2007a: Development andvalidation of a highly sensitive economic enzymeimmunoassayfor prolact<strong>in</strong> determ<strong>in</strong>ation <strong>in</strong> blood plasma of mithun(Bos frontalis) and its application dur<strong>in</strong>g milk let down andcyclicity. Anim Reprod Sci 99, 182–195.Mondal M, Rajkhowa C, Prakash BS, 2007b: Highly sensitivesecond antibody format enzymeimmunoassays for determ<strong>in</strong>ationof estradiol-17b and total estrogen <strong>in</strong> blood plasmaof mithun (Bos frontalis). Zool Sci 24, 408–413.Mondal M, Rajkhowa C, Prakash BS, 2007c: Plasma growthhormone concentration <strong>in</strong> mithun (Bos frontalis) of differentages: relation to age and body weight. J Anim Physiol AnimNutr 91, 68–74.Pursley JR, Mee MO, Wiltbank MC, 1995: Synchronization ofovulation <strong>in</strong> dairy cows us<strong>in</strong>g PGF 2a and GnRH. Theriogenology44, 915–923.Raud HR, Kiddy CA, Odell WD, 1971: The effect of stressupon the determ<strong>in</strong>ation of serum prolact<strong>in</strong> by radioimmunoassay.Proc Soc Expt Biol Med 136, 689–704.Sarkar M, 2004: Endocr<strong>in</strong>e changes dur<strong>in</strong>g cyclicity and tim<strong>in</strong>gof ovulation dur<strong>in</strong>g spontaneous and synchronised estrus <strong>in</strong>yaks. PhD Thesis, Submitted to National Dairy ResearchInstitute, Karnal, Haryana, India.Sarkar M, Prakash BS, 2005a: Tim<strong>in</strong>g of ovulation <strong>in</strong> relationto onset of estrus and LH peak <strong>in</strong> yak (Poephagus grunniensL.). Anim Reprod Sci 86, 353–362.Sarkar M, Prakash BS, 2005b: Synchronisation of ovulation <strong>in</strong>yaks (Poephagus grunniens L.) us<strong>in</strong>g PGF 2a and GnRH.Theriogenology 63, 2494–2503.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


<strong>Reproduction</strong> Augmentation <strong>in</strong> Yak and Mithun 223Sarkar M, Prakash BS, 2005c: Circadian variations <strong>in</strong> plasmalevels of melaton<strong>in</strong> and prolact<strong>in</strong> dur<strong>in</strong>g breed<strong>in</strong>g and nonbreed<strong>in</strong>gseasons <strong>in</strong> yak (Poephagus grunniens L.). AnimReprod Sci 90, 149–162.Sarkar M, Prakash BS, 2006: Application of sensitiveenzymeimmunoassays for oxytoc<strong>in</strong> and prolact<strong>in</strong> determ<strong>in</strong>ation<strong>in</strong> blood plasma of yaks (Poephagus grunniens L.)dur<strong>in</strong>g milk let down and cyclicity. Theriogernology 65,499–516.Sarkar M, Borah S, Chakraborty P, Deka BC, Sharma BC,Duttaborah B, Borah S, Baruah K, Ramesha KP, PourochottamneR, Kataktakware M, Sarvanan BC, SenguptaDH, Das S, Prakash BS, Bhattacharya M, 2006a: Calvesborn from anestrus yak (Poephagus grunniens L.) us<strong>in</strong>govsynch and superovulation treatment. Zool Sci 23, 721–725.Sarkar M, Meyer HHD, Prakash BS, 2006b: Is the yak(Poephagus grunniens L.) really a seasonal breeder? Theriogenology65, 721–730.Sarkar M, Chakraborty P, Sharma BC, Deka BC, DuttaborahBK, Mohanty TK, Prakash BS, 2007a: Assessment ofsuperovulatory responses <strong>in</strong> terms of palpable corpora luteaand embryo recovery us<strong>in</strong>g plasma progesterone <strong>in</strong> yaks(Poephagus grunniens L.). Res Vet Sci, <strong>in</strong> press.Sarkar M, Das BC, Duttaborah BK, Kumar V, Mohan K,Bhattacharya M, Prakash BS, 2007b: Application of sensitiveenzymeimmunoassay for determ<strong>in</strong>ation of cortisol <strong>in</strong>blood plasma of yaks (Poephagus grunniens L.). Gen CompEndocr<strong>in</strong>ol 154, 85–90.Sarkar M, Duttaborah BK, Kumar V, Mohan K, Prakash BS,2007c: Plasma concentrations of 13,14-dihydro-15-keto-PGF 2a and progesterone dur<strong>in</strong>g periparturient period <strong>in</strong>yaks (Poephagus grunniens L.). Zoo Sci (communicated).Sarkar M, Nandankar UA, Duttaborah BK, Bhattacharya M,Prakash BS, 2008. Plasma growth hormone concentrations<strong>in</strong> female yak (Poephagus grunniens L.) of different ages:relations with age and body weight. Lives Sci 115, 313–318.Sarkar M, Sengupta DH, Duttaborah B, Rajkhoa J, Bora S,Bandopadhyay S, Ghosh M, Ahmed FA, Saikia P, MohonK, Prakash BS, 2007e: Efficacy of heatsynch protocol for<strong>in</strong>duction of estrus, synchronization of ovulation and timedartificial <strong>in</strong>sem<strong>in</strong>ation <strong>in</strong> yaks (Poephagus grunniens L.).Anim Reprod Sci 104, 306–312.Short RE, Bellows RA, 1971: Relationships between weightga<strong>in</strong>s, age at puberty, and reproductive performance <strong>in</strong>heifers. J Anim Sci 32, 127–131.S<strong>in</strong>gh J, Madan ML, 1993: RIA of prolact<strong>in</strong> as related tocircadian changes <strong>in</strong> buffaloes. Buffalo J 9, 159–164.T<strong>in</strong>dal JS, 1974: Hypothalamic control of secretion and releaseof prolact<strong>in</strong>. J Reprod Fertil 39, 437–461.Tucker HA, 1971: Hormonal response to milk<strong>in</strong>g. J Anim Sci32, 137–146.Wiener G, Jianl<strong>in</strong> H, Ruijun L, 2003: The Yak, 2nd edn. RAPpublication 2003 ⁄ 06, FAO, Bangkok, Thailand.Yu SJ, Huang TM, Chen BX, 1993: Reproductive patterns ofthe yak. I. Reproductive phenomena of the female yak. BrVet J 149, 579–583.Zagdsuren Y, Davaa M, Magash A, 1997: Superovulation <strong>in</strong>female yaks. In: Proceed<strong>in</strong>gs of the 2nd InternationalCongress on Yak, X<strong>in</strong><strong>in</strong>g, Ch<strong>in</strong>a, pp. 193–194.Author’s address (for correspondence): BS Prakash, Dairy CattlePhysiology Division, NDRI, Karnal-13200, Haryana, India. E-mail:bsp1001@rediffmail.comConflict of <strong>in</strong>terest: The authors declare no conflict of <strong>in</strong>terests.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 224–231 (2008); doi: 10.1111/j.1439-0531.2008.01166.xISSN 0936-6768Follicle Development <strong>in</strong> MaresFX Donadeu 1 and HG Pedersen 21 Rosl<strong>in</strong> Institute, Royal (Dick) School of Veter<strong>in</strong>ary Studies, University of Ed<strong>in</strong>burgh, Ed<strong>in</strong>burgh, UK; 2 Veter<strong>in</strong>ary <strong>Reproduction</strong> and Obstetrics,Department of Large Animal Sciences, Faculty of Life Sciences, University of Copenhagen, Copenhagen, DenmarkContentsThe mare provides a unique experimental model for study<strong>in</strong>gfollicle development <strong>in</strong> monovular species. Development ofantral follicles <strong>in</strong> horses is characterized by the periodic growthof follicular waves which often <strong>in</strong>volve the selection of a s<strong>in</strong>gledom<strong>in</strong>ant follicle. If properly stimulated, the dom<strong>in</strong>ant folliclewill complete development and eventually ovulate a fertileoocyte. Regulation of follicular wave emergence and follicleselection <strong>in</strong>volves an <strong>in</strong>terplay between circulat<strong>in</strong>g gonadotrop<strong>in</strong>sand follicular factors that ensures that <strong>in</strong>dividual folliclesare properly stimulated to grow (or to regress) at any givenstage of follicular wave development. Periodic development offollicular waves cont<strong>in</strong>uously occurs dur<strong>in</strong>g most of post-natallife <strong>in</strong> the mare and is <strong>in</strong>fluenced by factors such as stage ofoestrous cycle, season, pregnancy, age, breed and <strong>in</strong>dividual sothat different types of follicular waves (m<strong>in</strong>or or major,ovulatory or anovulatory) and different levels of activitywith<strong>in</strong> waves may develop under different physiologicalconditions. Changes <strong>in</strong> gonadotrop<strong>in</strong> levels and ⁄ or <strong>in</strong> thesensitivity of follicles to circulat<strong>in</strong>g gonadotrop<strong>in</strong>s seemto account largely for these physiological variations <strong>in</strong> follicledevelopment.IntroductionAlthough scientific <strong>in</strong>terest <strong>in</strong> equ<strong>in</strong>e follicles existed bythe 1920s (Seaborne 1925), detailed studies on equ<strong>in</strong>efollicle dynamics did not beg<strong>in</strong> until 50 years later underthe pioneer<strong>in</strong>g lead of OJ G<strong>in</strong>ther at the University ofWiscons<strong>in</strong> (reviewed <strong>in</strong> G<strong>in</strong>ther 1979). Dur<strong>in</strong>g the early1980s, ultrasonography became available for equ<strong>in</strong>etheriogenology (Palmer and Driancourt 1980). Transrectalultrasonography comb<strong>in</strong>ed with systemic hormonemeasurements were subsequently used extensivelyto understand equ<strong>in</strong>e follicle dynamics. The more recent<strong>in</strong>troduction (mid-1990s) of the technique of ultrasoundguidedtransvag<strong>in</strong>al ovarian puncture provided scientistsunprecedented access to the live equ<strong>in</strong>e ovary andallowed the target<strong>in</strong>g of <strong>in</strong>dividual follicles for collectionof follicle samples, <strong>in</strong>jection of test substances orexperimental manipulation of follicles (Gastal et al.1997; Gerard and Monget 1998; Donadeu and G<strong>in</strong>ther2001; Martoriati et al. 2003). The experimental use ofthese procedures has led to extraord<strong>in</strong>ary advances <strong>in</strong>the knowledge of equ<strong>in</strong>e ovarian physiology dur<strong>in</strong>g thelast 15 years (reviewed <strong>in</strong> Beg and G<strong>in</strong>ther 2006;Donadeu and Watson 2007). Two important outcomesof this knowledge have been an improvement of reproductiveefficiency <strong>in</strong> mares (Squires 2006) and a greaterunderstand<strong>in</strong>g of follicular physiology <strong>in</strong> monovularspecies <strong>in</strong> general. In regard to the latter, considerationof the mare as a useful model for study<strong>in</strong>g ovarianprocesses dur<strong>in</strong>g human health and disease has recently<strong>in</strong>creased (G<strong>in</strong>ther et al. 2004c; Carnevale 2008).This review summarizes current knowledge on equ<strong>in</strong>efollicle development. Information on pre-antral andearly antral follicles is particularly scarce <strong>in</strong> the horse;therefore, relatively more focus has been placed on thelate stages of antral follicle development up to the preovulatorystage. Details on physiological and practicalaspects of the ovulatory process and subsequent formationof the corpus luteum can be found elsewhere(G<strong>in</strong>ther 1992; Boerboom and Sirois 2001; Squires 2006).Preantral Stages of Follicle DevelopmentDur<strong>in</strong>g early fetal life, primordial germ cells migrate tothe primitive gonadal ridge where they proliferate andsubsequently enter meiotic division to become arrested<strong>in</strong> prophase I as primary oocytes (G<strong>in</strong>ther 1992).Meiotic arrest cont<strong>in</strong>ues until atresia or until theprimary oocyte is stimulated by an LH surge dur<strong>in</strong>gpost-natal life. Based on histological studies of thedevelop<strong>in</strong>g fetal horse ovary, meiotic divisions beg<strong>in</strong> atabout day 70 of pregnancy (Deanesly 1975). Widespreadatresia characterizes the first oocytes to enter the meioticphase between days 73 and 150 of pregnancy with apeak at approximately day 100. Development of oocytes<strong>in</strong>to primordial follicles is not apparent until day 150(Deanesly 1975) and the number of primordial folliclescont<strong>in</strong>ues to <strong>in</strong>crease dur<strong>in</strong>g subsequent fetal life so thatseveral thousand of these follicles are conta<strong>in</strong>ed with<strong>in</strong>the ovaries at birth (G<strong>in</strong>ther 1992). Proliferation of thesomatic cell layer surround<strong>in</strong>g the primary oocyte leadsto the sequential development of primordial follicles<strong>in</strong>to primary, secondary and, f<strong>in</strong>ally, antral follicles,a process that beg<strong>in</strong>s <strong>in</strong> the fetus and cont<strong>in</strong>uesthroughout post-natal life. It is not known how folliclesare selected to grow from the pool of primordial, rest<strong>in</strong>gfollicles <strong>in</strong> the mare. Adult equ<strong>in</strong>e ovaries were reportedto conta<strong>in</strong> approximately 36 000 primordial follicles and100 grow<strong>in</strong>g follicles at any one time, with largevariability <strong>in</strong> actual follicle numbers between <strong>in</strong>dividualmares (Driancourt et al. 1982). Comparatively higherfollicle numbers (threefold) were found <strong>in</strong> the adultbov<strong>in</strong>e ovary. Proliferative activity of small pre-antralfollicles <strong>in</strong> pony mares was higher at the beg<strong>in</strong>n<strong>in</strong>g of theovulatory season than dur<strong>in</strong>g anoestrus or at the end ofthe ovulatory season, whereas follicular atresia wasunaffected by season (Driancourt et al. 1983).Antral Follicle Development and FollicularWavesThe equ<strong>in</strong>e follicle develops an antrum when it reachesapproximately 0.3 mm <strong>in</strong> diameter (Driancourt et al.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Follicles and Mares 2251982). Similar to pre-antral stages, little is known on thedevelopment of antral follicles before they reachapproximately 2 mm, the smallest diameter that canusually be detected by transrectal ultrasonography. Ithas been reported that the growth of an equ<strong>in</strong>e folliclefrom 0.1 to 1 mm takes approximately two oestrouscycles (Driancourt 1979) and that atresia dur<strong>in</strong>gthis early phase of antral development is rare (G<strong>in</strong>ther1992).As <strong>in</strong> other farm species and humans, the developmentof antral follicles <strong>in</strong> the horse is characterized bythe periodic growth of cohorts of follicles or follicularwaves (Sirois et al. 1989; Bergfelt and G<strong>in</strong>ther 1993).Characterization of follicular waves has <strong>in</strong>volved theultrasonic day-to-day identification of <strong>in</strong>dividual follicles(Bergfelt and G<strong>in</strong>ther 1993; Gastal et al. 1997) andthe use of a statistical method that avoids the need toma<strong>in</strong>ta<strong>in</strong> the identities of <strong>in</strong>dividual follicles dur<strong>in</strong>gserial ultrasound exam<strong>in</strong>ations (G<strong>in</strong>ther and Bergfelt1992; Donadeu and G<strong>in</strong>ther 2002b). Follicular waves<strong>in</strong> the horse can be identified <strong>in</strong> relation to follicles2 mm <strong>in</strong> diameter and larger; however, it is not knownwhether earlier antral and pre-antral stages are alsocharacterized by wave-like patterns of growth,a question that has also not been clarified <strong>in</strong> otherspecies (Mizunuma et al. 1999; McGee and Hsueh2000). Follicular waves and their regulation with anemphasis on normal oestrous cycles will be described <strong>in</strong>this and the next section. Follicular wave patternscharacteristic of other reproductive states will bedescribed <strong>in</strong> a separate section.Characteristics of a follicular wave. Follicular waveemergence has been normally def<strong>in</strong>ed for experimentalpurposes as occurr<strong>in</strong>g when the largest follicle reaches 6or 13 mm, depend<strong>in</strong>g on the study (G<strong>in</strong>ther et al.2003a). Identification of wave emergence often requiresday-to-day ultrasonic evaluation of the ovary usuallyafter aspiration of all follicles from previous waves.A follicular wave <strong>in</strong>itially <strong>in</strong>volves the simultaneousgrowth of a variable number of follicles at a commonrate of between 2 and 3 mm ⁄ day. A recent study<strong>in</strong>volv<strong>in</strong>g ablation of all follicles dur<strong>in</strong>g the middle ofan oestrous cycle <strong>in</strong> pony mares reported a mean ofapproximately 12 follicles emerg<strong>in</strong>g dur<strong>in</strong>g the commongrowth phase of the ablation-<strong>in</strong>duced wave (Gastalet al. 2004). Approximately two follicles emerged eachday dur<strong>in</strong>g the first 4 days after wave emergence with aprogressive decrease <strong>in</strong> the numbers of follicles emerg<strong>in</strong>gthereafter. The exact number of follicles emerg<strong>in</strong>g with<strong>in</strong>waves is affected by factors such as season (Donadeuand G<strong>in</strong>ther 2003).The phase of common follicle growth is followed bythe selection of a s<strong>in</strong>gle follicle (occasionally twofollicles) which is manifested as a deviation <strong>in</strong> diameterbetween the two largest follicles of the wave beg<strong>in</strong>n<strong>in</strong>gwhen the largest follicle reaches approximately 22 mm(G<strong>in</strong>ther et al. 2003a). Deviation beg<strong>in</strong>s a mean of7 days before ovulation and is characterized by thecont<strong>in</strong>uous growth (at an unchanged rate) of the largest(selected) follicle as a dom<strong>in</strong>ant follicle and thesimultaneous cease <strong>in</strong> growth and subsequent regressionof smaller, subord<strong>in</strong>ate follicles. Once it grows toapproximately 35–45 mm, the dom<strong>in</strong>ant follicle normallyeither ovulates or ceases to grow and beg<strong>in</strong>s toregress, depend<strong>in</strong>g on whether an ovulatory LH surgeoccurs. The establishment of dom<strong>in</strong>ance is mediated bya differential <strong>in</strong>crease <strong>in</strong> trophic support to the largestfollicle of a wave by mechanisms that will be expla<strong>in</strong>ed<strong>in</strong> more detail <strong>in</strong> the next section. This leads to profoundchanges <strong>in</strong> follicular cell function that are necessary forthe eventual full maturation of the follicle to anovulatory-competent state and that are reflected <strong>in</strong>dramatic changes <strong>in</strong> global gene expression, changesthat have begun to be characterized <strong>in</strong> other species,most notably cattle (Sisco et al. 2003; Fayad et al. 2004;Mihm et al. 2008).Dom<strong>in</strong>ance does not seem to be a pre-determ<strong>in</strong>edtrait among the follicles grow<strong>in</strong>g <strong>in</strong> a wave becausefollicle ablation studies have demonstrated that allfollicles have similar capacity to become dom<strong>in</strong>ant,a capacity that <strong>in</strong> subord<strong>in</strong>ate follicles is lost with<strong>in</strong>approximately 48 h after the beg<strong>in</strong>n<strong>in</strong>g of deviation(Gastal et al. 2004). The same studies showed that <strong>in</strong>approximately 61% of waves the first follicle toemerge at 6 mm ma<strong>in</strong>ta<strong>in</strong>s its size advantage oversmaller follicles through the common growth phaseand becomes dom<strong>in</strong>ant. The likelihood of the largestfollicle becom<strong>in</strong>g dom<strong>in</strong>ant <strong>in</strong>creases as it approachesthe expected diameter at the beg<strong>in</strong>n<strong>in</strong>g of deviation(Gastal et al. 2004). In a few <strong>in</strong>stances, yet, the largestfollicle ceases or slows down its growth dur<strong>in</strong>g thecommon growth phase and is replaced by the secondlargest follicle (or sometimes even a smaller follicle)which then becomes the dom<strong>in</strong>ant follicle.Types of follicular waves. Follicular waves have beenclassified as major waves (referred to <strong>in</strong> the literatureand throughout this review simply as follicular waves)or m<strong>in</strong>or waves, depend<strong>in</strong>g on whether they <strong>in</strong>volve thedevelopment of a readily identifiable dom<strong>in</strong>ant follicleor they produce only smaller follicles, respectively(G<strong>in</strong>ther 1993; Donadeu and G<strong>in</strong>ther 2002b). Thenumber of follicular waves dur<strong>in</strong>g an oestrous cyclevaries between species. In the horse, as <strong>in</strong> humans, onlyone or two major follicular waves develop dur<strong>in</strong>g eachcycle (G<strong>in</strong>ther et al. 2004c). A major wave (namedprimary wave) always emerges dur<strong>in</strong>g the middle of theequ<strong>in</strong>e oestrous cycle and produces the ovulatoryfollicle. Approximately 25% of <strong>in</strong>terovulatory <strong>in</strong>tervals<strong>in</strong>volve an additional major wave (secondary wave) thatdevelops dur<strong>in</strong>g the first half of the cycle (Sirois et al.1989; Bergfelt and G<strong>in</strong>ther 1993). The <strong>in</strong>cidence ofsecondary waves is significantly higher <strong>in</strong> some breedssuch as Thoroughbreds, and some of these waves mayproduce ovulations. The ability to ovulate <strong>in</strong> thepresence of high progesterone levels dur<strong>in</strong>g dioestrusseems to be unique to the horse (G<strong>in</strong>ther 1992). M<strong>in</strong>orwaves (largest follicle usually


226 FX Donadeu and HG PedersenRegulation of Follicular Wave DevelopmentEmergence of a follicular wave and subsequent selectionof a dom<strong>in</strong>ant follicle are under f<strong>in</strong>ely-tuned regulationby systemic mechanisms <strong>in</strong>volv<strong>in</strong>g changes <strong>in</strong> gonadotrop<strong>in</strong>levels and local mechanisms that <strong>in</strong>volve changes<strong>in</strong> follicular factor levels.Systemic regulation of follicular wave development.Unlike pre-antral follicles, antral follicles cannotdevelop without adequate gonadotrop<strong>in</strong> stimulation andthis has been experimentally shown <strong>in</strong> the mare(Pedersen et al. 2002; Imboden et al. 2006).Follicular waves <strong>in</strong> the mare, as <strong>in</strong> other species, aretemporally preceded by a stimulatory surge <strong>in</strong> circulat<strong>in</strong>gFSH (G<strong>in</strong>ther et al. 2003a). The acute dependence offollicular waves on FSH has been shown <strong>in</strong> mares by the<strong>in</strong>ability of follicles to grow beyond a diameter of 15 mmafter suppression of circulat<strong>in</strong>g FSH by systemic <strong>in</strong>jectionof follicular fluid (Bergfelt and G<strong>in</strong>ther 1985). Closefunctional relationships between FSH surges and follicularwaves <strong>in</strong>volve not only positive effects of FSH onfollicles but also negative effects of follicles on FSH. Thewave-associated FSH surge reaches a peak when thelargest follicle is approximately 13 mm <strong>in</strong> diameter(Gastal et al. 1997; Donadeu and G<strong>in</strong>ther 2001). Thefollow<strong>in</strong>g decl<strong>in</strong>e <strong>in</strong> FSH results from an <strong>in</strong>crease <strong>in</strong>circulat<strong>in</strong>g <strong>in</strong>hib<strong>in</strong>, presumably <strong>in</strong>hib<strong>in</strong>-A (Watson andAl-zi’abi 2002), contributed ma<strong>in</strong>ly by the three largestfollicles of the wave as they grow above 13 mm(Donadeu and G<strong>in</strong>ther 2001). The decl<strong>in</strong><strong>in</strong>g FSHconcentrations cont<strong>in</strong>ue to support growth of thefollicles of the wave until the largest (future dom<strong>in</strong>ant)follicle reaches the expected diameter at the beg<strong>in</strong>n<strong>in</strong>g ofdeviation (approximately 22 mm). At that time, circulat<strong>in</strong>gFSH levels become too low to ma<strong>in</strong>ta<strong>in</strong> thegrowth of the follicles of the wave. The low FSH, yet,does not restrict the growth of the future dom<strong>in</strong>antfollicle which by that time has acquired the ability tomore efficiently use circulat<strong>in</strong>g gonadotrop<strong>in</strong>s forgrowth (G<strong>in</strong>ther et al. 2003a). The differential responsesof the largest and smaller follicles of a wave togonadotrop<strong>in</strong>s result <strong>in</strong> the <strong>in</strong>itiation of diameterdeviation. Cont<strong>in</strong>uous suppression of FSH throughoutdeviation ensures the morphological and functionaldemise of subord<strong>in</strong>ate follicles and is attributable toproduction of high levels of <strong>in</strong>hib<strong>in</strong> and oestradiol bythe dom<strong>in</strong>ant follicle (Gastal et al. 1999; Donadeu andG<strong>in</strong>ther 2001). The critical role of low FSH levels <strong>in</strong> thedeviation mechanism <strong>in</strong> mares is illustrated by thedisruption of the deviation mechanism after adm<strong>in</strong>istrationof FSH (Squires 2006) or immunization aga<strong>in</strong>st<strong>in</strong>hib<strong>in</strong> (McCue et al. 1992) lead<strong>in</strong>g to the developmentof multiple ovulatory follicles.While FSH is particularly important for folliculargrowth before deviation, LH becomes more criticaldur<strong>in</strong>g deviation. This has been demonstrated by studiesshow<strong>in</strong>g that experimental suppression of LH <strong>in</strong> cycl<strong>in</strong>gmares leads to the regression of the dom<strong>in</strong>ant follicleearly dur<strong>in</strong>g its development (Bergfelt et al. 2001), and isconsistent with an <strong>in</strong>crease <strong>in</strong> circulat<strong>in</strong>g LH before thebeg<strong>in</strong>n<strong>in</strong>g of deviation (Bergfelt et al. 2001). The studyof LH–follicle <strong>in</strong>terrelationships dur<strong>in</strong>g the anovulatoryseason has revealed a direct temporal relationshipbetween an <strong>in</strong>crease <strong>in</strong> circulat<strong>in</strong>g LH, but not FSH,and the development of major waves, further <strong>in</strong>dicat<strong>in</strong>gthat an <strong>in</strong>crease <strong>in</strong> LH plays a major role <strong>in</strong> stimulat<strong>in</strong>gthe development of dom<strong>in</strong>ant follicles (reviewed <strong>in</strong>Donadeu and Watson 2007). An additional conclusionwas that circulat<strong>in</strong>g LH concentrations above thoserequired for growth of the dom<strong>in</strong>ant follicle are requiredfor the development of ovulatory competence, i.e. forthe dom<strong>in</strong>ant follicle to become fully responsive to anLH surge (Donadeu and Watson 2007).Studies <strong>in</strong> cattle have shown that LH receptorexpression differentially <strong>in</strong>creases <strong>in</strong> granulosa and thecacells of the early dom<strong>in</strong>ant follicle (Bao and Garverick1998; Beg and G<strong>in</strong>ther 2006). These f<strong>in</strong>d<strong>in</strong>gs areconsistent with those <strong>in</strong> other species (Richards 2001).Although not critically exam<strong>in</strong>ed <strong>in</strong> relation to thebeg<strong>in</strong>n<strong>in</strong>g of deviation, higher LH receptor levels havebeen reported <strong>in</strong> dom<strong>in</strong>ant-size follicles than <strong>in</strong> smallerfollicles <strong>in</strong> the horse (Fay and Douglas 1987; Goudetet al. 1999). Taken together, the f<strong>in</strong>d<strong>in</strong>gs on LH levelsand LH receptor expression dur<strong>in</strong>g a follicular wave <strong>in</strong>mares are consistent with the conclusion that deviation<strong>in</strong>volves a critical <strong>in</strong>crease <strong>in</strong> the dependence of thedom<strong>in</strong>ant follicle on LH.Limited data exist on the <strong>in</strong>volvement of substancesother than gonadotrop<strong>in</strong>s <strong>in</strong> the endocr<strong>in</strong>e regulation ofantral follicles <strong>in</strong> mares. Based on observed direct effectson follicle growth or on the presence of specific receptors<strong>in</strong> equ<strong>in</strong>e ovaries, positive roles on follicle growth havebeen suggested for circulat<strong>in</strong>g levels of substances<strong>in</strong>clud<strong>in</strong>g growth hormone (Cochran et al. 1999), dopam<strong>in</strong>e(K<strong>in</strong>g et al. 2005) and prolact<strong>in</strong> (Thompson et al.1997).Although follicular <strong>in</strong>sul<strong>in</strong>-like growth factor-1(IGF-1) <strong>in</strong> mares largely derives from the systemiccirculation, its bioactivity is regulated mostly throughlocal mechanisms (Watson et al. 2004) and the role ofIGF-1 is therefore considered <strong>in</strong> the next section.Local regulation of follicular wave development. Localregulation of follicle development <strong>in</strong> monovular specieshas been studied extensively <strong>in</strong> relation to follicleselection (reviewed <strong>in</strong> Fortune et al. 2004; Beg andG<strong>in</strong>ther 2006; Knight and Glister 2006). A variety ofprote<strong>in</strong> and steroid factors <strong>in</strong>clud<strong>in</strong>g members of theIGF family, oestradiol, <strong>in</strong>hib<strong>in</strong>s and activ<strong>in</strong>s, follistat<strong>in</strong>and vascular endothelial growth factor (VEGF) are<strong>in</strong>volved. In general, these factors act, often <strong>in</strong> aparacr<strong>in</strong>e manner, to either enhance or dim<strong>in</strong>ish thetrophic effects of gonadotrop<strong>in</strong>s on follicular cellsthrough a variety of mechanisms. Differential changes<strong>in</strong> the levels of specific factors between follicles thusensure the cont<strong>in</strong>uous development of the dom<strong>in</strong>antfollicle and the regression of subord<strong>in</strong>ate follicles dur<strong>in</strong>gdeviation.A differential <strong>in</strong>crease <strong>in</strong> the levels of oestradiol, IGF-1,activ<strong>in</strong>-A and <strong>in</strong>hib<strong>in</strong>-A <strong>in</strong> the future dom<strong>in</strong>ant folliclewas associated with the beg<strong>in</strong>n<strong>in</strong>g of deviation <strong>in</strong> mares,whereas differentially elevated levels of progesteroneand <strong>in</strong>hib<strong>in</strong>-B occurred later dur<strong>in</strong>g the development ofthe dom<strong>in</strong>ant follicle (Donadeu and G<strong>in</strong>ther 2002a).Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Follicles and Mares 227Studies <strong>in</strong>volv<strong>in</strong>g <strong>in</strong>trafollicular factor <strong>in</strong>jection <strong>in</strong> mareshave provided <strong>in</strong>sight <strong>in</strong>to the complex <strong>in</strong>terrelationshipsbetween these factors dur<strong>in</strong>g follicle deviation(G<strong>in</strong>ther et al. 2005). It has been concluded thatalthough all these factors are likely <strong>in</strong>volved <strong>in</strong> thedevelopment of the dom<strong>in</strong>ant follicle after the beg<strong>in</strong>n<strong>in</strong>gof deviation, only IGF-1 is <strong>in</strong>volved <strong>in</strong> the <strong>in</strong>itiation ofdeviation by, among other actions, regulat<strong>in</strong>g the levelsof other growth factors <strong>in</strong> the dom<strong>in</strong>ant follicle (Beg andG<strong>in</strong>ther 2006). This is consistent with the particularlyimportant role of the IGF-1 system <strong>in</strong> follicle selection<strong>in</strong> other species (Mazerbourg et al. 2003). Results froma series of <strong>in</strong> vivo experiments have conv<strong>in</strong>c<strong>in</strong>glyconfirmed the critical role of the IGF system <strong>in</strong> follicleselection <strong>in</strong> mares. Injection of IGF-1 <strong>in</strong>to the secondlargest or a smaller follicle at the beg<strong>in</strong>n<strong>in</strong>g of deviationchanged its fate from subord<strong>in</strong>ate to co-dom<strong>in</strong>antresult<strong>in</strong>g <strong>in</strong> the development of multiple ovulatoryfollicles (G<strong>in</strong>ther et al. 2004a; b; Gastal et al. 2007).Conversely, <strong>in</strong>jection of IGF b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> (IGFBP)-3<strong>in</strong>to the largest follicle at the beg<strong>in</strong>n<strong>in</strong>g of deviationresulted <strong>in</strong> the follicle regress<strong>in</strong>g and be<strong>in</strong>g replaced bythe second largest follicle which became dom<strong>in</strong>ant(G<strong>in</strong>ther et al. 2004a). At least four types of IGFBPs(IGFBP-2, 3, 4 and 5), which negatively regulate IGFactivity, have been identified <strong>in</strong> equ<strong>in</strong>e follicles and theconcentrations of three of these (IGFBP-2, 4 and 5) arecorrelated negatively with those of IGF-1 dur<strong>in</strong>g follicledevelopment (Gerard and Monget 1998; Bridges et al.2002). Consistent with the stimulatory role of IGF-1 <strong>in</strong>the development of ovulatory follicles, reduced levels ofbioactive IGF-1 are thought to be <strong>in</strong>volved <strong>in</strong> thedevelopmental deficiencies of dom<strong>in</strong>ant follicles dur<strong>in</strong>gthe spr<strong>in</strong>g transition that prevent them from acquir<strong>in</strong>govulatory competence (Acosta et al. 2004; Watson et al.2004).An additional factor that has begun to be explored <strong>in</strong>relation to follicle selection <strong>in</strong> horses is VEGF. Thisangiogenic factor has been shown to be necessary forfollicle development <strong>in</strong> other species (Fraser and Wulff2001). VEGF levels differentially <strong>in</strong>crease <strong>in</strong> the dom<strong>in</strong>antfollicle <strong>in</strong> horses (G<strong>in</strong>ther et al. 2004b), and this<strong>in</strong>crease is thought to be mediated, at least partly, byIGF-1. VEGF is likely <strong>in</strong>volved <strong>in</strong> the reported <strong>in</strong>crease<strong>in</strong> vascularization of the future dom<strong>in</strong>ant follicle beforethe beg<strong>in</strong>n<strong>in</strong>g of deviation (Acosta et al. 2004) whichpresumably <strong>in</strong>creases the availability of circulat<strong>in</strong>ggonadotrop<strong>in</strong>s to the follicle. The reduced levels offollicular VEGF and low vascularity of the wall ofdom<strong>in</strong>ant follicles dur<strong>in</strong>g the spr<strong>in</strong>g transition relative tothe ovulatory season (Watson and Al-zi’abi 2002)underscore the critical role of VEGF <strong>in</strong> the developmentof the ovulatory follicle <strong>in</strong> horses.Effects of Different Physiological Conditions onFollicle Development <strong>in</strong> MaresEffects of season. The effects of season on follicularactivity <strong>in</strong> mares have been studied <strong>in</strong> considerabledetail. Important variations <strong>in</strong> follicular activity occurnot only between the ovulatory and anovulatory seasonsbut also between different periods with<strong>in</strong> each season.Studies <strong>in</strong> pony mares us<strong>in</strong>g follicle ablation to facilitatethe identification of <strong>in</strong>dividual follicular waves revealedthat, as dur<strong>in</strong>g the ovulatory season, follicular wavesperiodically occur dur<strong>in</strong>g the anovulatory season despitethe reduced levels of follicle development (Donadeu andG<strong>in</strong>ther 2002b, 2003; G<strong>in</strong>ther et al. 2003b). Only m<strong>in</strong>orwaves (largest follicle 12 mm with<strong>in</strong> waves (means of 3.2 and 11.5follicles <strong>in</strong> waves develop<strong>in</strong>g dur<strong>in</strong>g deep anoestrous andthe early spr<strong>in</strong>g transition, respectively). Althoughdom<strong>in</strong>ant follicles dur<strong>in</strong>g transition may not grow tothe diameters typical of ovulatory follicles, <strong>in</strong> the samestudy transitional waves produced more follicles thanwaves develop<strong>in</strong>g dur<strong>in</strong>g the ovulatory season (means of11.5 and 6.0 follicles >12 mm, respectively) attest<strong>in</strong>g tothe high levels of follicular activity even <strong>in</strong> the absenceof ovulation <strong>in</strong> transitional mares (Donadeu and G<strong>in</strong>ther2003).Based on consistent temporal relationships betweenfollicles and circulat<strong>in</strong>g hormones, it has been concludedthat the differences <strong>in</strong> follicle development betweendifferent periods of the anovulatory season as well as thedeficient development of dom<strong>in</strong>ant follicles dur<strong>in</strong>g thespr<strong>in</strong>g transition relative to the ovulatoy season are notattributable to deficient circulat<strong>in</strong>g FSH levels butrather to changes <strong>in</strong> LH and, possibly, differences <strong>in</strong>follicular sensitivity to gonadotrop<strong>in</strong>s (reviewed <strong>in</strong>Donadeu and Watson 2007).Season-related effects on follicular activity have alsobeen reported between the two halves of the ovulatoryseason, with higher levels of activity dur<strong>in</strong>g the first halfof the season due to higher <strong>in</strong>cidence of both secondarywaves and m<strong>in</strong>or waves associated with higher gonadotrop<strong>in</strong>levels (G<strong>in</strong>ther 1992, 1993).Effects of pregnancy and parturition. Considerableresearch work is needed to better characterize folliculardynamics and associated regulatory mechanisms dur<strong>in</strong>gand follow<strong>in</strong>g pregnancy <strong>in</strong> the mare. Based on comb<strong>in</strong>eddata from ultrasound and rectal palpation studies,follicular dynamics dur<strong>in</strong>g the first half of pregnancyare similar to those occurr<strong>in</strong>g dur<strong>in</strong>g the first half ofthe anovulatory season, with an <strong>in</strong>itial period of variableactivity (between days 11 and 40 of pregnancy) characterizedby the periodic development of major waves or,more commonly, development of sporadic major wavesor only m<strong>in</strong>or waves (G<strong>in</strong>ther and Bergfelt 1992),followed by a pronounced decrease <strong>in</strong> follicular activity<strong>in</strong> all mares between days 50 and 140 of pregnancy sothat the diameter of the largest follicle does not exceed15 mm by day 140 (Squires et al. 1974). A decrease <strong>in</strong>follicular activity has also been reported after the firstone-third of pregnancy <strong>in</strong> cattle (G<strong>in</strong>ther et al. 1996).Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


228 FX Donadeu and HG PedersenBased on reported hormone–follicle associations, deficientFSH levels do not seem to be responsible for thereduction <strong>in</strong> follicular growth <strong>in</strong> some mares betweendays 11 and 40 of pregnancy (G<strong>in</strong>ther and Bergfelt1992), an effect that may <strong>in</strong>stead be accounted for byreduced levels of LH (due to persistent progesteronenegative feedback), similar to the effects of the seasonalreduction <strong>in</strong> circulat<strong>in</strong>g LH on follicle growth dur<strong>in</strong>g thefall transitional period (G<strong>in</strong>ther et al. 2003b).The mechanisms responsible for the dramatic reduction<strong>in</strong> follicular growth dur<strong>in</strong>g mid and late pregnancy<strong>in</strong> mares have not been clarified but likely <strong>in</strong>volves atemporally associated decrease <strong>in</strong> circulat<strong>in</strong>g FSH(reviewed <strong>in</strong> G<strong>in</strong>ther 1992). This is different from theanovulatory season dur<strong>in</strong>g which changes <strong>in</strong> FSH levelsdo not seem to play a major role <strong>in</strong> the decrease <strong>in</strong>follicle growth dur<strong>in</strong>g deep anoestrus (Donadeu andWatson 2007). Reduced follicle numbers dur<strong>in</strong>g midpregnancyare also likely attributable to ovulation orlute<strong>in</strong>ization of follicles <strong>in</strong>to accessory corpora luteaunder the <strong>in</strong>fluence of chorionic gonadotrop<strong>in</strong> (G<strong>in</strong>ther1992). Further complexity <strong>in</strong>to the regulation of folliclegrowth dur<strong>in</strong>g pregnancy is provided by the observationthat the effects of season on hormones and folliclescont<strong>in</strong>ue to occur dur<strong>in</strong>g pregnancy (G<strong>in</strong>ther 1992).The natural pressure to produce a foal each year <strong>in</strong> aspecies with an 11-month-long pregnancy is reflected <strong>in</strong>an early post-partum ovulation <strong>in</strong> the mare (foal heat,typically with<strong>in</strong> 2 weeks of parturition). A steady<strong>in</strong>crease <strong>in</strong> the diameter of the largest follicle and<strong>in</strong> the numbers of follicles after parturition result<strong>in</strong>g<strong>in</strong> ovulation 14 days later was recently shown <strong>in</strong>Arabian mares (Gunduz et al. 2008). The post-partum<strong>in</strong>crease <strong>in</strong> follicular growth is <strong>in</strong>duced by an <strong>in</strong>crease <strong>in</strong>gonadotrop<strong>in</strong> secretion at the time of parturition (H<strong>in</strong>eset al. 1987; G<strong>in</strong>ther et al. 1994). Studies have shown thatthe follicular response to parturition may vary <strong>in</strong><strong>in</strong>dividual horses or different types of horses, and maynot readily occur <strong>in</strong> primiparous mares (Nagy et al.1998), <strong>in</strong> the presence of a nurs<strong>in</strong>g foal (G<strong>in</strong>ther 1992) ordur<strong>in</strong>g the w<strong>in</strong>ter, when the negative effects of seasonmay prevail over the positive effects of parturition(G<strong>in</strong>ther et al. 1994).Follicle development before puberty. Follicle development<strong>in</strong> spr<strong>in</strong>g-born pre-puberal pony fillies has recentlybeen studied <strong>in</strong> detail (Nogueira and G<strong>in</strong>ther 2004),add<strong>in</strong>g to limited <strong>in</strong>formation on follicle profiles fromearlier studies (reviewed <strong>in</strong> G<strong>in</strong>ther 1992). Follicularactivity dur<strong>in</strong>g 2–10 months of life was characterized bya progressive <strong>in</strong>crease <strong>in</strong> mean follicle diameter (fromapproximately 6 to 10 mm) and mean follicle numbers(from 3 to 17 follicles) between 2 and 5 months of age,a short plateau <strong>in</strong> activity co<strong>in</strong>cid<strong>in</strong>g with the w<strong>in</strong>termonths and a re-<strong>in</strong>itiation of follicle growth after7 months of age lead<strong>in</strong>g to the onset of the firstovulatory season <strong>in</strong> spr<strong>in</strong>g (Nogueira and G<strong>in</strong>ther2004). Changes <strong>in</strong> follicle activity dur<strong>in</strong>g the first yearof life were positively correlated with changes <strong>in</strong>circulat<strong>in</strong>g gonadotrop<strong>in</strong>s, consistent with a regulatoryrole of season on gonadotrop<strong>in</strong> and follicular activitybeg<strong>in</strong>n<strong>in</strong>g early dur<strong>in</strong>g life <strong>in</strong> mares. Follicular growthbefore puberty was characterized by the development of(m<strong>in</strong>or) follicular waves. Remarkably, these waves werenot temporally associated with statistically significantcirculat<strong>in</strong>g FSH surges, an observation that warrantsfurther <strong>in</strong>vestigation.Effects of ag<strong>in</strong>g on follicular activity. As highlighted <strong>in</strong> arecent review, many age-related changes <strong>in</strong> follicularactivity <strong>in</strong> the horse resemble those occurr<strong>in</strong>g <strong>in</strong> humans(Carnevale 2008). Follicular activity dur<strong>in</strong>g oestrouscycles beg<strong>in</strong>s to decrease <strong>in</strong> mares 20 years of age orolder eventually lead<strong>in</strong>g to a cease <strong>in</strong> ovarian activity(Carnevale et al. 1993, 1994). Interovulatory <strong>in</strong>tervalsfirst become longer <strong>in</strong> these mares due to longerfollicular phases associated with a primary follicularwave that emerges later and conta<strong>in</strong>s less follicles. Inaddition, the ovulatory LH surge is less pronounced <strong>in</strong>the older mares, and there is a higher <strong>in</strong>cidence ofultrasonically atypical ovulations characterized by acentral hypoechogenic area at the ovulatory site. Thereduction <strong>in</strong> follicular activity and frequency of ovulation<strong>in</strong> mares ‡20 years old is associated with an overallelevation <strong>in</strong> concentrations of FSH and LH dur<strong>in</strong>g thefollicular phase, a phenomenon that also occurs dur<strong>in</strong>gthe peri-menopausal period <strong>in</strong> women. This is eventuallyassociated with persistent ovarian <strong>in</strong>activity with follicles


Follicles and Mares 229dom<strong>in</strong>ant follicles and the simultaneous regression ofsubord<strong>in</strong>ate follicles dur<strong>in</strong>g follicle deviation.Follicular waves occur throughout post-natal life <strong>in</strong>the horse until follicles irreversibly cease to grow atapproximately ‡ 20 years of age. The levels of follicularactivity are affected by factors such as stage of theoestrous cycle, season, pregnancy, age, breed and<strong>in</strong>dividual. This results <strong>in</strong> variations <strong>in</strong> follicular wavepatterns that most notably <strong>in</strong>clude the production orabsence of a dom<strong>in</strong>ant follicle (major or m<strong>in</strong>or waves)and the development of ovulatory-competent or ovulatory-<strong>in</strong>competentdom<strong>in</strong>ant follicles. Available <strong>in</strong>formation<strong>in</strong>dicates that these physiological variations <strong>in</strong>follicular wave patterns are driven by changes <strong>in</strong>circulat<strong>in</strong>g levels of FSH and ⁄ or LH and by localchanges <strong>in</strong> responsiveness of follicles to gonadotrop<strong>in</strong>s.Yet, considerable work is needed to better understandthe mechanisms by which follicle development is regulateddur<strong>in</strong>g different physiological conditions outsidethe oestrous cycle <strong>in</strong> mares.ReferencesAcosta TJ, Beg MA, G<strong>in</strong>ther OJ, 2004: Aberrant blood flowarea and plasma gonadotrop<strong>in</strong> concentrations dur<strong>in</strong>g thedevelopment of dom<strong>in</strong>ant-sized transitional anovulatoryfollicles <strong>in</strong> mares. Biol Reprod 71, 637–642.Bao B, Garverick HA, 1998: Expression of steroidogenicenzyme and gonadotrop<strong>in</strong> receptor genes <strong>in</strong> bov<strong>in</strong>e folliclesdur<strong>in</strong>g ovarian follicular waves: a review. J Anim Sci 76,1903–1921.Beg MA, G<strong>in</strong>ther OJ, 2006: Follicle selection <strong>in</strong> cattle andhorses: role of <strong>in</strong>trafollicular factors. <strong>Reproduction</strong> 132,365–377.Bergfelt DR, G<strong>in</strong>ther OJ, 1985: Delayed follicular developmentand ovulation follow<strong>in</strong>g <strong>in</strong>hibition of FSH with equ<strong>in</strong>efollicular fluid <strong>in</strong> the mare. Theriogenology 24, 99–108.Bergfelt DR, G<strong>in</strong>ther OJ, 1993: Relationships between FSHsurges and follicular waves dur<strong>in</strong>g the estrous cycle <strong>in</strong> mares.Theriogenology 39, 781–796.Bergfelt DR, Gastal EL, G<strong>in</strong>ther OJ, 2001: Response ofestradiol and <strong>in</strong>hib<strong>in</strong> to experimentally reduced lute<strong>in</strong>iz<strong>in</strong>ghormone dur<strong>in</strong>g follicle deviation <strong>in</strong> mares. Biol Reprod 65,426–432.Boerboom D, Sirois J, 2001: Equ<strong>in</strong>e P450 cholesterol side-cha<strong>in</strong>cleavage and 3 beta-hydroxysteroid dehydrogenase ⁄ delta(5)-delta(4) isomerase: molecular clon<strong>in</strong>g and regulation of theirmessenger ribonucleic acids <strong>in</strong> equ<strong>in</strong>e follicles dur<strong>in</strong>g theovulatory process. Biol Reprod 64, 206–215.Bridges TS, Davidson TR, Chamberla<strong>in</strong> CS, Geisert RD,Spicer LJ, 2002: Changes <strong>in</strong> follicular fluid steroids, <strong>in</strong>sul<strong>in</strong>likegrowth factors (IGF) and IGF-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> concentration,and proteolytic activity dur<strong>in</strong>g equ<strong>in</strong>e folliculardevelopment. J Anim Sci 80, 179–190.Carnevale EM, 2008: The mare model for follicular maturationand reproductive ag<strong>in</strong>g <strong>in</strong> the woman. Theriogenology69, 23–30.Carnevale EM, Bergfelt DR, G<strong>in</strong>ther OJ, 1993: Ag<strong>in</strong>g effectson follicular activity and concentrations of FSH, LH, andprogesterone <strong>in</strong> mares. Anim Reprod Sci 31, 287–299.Carnevale EM, Bergfelt DR, G<strong>in</strong>ther OJ, 1994: Follicularactivity and concentrations of FSH and LH associated withsenescence <strong>in</strong> mares. Anim Reprod Sci 35, 231–246.Carnevale EM, Hermenet MJ, G<strong>in</strong>ther OJ, 1997: Age andpasture effects on vernal transition <strong>in</strong> mares. Theriogenology47, 1009–1018.Cochran RA, Leonardi-Cattolica AA, Sullivan MR, K<strong>in</strong>caidLA, Leise BS, Thompson DL Jr, Godke RA, 1999: Theeffects of equ<strong>in</strong>e somatotrop<strong>in</strong> (eST) on follicular developmentand circulat<strong>in</strong>g plasma hormone profiles <strong>in</strong> cyclicmares treated dur<strong>in</strong>g different stages of the estrous cycle.Domest Anim Endocr<strong>in</strong>ol 16, 57–67.Deanesly R, 1975: Germ cell development and the meioticprophase <strong>in</strong> the fetal horse ovary. J Reprod Fertil Suppl 23,547–552.Donadeu FX, G<strong>in</strong>ther OJ, 2001: Effect of number anddiameter of follicles on plasma concentrations of <strong>in</strong>hib<strong>in</strong>and FSH <strong>in</strong> mares. <strong>Reproduction</strong> 121, 897–903.Donadeu FX, G<strong>in</strong>ther OJ, 2002a: Changes <strong>in</strong> Concentrationsof follicular fluid factors dur<strong>in</strong>g follicle selection <strong>in</strong> mares.Biol Reprod 66, 1111–1118.Donadeu FX, G<strong>in</strong>ther OJ, 2002b: Follicular waves andcirculat<strong>in</strong>g concentrations of gonadotroph<strong>in</strong>s, <strong>in</strong>hib<strong>in</strong> andoestradiol dur<strong>in</strong>g the anovulatory season <strong>in</strong> mares. <strong>Reproduction</strong>124, 875–885.Donadeu FX, G<strong>in</strong>ther OJ, 2003: Interactions of follicularfactors and season <strong>in</strong> the regulation of circulat<strong>in</strong>g concentrationsof gonadotroph<strong>in</strong>s <strong>in</strong> mares. <strong>Reproduction</strong> 125,743–750.Donadeu FX, Watson ED, 2007: Seasonal changes <strong>in</strong> ovarianactivity: lessons learnt from the horse. Anim Reprod Sci 100,225–242.Driancourt MA, 1979: Follicular k<strong>in</strong>etics <strong>in</strong> the mare ovary.Ann Biol Anim Biochim Biophys 19, 1443–1453.Driancourt MA, Paris A, Roux C, Mariana JC, Palmer E,1982: Ovarian follicular populations <strong>in</strong> pony and saddletypemares. Reprod Nutr Dev 22, 1035–1047.Driancourt MA, Prunier A, Palmer E, Mariana J, 1983:Seasonal effects on ovarian follicular development <strong>in</strong> ponymares. Reprod Nutr Dev 23, 207–215.Fay JE, Douglas RH, 1987: Changes <strong>in</strong> thecal and granulosacell LH and FSH receptor content associated with follicularfluid and peripheral plasma gonadotroph<strong>in</strong> and steroidhormone concentrations <strong>in</strong> preovulatory follicles of mares. JReprod Fertil Suppl 35, 169–181.Fayad T, Levesque V, Sirois J, Silversides DW, Lussier JG, 2004:Gene expression profil<strong>in</strong>g of differentially expressed genes <strong>in</strong>granulosa cells of bov<strong>in</strong>e dom<strong>in</strong>ant follicles us<strong>in</strong>g suppressionsubtractive hybridization. Biol Reprod 70, 523–533.Fortune JE, Rivera GM, Yang MY, 2004: Follicular development:the role of the follicular microenvironment <strong>in</strong> selectionof the dom<strong>in</strong>ant follicle. Anim Reprod Sci 82–83, 109–126.Fraser HM, Wulff C, 2001: Angiogenesis <strong>in</strong> the primate ovary.Reprod Fertil Dev 13, 557–566.Gastal EL, Gastal MO, Bergfelt DR, G<strong>in</strong>ther OJ, 1997: Roleof diameter differences among follicles <strong>in</strong> selection of afuture dom<strong>in</strong>ant follicle <strong>in</strong> mares. Biol Reprod 57, 1320–1327.Gastal EL, Donadeu FX, Gastal MO, G<strong>in</strong>ther OJ, 1999:Echotextural changes <strong>in</strong> the follicular wall dur<strong>in</strong>g follicledeviation <strong>in</strong> mares. Theriogenology 52, 803–814.Gastal EL, Gastal MO, Beg MA, G<strong>in</strong>ther OJ, 2004: Interrelationshipsamong follicles dur<strong>in</strong>g the common-growthphase of a follicular wave and capacity of <strong>in</strong>dividual folliclesfor dom<strong>in</strong>ance <strong>in</strong> mares. <strong>Reproduction</strong> 128, 417–422.Gastal EL, Gastal MO, Donadeu FX, Acosta TJ, Beg MA,G<strong>in</strong>ther OJ, 2007: Temporal relationships among LH,estradiol, and follicle vascularization preced<strong>in</strong>g the firstcompared with later ovulations dur<strong>in</strong>g the year <strong>in</strong> mares.Anim Reprod Sci 102, 314–321.Gerard N, Monget P, 1998: Intrafollicular <strong>in</strong>sul<strong>in</strong>-like growthfactor-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> levels <strong>in</strong> equ<strong>in</strong>e ovarian folliclesdur<strong>in</strong>g preovulatory maturation and regression. BiolReprod 58, 1508–1514.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


230 FX Donadeu and HG PedersenG<strong>in</strong>ther OJ, 1979: Reproductive Biology of the Mare: Basicand Applied Aspects, 1st edn. Equiservices Cross Pla<strong>in</strong>s,WI, USA.G<strong>in</strong>ther OJ, 1990: Folliculogenesis dur<strong>in</strong>g the transitionalperiod and early ovulatory season <strong>in</strong> mares. J Reprod Fertil90, 311–320.G<strong>in</strong>ther OJ, 1992: Reproductive Biology of the Mare: Basicand Applied Aspects, 2nd edn. Equiservices Publish<strong>in</strong>gCross Pla<strong>in</strong>s, WI, USA.G<strong>in</strong>ther OJ, 1993: Major and m<strong>in</strong>or follicular waves dur<strong>in</strong>g theequ<strong>in</strong>e estrous cycle. J Eq Vet Sci 13, 18–25.G<strong>in</strong>ther OJ, Bergfelt DR, 1992: Associations between FSHconcentrations and major and m<strong>in</strong>or follicular waves <strong>in</strong>pregnant mares. Theriogenology 38, 807–821.G<strong>in</strong>ther OJ, Bergfelt DR, 1993: Growth of small follicles andconcentrations of FSH dur<strong>in</strong>g the equ<strong>in</strong>e oestrous cycle. JReprod Fert 99, 105–111.G<strong>in</strong>ther OJ, Baucus KL, Bergfelt DR, 1994: Follicular andFSH responses to parturition dur<strong>in</strong>g the anovulatory season<strong>in</strong> mares. Theriogenology 41, 613–627.G<strong>in</strong>ther OJ, Kot K, Kulick LJ, Mart<strong>in</strong> S, Wiltbank MC, 1996:Relationships between FSH and ovarian follicular wavesdur<strong>in</strong>g the last six months of pregnancy <strong>in</strong> cattle. J ReprodFert 108, 271–279.G<strong>in</strong>ther OJ, Beg MA, Donadeu FX, Bergfelt DR, 2003a:Mechanism of follicle deviation <strong>in</strong> monovular farm species.Anim Reprod Sci 78, 239–257.G<strong>in</strong>ther OJ, Woods BG, Meira C, Beg MA, Bergfelt DR,2003b: Hormonal mechanism of follicle deviation as <strong>in</strong>dicatedby major versus m<strong>in</strong>or follicular waves dur<strong>in</strong>g thetransition <strong>in</strong>to the anovulatory season <strong>in</strong> mares. <strong>Reproduction</strong>126, 653–660.G<strong>in</strong>ther OJ, Bergfelt DR, Beg MA, Meira C, Kot K, 2004a:In vivo effects of an <strong>in</strong>trafollicular <strong>in</strong>jection of <strong>in</strong>sul<strong>in</strong>-likegrowth factor 1 on the mechanism of follicle deviation <strong>in</strong>heifers and mares. Biol Reprod 70, 99–105.G<strong>in</strong>ther OJ, Gastal EL, Gastal MO, Checura CM, Beg MA,2004b: Dose–response study of <strong>in</strong>trafollicular <strong>in</strong>jection of<strong>in</strong>sul<strong>in</strong>-like growth factor-I on follicular fluid factorsand follicle dom<strong>in</strong>ance <strong>in</strong> mares. Biol Reprod 70, 1063–1069.G<strong>in</strong>ther OJ, Gastal EL, Gastal MO, Bergfelt DR, BaerwaldAR, Pierson RA, 2004c: Comparative study of the dynamicsof follicular waves <strong>in</strong> mares and women. Biol Reprod 71,1195–1201.G<strong>in</strong>ther OJ, Gastal EL, Gastal MO, Beg MA, 2005: In vivoeffects of pregnancy-associated plasma prote<strong>in</strong>-A, activ<strong>in</strong>-Aand vascular endothelial growth factor on other follicularfluidfactors dur<strong>in</strong>g follicle deviation <strong>in</strong> mares. <strong>Reproduction</strong>129, 489–496.Goudet G, Bel<strong>in</strong> F, Bezard J, Gerard N, 1999: Intrafollicularcontent of lute<strong>in</strong>iz<strong>in</strong>g hormone receptor, {alpha}-<strong>in</strong>hib<strong>in</strong>,and aromatase <strong>in</strong> relation to follicular growth, estrous cyclestage, and oocyte competence for <strong>in</strong> vitro maturation <strong>in</strong> themare. Biol Reprod 60, 1120–1127.Gunduz MC, Kasikci G, Ekiz B, 2008: Follicular and steroidhormone changes <strong>in</strong> Arabian mares <strong>in</strong> the postpartumperiod. Anim Reprod Sci (<strong>in</strong> press).H<strong>in</strong>es KK, Fitzgerald BP, Loy RG, 1987: Effect of pulsatilegonadotroph<strong>in</strong> release on mean serum LH and FSH<strong>in</strong> peri-parturient mares. J Reprod Fertil Suppl 35, 635–640.Imboden I, Janett F, Burger D, Crowe MA, Hassig M, Thun R,2006: Influence of immunization aga<strong>in</strong>st GnRH on reproductivecyclicity and estrous behavior <strong>in</strong> the mare. Theriogenology66, 1866–1875.K<strong>in</strong>g SS, Campbell AG, Dille EA, Roser JF, Murphy LL,Jones KL, 2005: Dopam<strong>in</strong>e receptors <strong>in</strong> equ<strong>in</strong>e ovariantissues. Domest Anim Endocr<strong>in</strong>ol 28, 405–415.Knight PG, Glister C, 2006: TGF-{beta} superfamily membersand ovarian follicle development. <strong>Reproduction</strong> 132, 191–206.Martoriati A, Duchamp G, Gerard N, 2003: In vivo effect ofepidermal growth factor, <strong>in</strong>terleuk<strong>in</strong>-1{beta}, and <strong>in</strong>terleuk<strong>in</strong>-1RAon equ<strong>in</strong>e preovulatory follicles. Biol Reprod 68,1748–1754.Mazerbourg S, Bondy CA, Zhou J, Monget P, 2003: The<strong>in</strong>sul<strong>in</strong>-like growth factor system: a key determ<strong>in</strong>ant role <strong>in</strong>the growth and selection of ovarian follicles? A comparativespecies study. Reprod Domest Anim 38, 247–258.McCue PM, Carney NJ, Hughes JP, Rivier J, Vale W, LasleyBL, 1992: Ovulation and embryo recovery rates follow<strong>in</strong>gimmunization of mares aga<strong>in</strong>st an <strong>in</strong>hib<strong>in</strong> alpha-subunitfragment. Theriogenology 38, 823–831.McGee EA, Hsueh AJW, 2000: Initial and cyclic recruitmentof ovarian follicles. Endocr<strong>in</strong>e Rev 21, 200–214.Mihm M, Baker PJ, Flem<strong>in</strong>g LM, Monteiro AM,O’Shaughnessy PJ, 2008: Differentiation of the bov<strong>in</strong>edom<strong>in</strong>ant follicle from the cohort upregulates mRNAexpression for new tissue development genes. <strong>Reproduction</strong>135, 253–265.Mizunuma H, Liu X, Andoh K, Abe Y, Kobayashi J, YamadaK, Yokota H, Ibuki Y, Hasegawa Y, 1999: Activ<strong>in</strong> fromsecondary follicles causes small preantral follicles torema<strong>in</strong> dormant at the rest<strong>in</strong>g stage. Endocr<strong>in</strong>ology 140,37–42.Nagy P, Huszenicza G, Juhasz J, Kulcsar M, Solti L, Reiczigel J,Abavary K, 1998: Factors <strong>in</strong>fluenc<strong>in</strong>g ovarian activity andsexual behavior of postpartum mares under farm conditions.Theriogenology 50, 1109–1119.Nogueira GP, G<strong>in</strong>ther OJ, 2004: Dynamics of follicle populationsand gonadotrop<strong>in</strong> concentrations <strong>in</strong> fillies age two toten months. Equ<strong>in</strong>e Vet J 32, 482–488.Palmer E, Driancourt MA, 1980: Use of ultrasonic echography<strong>in</strong> equ<strong>in</strong>e gynecology. Theriogenology 13, 203–216.Pedersen HG, Berrocal B, Thomson SRM, Watson ED, 2002:Gonadotropic control of follicular growth <strong>in</strong> the mare.Theriogenology 58, 465–468.Richards JS, 2001: Perspective: the ovarian follicle –a perspective <strong>in</strong> 2001. Endocr<strong>in</strong>ology 142, 2184–2193.Seaborne E, 1925: The oestrous cycle of the mare and someassociated phenomena. Anat Rec 30, 277–286.Sirois J, Ball BA, Fortune JE, 1989: Patterns of growth andregression of ovarian follicles dur<strong>in</strong>g the oestrous cycle andafter hemiovariectomy <strong>in</strong> mares. Equ<strong>in</strong>e Vet J 8(Suppl.), 43–48.Sisco B, Hagemann LJ, Shell<strong>in</strong>g AN, Pfeffer PL, 2003:Isolation of genes differentially expressed <strong>in</strong> dom<strong>in</strong>ant andsubord<strong>in</strong>ate bov<strong>in</strong>e follicles. Endocr<strong>in</strong>ology 144, 3904–3913.Squires EL, 2006: Superovulation <strong>in</strong> mares. Veter Cl<strong>in</strong> N Am:Eq Pract 22, 819–830.Squires EL, Garcia MC, G<strong>in</strong>ther OJ, 1974: Effects ofpregnancy and hysterectomy on the ovaries of pony. JAnim Sci 38, 823–830.Thompson DL Jr, Hoffman R, DePew CL, 1997: Prolact<strong>in</strong>adm<strong>in</strong>istration to seasonally anestrous mares: reproductive,metabolic, and hair-shedd<strong>in</strong>g responses. J Anim Sci 75,1092–1099.Vanderwall DKW, GL, 1990: Age-related subfertility <strong>in</strong> themare. In: Proc 35th Annual Conv American Assoc Equ<strong>in</strong>ePractitioners, 85–89.Watson ED, Al-zi’abi MO, 2002: Characterization of morphologyand angiogenesis <strong>in</strong> follicles of mares dur<strong>in</strong>g spr<strong>in</strong>gtransition and the breed<strong>in</strong>g season. <strong>Reproduction</strong> 124, 227–234.Watson ED, Heald M, Tsigos A, Leask R, Steele M, GroomeNP, Riley SC, 2002: Plasma FSH, <strong>in</strong>hib<strong>in</strong> A and <strong>in</strong>hib<strong>in</strong>isoforms conta<strong>in</strong><strong>in</strong>g pro- and -alphaC dur<strong>in</strong>g w<strong>in</strong>ter anoes-Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Follicles and Mares 231trus, spr<strong>in</strong>g transition and the breed<strong>in</strong>g season <strong>in</strong> mares.<strong>Reproduction</strong> 123, 535–542.Watson ED, Bae SE, Thomassen R, Thomson SR, Woad K,Armstrong DG, 2004: Insul<strong>in</strong>-like growth factors-I and -IIand <strong>in</strong>sul<strong>in</strong>-like growth factor-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>-2 <strong>in</strong> dom<strong>in</strong>antequ<strong>in</strong>e follicles dur<strong>in</strong>g spr<strong>in</strong>g transition and theovulatory season. <strong>Reproduction</strong> 128, 321–329.Author’s address (for correspondence): FX Donadeu, Rosl<strong>in</strong> Institute,Royal (Dick) School of Veter<strong>in</strong>ary Studies, University of Ed<strong>in</strong>burgh,Ed<strong>in</strong>burgh, UK. E-mail: xavier.donadeu@ed.ac.ukConflict of <strong>in</strong>terest: All authors declare no conflict of <strong>in</strong>terests.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 232–237 (2008); doi: 10.1111/j.1439-0531.2008.01167.xISSN 0936-6768Prote<strong>in</strong>s Associated With the Early Intrauter<strong>in</strong>e Equ<strong>in</strong>e ConceptusMA Hayes 1 , BA Qu<strong>in</strong>n 1 , ND Keirstead 1 , P Katavolos 1 , RO Waelchli 2 and KJ Betteridge 21 Departments of Pathobiology and 2 Biomedical Sciences, Ontario Veter<strong>in</strong>ary College, University of Guelph, Guelph, ON, CanadaContentsA critical period of early gestation <strong>in</strong> the mare <strong>in</strong>volves theimmobilization (fixation) of the encapsulated conceptus ataround days 16–17. We compared the major prote<strong>in</strong>s <strong>in</strong> thenormal equ<strong>in</strong>e embryonic capsule and endometrial secretionsaround the period of fixation with those from pregnancies <strong>in</strong>the process of term<strong>in</strong>ation <strong>in</strong>duced by adm<strong>in</strong>istration of ananalogue of prostagland<strong>in</strong> F 2a (PGF 2a ). Uterocal<strong>in</strong> andb 2 -microglobul<strong>in</strong> (b 2 M) associated with the embryonic capsulewere proteolytically converted to smaller forms dur<strong>in</strong>g thefixation period. These conversions were similar <strong>in</strong> conceptusesfrom control and treated mares. A 17 kDa cationic prote<strong>in</strong>identified as a secretory phospholipase A2 (sPLA2) type IIAwas detected bound to normal capsules but <strong>in</strong>creased substantially<strong>in</strong> response to PGF 2a . Two forms of uteroglob<strong>in</strong> weredist<strong>in</strong>guished by partial am<strong>in</strong>o acid sequences of 6 kDabands <strong>in</strong> flush fluids from normal pregnant uteri. Afteradm<strong>in</strong>istration of PGF 2a one immunoreactive form of uteroglob<strong>in</strong>was preferentially <strong>in</strong>creased. These studies demonstratethat failure of pregnancy <strong>in</strong> this model is associated with an<strong>in</strong>crease <strong>in</strong> secretory phospholipase <strong>in</strong> the capsule and a change<strong>in</strong> the forms of uteroglob<strong>in</strong> <strong>in</strong> the uter<strong>in</strong>e secretions.IntroductionEmbryonic loss before placentation accounts for asubstantial proportion of pregnancy failures <strong>in</strong> mares(Ball 1988; Baker et al. 1993; Carnevale et al. 2000;Morris and Allen 2002). In Thoroughbred studs <strong>in</strong>Newmarket, UK, 16–17% of pregnancies diagnosed byultrasound at about day 15 are subsequently lost, mostbetween days 15 and 35 (Morris and Allen 2002).Dur<strong>in</strong>g the second and third weeks of gestation, thenormal conceptus (embryo proper, the extra-embryonicmembranes and their conta<strong>in</strong>ed fluid) expands rapidlyand migrates <strong>in</strong> the uterus until around days 16–17 whenit becomes ‘fixed’ at the site of subsequent placentation(G<strong>in</strong>ther et al. 1985). A better understand<strong>in</strong>g of themolecular <strong>in</strong>teractions between the conceptus andendometrium might improve methods for diagnosisand treatment of mares that are prone to los<strong>in</strong>g theconceptus dur<strong>in</strong>g this stage.The embryonic capsule, which is essential for survivalof the embryo (Stout et al. 2005), is composed largely ofacellular glycoprote<strong>in</strong> produced ma<strong>in</strong>ly by the trophoblast(Albihn et al. 2003). It also conta<strong>in</strong>s some maternally-secretedprote<strong>in</strong>s, notably uterocal<strong>in</strong>-p19 which isimplicated <strong>in</strong> transport of small hydrophobic substancesacross the capsule (Crossett et al. 1998; Stewart et al.2000; Kennedy 2005). Before fixation, the yolk-sac wallconta<strong>in</strong>s large amounts of GM2-activator prote<strong>in</strong>(GM2AP) suggest<strong>in</strong>g that this prote<strong>in</strong> plays a role <strong>in</strong>transport of glycolipids or phospholipids while thecapsule is <strong>in</strong>tact (Qu<strong>in</strong>n et al. 2006). Dur<strong>in</strong>g the fixationperiod, sialic acid decreases <strong>in</strong> the capsule (Oriol et al.1993a,b) expos<strong>in</strong>g fewer negatively charged galactoseand N-acetylgalactose residues of the major core type 1O-glycan (Arar et al. 2007). This change might beimportant <strong>in</strong> chang<strong>in</strong>g the permeability or ‘stick<strong>in</strong>ess’ ofthe capsule. The capsule also conta<strong>in</strong>s b 2 -microglobul<strong>in</strong>(b 2 M) which undergoes limited proteolysis dur<strong>in</strong>g thefixation period; the role of this conversion is unknown(Qu<strong>in</strong>n et al. 2007). While such changes <strong>in</strong> the capsulemight play some functional role <strong>in</strong> the process offixation, they might also signify deterioration of thecapsule <strong>in</strong> preparation for its removal which is consideredto be completed by about day 21 (Betteridge 2007).To learn more about the molecular events associatedwith fixation dur<strong>in</strong>g successful early pregnancy, we havecompared prote<strong>in</strong>s <strong>in</strong> the capsule, yolk sac and uterusdur<strong>in</strong>g the fixation period <strong>in</strong> normal gestation (Qu<strong>in</strong>net al. 2007) with those <strong>in</strong> pregnancies <strong>in</strong>duced to fail byadm<strong>in</strong>istration of a luteolytic dose of a prostagland<strong>in</strong>F 2a analogue (Chu et al. 1997; Betteridge et al. 2006). Inthis paper, we further characterized the effects ofluteolysis on p19-uterocal<strong>in</strong> and a secretory phospholipaseassociated with the capsule, and on the forms ofuteroglob<strong>in</strong> <strong>in</strong> the uter<strong>in</strong>e fluids.Materials and MethodsSeventeen mares were used accord<strong>in</strong>g to a researchprotocol approved by the Animal Care Committee ofthe University of Guelph. They were monitored foroestrus and bred by artificial <strong>in</strong>sem<strong>in</strong>ation with freshsemen. Ovarian follicles were monitored daily by transrectalultrasonography so that conceptuses collectedcould be aged correctly to with<strong>in</strong> 0.5 day; the day thatovulation was first detected was designated as day 0.Some mares were treated on day 12 or 14 with aprostagland<strong>in</strong> PGF2a analogue (Estrumate) to <strong>in</strong>duceluteolysis (Chu et al. 1997; Betteridge et al. 2006).Between days 15 and 18 post-ovulation conceptuseswere flushed from a mare’s uterus by transcervicaluter<strong>in</strong>e lavage with 1 l volumes of phosphate-bufferedsal<strong>in</strong>e (PBS; pH 7.4) us<strong>in</strong>g the method previouslydescribed by Waelchli and Betteridge (1996). Varioussamples (capsule, yolk-sac fluid, yolk-sac wall, uter<strong>in</strong>eflush fluids and endometrial biopsies) were collected forproteomic analysis as previously described (Qu<strong>in</strong>n et al.2007). The endometrial samples were bisected; one halfwas flash-frozen and stored <strong>in</strong> liquid nitrogen, the otherhalf was stored <strong>in</strong> RNAlater (Ambion, Aust<strong>in</strong>, TX,USA) for RT-PCR. The capsule was dissected, washedthree times <strong>in</strong> PBS to release loosely bound prote<strong>in</strong>s, andcut <strong>in</strong>to portions for electrophoresis. The yolk-sac wallwas dissected from the embryonic disc and separated<strong>in</strong>to bilam<strong>in</strong>ar and trilam<strong>in</strong>ar portions for analysis. TheÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Prote<strong>in</strong>s <strong>in</strong> Early Equ<strong>in</strong>e Conceptuses 233uter<strong>in</strong>e flush and yolk-sac fluids were centrifuged toremove cells and particulates. Flush fluid aliquots(100 ml) were concentrated to 2 ml by centrifugalultrafiltration at a 5 kDa cutoff (Amicon Ultra, MilliporeCorporation, Bedford, MA, USA) and stored at)70°C until they were analysed. Uter<strong>in</strong>e biopsies andyolk-sac wall tissues were homogenized <strong>in</strong> 0.5 ml PBSconta<strong>in</strong><strong>in</strong>g prote<strong>in</strong>ase <strong>in</strong>hibitors (Complete M<strong>in</strong>i, RocheDiagnostics, Mannheim, Germany).Prote<strong>in</strong>s of <strong>in</strong>terest were compared by one- and twodimensionalSDS–PAGE and Western blots us<strong>in</strong>g methodsdescribed previously (Brooks et al. 2003; Qu<strong>in</strong>n et al.2007). Prote<strong>in</strong>s <strong>in</strong> gels were identified by MALDI-TOFand MS ⁄ MS am<strong>in</strong>o acid sequenc<strong>in</strong>g of <strong>in</strong>-gel tryps<strong>in</strong>digests (Brooks et al. 2003) us<strong>in</strong>g MS-fit (Prote<strong>in</strong>Prospector,University of California, San Francisco, CA,USA) and Scaffold (Proteome Software, Portland, OR,USA). Direct am<strong>in</strong>o acid sequences determ<strong>in</strong>ed bytandem mass spectrometry at the Advanced Prote<strong>in</strong>Technology Center, Hospital for Sick Children (Toronto,ON, Canada) (Lillie et al. 2006) were compared forsimilarity with sequences of Equus caballus via the NCBInon-redundant prote<strong>in</strong> sequence database. Equalamounts of orig<strong>in</strong>al material were loaded onto gels andcomparisons made based on band density, and the sizedeterm<strong>in</strong>ed us<strong>in</strong>g pre-sta<strong>in</strong>ed broad range molecularweight markers [Bio-Rad (Hercules, CA, USA) or NewEngland Biolabs (Ipswich, MA, USA) P7701S]. Differences<strong>in</strong> amounts of uteroglob<strong>in</strong> <strong>in</strong> uter<strong>in</strong>e flush fluidswere also compared by quantitative 2D-difference-<strong>in</strong>-gelelectrophoresis (2D-DIGE) us<strong>in</strong>g the methods described(Hobson et al. 2007). Gels were scanned with aTyphoon 9410 imager (excitation ⁄ emission filter of488 nm ⁄ 520 nm for Cy2, 532 nm ⁄ 580 nm for Cy3, and633 nm ⁄ 670 nm for Cy5). Statistical and quantitativeanalyses of spot changes on images were completed <strong>in</strong>triplicate samples from each group us<strong>in</strong>g DeCyder 6.5software (GE Healthcare, Uppsala, Sweden).Prote<strong>in</strong> concentrations <strong>in</strong> samples were determ<strong>in</strong>ed bythe Biorad prote<strong>in</strong> assay (Bradford 1976). Prote<strong>in</strong>s fromsome gels were transferred to nitrocellulose membranesfor immunoblot analysis, as described previously (Qu<strong>in</strong>net al. 2007). Specific b<strong>in</strong>d<strong>in</strong>g was detected by <strong>in</strong>cubationwith rabbit antiserum aga<strong>in</strong>st recomb<strong>in</strong>ant equ<strong>in</strong>ep19 ⁄ uterocal<strong>in</strong> from which the GST fusion partnerhad been removed (Kennedy 2005), or aga<strong>in</strong>st asynthetic peptide of uteroglob<strong>in</strong> ⁄ Clara cell secretoryprote<strong>in</strong> (Katavolos 2006), or equ<strong>in</strong>e secretory phopspholipaseA2 (gi: 126513263). Anti-peptide antibodieswere raised <strong>in</strong> rabbits immunized aga<strong>in</strong>st peptidesconjugated to keyhole limpet hemocyan<strong>in</strong> (KLH)(Pacific Immunology, Ramona, CA, USA). The peptideimmunogens were KEATSSYGFYGC from the predictedsPLA2 from equ<strong>in</strong>e endometrium (gi: 126513263)and EPSKPDADMKAATTQLKTLV correspond<strong>in</strong>gto equ<strong>in</strong>e secretoglob<strong>in</strong> 1A1 (gi: 126352306) (Katavolos2006).The am<strong>in</strong>o acid sequence of equ<strong>in</strong>e sPLA2 wasdeterm<strong>in</strong>ed from complementary DNA (cDNA) derivedfrom endometrium from pregnant mares. Tissue expressionof the putative sPLA 2 gene was detected qualitativelyby RT-PCR us<strong>in</strong>g <strong>in</strong>dividual endometrial biopsiespreviously stored <strong>in</strong> RNAlater. Total RNA was isolatedus<strong>in</strong>g TRIzol Ò reagent (Ambion), and a phenol-guanid<strong>in</strong>eisothiocyanate protocol (Chomczynski and Sacchi1987). Messenger RNA was copied <strong>in</strong>to cDNA us<strong>in</strong>g theThermoscript RT-PCR system accord<strong>in</strong>g to the manufacturer’srecommended protocol (Invitrogen, Burl<strong>in</strong>gton,ON, Canada). Amplification of sPLA 2 wasperformed us<strong>in</strong>g Plat<strong>in</strong>um Taq DNA polymerase (Invitrogen)accord<strong>in</strong>g to the manufacturer’s recommendations,us<strong>in</strong>g 0.5 lM of forward primer(GGTCCAGGGGCATTTGCG) and reverse primer(AACTTGGATCGGGCAGGGAG). PCR productswere electrophoresed on a 2% agarose gel and sta<strong>in</strong>edwith ethidium bromide. PCR amplicons were pooledand sequenced to exam<strong>in</strong>e the possibility of s<strong>in</strong>glenucleotide polymorphisms (SNPs) with<strong>in</strong> the cod<strong>in</strong>gregion, <strong>in</strong>clud<strong>in</strong>g the presence of heterozygotes. DNAsequenc<strong>in</strong>g was performed at the Guelph MolecularSupercentre (Guelph, ON, Canada). Nucleotide sequenceswere compared aga<strong>in</strong>st sequences at NCBI<strong>in</strong>clud<strong>in</strong>g Equus caballus build 1.1 (Eca1.1) genomebased on the 6.8X WGS assembly of the domestic horsegenome—EquCab1 (Broad Institute, Cambridge, MA,USA).ResultsBefore fixation, the major prote<strong>in</strong> bands visible byreduced 1D SDS–PAGE <strong>in</strong> extracts of normal embryoniccapsule were uterocal<strong>in</strong> ⁄ p19 and the <strong>in</strong>tact p10form of b 2 M (Fig. 1). This was consistent with previousstudies of conceptuses from normal gestation, andconfirmed by Western blot, MALDI-TOF and MS ⁄ MSsequence determ<strong>in</strong>ation of <strong>in</strong>-gel tryps<strong>in</strong> digests. Uterocal<strong>in</strong>bands or spots <strong>in</strong> the normal capsule werevariable, <strong>in</strong> part due to degradation to smaller formsD14ND16TD16ND18 D18T Np19p17p8, p10p5, p6Fig. 1. Silver-sta<strong>in</strong>ed 15% SDS–PAGE reduc<strong>in</strong>g gel of prote<strong>in</strong>s elutedfrom equ<strong>in</strong>e embryonic capsules before and after conceptus fixation atabout day 16. Lane 1, MW markers; lanes 2, 4 and 6 are from normalpregnancies; lanes 3 and 5 are from mares treated with a luteolytic doseof a prostagland<strong>in</strong> F 2a analogue and show <strong>in</strong>creased amounts of p17,identified as secretory phospholipase A2 (sPLA2). Luteolysis had lesseffect on the proteolytic products of b 2 M (p10 to p8) and uterocal<strong>in</strong>(p19 to p5 and p6)Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


234 MA Hayes, BA Qu<strong>in</strong>n, ND Keirstead, P Katavolos, RO Waelchli and KJ Betteridgewith tryps<strong>in</strong> fragment masses consistent with uterocal<strong>in</strong>.By day 18, most of the p10 form of b 2 M <strong>in</strong> the capsulehad been converted to the p8 form (D9-b 2 M) that lacksn<strong>in</strong>e am<strong>in</strong>o acids from the N-term<strong>in</strong>us (Qu<strong>in</strong>n et al.2007).A band of 17 kDa (p17) was observed around thetime of fixation <strong>in</strong> normal capsules (Fig. 1), consistentwith previous studies (Qu<strong>in</strong>n et al. 2007). This wasmarkedly <strong>in</strong>creased <strong>in</strong> capsules from mares that weretreated with PGF 2a (Fig. 1). The p17 band of a day 18conceptus from a treated mare was tentatively identifiedas a secretory phospholipase A2 (sPLA2) based onhomology of two partial sequences (K)LLNYKFSYRand (K)YQYYNNK determ<strong>in</strong>ed by MS ⁄ MS. Thesesequences were most similar to a rat sPLA2 (gi: 220858),the cDNA sequence of which was used to identify as<strong>in</strong>gle cDNA sequence from an equ<strong>in</strong>e articular cartilagecDNA library (gi: 57706753). Primers designed from thissequence were used to amplify a complete cDNAsequence from cDNA prepared by RT-PCR of RNAextracted from endometrium from a normal pregnantmare (NM_001100113). This is 100% identical with acod<strong>in</strong>g sequence <strong>in</strong> chromosome 2 of the horse genome(chr2:32592684–32595740). The predicted am<strong>in</strong>o acidsequence <strong>in</strong>cluded the two partial sequences we identifiedby MS ⁄ MS of the p17 band. Also, MALDI-TOFanalysis of tryps<strong>in</strong> digested p17 matched six peptidemasses of a theoretical digest of the predicted sPLA2prote<strong>in</strong> from our cDNA sequence. The predictedsequence also had homology with an N-term<strong>in</strong>alsequence (XXLXFXKMIXLMTGKQAT) reported fora 17 kDa progesterone-dependent secreted phospholipase<strong>in</strong> the equ<strong>in</strong>e uterus (Beier-Hellwig et al. 1995).Polyclonal antibodies generated aga<strong>in</strong>st a syntheticpeptide (KEATSSYGFYGC) of our sequence alsorecognized p17 <strong>in</strong> immunoblots of capsules from normaland treated mares (not shown).The p17 band was not detected <strong>in</strong> 2D-PAGE ofcontrol capsules (Fig. 2a,b). However, <strong>in</strong> capsules fromtreated mares, a s<strong>in</strong>gle 17 kDa spot was present at pI 9.8on days 16 (Fig. 2c) and 18 (not shown).By 2D-PAGE of pooled days 13–15 control capsuleextracts, a 19 kDa uterocal<strong>in</strong> spot was observed at pI 9.5(Fig. 2a). The p19 spot was less prom<strong>in</strong>ent <strong>in</strong> control(Fig. 2b) and treated days 16 and 18 capsules (Fig. 2c),whereas a doublet of smaller spots observed at pI 9.5were consistent with uterocal<strong>in</strong> determ<strong>in</strong>ed by MS ⁄ MSsequences (Fig. 2b).Before fixation (Fig. 2a), there were three prom<strong>in</strong>ent10 kDa bands and one 8 kDa band <strong>in</strong> the pI rangeof 5.0–6.5 that were identified as forms of b 2 M byMS ⁄ MS peptide sequences of <strong>in</strong>-gel tryps<strong>in</strong> digests.After fixation, the 10 kDa forms of b 2 M decreasedwhereas the 8 kDa form became more prom<strong>in</strong>ent(Fig. 2b).To evaluate the changes <strong>in</strong> uter<strong>in</strong>e production ofsPLA2, we exam<strong>in</strong>ed concentrated uter<strong>in</strong>e flush fluidsamples from control and treated pregnancies. At day 16of gestation, flush samples from control mares conta<strong>in</strong>edvariable amounts of a 19 kDa band (Fig. 3a), consistentwith uterocal<strong>in</strong> by immunoblott<strong>in</strong>g (Qu<strong>in</strong>n et al. 2007)and by MALDI-TOF analysis of <strong>in</strong>-gel tryps<strong>in</strong> digests.By comparison, there was a less abundant band atpI 5 7 11p19p17p10p19p17p10p19p17p10Fig. 2. Silver-sta<strong>in</strong>ed 2D SDS–PAGE reduc<strong>in</strong>g gel of prote<strong>in</strong>s elutedfrom equ<strong>in</strong>e embryonic capsules. (a) Controls pooled from days 13.5 to15.5; (b) control from day 16; (c) treated from day 166 kDa (Fig. 3a), that migrated to the position ofimmunoreactive uteroglob<strong>in</strong> also found <strong>in</strong> previousstudies of uter<strong>in</strong>e flush samples (Qu<strong>in</strong>n et al. 2007).Uteroglob<strong>in</strong>s migrate faster than expected for theircalculated molecular masses (Mu¨ller-Scho¨ttle et al.2002; Mukherjee et al. 2007). The p6 uteroglob<strong>in</strong> bandswere moderately <strong>in</strong>creased <strong>in</strong> <strong>in</strong>tensity <strong>in</strong> the silversta<strong>in</strong>ed gels of uter<strong>in</strong>e flush fluids from mares that weretreated with the PGF 2a analogue compared with normalmares (Fig. 3a). However, <strong>in</strong> Western blots with polyclonalantibodies raised aga<strong>in</strong>st the EPSKPDADMKAATTQLKTLV, uteroglob<strong>in</strong> bands gave a markedlyÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Prote<strong>in</strong>s <strong>in</strong> Early Equ<strong>in</strong>e Conceptuses 235pI 3 11p19p17p101 2 3 4 5 6 7 8p6pI 5.0 pI 5.8Fig. 4. Silver-sta<strong>in</strong>ed 2D SDS–PAGE reduc<strong>in</strong>g gel of prote<strong>in</strong>s <strong>in</strong>uter<strong>in</strong>e flush collected from a control mare on day 16.5. Two bands <strong>in</strong>the 6 kDa region were identified by MS ⁄ MS sequenc<strong>in</strong>g as forms ofuteroglob<strong>in</strong>. The major band at pI 4.9–5.0 and the m<strong>in</strong>or band at pI5.8–6.0 both conta<strong>in</strong>ed sequences of products of two uteroglob<strong>in</strong> genes(see Table 1)Fig. 3. Silver-sta<strong>in</strong> (a) and immunoblot with antibody to a syntheticuteroglob<strong>in</strong> peptide (b) of 15% SDS-PAGE reduc<strong>in</strong>g gel of prote<strong>in</strong>s <strong>in</strong>uter<strong>in</strong>e flush samples collected on day 16.5 from control (lanes 1–4)and PGF 2a -treated (lanes 5–8) pregnant maresstronger signal <strong>in</strong> treated mares than <strong>in</strong> normal mares(Fig. 3b). This suggested that the form of uteroglob<strong>in</strong>that <strong>in</strong>creased after PGF 2a was different from theuteroglob<strong>in</strong> band <strong>in</strong> the normal pregnant uterus. By2D PAGE, the p6 bands migrated as a major form at pI4.9–5.0 and a m<strong>in</strong>or form at pI 5.8–6.0 (Fig. 4), both ofwhich were identified as uteroglob<strong>in</strong> by MS ⁄ MSsequenc<strong>in</strong>g of <strong>in</strong>-gel tryps<strong>in</strong> digests (Table 1). Bothspots were composed of two forms, based on am<strong>in</strong>o acidsequences predicted from two uteroglob<strong>in</strong> horse genomicsequences (GenBank 124072669). Two peptides ofmasses 932.547 (KTLVDFLPKN) and 1292.622(AVEPFKPDADMKA) were consistent with horsep6uteroglob<strong>in</strong> reference sequence NP_001075327 andsecretoglob<strong>in</strong> ⁄ CCSP of equ<strong>in</strong>e lung orig<strong>in</strong> (AAW83215,AAW83216, AAW83215 and AAW83218). By comparison,two other sequenced peptides of masses 1362.712(KLMDKIAKSPLCA) and 1391.705 (KLMDKIVESPLCA) present <strong>in</strong> the major pI 4.9–5.0 spot <strong>in</strong>dicatedthat two secretoglob<strong>in</strong>s <strong>in</strong> the whole genome shotgunsequence of the horse (gi 124072669) were expressed.The <strong>in</strong>crease <strong>in</strong> immunoreactive uteroglob<strong>in</strong> after luteolysiswas attributed to a selective <strong>in</strong>crease <strong>in</strong> thesecretoglob<strong>in</strong> 1A1 form (NP_001075327) that <strong>in</strong>cludesthe peptide with tyros<strong>in</strong>e rather than alan<strong>in</strong>e at position59 used to generate the antibody (Katavolos 2006).The m<strong>in</strong>or spot (pI 5.8–6.0) was <strong>in</strong>creased threefold(p = 0.025) at day 18 <strong>in</strong> the treated mares <strong>in</strong> comparisonwith control mares (N = 3 per group), as quantifiedby 2D-DIGE.DiscussionThese studies were conducted to identify potential prote<strong>in</strong>biomarkers that might dist<strong>in</strong>guish successful and unsuccessfulfixation of the equ<strong>in</strong>e conceptus, or that might helpto expla<strong>in</strong> the molecular basis of these processes. For thisTable 1. Am<strong>in</strong>o acid sequences of two forms of uteroglob<strong>in</strong> <strong>in</strong> uter<strong>in</strong>e flush fluid from a day 15 normal pregnancySpot A,pISpot B,pIMeasuredmassPeptide sequence determ<strong>in</strong>edby MALDI-TOF and MS ⁄ MSSimilarity with knownuteroglob<strong>in</strong> ⁄ CCSP sequences4.9–5.0 5.8–6.0 gi:20385506,gi:126352306gi:58978622, gi:58978613,gi:58978651, gi:58978641+ + 932.551 (K)TLVDFLPK(N) Yes Yes+ 1275.740 (K)TLVDFLPKNTK(D) Yes No+ + 1292.625 (A)VEPFKPDADMK(A) Yes Yes+ 1362.712 (K)LMDKIAKSPLCA(–) Yes No+ + 1391.705 (K)LMDKIVESPLCA(–) No Yes+ 1832.060 (K)TLVDFLPKNTKDSILK(L) Yes NoÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


236 MA Hayes, BA Qu<strong>in</strong>n, ND Keirstead, P Katavolos, RO Waelchli and KJ Betteridgecomparison, we used an experimental model of PGF 2a -<strong>in</strong>duced luteolysis that leads to failure of fixation anddelayed loss of sialic acid from the embryonic capsule(Chu et al. 1997; Betteridge et al. 2006). These studiesfollow on from a previous demonstration that b 2 Massociated with the normal capsule is truncated to an8 kDa form (D9-b 2 M) around the time of fixation (Qu<strong>in</strong>net al. 2007). The present studies show that b 2 M exists asmultiple 10 kDa forms and one 8 kDa form that can beseparated by 2D-PAGE, and that luteolysis has only am<strong>in</strong>or <strong>in</strong>fluence on proteolysis or b<strong>in</strong>d<strong>in</strong>g of b 2 M associatedwith the capsule.The most obvious luteolysis-associated alteration <strong>in</strong>capsule prote<strong>in</strong>s was an <strong>in</strong>crease <strong>in</strong> a 17 kDa secretoryphospholipase A2 (sPLA2) classified as type IIA on thebasis of am<strong>in</strong>o acid and cDNA sequence homology of thecDNA sequence we obta<strong>in</strong>ed from pregnant endometrium.Previous reports have noted the presence of apartially characterized PLA2 <strong>in</strong> the normal pregnantuterus (Beier-Hellwig et al. 1995; Qu<strong>in</strong>n et al. 2007) butits function there is unknown. PLA2s are a complexgroups of enzymes that cleave glycerophospholipids atthe sn-2 position and release a free fatty acid, most oftenarachidonic acid which is the cyclooxygenase substrateprecursor of various eicosanoids. PLA2s can thereby havediverse signall<strong>in</strong>g roles that are especially important <strong>in</strong><strong>in</strong>flammation and haemostasis, and <strong>in</strong> the regulation ofovarian function, pregnancy and parturition (Tithof et al.2007). Secretory PLA2-IIA has a high pI and b<strong>in</strong>ds toanionic phospholipid <strong>in</strong>terface rather than phospholipids<strong>in</strong> <strong>in</strong>tact cell membranes (Beers et al. 2003; Birts et al.2007). Human sPLA2-IIA also has an ability to b<strong>in</strong>d tohepar<strong>in</strong>-sulphate proteoglycans (Birts et al. 2007). Whilethe functions of sPLA2-IIA are still unclear, the evidencesupports the view that they relate to its role as an <strong>in</strong>nateimmune prote<strong>in</strong> <strong>in</strong>volved <strong>in</strong> the catabolism of cell debris(bacteria, apoptotic cells) <strong>in</strong> extracellular locations (Birtset al. 2007). Equ<strong>in</strong>e uter<strong>in</strong>e sPLA2 characterized <strong>in</strong> ourstudies also has a high pI of 9.8, b<strong>in</strong>ds to the embryoniccapsule and <strong>in</strong>creases when fixation has been blocked byadm<strong>in</strong>istration of PGF 2a . It is therefore plausible thatsPLA2 contributes to the imm<strong>in</strong>ent removal and degradationof the capsule or the conceptus.Uterocal<strong>in</strong> is a well characterized lipocal<strong>in</strong> secreted byendometrial glands <strong>in</strong> the luteal stage of the ovariancycle and <strong>in</strong> the early stages of normal pregnancy <strong>in</strong>equids (Stewart et al. 2000; Suire et al. 2001; Kennedy2005). Uterocal<strong>in</strong> is also a highly cationic prote<strong>in</strong>(pI 9.4 similar to that of sPLA2) (Fig. 2) and thisproperty likely expla<strong>in</strong>s why it also b<strong>in</strong>ds to theembryonic capsule. Recent evidence suggests that uterocal<strong>in</strong>is <strong>in</strong>volved <strong>in</strong> the transport of small lipophilic,ma<strong>in</strong>ly nutrient, substances across the glycan capsuleand <strong>in</strong>to the yolk sac (Suire et al. 2001; Kennedy 2005;Qu<strong>in</strong>n et al. 2007). Amounts <strong>in</strong> the uter<strong>in</strong>e flush werelower after treatment with PGF 2a , consistent withprogesterone dependence (Suire et al. 2001). Theamounts present <strong>in</strong> the capsule were variable and bydays 16 and 18, some had been proteolytically convertedto smaller fragments of approximately 10 kDa (Fig. 2b).It appears likely that this alteration is part of the processof degradation of the capsule rather than a processrelevant to its function.Observations <strong>in</strong> the present study <strong>in</strong>dicate that thereare at least two uteroglob<strong>in</strong> genes expressed <strong>in</strong> theequ<strong>in</strong>e uterus dur<strong>in</strong>g early pregnancy. The form thatwas more consistent immunoreactively with equ<strong>in</strong>euteroglob<strong>in</strong> (Mu¨ller-Scho¨ttle et al. 2002) was <strong>in</strong>creasedsubstantially after luteolysis. Uteroglob<strong>in</strong> was firstidentified as a low molecular weight secreted uter<strong>in</strong>eprote<strong>in</strong> <strong>in</strong> rabbits and is the found<strong>in</strong>g member of thelarge secretoglob<strong>in</strong> superfamily of prote<strong>in</strong>s that also<strong>in</strong>cludes Clara cell secretory prote<strong>in</strong>s (CCSP) (Beier2000; Mu¨ller-Scho¨ttle et al. 2002; Mukherjee et al.2007). These prote<strong>in</strong>s are expressed <strong>in</strong> various tissues,ma<strong>in</strong>ly <strong>in</strong> the lung and uterus. Uteroglob<strong>in</strong> (secretoglob<strong>in</strong>1A1) is expressed <strong>in</strong> various mammals <strong>in</strong>clud<strong>in</strong>g thehorse (Mu¨ller-Scho¨ttle et al. 2002) and there are threeclosely similar genes <strong>in</strong> the horse genome, located onchromosome 12. Previous studies showed that theuteroglob<strong>in</strong> that is detected <strong>in</strong> immunoblots with anantibody to a peptide sequence of uteroglob<strong>in</strong> ⁄ secretoglob<strong>in</strong>1A1 is more abundant <strong>in</strong> the non-pregnant uterusthan dur<strong>in</strong>g the fixation period of normal pregnancy(Qu<strong>in</strong>n et al. 2007). Uteroglob<strong>in</strong> has a small lipophilicb<strong>in</strong>d<strong>in</strong>g pocket (von der Decken et al. 2005) and canb<strong>in</strong>d prostagland<strong>in</strong>s such as PGF 2a (Mukherjee et al.2007) but the significance of these properties is unclear.Secretoglob<strong>in</strong>s can b<strong>in</strong>d various small lipophilic molecules<strong>in</strong>clud<strong>in</strong>g ret<strong>in</strong>oids and polychlor<strong>in</strong>ated biphenyls(von der Decken et al. 2005; Mukherjee et al. 2007).CCSP has anti-<strong>in</strong>flammatory functions <strong>in</strong> the respiratorytract, where it might b<strong>in</strong>d lipid mediators andsPLA2 (Mukherjee et al. 2007). Further studies arerequired to determ<strong>in</strong>e whether there is a relationshipbetween the <strong>in</strong>creases <strong>in</strong> sPLA2 and uteroglob<strong>in</strong> <strong>in</strong>response to luteolysis.These studies have employed analytical methods witha level of sensitivity most suitable for demonstration ofchanges <strong>in</strong> the most abundant prote<strong>in</strong>s. It is expectedthat there are also alterations <strong>in</strong> many cytok<strong>in</strong>es andenzymes that are not apparent <strong>in</strong> these approaches.Moreover, it is still unclear whether the modifications <strong>in</strong>prote<strong>in</strong>s and glycans that can be observed <strong>in</strong> the capsuledur<strong>in</strong>g and after the period of fixation are functionallyimportant <strong>in</strong> <strong>in</strong>teractions between the endometrium andconceptus. Some of these changes might be part of thenormal removal of the capsule. However, the <strong>in</strong>crease <strong>in</strong>sPLA2-type IIA <strong>in</strong> the capsule and a change <strong>in</strong> theexpression of uteroglob<strong>in</strong> genes might have a role <strong>in</strong> thepend<strong>in</strong>g demise of the conceptus <strong>in</strong> some circumstances.Alternatively, these changes might reflect the return tothe non-pregnant post-luteolysis condition.AcknowledgementsThis research was supported by the Natural Sciences and Eng<strong>in</strong>eer<strong>in</strong>gResearch Council of Canada (NSERC); the Grayson Jockey ClubResearch Foundation Inc.; Equ<strong>in</strong>e Guelph; The E.P. Taylor researchFoundation; and the Ontario M<strong>in</strong>istry of Agriculture, Food and RuralAffairs (OMAFRA). Natalie Keirstead was supported by a Fellowshipfrom the Canadian Institutes of Health Research (CIHR). We thankDorothee Bienzle, Jeff Caswell, Gordon Kirby and John Lumsden foruse of analytical equipment funded by the Canadian Foundation forInnovation (CFI). We are grateful for technical advice of DavidHobson and Paul Huber <strong>in</strong> relation to DIGE methods, and for MSanalysis by Li Zhang at the Advanced Prote<strong>in</strong> Technology Centre,Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Prote<strong>in</strong>s <strong>in</strong> Early Equ<strong>in</strong>e Conceptuses 237Hospital for Sick Children, Toronto, Ontario. The assistance of CaraLategan, Mandy Mulder and Garrett Jack and staff of the ArkellResearch Station is appreciated.ReferencesAlbihn A, Waelchli RO, Samper J, Oriol JG, Croy BA,Betteridge KJ, 2003: Production of capsular material byequ<strong>in</strong>e trophoblast transplanted <strong>in</strong>to immunodeficient mice.<strong>Reproduction</strong> 125, 855–863.Arar S, Chan KH, Qu<strong>in</strong>n BA, Waelchli RO, Hayes MA,Betteridge KJ, Monteiro MA, 2007: Desialylation of coretype 1 O-glycan <strong>in</strong> the equ<strong>in</strong>e embryonic capsule co<strong>in</strong>cideswith immobilization of the conceptus <strong>in</strong> the uterus. CarbohydrRes 342, 1110–1115.Baker CB, Little TV, McDowell KJ, 1993: The live foal<strong>in</strong>g rateper cycle <strong>in</strong> mares. Equ<strong>in</strong>e Vet J 15, 28–30.Ball BA, 1988: Embryonic loss <strong>in</strong> mares: <strong>in</strong>cidence, possiblecauses and diagnostic considerations. Vet Cl<strong>in</strong>ics N AmEqu<strong>in</strong>e Pract 4, 263–290.Beers SA, Buckland AG, Giles N, Gelb MH, Wilton DC,2003: Effect of tryptophan <strong>in</strong>sertions on the properties of thehuman group IIA phospholipase A2: mutagenesis producesan enzyme with characteristics similar to those of the humangroup V phospholipase A2. Biochemistry 42, 7326–7338.Beier HM, 2000: The discovery of uteroglob<strong>in</strong> and itssignificance for reproductive biology and endocr<strong>in</strong>ology.Ann N Y Acad Sci 923, 9–24.Beier-Hellwig K, Kremer H, Bonn B, L<strong>in</strong>dner D, Beier HM,1995: Partial sequenc<strong>in</strong>g and identification of three prote<strong>in</strong>sfrom equ<strong>in</strong>e uter<strong>in</strong>e secretion regulated by progesterone.Reprod Domest Anim 30, 295–298.Betteridge KJ, 2007: Equ<strong>in</strong>e embryology: an <strong>in</strong>ventory ofunanswered questions. Theriogenology 68(Suppl 1), S9–S21.Betteridge KJ, Waelchli RO, Christie HL, Raeside JI, Qu<strong>in</strong>nBA, Hayes MA, 2006: Effects of luteolysis on conceptusdevelopment dur<strong>in</strong>g the second and third weeks of pregnancy<strong>in</strong> the mare. Anim Reprod Sci 91, 395–398.Birts CN, Barton CH, Wilton DC, 2007: A catalytically<strong>in</strong>dependentphysiological function for human acute phaseprote<strong>in</strong> group IIA phospholipase A2: cellular uptake facilitatescell debris removal. J Biol Chem 283, 5034–5045.Bradford MM, 1976: A rapid and sensitive method for thequantitation of microgram quantities of prote<strong>in</strong> utiliz<strong>in</strong>g thepr<strong>in</strong>ciple of prote<strong>in</strong>-dye b<strong>in</strong>d<strong>in</strong>g. Anal Biochem 72, 248–254.Brooks AS, Hammermueller J, DeLay JP, Hayes MA, 2003:Expression and secretion of ficol<strong>in</strong> beta by porc<strong>in</strong>e neutrophils.Biochim Biophys Acta 1624, 36–45.Carnevale EM, Ramirez RJ, Squires EL, Alvarenga MA,Vanderwall DK, McCue PM, 2000: Factors affect<strong>in</strong>gpregnancy rates and early embryonic death after equ<strong>in</strong>eembryo transfer. Theriogenology 54, 965–979.Chomczynski P, Sacchi N, 1987: S<strong>in</strong>gle-step method of RNAisolation by acid guanid<strong>in</strong>ium thiocyanate-phenol-chloroformextraction. Anal Biochem 162, 156–159.Chu JW, Sharom FJ, Oriol JG, Betteridge KJ, Cleaver BD,Sharp DC, 1997: Biochemical changes <strong>in</strong> the equ<strong>in</strong>e capsulefollow<strong>in</strong>g prostagland<strong>in</strong>-<strong>in</strong>duced pregnancy failure. MolReprod Dev 46, 286–295.Crossett B, Suire S, Herrler A, Allen WR, Stewart F, 1998:Transfer of a uter<strong>in</strong>e lipocal<strong>in</strong> from the endometrium of themare to the develop<strong>in</strong>g equ<strong>in</strong>e conceptus. Biol Reprod 59,483–490.von der Decken V, Delbruck H, Herrler A, Beier HM, FischerR, Hoffmann KM, 2005: Recomb<strong>in</strong>ant bov<strong>in</strong>e uteroglob<strong>in</strong>at 1.6 A resolution: a prelim<strong>in</strong>ary X-ray crystallographicanalysis. Acta Crystallogr F 61, 499–502.G<strong>in</strong>ther OJ, Bergfelt DR, Leith GS, Scraba ST, 1985:Embryonic loss <strong>in</strong> mares: <strong>in</strong>cidence and ultrasonic morphology.Theriogenology 24, 73–86.Hobson DJ, Rupa P, Diaz GJ, Zhang H, Yang M, M<strong>in</strong>e Y,Turner PV, Kirby GM, 2007: Proteomic analysis ofovomucoid hypersensitivity <strong>in</strong> mice by two-dimensionaldifference gel electrophoresis (2D-DIGE). Food ChemToxicol 45, 2372–2380.Katavolos P2006: The role of Clara cells <strong>in</strong> the pathogenesis ofequ<strong>in</strong>e recurrent airway obstruction. PhD Thesis, Universityof Guelph.Kennedy MW, 2005: Uterocal<strong>in</strong> – provider of essential lipidsand am<strong>in</strong>o acids to the pre-placentation equ<strong>in</strong>e conceptus.Havemeyer Found Monogr Ser 16, 53–56.Lillie BN, Hammermueller JD, Mac<strong>in</strong>nes JI, Jacques M,Hayes MA, 2006: Porc<strong>in</strong>e mannan-b<strong>in</strong>d<strong>in</strong>g lect<strong>in</strong> A b<strong>in</strong>ds toAct<strong>in</strong>obacillus suis and Haemophilus parasuis. Dev ComImmunol 30, 954–965.Morris LH, Allen WR, 2002: Reproductive efficiency of<strong>in</strong>tensively managed Thoroughbred mares <strong>in</strong> Newmarket.Equ<strong>in</strong>e Vet J 34, 51–60.Mukherjee AB, Zhang Z, Chilton BS, 2007: Uteroglob<strong>in</strong>: asteroid-<strong>in</strong>ducible immunomodulatory prote<strong>in</strong> that foundedthe secretoglob<strong>in</strong> superfamily. Endocr Rev 28, 707–725.Mu¨ller-Scho¨ttle F, Bogusz A, Grotz<strong>in</strong>ger J, Herrler A,Krusche CA, Beier-Hellwig K, Beier HM, 2002: Full-lengthcomplementary DNA and the derived am<strong>in</strong>o acid sequenceof horse uteroglob<strong>in</strong>. Biol Reprod 66, 1723–1728.Oriol JG, Betteridge KJ, Clarke AJ, Sharom FJ, 1993a:Muc<strong>in</strong>-like glycoprote<strong>in</strong>s <strong>in</strong> the equ<strong>in</strong>e embryonic capsule.Mol Reprod Dev 34, 255–265.Oriol JG, Sharom FJ, Betteridge KJ, 1993b: Developmentallyregulated changes <strong>in</strong> the glycoprote<strong>in</strong>s of the equ<strong>in</strong>eembryonic capsule. J Reprod Fertil 99, 653–664.Qu<strong>in</strong>n BA, Caswell DE, Lillie BN, Waelchli RO, BetteridgeKJ, Hayes MA, 2006: The GM2-activator prote<strong>in</strong> is a majorprote<strong>in</strong> expressed by the encapsulated equ<strong>in</strong>e trophoblast.Anim Reprod Sci 91, 391–394.Qu<strong>in</strong>n BA, Hayes MA, Waelchli RO, Kennedy MW, BetteridgeKJ, 2007: Changes <strong>in</strong> major prote<strong>in</strong>s <strong>in</strong> the embryonic capsuledur<strong>in</strong>g immobilization (fixation) of the conceptus <strong>in</strong> the thirdweek of pregnancy <strong>in</strong> the mare. <strong>Reproduction</strong> 134, 161–170.Stewart F, Kennedy MW, Suire S, 2000: A novel uter<strong>in</strong>elipocal<strong>in</strong> support<strong>in</strong>g pregnancy <strong>in</strong> equids. Cell Mol Life Sci57, 1373–1378.Stout TA, Meadows S, Allen WR, 2005: Stage-specificformation of the equ<strong>in</strong>e blastocyst capsule is <strong>in</strong>strumentalto hatch<strong>in</strong>g and to embryonic survival <strong>in</strong> vivo. Anim ReprodSci 87, 269–281.Suire S, Stewart F, Beauchamp J, Kennedy MW, 2001:Uterocal<strong>in</strong>, a lipocal<strong>in</strong> provision<strong>in</strong>g the preattachmentequ<strong>in</strong>e conceptus: fatty acid and ret<strong>in</strong>ol b<strong>in</strong>d<strong>in</strong>g properties,and structural characterization. Biochem J 356, 369–376.Tithof PK, Roberts MP, Guan W, Elgayyar M, Godk<strong>in</strong> JD,2007: Dist<strong>in</strong>ct phospholipase A2 enzymes regulate prostagland<strong>in</strong>E2 and F2alpha production by bov<strong>in</strong>e endometrialepithelial cells. Reprod Biol Endocr<strong>in</strong>ol 5, 16.Waelchli RO, Betteridge KJ, 1996: Osmolality of equ<strong>in</strong>eblastocyst fluid from days 11 to 25 of pregnancy. ReprodFertil Dev 8, 981–988.Author’s address (for correspondence): MA Hayes, Department ofPathobiology, Ontario Veter<strong>in</strong>ary College, University of Guelph,Guelph, Ontario, Canada N1G2W1. E-mail: ahayes@uoguelph.caConflict of <strong>in</strong>terest: MA Hayes declares no conflict of <strong>in</strong>terest; therema<strong>in</strong><strong>in</strong>g authors have not declared any conflict of <strong>in</strong>terests.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 238–244 (2008); doi: 10.1111/j.1439-0531.2008.01168.xISSN 0936-6768Heat Stress, the Follicle, and Its Enclosed Oocyte: Mechanisms and PotentialStrategies to Improve Fertility <strong>in</strong> Dairy CowsZ RothDepartment of Animal Science, Faculty of Agricultural, Food and Environmental Quality Sciences, The Hebrew University, Rehovot, IsraelContentsReduced reproductive performance of lactat<strong>in</strong>g cows dur<strong>in</strong>gthe summer is associated with decreased thermoregulatorycompetence due to <strong>in</strong>tensive genetic selection for high milkproduction. This review exam<strong>in</strong>es the immediate and delayedeffects of heat stress on follicular function and describes somepotential strategies for their alleviation. It focuses on how heatstress affects the follicle and its enclosed oocyte, suggest<strong>in</strong>gthat perturbations <strong>in</strong> the follicular microenvironment, towhich the oocytes are exposed for long periods of development,reduce their developmental competence. Among thepotential alterations are reduction <strong>in</strong> gonadotrop<strong>in</strong> secretion,alteration <strong>in</strong> follicular growth, attenuation of dom<strong>in</strong>ance, anddisruption of steroidogenesis. Evaporative cool<strong>in</strong>g methodsare the most common strategy used to alleviate the effect ofheat stress; however, there is a compell<strong>in</strong>g need to f<strong>in</strong>dadditional ways to improve fertility dur<strong>in</strong>g the summer andautumn. Hormonal treatment to enhance removal of theimpaired follicles by synchronization of follicular waves withGnRH and PGF2a is suggested. An alternative method isstimulation of follicular growth by a brief treatment with bSTor FSH. Other strategies, such as timed AI and embryotransfer, have been recently used, mak<strong>in</strong>g the optimization ofembryo cryopreservation procedures highly relevant. Protectionof the ovarian pool of oocytes from thermal stress vianutritional manipulations or adm<strong>in</strong>istration of antioxidants orother survival factors should also be considered. A betterunderstand<strong>in</strong>g of the underly<strong>in</strong>g mechanisms by which heatstress impairs fertility may lead to the development ofadditional approaches to alleviate these effects.IntroductionReduced reproductive performance of lactat<strong>in</strong>g cowsdur<strong>in</strong>g the summer has been well-documented and isassociated with decreased thermoregulatory competenceof the animal due to <strong>in</strong>tensive genetic selection for highmilk production. The most common strategy to alleviatethe effect of heat stress <strong>in</strong> dairy farms is to provide shadeand evaporative cool<strong>in</strong>g systems, based on a comb<strong>in</strong>ationof spr<strong>in</strong>kl<strong>in</strong>g and ventilation, to enable animals toma<strong>in</strong>ta<strong>in</strong> normothermia. This approach can reducebody temperature and <strong>in</strong>crease milk production; however,its effect on summer fertility is limited (Hansen1997).Dur<strong>in</strong>g the autumn, although the weather is coolerand the cows are no longer subjected to thermal stress,conception rates rema<strong>in</strong> low (Hansen 1997; Zeron et al.2001). Hyperthermia can directly disrupt concomitantfollicular function with adverse carry-over effects on itscompetence (Roth et al. 2000a,b). These <strong>in</strong>clude alterations<strong>in</strong> follicular development, <strong>in</strong>clud<strong>in</strong>g depression ofdom<strong>in</strong>ance, impairment of follicular steroidogenesis andalterations <strong>in</strong> gonadotrop<strong>in</strong> secretion (Wolfenson et al.2000). Given the complexity of the follicles’ activities,their <strong>in</strong>terdependence and their <strong>in</strong>teractions, perturbation<strong>in</strong> one component may disrupt oocyte developmentalcompetence (Webb and Campbell 2007). This reviewdescribes the immediate and delayed effects of heatstress on follicular function and some potential ways toalleviate them. It emphasizes the importance of theendocr<strong>in</strong>e milieu and follicular microenvironment towhich the ovarian pool of oocytes is exposed anddescribes potential impairments <strong>in</strong> the follicle-enclosedoocyte upon heat shock, which subsequently affect itsdevelopmental competence.Immediate and Delayed Effects of Heat Stresson Follicular FunctionHeat stress and follicular dynamicsThe use of ultrasonography <strong>in</strong> recent years has demonstratedthat expos<strong>in</strong>g dairy cows to elevated temperaturesalters follicular growth and function. Immediateeffects of heat stress <strong>in</strong>clude reduced size of the first- andsecond-wave dom<strong>in</strong>ant follicles (Bad<strong>in</strong>ga et al. 1993;Wilson et al. 1998a,b) and attenuation of dom<strong>in</strong>ance, asreflected by an <strong>in</strong>creased number of large-sized follicles(Bad<strong>in</strong>ga et al. 1993, 1994; Wolfenson et al. 1995; Rothet al. 2000a) and delayed regression of subord<strong>in</strong>atefollicles (Wilson et al. 1998b; Roth et al. 2000a). Moreover,heat stress impairs the growth of medium-sizedfollicles (6–9 mm), as <strong>in</strong>dicated by the earlier emergenceand delayed decrease of the second follicular wave(Roth et al. 2000a). Such alterations may lead to earlyemergence of the pre-ovulatory follicle and <strong>in</strong>creasedduration of dom<strong>in</strong>ance (Wolfenson et al. 1995), both ofwhich are negatively associated with conception rate(Mihm et al. 1994). The picture regard<strong>in</strong>g the effects ofheat stress on follicular recruitment is less clear (Wolfensonet al. 2000). A recent study <strong>in</strong> goats has <strong>in</strong>dicatedthat follicles exposed to heat stress dur<strong>in</strong>g recruitmentregress and never develop to large sizes or ovulate(Ozawa et al. 2005).Heat stress affects not only the antral folliclesemerg<strong>in</strong>g <strong>in</strong> the follicular wave, but also the ovarianpool of small antral follicles, result<strong>in</strong>g <strong>in</strong> carry-overeffects on follicular function. Growth of medium-sizedfollicles was attenuated <strong>in</strong> cows exposed to direct solarradiation <strong>in</strong> the previous oestrous cycle (Roth et al.2000a). The stage of follicular development that issusceptible to thermal stress has not been preciselydef<strong>in</strong>ed, but Roth et al. (2000a) <strong>in</strong>troduced evidencesuggest<strong>in</strong>g that early antral follicles of approximately0.5–1.0 mm <strong>in</strong> diameter are sensitive to heat stress.Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Follicular and Oocyte Competence under Heat Stress 239The endocr<strong>in</strong>e background govern<strong>in</strong>g alterations <strong>in</strong>follicular dynamics and depression of dom<strong>in</strong>ance underheat stress is not fully understood. Accumulat<strong>in</strong>gevidence <strong>in</strong>dicates alterations <strong>in</strong> systemic gonadotrop<strong>in</strong>,<strong>in</strong>hib<strong>in</strong> and steroid concentrations, as well as <strong>in</strong> steroidogenesis(for review, see Wolfenson et al. 2000). Heat<strong>in</strong>duced<strong>in</strong>creases <strong>in</strong> the number of medium-sizedfollicles (i.e. immediate effect) were associated with<strong>in</strong>creased concentrations of follicle-stimulat<strong>in</strong>g hormone(FSH) and reduced concentrations of <strong>in</strong>hib<strong>in</strong> <strong>in</strong> theplasma (Wolfenson et al. 1995; Roth et al. 2000a).Inhib<strong>in</strong> and oestradiol synergistically affect FSH secretion(Kaneko et al. 1995). Thus, heat-<strong>in</strong>duced impairmentof granulosa cell function, the ma<strong>in</strong> source ofplasma oestradiol and <strong>in</strong>hib<strong>in</strong>, can lead to <strong>in</strong>creasedFSH concentrations <strong>in</strong> the plasma (Roth et al. 2000a).Follicular steroid production under heat stressSeasonal studies report that lower steroid concentrations<strong>in</strong> the follicular fluid obta<strong>in</strong>ed from large folliclesdur<strong>in</strong>g the hot season are associated with reducedviability of granulosa cells and impaired aromataseactivity (Bad<strong>in</strong>ga et al. 1993; Wolfenson et al. 1995).Bridges et al. (2005) reported that follicle piecesobta<strong>in</strong>ed from heat-stressed cows secrete lower levelsof androstenedione and oestradiol upon gonadotrop<strong>in</strong>stimulation. Similarly, thecal cells <strong>in</strong>cubated at hightemperatures or collected from heat-stressed cows produceless androstenedione when stimulated by LH, butnot by forskol<strong>in</strong>, imply<strong>in</strong>g a disruption <strong>in</strong> LH receptorfunction upon heat stress (Wolfenson et al. 1995; Rothet al. 2001b). Nonetheless, expression of the mRNAencod<strong>in</strong>g LH receptors <strong>in</strong> bov<strong>in</strong>e thecal cells did notprovide evidence of such alterations (Roth et al. 2000b).In contrast, a recent study <strong>in</strong> goat reported decreasedexpression of LH receptors <strong>in</strong> heat-stressed follicles(Ozawa et al. 2005). Thus, many aspects of the molecularand cellular mechanisms underly<strong>in</strong>g heat-<strong>in</strong>duceddisruption of steroidogenesis rema<strong>in</strong> to be elucidated.Alterations of steroidogenic capacity <strong>in</strong>duced bysummer heat stress carry over to the f<strong>in</strong>al stage offollicle development, as evidenced by reduced androstenedioneproduction by thecal cells and low oestradiolconcentrations <strong>in</strong> follicular fluid collected from dom<strong>in</strong>antfollicles <strong>in</strong> the autumn (Wolfenson et al. 1997). Inagreement with this, decreased oestradiol and androstenedioneproduction was recorded <strong>in</strong> granulosa andthecal cells obta<strong>in</strong>ed from follicles three to four weeksafter acute heat stress (Roth et al. 2000a). Similarly,oestradiol content <strong>in</strong> the follicular fluid aspirated fromcows was relatively low <strong>in</strong> late summer and <strong>in</strong>creasedthroughout the autumn (Roth et al. 2004). Thus theextent of the heat stress effects on follicular function istransient as also reflected by the spontaneous improvementof fertility throughout autumn and early w<strong>in</strong>ter(Zeron et al. 2001).A strategy to enhance the removal of impairedfollicles and the reappearance of apparently normalfollicles was suggested: <strong>in</strong>duction of frequent follicularwaves by hormonal treatment (i.e. GnRH followed byPGF2a (counteracted the effect of summer heat stress(Guzeloglu et al. 2001). Apparently, stimulation ofgonadotrop<strong>in</strong>s by GnRH has a beneficial effect ondevelop<strong>in</strong>g follicles s<strong>in</strong>ce <strong>in</strong>duction of frequent follicularwaves dur<strong>in</strong>g the autumn <strong>in</strong>creased oestradiol content <strong>in</strong>pre-ovulatory follicles aspirated from previously heatstressedcows (Roth et al. 2004).Among the potential adverse effects associated withlow oestradiol levels are impairment of oestrus durationand <strong>in</strong>tensity, <strong>in</strong>creased <strong>in</strong>cidence of anoestrus, silentovulation, and a reduced number of mounts (Gwazdauskaset al. 1981). Poor oestrus detection can beimproved by us<strong>in</strong>g modern aids such as the Heat-Watchsystem, a radio-telemetric pressure transducer, pedometricoestrus detection or alternatively, utilization ofovulation-synchronization protocols with fixed-timeartificial <strong>in</strong>sem<strong>in</strong>ation (AI) procedures (Moore andThatcher 2006). Nevertheless, these procedures cannotovercome the negative effects of heat stress onconception.Reduced oestradiol concentrations <strong>in</strong> the blood dur<strong>in</strong>gthe follicular phase may also affect the pre-ovulatoryLH surge which, <strong>in</strong> turn, disrupts the cascade of eventslead<strong>in</strong>g to oocyte ovulation. This is demonstrated by thelow amplitude of the GnRH-<strong>in</strong>duced LH surge <strong>in</strong> heatstressedcows express<strong>in</strong>g low oestradiol concentration(Gilad et al. 1993). Nonetheless, studies <strong>in</strong> which GnRHwas adm<strong>in</strong>istered at the onset of oestrus (Ullah et al.1996; Kaim et al. 2003) <strong>in</strong>creased conception rates <strong>in</strong>heat-stressed primiparous cows, but this <strong>in</strong>crease wasless pronounced <strong>in</strong> multiparous cows.Heat Stress and Oocyte DevelopmentalCompetenceIn-vivo and <strong>in</strong>-vitro studies support the view that bov<strong>in</strong>eoocytes are susceptible to thermal stress at variousstages of follicular development. Perturbation <strong>in</strong> thephysiology of the follicle-enclosed oocyte dur<strong>in</strong>g thelengthy period of follicular development could potentiallylead to an oocyte with reduced competence forfertilization and subsequent development.Oocytes harvested from cows dur<strong>in</strong>g the summerexhibit a reduced ability to develop to the blastocyststage after <strong>in</strong>-vitro fertilization (Rocha et al. 1998; Al-Katanani et al. 2002) or chemical activation (Zeronet al. 2001). Cleavage to the two- and four-cell stagesfollow<strong>in</strong>g chemical activation was delayed <strong>in</strong> bov<strong>in</strong>eoocytes collected dur<strong>in</strong>g the hot season (May–November)relative to oocytes collected dur<strong>in</strong>g the cold season(December–April) (Aroyo et al. 2007b). The tim<strong>in</strong>g offirst cleavage (early vs delayed) is considered to havemajor long-last<strong>in</strong>g effects on subsequent embryonicdevelopmental potential (Fenwick et al. 2002), whichmay expla<strong>in</strong> the <strong>in</strong>ferior developmental competence ofoocytes collected dur<strong>in</strong>g the summer.Elevated air temperatures before oestrus have beenassociated with reduced fertility (Al-Katanani et al.1999; Chebel et al. 2004). Intensive cool<strong>in</strong>g from 1 daybefore oestrus to 8 days post-AI did not improve theconception rate of cows <strong>in</strong> the summer (Her et al. 1988).Similarly, us<strong>in</strong>g a fan-and-fogger cool<strong>in</strong>g system 42 daysbefore oocyte collection did not <strong>in</strong>crease the proportionof oocytes that developed to the blastocyst stage (Al-Katanani et al. 2002). Thus, either the oocytes wereÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


240 Z Rothalready compromised prior to the period of heat-stressrelief or the cool<strong>in</strong>g was not efficient enough to decreaseheat stress. A study performed from late summer toearly w<strong>in</strong>ter <strong>in</strong>dicated that a period of two to threeoestrous cycles is required for recovery from heatdamage and appearance of competent oocytes (Rothet al. 2001a). In support of this form of recovery,<strong>in</strong>duction of maternal hyperthermia <strong>in</strong> mice carried overthrough three pregnancy cycles, as expressed by a lowerpregnancy rate and smaller litter sizes <strong>in</strong> the first cycleand slight <strong>in</strong>creases <strong>in</strong> these parameters through thesecond and the third cycles after heat exposure (Aroyoet al. 2007a). It appears that not only the <strong>in</strong>dividualovulated oocyte, but also the ovarian pool of oocytescan be damaged dur<strong>in</strong>g heat exposure. Nevertheless, theexact follicular stage at which the enclosed oocyte issusceptible to thermal stress has not been def<strong>in</strong>ed.Germ<strong>in</strong>al-vesicle-stage oocytesMammalian oocytes are arrested at the prophase stageof the first meiotic division and acquire their meioticcompetence and fertilization potential <strong>in</strong> a stepwisemanner dur<strong>in</strong>g follicular development. In an attempt todeterm<strong>in</strong>e the effect of heat stress on germ<strong>in</strong>al-vesicle(GV)-stage oocytes, Payton et al. (2004) exam<strong>in</strong>ed theeffect of heat shock on cumulus oocyte complexes(COCs) held at the GV stage us<strong>in</strong>g S-roscovit<strong>in</strong>e, a cellcycle<strong>in</strong>hibitor. Exposure of GV-stage oocytes to 41°Cdid not impair GV breakdown but reduced the proportionof oocytes that progressed to metaphase II (MII)and was associated with further impairment of blastocystdevelopment. Similarly, <strong>in</strong> a recent study <strong>in</strong> mice,exposure of GV-stage oocytes to maternal hyperthermiadisrupted their developmental competence (Aroyo et al.2007a).The aforementioned concept of remov<strong>in</strong>g impairedfollicles also seems to be relevant to improv<strong>in</strong>g oocytequality, s<strong>in</strong>ce frequent follicle aspirations by an ovumpick-up procedure improved the morphology and developmentalcompetence of oocytes aspirated <strong>in</strong> the fallfollow<strong>in</strong>g the hot summer. Accord<strong>in</strong>gly, hormonaltreatment with FSH, which is known to stimulatefollicular growth, <strong>in</strong>creased the proportion of grade-1oocytes and the proportion that cleaved to the two-cellstage post-chemical activation. Similarly, short-termadm<strong>in</strong>istration of bov<strong>in</strong>e somatotrop<strong>in</strong> (bST) <strong>in</strong>creasedthe proportion of grade-1 oocytes, but it did notimprove cleavage rate (Roth et al. 2002). Nevertheless,neither treatment was able to <strong>in</strong>crease the rate ofblastocyst formation.MII-stage oocytesIn cattle, the process of oocyte maturation co<strong>in</strong>cideswith oestrus events, which are known to be impaired byheat stress, as already described. Expos<strong>in</strong>g animals toheat stress between the onset of oestrus and <strong>in</strong>sem<strong>in</strong>ationdisrupts subsequent embryonic development, withan <strong>in</strong>creased proportion of abnormal and retardedembryos (Putney et al. 1989; Ealy et al. 1993). In-vitrostudies also <strong>in</strong>dicate that exposure of cultured COCs toelevated temperatures dur<strong>in</strong>g the first 12 h of maturationdecreases their cleavage rate (Roth et al. 2004; Rothand Hansen 2005) and the proportion of oocytes thatdevelop <strong>in</strong>to blastocysts (Edwards and Hansen 1997;Roth and Hansen 2004a,b; Ju et al. 2005). Even thoughthe underly<strong>in</strong>g mechanism is not entirely clear, heatstress may directly affect the oocyte or mediate negativeeffects through an impaired follicular environment (i.e.follicular fluid steroid content) and the impaired functionof surround<strong>in</strong>g cumulus cells. Lenz et al. (1983)showed that cultur<strong>in</strong>g COCs at 41°C for 24 h alters theirfunction, reduces cumulus-cell hyaluronic acid production,and impedes meiosis resumption. More recentstudies have reported that heat shock impairs bothnuclear and cytoplasmic maturation events, such astranslocation of cortical granules to the oolemma(Payton et al. 2004), cytoskeleton rearrangement (Rothand Hansen 2005), and sp<strong>in</strong>dle formation (Ju et al.2005; Roth and Hansen 2005). Similarly, heat-shock<strong>in</strong>ducedperturbations of the sp<strong>in</strong>dle apparatus havebeen reported <strong>in</strong> mature porc<strong>in</strong>e oocytes (Ju and Tseng2004) and <strong>in</strong> parthenogenetically activated bov<strong>in</strong>eoocytes (Tseng et al. 2004). Such alterations maypotentially lead to <strong>in</strong>complete nuclear maturation (Paytonet al. 2004; Roth and Hansen 2005), fertilizationfailure, and ⁄ or abnormal zygote formation. Edwardset al. (2005) reported that cultur<strong>in</strong>g oocytes at 41°Cdoes not compromise their nuclear and cytoplasmicmaturation but accelerates the process, and they suggestedthat fertilization performed 5 h earlier canattenuate the deleterious effect on oocyte development.Roth and Hansen (2005) reported that most heatshockedoocytes fail to undergo maturation and fertilizationand are arrested at stages MI through MII.Moreover, heat-shock-<strong>in</strong>duced apoptosis has been documentedfor bov<strong>in</strong>e oocytes exposed to elevated temperaturesdur<strong>in</strong>g maturation, <strong>in</strong> which an <strong>in</strong>creasedproportion of oocytes express high caspase activity andTUNEL-positive reactions (Roth and Hansen 2004a,b).Similar f<strong>in</strong>d<strong>in</strong>gs have been recently reported for porc<strong>in</strong>eoocytes (Tseng et al. 2006). S<strong>in</strong>ce the anti-apoptoticmolecule sph<strong>in</strong>gos<strong>in</strong>e 1-phosphate (S1P) blocked theeffect of heat shock on progression through meiosis(Roth and Hansen 2004b), it is reasonable to assumethat heat-shock-<strong>in</strong>duced apoptosis is functionallyrelated to the <strong>in</strong>hibition of meiotic progression. Thesef<strong>in</strong>d<strong>in</strong>gs <strong>in</strong>dicate that more than one mechanism is<strong>in</strong>volved <strong>in</strong> <strong>in</strong>duc<strong>in</strong>g adverse heat-stress effects <strong>in</strong> theoocyte.Heat-<strong>in</strong>duced Oxidative Stress and OocyteCompetenceHeat-shock-<strong>in</strong>duced oxidative stress is <strong>in</strong>volved <strong>in</strong> earlyembryonic loss <strong>in</strong> mice, when stress is imposed at thezygote stage (Ozawa et al. 2002, 2004; Matsuzuka et al.2004, 2005). Whether it is also <strong>in</strong>volved earlier <strong>in</strong>disrupt<strong>in</strong>g the bov<strong>in</strong>e follicle-enclosed oocyte rema<strong>in</strong>sunclear. Oxidants such as H 2 O 2 can <strong>in</strong>duce meiotic cellcyclearrest as well as morphological changes characteristicof apoptosis <strong>in</strong> a stage-dependent manner,particularly <strong>in</strong> immature oocytes (Chaube et al. 2005).Reactive oxygen species (ROS) are essential, and serveas key signal<strong>in</strong>g molecules <strong>in</strong> the resumption of meiosisÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Follicular and Oocyte Competence under Heat Stress 241(Takami et al. 1999), whereas excessive ROS levelswith<strong>in</strong> the follicle are associated with <strong>in</strong>creased cytoplasmicdefects and abnormal chromosomal segregation(Van Blerkom et al. 1997).Given the importance of the oxidative status of theoocyte, and the fact that bov<strong>in</strong>e oocytes are rich <strong>in</strong> fattyacids (McEvoy et al. 2000), membrane fatty-acid compositionis considered a major factor <strong>in</strong> determ<strong>in</strong><strong>in</strong>g theoocyte’s fate (Matorras et al. 1998). Seasonally <strong>in</strong>creasedvariation <strong>in</strong> the fatty-acid profiles of thefollicular fluid and oocytes is associated with reduceddevelopment of the oocyte (Zeron et al. 2001). In the hotsummer, the proportions of saturated fatty acids <strong>in</strong>oocyte and granulosa cells are higher than those ofmono- and polyunsaturated fatty acids, imply<strong>in</strong>greduced oxidative status of the oocyte. Thus, adm<strong>in</strong>istrationof antioxidants to reduce fatty-acid oxidation isone suggested strategy to stabilize the oocyte membraneupon heat stress. A recent study <strong>in</strong> mice providesevidence that adm<strong>in</strong>istration of the antioxidant epigallocatech<strong>in</strong>gallate (EGCG; 100 mg ⁄ kg body weight)before <strong>in</strong>duction of hyperthermia <strong>in</strong>creases the proportionof <strong>in</strong>-vivo-derived zygotes that reach the blastocyststage (Aroyo et al. 2006). Similarly, <strong>in</strong>-vitro maturationwith polyphenols improved the production of bov<strong>in</strong>eembryos (Wang et al. 2007). A beneficial effect was alsoachieved by feed<strong>in</strong>g lactat<strong>in</strong>g cows the antioxidantb-carotene (400 mg ⁄ day) for 90 days before <strong>in</strong>sem<strong>in</strong>ation(Arechiga et al. 1998). Apparently, unlike forembryos (Hansen 2007b), antioxidant adm<strong>in</strong>istrationcan alleviate the effect of thermal stress on the ovarianpool of oocytes.Accumulat<strong>in</strong>g evidence suggests that fat supplementation<strong>in</strong>fluences reproductive processes that are notdirectly related to energy balance and has beneficialeffects on the follicle, oocyte, embryo, and uterus(Mattos et al. 2000; Thatcher et al. 2003). Alterations<strong>in</strong> fatty-acid composition of the oocyte might improveits developmental capacity. Zeron et al. (2002) reportedthat feed<strong>in</strong>g ewes a diet supplemented with Ca salt offish oil <strong>in</strong>creases the proportion of polyunsaturated fattyacids <strong>in</strong> the plasma and cumulus cells and improvesoocyte quality, as determ<strong>in</strong>ed by membrane <strong>in</strong>tegrity.Feed<strong>in</strong>g cows a diet enriched <strong>in</strong> unsaturated fatty acidsdur<strong>in</strong>g the summer did not have a beneficial effect onoocyte quality (Bilby et al. 2006), whereas a high level offeed<strong>in</strong>g (Adamiak et al. 2005) or a high level of fat <strong>in</strong> thediet (Fouladi-Nashta et al. 2007) improved oocytedevelopmental competence. Given the potential ofnutritional manipulation, further research is warrantedto identify specific fatty acids and fat levels <strong>in</strong> diets thatmay have a beneficial effect on the ovarian pool ofoocytes.Heat Stress and Early Embryonic DevelopmentStudies <strong>in</strong>dicate that embryonic loss under heat stress isdue to the sensitivity of early embryos to elevatedtemperatures. Embryos at early developmental stagesare more susceptible to thermal stress and become moreresistant dur<strong>in</strong>g later developmental phases (Hansen2007a,b). Exposure of cows to heat stress on day 1 (butnot on days 3, 5, or 7) after oestrus decreased thedevelopment and viability of embryos on day 8 (Ealyet al. 1993). Exposure of cows to elevated temperaturesbetween onset of oestrus and <strong>in</strong>sem<strong>in</strong>ation (Putney et al.1988) decreased subsequent embryo development. Similarly,<strong>in</strong>duction of heat shock <strong>in</strong> vitro blocked thedevelopment of two-cell-stage embryos but had only amoderate effect on four- to eight-cell-stage embryos anda limited effect on the morulae (Hansen 2007a). Nevertheless,the mechanism by which the develop<strong>in</strong>g embryoacquires heat resistance to cellular disruption caused byelevated temperatures is not known (for reviews, see Ju2005; Hansen 2007a).Given that oocytes and early embryos are particularlysensitive to heat stress, embryo-transfer procedures thatbypass the effects of heat stress on early embryonicdevelopment have been attempted (see review, Rutledge2001; Hansen 2007b). One major limitation of thisapproach is the poor survival of embryos follow<strong>in</strong>gfreez<strong>in</strong>g. Transferr<strong>in</strong>g <strong>in</strong>-vivo-derived frozen-thawedembryos <strong>in</strong>creased pregnancy rates for recipient cowsrelative to traditionally <strong>in</strong>sem<strong>in</strong>ated cows (Putney et al.1989). Similarly, the percentage of pregnancies dur<strong>in</strong>gthe hot season was greater for cows receiv<strong>in</strong>g <strong>in</strong>-vitroderivedfresh embryos but not for those receiv<strong>in</strong>g frozenembryos (Ambrose et al. 1999; Drost et al. 1999).Moreover, use of vitrification did not improve thesurvival of the transferred embryos (Al-Katanani et al.2002). There is thus a compell<strong>in</strong>g need to optimizeprocedures for embryo cryopreservation.Currently, fresh or <strong>in</strong>-vivo-derived frozen embryos arethe ma<strong>in</strong> source for embryo-production programs.Therefore, treatments that protect the ovarian pool ofoocytes can <strong>in</strong>crease the number of competent oocytesavailable for embryo transfer. A protective effect of S1Phas been reported for bov<strong>in</strong>e oocytes exposed to heatshock dur<strong>in</strong>g maturation (Roth and Hansen 2004b,2005), as reflected by the reduced proportion of oocytesthat undergo apoptosis and the <strong>in</strong>creased number thatreach the MII stage and develop <strong>in</strong>to blastocysts. Theseembryos seem to have a high potential for development,s<strong>in</strong>ce embryos derived from heat-shocked and S1Pprotectedoocytes were of the same quality as those fromnon-stressed oocytes, as determ<strong>in</strong>ed by their total cellnumber, the proportion of apoptotic cells, and the<strong>in</strong>tensity of caspase activity (Roth and Hansen 2004b).Moreover, pups developed from heat-stressed mice didnot differ from control pups <strong>in</strong> their learn<strong>in</strong>g potentialor episodic memory, as determ<strong>in</strong>ed by behavioural andrecognition tests (Aroyo et al. 2007a).SummaryHyperthermia can impair cellular function <strong>in</strong> varioustissues of the reproductive system. However, disruptionof the follicle and its enclosed oocyte seems to be apivotal factor <strong>in</strong> the complex mechanism via which heatstress impairs fertility. This <strong>in</strong>cludes alterations <strong>in</strong> theendocr<strong>in</strong>e milieu and follicular microenvironment towhich the ovarian pool of oocytes is exposed, lead<strong>in</strong>g totheir decreased developmental competence. Hyperthermiacan directly disrupt follicular function, but a carryovereffect on the follicle and its enclosed oocyte is alsoevident.Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


242 Z RothEvaporative cool<strong>in</strong>g systems used to ma<strong>in</strong>ta<strong>in</strong> normothermia<strong>in</strong> high-lactat<strong>in</strong>g cows are likely to rema<strong>in</strong>obligatory <strong>in</strong> dairy management dur<strong>in</strong>g the hot season.However, s<strong>in</strong>ce they only partially restore normalfertility, there is a compell<strong>in</strong>g need for additionalapproaches. Beneficial strategies might <strong>in</strong>volve hormonaltreatment to enhance the removal of impairedfollicles, for example: synchronization of recurr<strong>in</strong>gfollicular waves with the use of GnRH and PGF2a, orstimulation of follicular growth with brief treatments ofbST or small doses of FSH. More precise treatmentsdesigned to mitigate the deleterious effect on theendocr<strong>in</strong>e milieu and to <strong>in</strong>crease post-<strong>in</strong>sem<strong>in</strong>ationplasma progesterone should be further exam<strong>in</strong>ed. Otherstrategies, such as timed embryo transfer, have beenmoderately successful. Therefore, optimiz<strong>in</strong>g the proceduresfor cryopreservation might allow produc<strong>in</strong>gembryos dur<strong>in</strong>g the cool months of the year, when theoocytes are not subjected to heat damage. Protection ofthe ovarian oocyte pool from thermal stress throughnutritional manipulations, adm<strong>in</strong>istration of ROSand ⁄ or other survival factors warrant further <strong>in</strong>vestigation.AcknowledgementsThe author thanks his colleagues, D. Wolfenson from the HebrewUniversity of Jerusalem and P.J. Hansen from the University ofFlorida, for their contribution to the study of reproduction <strong>in</strong> heatstressedcows which was discussed <strong>in</strong> this paper.ReferencesAdamiak J, Mackie K, Watt RG, Webb R, S<strong>in</strong>clair KD, 2005:Impact of nutrition on oocyte quality: cumulative effects ofbody composition and diet lead<strong>in</strong>g to hyper<strong>in</strong>sul<strong>in</strong>emia <strong>in</strong>cattle. Biol Reprod 73, 918–926.Al-Katanani YM, Webb DW, Hansen PJ, 1999: Factorsaffect<strong>in</strong>g seasonal variation <strong>in</strong> 90-day non-return rate to firstservice <strong>in</strong> lactat<strong>in</strong>g Holste<strong>in</strong> cows <strong>in</strong> a hot climate. J DairySci 82, 2611–2616.Al-Katanani YM, Paula-Lopes FF, Hansen PJ, 2002: Effect ofseason and exposure to heat stress on oocyte competence <strong>in</strong>Holste<strong>in</strong> cows. J Dairy Sci 85, 390–396.Ambrose JD, Drost M, Monson RL, Rutledge JJ, Leibfried-Rutledge ML, Thatcher MJ, Kassa T, B<strong>in</strong>elli M, Hansen PJ,Chenoweth PJ, Thatcher WW, 1999: Efficacy of timedembryo transfer with fresh and frozen <strong>in</strong> vitro producedembryos to <strong>in</strong>crease pregnancy rates <strong>in</strong> heat-stressed dairycattle. J Dairy Sci 82, 2369–2376.Arechiga CF, Staples CR, McDowell LR, Hansen PJ, 1998:Effects of timed <strong>in</strong>sem<strong>in</strong>ation and supplemental beta-caroteneon reproduction and milk yield of dairy cows underheat stress. J Dairy Sci 81, 390–402.Aroyo A, Yav<strong>in</strong> S, Roth Z, Arav A, 2006: Pretreatment withantioxidant epigallocatech<strong>in</strong> gallate (EGCG) counteractsthe negative effects of maternal hyperthermia on embryonicdevelopment <strong>in</strong> mice. Reprod Fertil Dev 18, 185. Abstract156).Aroyo A, Yav<strong>in</strong> S, Arav A, Roth Z, 2007a: Maternalhyperthermia disrupts developmental competence of follicle-enclosedoocytes: <strong>in</strong> vivo and ex vivo studies <strong>in</strong> mice.Theriogenology 67, 1013–1021.Aroyo A, Yav<strong>in</strong> S, Roth Z, Arav A, 2007b: H<strong>in</strong>der<strong>in</strong>g ofcleavage tim<strong>in</strong>g <strong>in</strong> bov<strong>in</strong>e parthenotes dur<strong>in</strong>g the hot season.Reprod Fertil Dev 19, 203. Abstract 173).Bad<strong>in</strong>ga L, Thatcher WW, Diaz T, Drost M, Wolfenson D,1993: Effect of environmental heat stress on folliculardevelopment and steroidogenesis <strong>in</strong> lactat<strong>in</strong>g Holste<strong>in</strong> cows.Theriogenology 39, 797–810.Bad<strong>in</strong>ga L, Thatcher WW, Wilcox CJ, Morris G, Entwistle K,Wolfenson D, 1994: Effect of season on follicular dynamicsand plasma concentrations of oestradiol-17ß, progesteroneand lute<strong>in</strong>iz<strong>in</strong>g hormone <strong>in</strong> lactat<strong>in</strong>g Holste<strong>in</strong> cows. Theriogenology42, 1263–1274.Bilby TR, Block J, do Amaral BC, Sa Filho O, Silvestre FT,Hansen PJ, Staples CR, Thatcher WW, 2006: Effects ofdietary unsaturated fatty acids on oocyte quality andfollicular development <strong>in</strong> lactat<strong>in</strong>g dairy cows <strong>in</strong> summer.J Dairy Sci 89, 3891–3903.Bridges PJ, Brusie MA, Fortune JE, 2005: Elevated temperature(heat stress) <strong>in</strong> vitro reduces androstenedione andestradiol and <strong>in</strong>creases progesterone secretion by follicularcells from bov<strong>in</strong>e dom<strong>in</strong>ant follicles. Domest Anim Endocr<strong>in</strong>ol29, 508–522.Chaube SK, Prasad PV, Thakur SC, Shrivastav TG, 2005:Hydrogen peroxide modulates meiotic cell cycle and <strong>in</strong>ducesmorphological features characteristic of apoptosis <strong>in</strong> ratoocytes cultured <strong>in</strong> vitro. Apoptosis 10, 863–874.Chebel RC, Santos JE, Reynolds JP, Cerri RL, Juchem SO,Overton M, 2004: Factors affect<strong>in</strong>g conception rate afterartificial <strong>in</strong>sem<strong>in</strong>ation and pregnancy loss <strong>in</strong> lactat<strong>in</strong>g dairycows. Anim Reprod Sci 84, 239–255.Drost M, Ambrose JD, Thatcher M-J, Cantrell CK, WolfsdorfKE, Hasler JF, Thatcher WW, 1999: Conception rates afterartificial <strong>in</strong>sem<strong>in</strong>ation or embryo transfer <strong>in</strong> lactat<strong>in</strong>g dairycows dur<strong>in</strong>g summer <strong>in</strong> Florida. Theriogenology 52, 1161–1167.Ealy AD, Drost M, Hansen PJ, 1993: Developmental changes<strong>in</strong> embryonic resistance to adverse effects of maternal heatstress <strong>in</strong> cows. J Dairy Sci 76, 2899–2905.Edwards JL, Hansen PJ, 1997: Differential responses of bov<strong>in</strong>eoocytes and preimplantation embryos to heat shock. MolReprod Dev 46, 138–145.Edwards JL, Saxton AM, Lawrence JL, Payton RR, DunlapJR, 2005: Exposure to a physiologically relevant elevatedtemperature hastens <strong>in</strong> vitro maturation <strong>in</strong> bov<strong>in</strong>e oocytes.J Dairy Sci 88, 4326–4333.Fenwick J, Platteau P, Murdoch AP, Herbert M, 2002: Timefrom <strong>in</strong>sem<strong>in</strong>ation to first cleavage predicts developmentalcompetence of human preimplantation embryos <strong>in</strong> vitro.Hum Reprod 17, 407–412.Fouladi-Nashta AA, Gutierrez CG, Gong JG, GarnsworthyPC, Webb R, 2007: Impact of dietary fatty acids on oocytequality and development <strong>in</strong> lactat<strong>in</strong>g dairy cows. BiolReprod 77, 9–17.Gilad E, Meidan R, Berman A, Graber Y, Wolfenson D,1993: Effect of heat stress on tonic and GnRH-<strong>in</strong>ducedgonadotroph<strong>in</strong> secretion <strong>in</strong> relation to concentration ofoestradiol <strong>in</strong> plasma of cyclic cows. J Reprod Fertil 99,315–321.Guzeloglu A, Ambrose JD, Kassa T, Diaz T, Thatcher MJ,Thatcher WW, 2001: Long-term follicular dynamics andbiochemical characteristics of dom<strong>in</strong>ant follicles <strong>in</strong> dairycows subjected to acute heat stress. Anim Reprod Sci 66, 15–34.Gwazdauskas FC, Thatcher WW, Kiddy CA, Paape MJ,Wilcox CZ, 1981: Hormonal patterns dur<strong>in</strong>g heat stressfollow<strong>in</strong>g PGF2a-tham salt <strong>in</strong>duced luteal regression <strong>in</strong>heifers. Theriogenology 16, 271–285.Hansen PJ, 1997: Effects of environment of bov<strong>in</strong>e reproduction.In: Youngquist RS (ed.), Current Therapy <strong>in</strong> LargeAnimal Theriogenology. Saunders WB, Philadelphia, pp.403–415.Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Follicular and Oocyte Competence under Heat Stress 243Hansen PJ, 2007a: To be or not to be; determ<strong>in</strong>ants ofembryonic survival follow<strong>in</strong>g heat shock. Theriogenology68, 40–48. (Review).Hansen PJ, 2007b: Exploitation of genetic and physiologicaldeterm<strong>in</strong>ants of embryonic resistance to elevated temperatureto improve embryonic survival <strong>in</strong> dairy cattle dur<strong>in</strong>gheat stress. Theriogenology 68, 242–249. (Review).Her E, Wolfenson D, Flamenbaum I, Folman Y, Kaim M,Berman A, 1988: Thermal productive, and reproductiveresponses of high yield<strong>in</strong>g cows exposed to short termcool<strong>in</strong>g <strong>in</strong> summer. J Dairy Sci 71, 1085–1092.Ju JC, 2005: Cellular responses of oocyte and embryos underthermal stress: h<strong>in</strong>ts to molecular signal<strong>in</strong>g. Anim Reprod 2,79–90.Ju JC, Tseng JK, 2004: Nuclear and cytoskeletal alterations of<strong>in</strong> vitro matured porc<strong>in</strong>e oocytes under hyperthermia. MolReprod Dev 68, 125–133.Ju JC, Jiang S, Tseng JK, Parks JE, Yang X, 2005: Heat shockreduces developmental competence and alters sp<strong>in</strong>dle configurationof bov<strong>in</strong>e oocytes. Theriogenology 64, 1677–1689.Kaim M, Bloch A, Wolfenson D, Braw-Tal R, Rosenberg M,Voet H, Folman Y, 2003: Effects of GnRH adm<strong>in</strong>istered tocows at the onset of estrus on tim<strong>in</strong>g of ovulation, endocr<strong>in</strong>eresponses, and conception. J Dairy Sci 86, 2012–2021.Kaneko H, Nakanishi Y, Akagi S, Arai K, Taya K, WatanabeG, Sasamoto S, Hasegawa Y, 1995: Immunoneutralizationof <strong>in</strong>hib<strong>in</strong> and oestradiol dur<strong>in</strong>g the follicular phase of theoestrous cycle <strong>in</strong> cows. Biol Reprod 53, 931–939.Lenz RW, Ball GD, Leibfried ML, Ax RL, First NL, 1983: Invitro maturation and fertilization of bov<strong>in</strong>e oocytes aretemperature-dependent processes. Biol Reprod 29, 173–179.Matorras R, Ruiz JI, Mendoza R, Ruiz N, Sanjurjo P,Rodriguez-Escudero FJ, 1998: Fatty acid composition offertilization-failed human oocytes. Hum Reprod 13, 2227–2230.Matsuzuka T, Ozawa M, Hirabayashi M, Ushitani A, KanaiY, 2004: Developmental competence and glutathione contentof maternally heat-stressed mouse oocytes and zygotes.Anim Sci J 75, 117–124.Matsuzuka T, Ozawa M, Nakamura A, Ushitani A, HirabayashiM, Kanai Y, 2005: Effects of heat stress on the redoxstatus <strong>in</strong> the oviduct and early embryonic development <strong>in</strong>mice. J Reprod Dev 51, 281–287.Mattos R, Staples CR, Thatcher WW, 2000: Effects of dietaryfatty acids on reproduction <strong>in</strong> rum<strong>in</strong>ants. Rev Reprod 5, 38–45. (Review).McEvoy TG, Coull GD, Broadbent PJ, Hutch<strong>in</strong>son JS,Speake BK, 2000: Fatty acid composition of lipids <strong>in</strong>immature cattle, pig and sheep oocytes with <strong>in</strong>tact zonapellucida. J Reprod Fertil 118, 163–170.Mihm M, Baguisi A, Boland MP, Roche JF, 1994: Associationbetween the duration of dom<strong>in</strong>ance of the ovulatory follicleand pregnancy rate <strong>in</strong> beef heifers. J Reprod Fertil 102, 123–130.Moore K, Thatcher WW, 2006: Major advances associatedwith reproduction <strong>in</strong> dairy cattle. J Dairy Sci 89, 1254–1266.Ozawa M, Hirabayashi M, Kanai Y, 2002: Developmentalcompetence and oxidative state of mouse zygotes heatstressedmaternally or <strong>in</strong> vitro. <strong>Reproduction</strong> 124, 683–689.Ozawa M, Matsuzuka T, Hirabayashi M, Kanai Y, 2004:Redox status of the oviduct and Cdc2 activity <strong>in</strong> 2-cell stageembryos <strong>in</strong> heat-stressed mice. Biol Reprod 71, 291–296.Ozawa M, Tabayashi D, Latief TA, Shimizu T, Oshima I,Kanai Y, 2005: Alterations <strong>in</strong> follicular dynamics andsteroidogenic abilities <strong>in</strong>duced by heat stress dur<strong>in</strong>g follicularrecruitment <strong>in</strong> goats. <strong>Reproduction</strong> 129, 621–630.Payton RR, Romar R, Coy P, Saxton AM, Lawrence JL,Edwards JL, 2004: Susceptibility of bov<strong>in</strong>e germ<strong>in</strong>al vesiclestageoocytes from antral follicles to direct effects of heatstress <strong>in</strong> vitro. Biol Reprod 71, 1303–1308.Putney DJ, Drost M, Thatcher WW, 1988: Embryonicdevelopment <strong>in</strong> superovulated dairy cattle exposed toelevated ambient temperatures between days 1–7 post<strong>in</strong>sem<strong>in</strong>ation. Theriogenology 33, 195–209.Putney DJ, Drost M, Thatcher WW, 1989: Influence ofsummer heat stress on pregnancy rates of lactat<strong>in</strong>g dairycattle follow<strong>in</strong>g embryo transfer or artificial <strong>in</strong>sem<strong>in</strong>ation.Theriogenology 31, 765–778.Rocha A, Randel RD, Broussard JR, Lim JM, Blair RM,Roussel JD, Godke RA, Hansel W, 1998: High environmentaltemperature and humidity decrease oocyte quality <strong>in</strong>Bos taurus but not <strong>in</strong> Bos <strong>in</strong>dicus cows. Theriogenology 49,657–665.Roth Z, Hansen PJ, 2004a: Involvement of apoptosis <strong>in</strong> heat<strong>in</strong>duceddisruption of competence of bov<strong>in</strong>e oocyte. BiolReprod 71, 1898–1906.Roth Z, Hansen PJ, 2004b: Sph<strong>in</strong>gos<strong>in</strong>e-1-Phosphate protectscultured bov<strong>in</strong>e oocytes from physiologically-relevant thermalstress. Biol Reprod 71, 2072–2078.Roth Z, Hansen PJ, 2005: Disruption of nuclear maturationand rearrangement of cytoskeletal elements <strong>in</strong> bov<strong>in</strong>eoocytes exposed to heat shock dur<strong>in</strong>g maturation. <strong>Reproduction</strong>129, 235–244.Roth Z, Meidan R, Braw-Tal R, Wolfenson D, 2000a:Immediate and delayed effects of heat stress on folliculardevelopment and its association with plasma FSH and<strong>in</strong>hib<strong>in</strong> concentration <strong>in</strong> cows. J Reprod Fertil 120, 83–90.Roth Z, Meidan R, Preger E, Levi N, Braw-Tal R, WolfensonD 2000b: Delayed effects of heat stress on steroidogenesis <strong>in</strong>bov<strong>in</strong>e preovulatory follicles. 14th International Congresson Animal <strong>Reproduction</strong>, Stockholm, Sweden, Abst. 1, 15.Roth Z, Arav A, Bor A, Zeron Y, Braw-Tal R, WolfensonD, 2001a: Improvement of quality of oocytes collected <strong>in</strong>the autumn by enhanced removal of impaired folliclesfrom previously heat-stressed cows. <strong>Reproduction</strong> 122,737–744.Roth Z, Meidan R, Braw-Tal R, Shaham-Albalancy A,Wolfenson D, 2001b: Delayed effect of heat stress onsteroidogenesis <strong>in</strong> bov<strong>in</strong>e medium-size and preovulatoryfollicles. <strong>Reproduction</strong> 121, 745–751.Roth Z, Arav A, Bor A, Zeron Y, Braw-Tal R, Wolfenson D,2002: Effect of treatment with FSH or bST on the quality ofoocytes aspirated <strong>in</strong> the autumn from previously heatstressedcows. J Dairy Sci 85, 1398–1405.Roth Z, Bor A, Braw-Tal R, Wolfenson D, 2004: Carry-overeffect of summer thermal stress on characteristics of the preovulatoryfollicle of lactat<strong>in</strong>g cows. J Therm Biol 29, 681–685.Rutledge JJ, 2001: Use of embryo transfer and IVF to bypasseffects of heat stress. Theriogenology 55, 105–111. (Review).Takami M, Preston SL, Toyloy VA, Behrman HR, 1999:Antioxidants reversibly <strong>in</strong>hibit the spontaneous resumptionof meiosis. Am J Physiol 276, 684–688.Thatcher WW, Guzeloglu A, Meikle A, Kamimura S, Bilby T,Kowalski AA, Bad<strong>in</strong>ga L, Persh<strong>in</strong>g R, Bartolome J, SantosJE, 2003: Regulation of embryo survival <strong>in</strong> cattle. ReprodSuppl 61, 253–266. (Review).Tseng JK, Chen CH, Chou PC, Yeh SP, Ju JC, 2004:Influences of follicular size on parthenogenetic activationand <strong>in</strong> vitro heat shock on the cytoskeleton <strong>in</strong> cattle oocytes.Reprod Domest Anim 39, 146–153.Tseng JK, Tang PC, Ju JC, 2006: In vitro thermal stress<strong>in</strong>duces apoptosis and reduces development of porc<strong>in</strong>eparthenotes. Theriogenology 15, 1073–1082.Ullah G, Fuquay JW, Keawkhong T, Clark BL, Pogue DE,Murphey EJ, 1996: Effect of gonadotrop<strong>in</strong>-releas<strong>in</strong>g hormoneat estrus on subsequent luteal function and fertility <strong>in</strong>Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


244 Z Rothlactat<strong>in</strong>g Holste<strong>in</strong>s dur<strong>in</strong>g heat stress. J Dairy Sci 79, 1950–1953.Van Blerkom J, Antczak M, Schrader R, 1997: The developmentalpotential of the human oocyte is related to thedissolved oxygen content of follicular fluid: association withvascular endothelial growth factor levels and perifollicularblood flow characteristics. Hum Reprod 12, 1047–1055.Wang ZG, Yu SD, Xu ZR, 2007: Improvement <strong>in</strong> bov<strong>in</strong>eembryo production <strong>in</strong> vitro by treatment with green teapolyphenols dur<strong>in</strong>g <strong>in</strong> vitro maturation of oocytes. AnimReprod Sci 100, 22–31.Webb R, Campbell BK, 2007: Development of the dom<strong>in</strong>antfollicle: mechanisms of selection and ma<strong>in</strong>tenance of oocytequality. Soc Reprod Fertil Suppl 64, 141–163.Wilson SJ, Kirby CJ, Koenigsfeld AT, Keisler DH, Lucy MC,1998a: Effects of controlled heat stress on ovarian functionof dairy cattle. 2. Heifers. J Dairy Sci 81, 2132–2138.Wilson SJ, Marion RS, Spa<strong>in</strong> JN, Spiers DE, Keisler DH,Lucy MC, 1998b: Effects of controlled heat stress on ovarianfunction of dairy cattle. 1. Lactat<strong>in</strong>g cows. J Dairy Sci 81,2124–2131.Wolfenson D, Thatcher WW, Bad<strong>in</strong>ga L, Savio JD, Meidan R,Lew BJ, Braw-Tal R, Berman A, 1995: Effect of heat stresson follicular development dur<strong>in</strong>g the oestrous cycle <strong>in</strong>lactat<strong>in</strong>g dairy cattle. Biol Reprod 52, 1106–1113.Wolfenson D, Lew BJ, Thatcher WW, Graber Y, Meidan R,1997: Seasonal and acute heat stress effects on steroidproduction by dom<strong>in</strong>ant follicles <strong>in</strong> cows. Anim Reprod Sci47, 9–19.Wolfenson D, Roth Z, Meidan R, 2000: Impaired reproduction<strong>in</strong> heat-stressed cattle: basic and applied aspects. AnimReprod Sci 61, 535–547. (Review).Zeron Y, Ocheretny A, Kedar O, Borochov A, Sklan D, AravA, 2001: Seasonal changes <strong>in</strong> bov<strong>in</strong>e fertility: relation todevelopmental competence of oocytes, membrane propertiesand fatty acid composition of follicles. <strong>Reproduction</strong> 121,447–454.Zeron Y, Sklan D, Arav A, 2002: Effect of polyunsaturatedfatty acid supplementation on biophysical parameters andchill<strong>in</strong>g sensitivity of ewe oocytes. Mol Reprod Dev 61, 271–278.Author’s address (for correspondence): Z Roth, Department ofAnimal Science, Faculty of Agricultural, Food and EnvironmentalQuality Sciences, The Hebrew University, PO Box 12, Rehovot 76100,Israel. E-mail: roth@agri.huji.ac.ilConflict of <strong>in</strong>terest: The author declares no conflict of <strong>in</strong>terest.Research was supported <strong>in</strong> part by BARD grant F1–330–2002.Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 245–251 (2008); doi: 10.1111/j.1439-0531.2008.01169.xISSN 0936-6768Fertilization and Early Embryonic Development <strong>in</strong> the Porc<strong>in</strong>e Fallopian TubeK-P Bru¨ssow 1 ,JRátky 2 and H Rodriguez-Mart<strong>in</strong>ez 31 Department of Reproductive Biology, FBN Research Institute for the Biology of Farm <strong>Animals</strong>, Dummerstorf, Germany; 2 Department of Biology of<strong>Reproduction</strong>, Research Institute for Animal Breed<strong>in</strong>g and Nutrition, Herceghalom, Hungary; 3 Division of <strong>Reproduction</strong>, Faculty of Veter<strong>in</strong>aryMedic<strong>in</strong>e and Animal Science, Swedish University of Agricultural Sciences (SLU), Uppsala, SwedenContentsFertilization and early embryo development relies on acomplex <strong>in</strong>terplay between the Fallopian tube and thegametes before and after fertilization. Thereby the oviduct,as a dynamic reproductive organ, enables reception, transportand maturation of male and female gametes, theirfusion, and supports early embryo development. This paperreviews current knowledge regard<strong>in</strong>g physiological processesbeh<strong>in</strong>d the transport of boar spermatozoa, their storage <strong>in</strong>and release from the functional sperm reservoir (SR), and ofthe <strong>in</strong>teractions that newly ovulated oocytes play with<strong>in</strong> thetube dur<strong>in</strong>g their transport to the site of fertilization.Experimental evidence of an ovarian control on spermrelease from the SR is highlighted. Furthermore, the impactof oviductal secretion on sperm capacitation, oocyte maturation,fertilization and early embryo development isstressed.IntroductionThe oviduct (synonyms Fallopian tube, salp<strong>in</strong>x, uter<strong>in</strong>etube) firstly described by Gabriel Fallopius <strong>in</strong> 1561, wasconsidered a simple connection between the ovary andthe uterus for a long time. Today, the oviduct isregarded as one of the most dynamic reproductiveorgans, whose functional aspects are not yet fullyunderstood (Rodriguez-Mart<strong>in</strong>ez et al. 2001). The oviductprovides a favourable environment for spermatozoa,enabl<strong>in</strong>g their transport and ensur<strong>in</strong>g beneficialconditions dur<strong>in</strong>g storage while prepar<strong>in</strong>g them forfertilization; as well as oocytes, mak<strong>in</strong>g ovum pick-upand transport to the site of fertilization possible, whileensur<strong>in</strong>g their f<strong>in</strong>al maturation. Both fertilization andearly embryonic development take place with<strong>in</strong> theoviduct. This paper reviews physiological processesbeh<strong>in</strong>d the transport of boar spermatozoa, their storage<strong>in</strong> and release from the functional sperm reservoir (SR),and of the <strong>in</strong>teractions that newly ovulated oocytes playdur<strong>in</strong>g their transport to the site of fertilization. Someaspects are highlighted, such as experimental evidencefor an ovarian <strong>in</strong>fluence on sperm release from the SRand the relevance of oviductal secretion on spermcapacitation, oocyte maturation, fertilization and earlyembryo development.The OviductThe porc<strong>in</strong>e oviduct is approximately 25 cm <strong>in</strong> length(Bru¨ ssow 1985) and it is anatomically divided <strong>in</strong>to threema<strong>in</strong> segments, e.g. the <strong>in</strong>fundibulum, the ampulla andthe isthmus. Connect<strong>in</strong>g areas, i.e. the term<strong>in</strong>al section(ostium), the ampulla-isthmic-junction (AIJ) and theutero-tubal-junction (UTJ) are also dist<strong>in</strong>guished. Thehistoarchitecture of the Fallopian tube is simple, witha non-glandular mucosa (endosalp<strong>in</strong>x), covered with al<strong>in</strong><strong>in</strong>g epithelium with secretory and ciliated cells,a double-layered smooth muscle (myosalp<strong>in</strong>x) and acover<strong>in</strong>g serosa (mesosalp<strong>in</strong>x). Towards the ostium, thethickness of the <strong>in</strong>ternal, circular muscle becomesth<strong>in</strong>ner; the longitud<strong>in</strong>al mucosal plicae ga<strong>in</strong> complexityand the number of ciliated cells considerably<strong>in</strong>creases (Rodriguez-Mart<strong>in</strong>ez et al. 2001; Yaniz et al.2006). This histoarchitecture builds tubal compartmentswith apparently different roles, all with optimal environmentwith regards to pH, osmotic pressure, nutrients,specific secretory products and signal molecules(reviewed by Rodriguez-Mart<strong>in</strong>ez 2007). Thus, preparationof gametes, fertilization and early embryonicdevelopment are supported and regulated by theoviduct.Sperm Transport and the Tubal SpermReservoirAfter deposition of spermatozoa <strong>in</strong> the cervix by mat<strong>in</strong>gor AI, a sperm subpopulation is rapidly transportedthrough the uterus, whereas the majority is elim<strong>in</strong>atedfrom the uter<strong>in</strong>e lumen (Rodriguez-Mart<strong>in</strong>ez et al.2005). Spermatozoa that ascended the uterus <strong>in</strong> the firstphase of sperm transport colonize the UTJ and thecaudal isthmus (>10 4 spermatozoa), a segment thatacts as a functional pre-ovulatory sperm reservoir,temporarily arrest<strong>in</strong>g spermatozoa (up to 30 h) and,presumably, activat<strong>in</strong>g them at a given time (Rodriguez-Mart<strong>in</strong>ez et al. 2005). In the SR, most spermatozoama<strong>in</strong>ta<strong>in</strong> normal ultrastructure and viability (Rodriguez-Mart<strong>in</strong>ezet al. 1990; Mburu et al. 1997). Severalconcerted factors are thought to expla<strong>in</strong> the formationof the functional SR, <strong>in</strong>clud<strong>in</strong>g the narrowed lumen(Hunter 1984), viscous mucus (Johansson et al. 2000),lower temperature (Hunter and Nichol 1986), localenzymatic and ionic milieu (Rodriguez-Mart<strong>in</strong>ez et al.1991), selective b<strong>in</strong>d<strong>in</strong>g of spermatozoa to the epithelium(Fazeli et al. 1999), and specific tubal fluid components(Tienthai et al. 2004), all which primarily leadto sperm quiescence.The sequential release of a restricted number ofspermatozoa from the SR towards the AIJ ensuresfertilization of oocytes with<strong>in</strong> a time w<strong>in</strong>dow, even ifovulation lasts over a longer <strong>in</strong>terval. The periovulatoryprogression of spermatozoa is suggested to be a complexand concerted process <strong>in</strong>clud<strong>in</strong>g open<strong>in</strong>g of the lumenby a decrease <strong>in</strong> the hormonally-driven endosalp<strong>in</strong>gealoedema, dissolution of the hyaluronan (HA)-richÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


246 K-P Bru¨ ssow, J Rátky and H Rodriguez-Mart<strong>in</strong>ezTable 1. Sperm subpopulations (as %; LSM ± SE) <strong>in</strong> the porc<strong>in</strong>e SRat pre-, peri- and post-ovulatory oestrus (modified from Tienthai et al.2004)RetrievedSR-spermatozoa (%)Oestrous stage at sperm retrieval (accounted formspontaneous ovulation)Preovulatory()8 h)Periovulatory(±4 h)Postovulatory(+8 h)Number of sows 7 10 7Viable, non-capacitated 72.8 ± 6.9 * 69.0 ± 6.8 * 70.9 ± 7.4 *Viable, capacitated 4.9 ± 3.0 * 1.4 ± 3.0 * 13.7 ± 3.0 *Dead 15.5 ± 5.3 * 16.9 ± 5.2 * 12.8 ± 5.7 **p < 0.05.mucus, hyperactive sperm motility, <strong>in</strong>creased flow oftubal fluid and re-directed oviductal contractions(Rodriguez-Mart<strong>in</strong>ez et al. 1998). On the other hand,there is no evidence of a massive sperm release orcapacitation dur<strong>in</strong>g the pre- and periovulatory periods(Table 1), but rather a constant sperm release towardsthe site of fertilization <strong>in</strong> relation to the occurrence ofspontaneous ovulation (Mburu et al. 1997; Tienthaiet al. 2004).What Controls Sperm Release from the SR?It is yet to be determ<strong>in</strong>ed which are mechanism(s) that<strong>in</strong>duce the sperm release from the porc<strong>in</strong>e SR. The mostmentioned is the proposed presence of discrete signalsact<strong>in</strong>g as transduced endocr<strong>in</strong>e <strong>in</strong>formation from thepre-ovulatory Graafian follicles (Hunter et al. 1983;Hunter 1990), although experimental evidence back<strong>in</strong>gthis mechanism is yet scarce.Experimental evidence for an ovarian <strong>in</strong>fluence on spermrelease from the SRMicro<strong>in</strong>jection of exogenous progesterone or progesterone-richfollicular fluid (FF) under the serosal layer ofthe oviduct surround<strong>in</strong>g the SR or directly <strong>in</strong>to the SRprovoked a prom<strong>in</strong>ent release of spermatozoa and a34% <strong>in</strong>cidence of polyspermy vs 2% <strong>in</strong> controls,<strong>in</strong>jected with steroid-free FF or medium (Hunter 1972;Hunter et al. 1999). This has lead to the assumptionthat progesterone could be the key effector of spermrelease from the pig SR. Progesterone concentrationsare 100 to 1000 times higher <strong>in</strong> the FF than <strong>in</strong>peripheral blood (Eiler and Nalbandov 1977; Blo¨ dowet al. 1990), and thus could stimulate sperm releasefrom the SR when enter<strong>in</strong>g the oviduct at ovulation.However, FF does not (or only <strong>in</strong> a negligible amount,Hansen et al. 1991) enter the porc<strong>in</strong>e oviduct, asdemonstrated by our own experiments, where the entryof FF <strong>in</strong>to the tube was prevented by unilateralendoscopic ligation of the oviduct on the uter<strong>in</strong>e sideof the <strong>in</strong>fundibulum or FF-aspiration before ovulation(Bru¨ ssow et al. 1999a, 2003). Analyses of progesteroneconcentration <strong>in</strong> FF and oviductal fluid showed thatwhile progesterone levels were similarly low prior to orafter ovulation (0.09 ± 0.13 vs 0.12 ± 0.16 ng ⁄ ml) anddid not differ from the contra-lateral control side (groupFF aspiration: 0.29 ± 0.17 vs 0.24 ± 0.35 ng ⁄ ml;Table 2. Percentages of spermatozoa detected <strong>in</strong> the ampulla andisthmus of <strong>in</strong>sem<strong>in</strong>ated gilts (n = 12) where FF entry <strong>in</strong>to the oviductwas prevented by aspiration of the FF or ligation of the oviduct on theuter<strong>in</strong>e side of the <strong>in</strong>fundibulum vs sham-ligation (Bru¨ ssow et al. 2003)Group(treatment vs control)Spermatozoa (%) with<strong>in</strong> oviductalsectionAmpullaIsthmusOviduct ligation 0 * 100 *Intact control 35.5 * 64.5 *FF aspiration 0 * 100 *Intact control 29.6 * 70.4 *Sham ligation 42.1 57.9Intact control 45.7 54.3*p < 0.05 vs control.group oviduct ligation: 0.22 ± 0.19 vs 0.21 ±0.22 ng ⁄ ml); those <strong>in</strong> FF were significantly higher(269.7 ± 67.9 and 389.6 ± 226.5 ng ⁄ ml) albeit notreflected <strong>in</strong> the oviductal fluid. Likewise, Hansen et al.(1991) found only 0.03 to 0.07% of the progesteroneconcentration of the FF was present with<strong>in</strong> the oviductalflush<strong>in</strong>g. Therefore, although a direct role ofprogesterone <strong>in</strong> elicit<strong>in</strong>g sperm release from the SR isyet to be determ<strong>in</strong>ed, a series of <strong>in</strong>terest<strong>in</strong>g results haveemanated from studies test<strong>in</strong>g the <strong>in</strong>volvement of FF <strong>in</strong>the process of fertilization. The effect of withdraw<strong>in</strong>gthe FF by aspiration of pre-ovulatory follicles or byoviduct ligation before ovulation was compared for itsrelation to the way spermatozoa were distributed with<strong>in</strong>the oviduct right after ovulation vs <strong>in</strong> sham-manipulatedor <strong>in</strong>tact oviducts (Bru¨ ssow et al. 1999b, 2003).Both a ligature of the oviduct or the FF-aspirationreduced the proportion of spermatozoa with<strong>in</strong> theampullar ⁄ AIJ tubal segment (Table 2), thus suggest<strong>in</strong>gthe sperm release from the SR was constra<strong>in</strong>ed. However,it is yet to be clarified whether an absentprogesterone-rich FF was the factor beh<strong>in</strong>d this difference.The FF is able to stimulate <strong>in</strong> vivo fertilization, asdemonstrated <strong>in</strong> a follow-up study (Bru¨ ssow et al. 2003)<strong>in</strong> which 6 to 10 cumulus-oocyte-complexes (COCs)recovered from donor gilts were transferred <strong>in</strong>to theoviducts of <strong>in</strong>sem<strong>in</strong>ated recipient gilts – either togetherwith 0.6 ml of FF <strong>in</strong> one oviduct (FF group – local<strong>in</strong>fluence of FF) or together with 0.6 ml of PBS <strong>in</strong>to thecontra-lateral oviduct (PBS group – without FF <strong>in</strong>fluence,with<strong>in</strong>-animal controls). Another group of giltsserved as sham-operated control-group. The FF wascollected from both ovaries <strong>in</strong> the recipient gilts andpooled, but the COCs were rejected, to dim<strong>in</strong>ishconfound<strong>in</strong>g effects. From this experiment, it wasevident that similarly to controls, the presence of FFcomponentswith<strong>in</strong> the oviduct <strong>in</strong>creased fertilizationrate (Table 3). However, s<strong>in</strong>ce a direct effect of FF isalready excluded <strong>in</strong> vivo, ow<strong>in</strong>g to the fact that the FFdoes not significantly enter the oviduct, neither it is anobligatory carrier of porc<strong>in</strong>e oocytes at ovulation(Bru¨ ssow et al. 1998a, 1999a); the <strong>in</strong>fluence of FFcomponents on sperm migration and fertilization ispossibly generated by local counter-current transfer <strong>in</strong>tothe blood and lymph circulation, as suggested by HunterÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Fertilization <strong>in</strong> the Porc<strong>in</strong>e Fallopian Tube 247Table 3. In vivo fertilization and cleavage rates of porc<strong>in</strong>e cumulusoocyte-complexes(COCs) transferred together with FF or PBS(n = 35 gilts; Bru¨ ssow et al. 2003)FF-treatedoviductsPBS-treatedoviductsControloviductsNumber of oviducts (n) 24 22 24Number of COCs transferred (n) 206 144 –Number of oocytes recovered (n) 138 70 235Recovery rate (%) 67.0 * 45.5 * 73.4 aNumber of oocytes fertilized (n) 78 26 119Fertilization rate (%) 56.5 * 31.7 * 50.6 *Number of cleaved embryos (n) 76 26 113Cleavage rate b (%) 97.4 100 98.3a Relative to the number of ovulated follicles.b Relative to the number of fertilized oocytes.*p < 0.05.et al. (1983), Hunter (1996). However, such hypothesisis yet to be proven.Involvement of oocytes and their cumulus cells <strong>in</strong> spermrelease form the SRThat oocytes and their cumulus vestment divert spermatozoatowards the ova has been considered previously(Aust<strong>in</strong> and Walton 1960; Harper 1973; Bedfordand Kim 1985), but putative molecular mechanisms arestill speculative (Hunter 1993), and experimental evidenceof an association of oocytes with SR-spermrelease <strong>in</strong> the pig is miss<strong>in</strong>g. Therefore, we created amodel <strong>in</strong> which ova from donor gilts were transferred atthe periovulatory period <strong>in</strong>to only one oviduct – theother serv<strong>in</strong>g as a control – of bilaterally ovectomized(aspiration of oocytes from the follicle) recipient giltsthat previously underwent endoscopic <strong>in</strong>trauter<strong>in</strong>e<strong>in</strong>sem<strong>in</strong>ation (endo-IUI) with low sperm-doses <strong>in</strong>toboth uter<strong>in</strong>e horns (Bru¨ ssow et al. 2006). Such a modelavoids ovulation of the recipient’s COCs, and enablesassessment of how the transfer of ova would <strong>in</strong>fluencesperm release from the SR. The most important f<strong>in</strong>d<strong>in</strong>gof this study was that the presence of COCs <strong>in</strong> theoviduct significantly <strong>in</strong>creased the percentages of spermatozoa<strong>in</strong> the ampullar and isthmic segments, comparedto control oviducts. Both total numbers andproportions of spermatozoa were always higher, <strong>in</strong>dicat<strong>in</strong>gthat the presence of ova affected sperm releasefrom the SR (Table 4). However, this result does notexpla<strong>in</strong> which component(s) of the oocytes promptedthe spermatozoa to leave the SR.Which components are <strong>in</strong>volved <strong>in</strong> the sperm release formthe SR?One of the components of the oocytes that may<strong>in</strong>fluence spermatozoa to leave the SR could be glycosam<strong>in</strong>oglycans(GAGs), especially the non-sulfatedGAG hyaluronan (HA) (Tienthai et al. 2004; Liberdaet al. 2006). The HA, a major component of the porc<strong>in</strong>eextracellular matrix of the cumulus and zona pellucida(ZP) (Yokoo et al. 2002) is <strong>in</strong>creas<strong>in</strong>gly synthesized byCOCs dur<strong>in</strong>g cumulus expansion (Kimura et al. 2002;Yokoo et al. 2007). It is thought to participate <strong>in</strong> spermcapacitation and release towards the site of fertilization(Tienthai et al. 2000a). S<strong>in</strong>ce the level of HA <strong>in</strong> theporc<strong>in</strong>e ampullar fluid <strong>in</strong>creases around ovulation(Tienthai et al. 2000b), it is possible that additionalHA enters the oviduct together with the COCs. Therefore,we experimentally analysed both the <strong>in</strong>fluence oftransferred ovum quality (COCs or cumulus cellremovedoocytes) and addition of HA (COCs + HA,oocytes + HA) on fertilization and the numbers ofZP-accessory spermatozoa <strong>in</strong> previously ovectomizedendo-IUI gilts (Bru¨ ssow et al. 2003, 2006). Embryodevelopment was not affected by ova quality or exogenousHA. The numbers of accessory spermatozoa werehighest <strong>in</strong> those COCs transferred together with HA(COC+HA), which did not differ from controls(Table 5). Comparable results have been achieved <strong>in</strong>previous experiments (Bru¨ ssow et al. 2003) where additionof exogenous HA, together with cumulus-freeoocytes <strong>in</strong>creased fertilization rates and the number ofaccessory spermatozoa compared to oocytes or COCs.One explanation for these results was that wash<strong>in</strong>gand ⁄ or manipulation of COCs <strong>in</strong> vitro, removed componentsimportant for fertilization. An alternative<strong>in</strong>terpretation was that the proposed impact of FF viacounter-current transfer (Hunter 1972) on sperm releaseis miss<strong>in</strong>g <strong>in</strong> gilts from which FF was withdrawn.Summariz<strong>in</strong>g the experimental data, the higher numberof accessory spermatozoa may strengthen the evidencethat HA participates <strong>in</strong> the process of sperm releasefrom the SR (Rodriguez-Mart<strong>in</strong>ez et al. 2005; Liberdaet al. 2006).Ovum Transport and Fertilization <strong>in</strong> theOviductOvulation, i.e. the ‘flow out’ of a mature folliclereleas<strong>in</strong>g a COC <strong>in</strong>to the oviduct, is a multifacetedTable 4. Distribution of boar spermatozoa (mean ± SE) with<strong>in</strong>oviductal sections <strong>in</strong> gilts (n = 10) with transferred COCs (treatedoviducts) or without COCs (control oviducts) (Bru¨ ssow et al. 2006)Treated oviducts (withCOCs)Control oviducts (withoutCOCs)n % n %Total sperm count 63 869 100 85 049 100Sperm reservoir 52 696 82.5 ± 0.5 * 80 921 95.2 ± 0.06 *Isthmus 5699 8.9 ± 0.11 * 2754 3.2 ± 0.06 *Ampulla 5474 8.6 ± 0.11 * 1374 1.6 ± 0.04 *Isthmus + Ampulla 11 173 17.5 ± 0.15 * 4128 4.9 ± 0.07 **p < 0.01 (chi-square).Table 5. Mean numbers (LSMeans ± SEM) of blastomeres and ofaccessory spermatozoa with<strong>in</strong> the zona pellucida <strong>in</strong> embryos develop<strong>in</strong>g<strong>in</strong> vivo follow<strong>in</strong>g oocyte or COC transfer, with or withoutexogenous HA (Bru¨ ssow et al. 2006)GroupBlastomeresAccessoryspermatozoaOocyte 2.6 ± 0.2 29.1 ± 3.1 *Oocyte + HA 2.9 ± 0.2 26.2 ± 3.9 *COC 3.1 ± 0.2 22.2 ± 3.3 *COC + HA 2.9 ± 0.2 46.2 ± 5.0 *Control 2.9 ± 0.2 45.2 ± 3.1 **p < 0.05.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


248 K-P Bru¨ ssow, J Rátky and H Rodriguez-Mart<strong>in</strong>ez<strong>in</strong>flammatory-like process <strong>in</strong>itiated by a surge <strong>in</strong> gonadotropichormones, followed by structural changes <strong>in</strong> thefollicular wall and vascular microcirculation, consequenceof the action of proteolytic enzymes andprostagland<strong>in</strong>s (Espey and Lipner 1994). At ovulation,oocytes at the metaphase II stage are picked up by thecilia-covered fimbria and guided through the <strong>in</strong>fundibulumand ampulla (Oxenreider and Day 1965; Alanko1974). Thereby oocytes with their cumulus vestmentaggregate with<strong>in</strong> an ‘egg plug’ (Tanabe et al. 1949;Spald<strong>in</strong>g et al. 1955). The rate of ovum transporttowards the AIJ is rapid (30–45 m<strong>in</strong>, Andersen 1927;Alanko 1965), with the <strong>in</strong>itial speed of ovum transportcalculated as 76 mm ⁄ h (Bru¨ ssow 1985). It is still notclear how ovum pick-up and transport are regulated, asCOCs are conveyed aga<strong>in</strong>st a flow of tubal fluid.Possibly, this is accomplished by a concerted <strong>in</strong>teractionbetween cumulus cells and the extracellular matrix theysynthesize (ma<strong>in</strong>ly HA) which, by dramatically <strong>in</strong>creas<strong>in</strong>gthe size of the egg plug, makes ciliary beat<strong>in</strong>g(Talbot et al. 1999), as well as myosalp<strong>in</strong>geal peristalsis(Rodriguez-Mart<strong>in</strong>ez et al. 1982) very effective <strong>in</strong> mov<strong>in</strong>gthe ova to the AIJ. Cumulus cells disperse with<strong>in</strong> 6 hpost-ovulation (Hunter and Dziuk 1968; Hunter 1974;Bru¨ ssow et al. 1987); a process which is strongly<strong>in</strong>fluenced by the presence of spermatozoa <strong>in</strong> the oviduct(Rodriguez-Mart<strong>in</strong>ez et al. 2001).The fertilization process with<strong>in</strong> the AIJ <strong>in</strong>cludes theb<strong>in</strong>d<strong>in</strong>g of spermatozoa to the glycoprote<strong>in</strong> cover<strong>in</strong>g ofthe oocyte, the ZP-penetration, and f<strong>in</strong>ally the fusionwith the oolemma. This generates the cortical reactionand subsequent a ZP-harden<strong>in</strong>g to prevent polyspermicpenetrations, a very effective system <strong>in</strong> vivo, but still aserious constra<strong>in</strong> <strong>in</strong> vitro (Funahashi et al. 2000, 2001).Gamete recognition, b<strong>in</strong>d<strong>in</strong>g and fusion are highlyregulated processes <strong>in</strong>volv<strong>in</strong>g a number of biochemicalevents (Jansen et al. 2001; Miller and Burk<strong>in</strong> 2001; Rathet al. 2006). The chronological and cytological details offertilization and early embryonic development <strong>in</strong> the pighave been thoroughly described by Hunter (1974). Earlyembryo development up to the 4-cell stage occurs with<strong>in</strong>the Fallopian tube. Fertilized embryos (appearance oftwo pronuclei) can be observed approximately 5–8 hafter ovulation <strong>in</strong> the AIJ and isthmus and rema<strong>in</strong> for alonger time (up to 26 h) <strong>in</strong> the one-cell stage. The 2-cellembryo stage is of short duration (6–8 h) and with<strong>in</strong>further 6–8 h they develop <strong>in</strong>to 4-cell embryos. Earlycleavage of embryos occurs <strong>in</strong> the isthmus, and mostlyat the 4-cell stage they leave the oviduct 50–60 h postovulation(Hancock 1961; Alanko 1974; Hunter 1974;Bru¨ ssow 1985). Blastulation is impeded <strong>in</strong> the oviduct(Oxenreider and Day 1965; Rodriguez-Mart<strong>in</strong>ez et al.1985), for reasons still unknown.Data on the transport of fertilized and non-fertilizedova through the porc<strong>in</strong>e oviduct are scarce and orig<strong>in</strong>ateonly from experiments with either <strong>in</strong>sem<strong>in</strong>ated or non<strong>in</strong>sem<strong>in</strong>atedsows. Some <strong>in</strong>fluence of fertilized embryoson oviductal transport was observed (Bru¨ ssow andRa´tky 1996), contrary to other authors (Mwanza et al.2002). However, other factors may <strong>in</strong>fluence ovumtransport. As such, PMSG-<strong>in</strong>duced superovulation(SO) alters the distribution of embryos <strong>in</strong> the oviductwith retardation <strong>in</strong> the ampulla on d1 after hCG<strong>in</strong>duced ovulation and an accelerated transport on d3(Bru¨ ssow et al. 1987). It rema<strong>in</strong>s unclear if this alterationis due to the higher number of oocytes releasedand ⁄ or an altered hormonal environment <strong>in</strong> the oviductafter SO. Stress-<strong>in</strong>duced alteration of cortisol levels byfood deprivation (Mburu et al. 1998) or exogenous-ACTH adm<strong>in</strong>istration (Razdan et al. 2002; Brandt et al.2007) <strong>in</strong>fluenced embryo development and distributionwith<strong>in</strong> the oviduct, as well as disturbed sperm transport,estimated on accessory sperm count (Brandt et al. 2006).Contribution of Oviductal Fluid on SpermCapacitation, Fertilization and EmbryoDevelopmentOviductal fluid (ODF) is formed by selective transudationfrom the blood and specific secretion from theoviduct epithelium; it differs from blood plasma <strong>in</strong>terms of ionic composition, pH, osmolarity and macromolecularcontent, and it varies with the endocr<strong>in</strong>estatus dur<strong>in</strong>g the oestrous cycle (Leese 1988; Leese et al.2001). Two areas important to prepare sperm forfertilization, such as sperm quiescence (ensur<strong>in</strong>g spermatozoaare kept alive and potentially fertile) andcapacitation (a destabiliz<strong>in</strong>g process, particularly relatedto the apical sperm plasma membrane; see reviewby Rodriguez-Mart<strong>in</strong>ez 2007). They are subscribed tobe <strong>in</strong>fluenced by the ODF; namely the presence ofdifferent pH environments and the presence of GAGs.For the first named, data from our laboratories <strong>in</strong>dicatethe SR possesses (at least pre-ovulation) an acidic pH,with low levels of local bicarbonate (Rodriguez-Mart<strong>in</strong>ez2007), which might be responsible for ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>gsperm quiescence and which prevents sperm capacitation<strong>in</strong> the SR shortly before ovulation (Mburu et al.1996; Tienthai et al. 2004). Our current results suggest asignificant proportion of boar spermatozoa retrievedfrom the pre-ovulatory SR and exposed to homologousODF of either pre-, peri- or post-ovulatory oestroussows only capacitate, when exposed to post-ovulatoryODF (Tienthai et al. 2004). Capacitation is, however,triggered if these SR-spermatozoa were exposed tobicarbonate (at levels recorded to be present <strong>in</strong> theAIJ ⁄ ampulla <strong>in</strong> vivo, e.g. 33–35 mM; Tienthai et al.2004). Bicarbonate is, therefore, considered a specificeffector of the process not only <strong>in</strong> pigs but also <strong>in</strong> otherspecies, such as the bov<strong>in</strong>e (Bergqvist et al. 2006). Suchexposure <strong>in</strong>duces sperm membrane destabilization and,ultimately, primes them to acrosomal exocytosis, asdemonstrated <strong>in</strong> vitro, and expected <strong>in</strong> vivo. It istherefore postulated that the constant release of <strong>in</strong>dividualspermatozoa out of the SR is sufficient to <strong>in</strong>ducetheir capacitation when adequate levels of the effectorare encountered outside of the SR area (Rodriguez-Mart<strong>in</strong>ez et al. 2005), thus mark<strong>in</strong>g capacitation as aperiovulatory process <strong>in</strong> vivo (Hunter and Rodriguez-Mart<strong>in</strong>ez 2004).The pig ODF conta<strong>in</strong>s GAGs, either non-sulphated(HA) or sulphated (S-GAGs, e.g. chondroit<strong>in</strong> sulphate,dermatan sulphate, keratan sulphate, heparan sulphateand hepar<strong>in</strong>) (Tienthai et al. 2001; Buhi 2002). Themean concentrations of total S-GAGs <strong>in</strong> ODF differbetween isthmus and ampulla and vary also <strong>in</strong> relationÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Fertilization <strong>in</strong> the Porc<strong>in</strong>e Fallopian Tube 249to the time of the oestrous cycle, be<strong>in</strong>g higher <strong>in</strong> theisthmus than <strong>in</strong> the ampulla, probably ow<strong>in</strong>g to thelarger secretory capacity of the latter (Tienthai et al.2001). The S-GAG-levels <strong>in</strong>crease significantly <strong>in</strong> isthmusdur<strong>in</strong>g the pre-ovulatory oestrus, to decreasetowards metoestrus, occurr<strong>in</strong>g <strong>in</strong> both tubae, probablydue to the bilateral ovarian activity <strong>in</strong> this species. Thenon-sulphated GAG HA is also present <strong>in</strong> the porc<strong>in</strong>eODF and without segmental differences, but with atendency to <strong>in</strong>crease dur<strong>in</strong>g stand<strong>in</strong>g oestrus, becom<strong>in</strong>ghighest around ovulation. Especially <strong>in</strong> the ampullarema<strong>in</strong>s high dur<strong>in</strong>g metoestrus (Tienthai et al. 2001).Both hyaluronan synthases, hyaluronan-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>sand specific membrane receptors are present <strong>in</strong> theepithelial l<strong>in</strong><strong>in</strong>g, particularly <strong>in</strong> the SR (Tienthai et al.2003a,b), where mucus accumulates before ovulation(Johansson et al. 2000). As mentioned before, GAGs,HA <strong>in</strong> particular, seem to be <strong>in</strong>volved <strong>in</strong> sperm survival,capacitation and b<strong>in</strong>d<strong>in</strong>g to and release from the SR (seeRodriguez-Mart<strong>in</strong>ez 2007).In-vitro development of porc<strong>in</strong>e oocytes hampers stillfrom normal development as <strong>in</strong>dicated by polyspermicfertilization. Apparently it is caused by <strong>in</strong>sufficientmaturation with a deficient formation of cortical granulesand results <strong>in</strong> constra<strong>in</strong>ed ZP-reaction after firstsperm penetration. Such situation was not seen when weexam<strong>in</strong>ed <strong>in</strong> vivo-matured or <strong>in</strong> vivo-fertilized oocytes,suggest<strong>in</strong>g that either the f<strong>in</strong>al maturation <strong>in</strong>side the preovulatoryfollicle (Bru¨ ssow, unpublished observations)or <strong>in</strong>side the oviduct is needed for full competence of thenewly ovulated oocytes. The latter seems most relevantconsider<strong>in</strong>g the f<strong>in</strong>al maturation of the ZP, as establishedby Funahashi et al. (2000, 2001), implicat<strong>in</strong>g theODF <strong>in</strong>volvement <strong>in</strong> these processes.The ODF is prote<strong>in</strong>-rich and conta<strong>in</strong>s several oviductspecific-prote<strong>in</strong>s(OSP) (Buhi et al. 1997; Buhi 2002;Buhi and Alvarez 2003). It supports fertilization andearly embryo development. For example, Wollenhauptand Bru¨ ssow (1995) described a 97 kDa OSP which waspre-dom<strong>in</strong>antly present on days 1–3 after ovulation(6.8–10.3% of the total oviductal prote<strong>in</strong>), and onlyattached to oviduct-derived embryos but not to <strong>in</strong>trafollicularoocytes or <strong>in</strong> vitro-derived embryos (Bru¨ ssowet al. 1998b). A contribution of this OSP on embryodevelopment was demonstrated <strong>in</strong> vitro. Supplementationof the 97 kDa prote<strong>in</strong> <strong>in</strong>creased de novo prote<strong>in</strong>synthesis <strong>in</strong> <strong>in</strong> vivo matured embryos (Wollenhaupt et al.1997), reduced polyspermic penetration (Kouba et al.2000), and <strong>in</strong>creased cleavage and blastocyst rate(McCauley et al. 2003).However, not only the ODF <strong>in</strong>fluences the fate ofgametes <strong>in</strong> the oviduct, but also oocytes and spermatozoaalter the oviductal secretory proteomic profile. After<strong>in</strong> vitro <strong>in</strong>cubation of porc<strong>in</strong>e oviducts either with boarspermatozoa or COCs, altogether 34 prote<strong>in</strong>s wereregulated by the presence of spermatozoa (n = 20),oocytes (n = 5) or of both gametes (n = 9). Of theseprote<strong>in</strong>s, 18 were up-regulated and 16 were downregulated(Georgiou et al. 2005). With respect to functionalcategories, gene-regulation related to prote<strong>in</strong>production, ma<strong>in</strong>tenance and repair (41%), antioxidantsand radical scavengers (18%), metabolism (15%) andmiscellaneous (25%). These changes seem to provide afavourable microenvironment for gametes and preparethe oviduct milieu for embryo arrival.Conclud<strong>in</strong>g RemarksAlthough numerous <strong>in</strong>teractions between the Fallopiantube and gametes <strong>in</strong> pigs are recognized, our knowledgeis yet limited. Attempts have to be made to elucidate thef<strong>in</strong>e tun<strong>in</strong>g of <strong>in</strong>tra-follicular oocyte development andovulation with sperm release and capacitation. Therelationship between gametes with<strong>in</strong> the oviduct andoviductal secretion, and subsequent embryo developmentneeds further research. Increased knowledge aboutthe complex and dynamic processes with<strong>in</strong> the Fallopiantube should additionally improve assisted techniques<strong>in</strong>clud<strong>in</strong>g <strong>in</strong> vitro embryo production <strong>in</strong> sw<strong>in</strong>e.AcknowledgementsThe studies of the authors have been made possible by grants from theGerman BMELV, Hungarian OTKA, FORMAS and the SwedishFarmer’s Foundation for Agricultural Research (SLF), Stockholm.ReferencesAlanko M, 1965: The site of recovery and cleavage rate of pigova from the tuba uter<strong>in</strong>a. Nord Vet Med 17, 323–327.Alanko M, 1974: Fertilization and early development of ova <strong>in</strong>AI-gilts, with special reference to role of tubal spermconcentration. A cl<strong>in</strong>ical and experimental study. PhDThesis, University of Hels<strong>in</strong>ki.Andersen D, 1927: The rate of passage of the mammalianovum through various portions of the Fallopian tube. Am JPhysiol 82, 557–569.Aust<strong>in</strong> CR, Walton A, 1960: Fertilisation. In: Parkes AS (ed.),Marshal’s Physiology of <strong>Reproduction</strong>. Longmans Green,London, 1-Part 2, pp. 310–-416.Bedford JM, Kim HH, 1985: Cumulus oophorus as a spermsequester<strong>in</strong>g device, <strong>in</strong> vivo. J Exp Zool 265, 321–328.Bergqvist AS, Ballester J, Johannisson A, Herna´ndez M,Lundeheim N, Rodrı´guez-Martı´nez H, 2006: In vitrocapacitation of bull spermatozoa by oviductal fluid and itscomponents. Zygote 14, 259–273.Blo¨ dow G, Bergfeld J, Kitzig M, Bru¨ ssow K-P, 1990: Steroidhormone levels <strong>in</strong> follicular fluid of pigs with spontaneousoestrus and synchronised ovulation. Arch Exp Vet Med 44,611–620.Brandt Y, Lang A, Madej A, Rodriguez-Mart<strong>in</strong>ez H, E<strong>in</strong>arssonS, 2006: Impact of ACTH adm<strong>in</strong>istration on theoviductal sperm reservoir <strong>in</strong> sows: the local endocr<strong>in</strong>eenvironment and distribution of spermatozoa. AnimReprod Sci 92, 107–122.Brandt Y, Madej A, Rodriguez-Mart<strong>in</strong>ez H, E<strong>in</strong>arsson S,2007: Effects of exogenous ACTH dur<strong>in</strong>g oestrus on earlyembryo development and oviductal transport <strong>in</strong> the sow.Reprod Domest Anim 42, 118–125.Bru¨ ssow K-P, 1985: Distribution of oocytes <strong>in</strong> oviduct of giltsfollow<strong>in</strong>g synchronised ovulation. Mh Vet-Med 40, 264–268.Bru¨ ssow K-P, Rátky J, 1996: Distribution of ova with<strong>in</strong> theoviduct of gilts after ovulation. Reprod Domest Anim 31,305–306.Bru¨ ssow K-P, Kauffold M, Bergfeld J, 1987: Effects ofdifferent PMSG doses on ovarian response as well as ondistribution and quality of oocytes <strong>in</strong> oviduct of giltsfollow<strong>in</strong>g synchronization of ovulation. Mh Vet-Med 42,764–768.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


250 K-P Bru¨ ssow, J Rátky and H Rodriguez-Mart<strong>in</strong>ezBru¨ ssow K-P, Rátky J, Schneider F, Torner H, Kanitz W,Solti L, 1998a: Does follicular fluid have any importance <strong>in</strong>the transfer of porc<strong>in</strong>e oocytes <strong>in</strong>to the oviduct. Theriogenology49, 340.Bru¨ ssow K-P, Alm H, Rátky J, Wollenhaupt K, Torner H1998b: The <strong>in</strong>fluence of gonadotrop<strong>in</strong>s on porc<strong>in</strong>e folliculargrowth and early embryonic development. Proc 49th EAAPMeet<strong>in</strong>g, Warsaw, 1998, MPhP4.4, p. 127.Bru¨ ssow K-P, Rátky J, Schneider F, Torner H, Kanitz W,Solti L, 1999a: Effects of follicular fluid on the transport ofporc<strong>in</strong>e oocytes <strong>in</strong>to the oviduct at ovulation. ReprodDomest Anim 34, 423–429.Bru¨ ssow K-P, Rátky J, Torner H, Sarlo´s P, Solti L, 1999b:Contribution of porc<strong>in</strong>e follicular fluid <strong>in</strong> the process offertilization <strong>in</strong> vivo. Reprod Domest Anim 34, 139–145.Bru¨ ssow K-P, Rátky J, Torner H, Solti L, 2003: Possible roleof follicular fluid on porc<strong>in</strong>e <strong>in</strong> vivo fertilization. In: Sato E,Miyamoto H, Manabe N (eds), Animal Frontiers. HokutoPub Co. Ltd, Kyoto, pp. 89–95.Bru¨ ssow K-P, Torner H, Rátky J, Manabe N, Tuchscherer A,2006: Experimental evidence for the <strong>in</strong>fluence of cumulusoocyte-complexeson sperm release from the porc<strong>in</strong>e oviductalsperm reservoir. J Reprod Dev 52, 249–257.Buhi WC, 2002: Characterization and biological roles ofoviduct-specific, oestrogen-dependent glycoprote<strong>in</strong>. <strong>Reproduction</strong>123, 355–362.Buhi WC, Alvarez IM, 2003: Identification, characterizationand localization of three prote<strong>in</strong>s expressed by the porc<strong>in</strong>eoviduct. Therogenology 60, 225–238.Buhi WC, Alvarez IM, Kouba AJ, 1997: Oviductal regulationof fertilization and early embryonic development. J ReprodFertil 52, 285–300.Eiler H, Nalbandov AV, 1977: Sex steroids <strong>in</strong> the follicularfluid and blood plasma dur<strong>in</strong>g the estrous cycle of pigs.Endocr<strong>in</strong>ology 100, 331–338.Espey LL, Lipner H, 1994: Ovulation. In: Knobil E, Neill JD(eds), The Physiology of <strong>Reproduction</strong>. Raven Press, NewYork, pp. 725–780.Fazeli A, Duncan AE, Watson PF, Holt WV, 1999:Sperm-oviduct <strong>in</strong>teraction: <strong>in</strong>duction of capacitation andpreferential b<strong>in</strong>d<strong>in</strong>g of uncapacitated spermatozoa to oviductalepithelial cells <strong>in</strong> porc<strong>in</strong>e species. Biol Reprod 60,879–86.Funahashi H, Ekwall H, Rodriguez-Mart<strong>in</strong>ez H, 2000: Thezona reaction <strong>in</strong> porc<strong>in</strong>e oocytes fertilized <strong>in</strong> vivo and <strong>in</strong> vitroas seen with scann<strong>in</strong>g electron microscopy. Biol Reprod 63,1437–1442.Funahashi H, Ekwall H, Kikuchi K, Rodriguez-Mart<strong>in</strong>ez H,2001: Transmission electron microscopy studies of the zonareaction <strong>in</strong> pig oocytes fertilised <strong>in</strong> vivo and <strong>in</strong> vitro.<strong>Reproduction</strong> 122, 443–452.Georgiou AS, Sostaric E, Wong CH, Snijders APL, WrightPC, Moore HD, Fazeli A, 2005: Gametes alter theoviductal secretory proteome. Mol Cell Proteomics 4,1785–1796.Hancock JL, 1961: Fertilization <strong>in</strong> the pig. J Reprod Fertil 2,307–331.Hansen C, Srikandakumar A, Downey B, 1991: Presence offollicular fluid <strong>in</strong> the porc<strong>in</strong>e oviduct and its contribution tothe acrosome reaction. Mol Reprod Dev 30, 148–153.Harper MJK, 1973: Stimulation of sperm movement from theisthmus to the site of fertilization <strong>in</strong> the rabbit oviduct. BiolReprod 8, 369–377.Hunter RHF, 1972: Local action of progesterone lead<strong>in</strong>g topolyspermic fertilization <strong>in</strong> pigs. J Reprod Fertil 31, 433–444.Hunter RHF, 1974: Chronological and cytological details offertilization and early embryonic development <strong>in</strong> thedomestic pig, Sus scrofa. Anat Rec 178, 169–185.Hunter RHF, 1984: Preovulatory arrest and periovulatoryredistribution of competent spermatozoa <strong>in</strong> the isthmus ofthe pig oviduct. J Reprod Fertil 72, 203–211.Hunter RHF, 1990: Fertilization of pig eggs <strong>in</strong> vivo and <strong>in</strong>vitro. J Reprod Fertil 40, 211–226.Hunter RHF, 1993: Sperm:egg ratios and putative molecularsignals to modulate gamete <strong>in</strong>teractions <strong>in</strong> polytocousmammals. Mol Reprod Dev 35, 324–327.Hunter RHF, 1996: Ovarian control of very low sperm ⁄ eggratios at the commencement of mammalian fertilization toavoid polyspermy. Mol Reprod Dev 44, 417–422.Hunter RHF, Dziuk PJ, 1968: Sperm penetration of pig eggs<strong>in</strong> relation to the tim<strong>in</strong>g of ovulation and <strong>in</strong>sem<strong>in</strong>ation. JReprod Fertil 15, 199–208.Hunter RHF, Nichol R, 1986: A preovulatory temperaturegradient between the isthmus and ampulla of pig oviductsdur<strong>in</strong>g the phase of sperm storage. Reprod Fertil 77, 599–606.Hunter RHF, Rodriguez-Mart<strong>in</strong>ez H, 2004: Capacitation ofmammalian spermatozoa <strong>in</strong> vivo, with a specific focus onevents <strong>in</strong> the Fallopian tubes. Mol Reprod Dev 67, 243–250.Hunter RHF, Cook B, Poyser NL, 1983: Regulation ofoviduct function <strong>in</strong> pigs by local transfer of ovarian steroidsand prostagland<strong>in</strong>s: a mechanism to <strong>in</strong>fluence sperm transport.Eur J Obstet Gynecol Reprod Biol 14, 225–232.Hunter RHF, Petersen HH, Greve T, 1999: Ovarian follicularfluid, progesterone and Ca 2+ ion <strong>in</strong>fluences on sperm releasefrom the Fallopian tube reservoir. Mol Reprod Dev 54, 283–291.Jansen S, Ekhlasi-Hundrieser M, Toepfer-Petersen E, 2001:Sperm adhesion molecules: Structure and function. CellTissue Organs 168, 82–92.Johansson M, Tienthai P, Rodrı´guez-Martı´nez H, 2000:Histochemistry and ultrastructure of the <strong>in</strong>tralum<strong>in</strong>al mucus<strong>in</strong> the sperm reservoir of the pig oviduct. J Reprod Dev 46,183–192.Kimura N, Konno Y, Miyoshi K, Matsumoto H, Sato E,2002: Expression of hyaluronan synthases and CD44 messengerRNAs <strong>in</strong> porc<strong>in</strong>e cumulus-oocyte complexes dur<strong>in</strong>g<strong>in</strong> vitro maturation. Biol Reprod 66, 707–717.Kouba AJ, Abeydeera LR, Alvarez IM, Day BN, Buhi WC,2000: Effects of the porc<strong>in</strong>e oviduct-specific glycoprote<strong>in</strong> onfertilization, polyspermy, and embryonic development <strong>in</strong>vitro. Biol Reprod 63, 242–250.Leese HJ, 1988: The formation and function of oviduct fluid. JReprod Fertil 82, 843–856.Leese HJ, Tay JI, Reischl J, Down<strong>in</strong>g SJ, 2001: Formation ofFallopian tubal fluid: role of a neglected epithelium.<strong>Reproduction</strong> 121, 339–346.Liberda J, Manaskova P, Prelovska L, Ticha M, Jonakova V,2006: Saccharide-mediated <strong>in</strong>teractions of boar sperm surfaceprote<strong>in</strong>s with components of the porc<strong>in</strong>e oviduct. JReprod Immunol 71, 112–25.Mburu JN, E<strong>in</strong>arsson S, Lundeheim N, Rodriguez-Mart<strong>in</strong>ezH, 1996: Distribution, number and membrane <strong>in</strong>tegrity ofspermatozoa <strong>in</strong> the pig oviduct <strong>in</strong> relation to spontaneousovulation. Anim Reprod Sci 45, 109–121.Mburu JN, Rodriguez-Mart<strong>in</strong>ez H, E<strong>in</strong>arsson S, 1997:Changes <strong>in</strong> sperm ultrastructure and localization <strong>in</strong> theporc<strong>in</strong>e oviduct around ovulation. Anim Reprod Sci 47,137–148.Mburu JN, E<strong>in</strong>arsson S, K<strong>in</strong>dahl H, Madej A, Rodriguez-Mart<strong>in</strong>ez H, 1998: Effects of postovulatory food deprivationon oviductal sperm concentration, embryo development andhormonal profiles <strong>in</strong> the pig. Anim Reprod Sci 52, 221–234.McCauley TC, Buhi WC, Wu GM, Mao J, Caamano JN,Didion BA, Day BN, 2003: Oviduct-specific glycoprote<strong>in</strong>modulates sperm-zona b<strong>in</strong>d<strong>in</strong>g and improves efficiency ofporc<strong>in</strong>e fertilization <strong>in</strong> vitro. Biol Reprod 69, 828–834.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Fertilization <strong>in</strong> the Porc<strong>in</strong>e Fallopian Tube 251Miller DJ, Burk<strong>in</strong> HR, 2001: Gamete adhesion molecules.<strong>Reproduction</strong> 58, 147–158.Mwanza M, Razdan P, Hulte´n F, E<strong>in</strong>arsson S, 2002: Transportof fertilized and unfertilized ova <strong>in</strong> sows. Anim ReprodSci 74, 69–74.Oxenreider SL, Day BN, 1965: Transport and cleavage of ova<strong>in</strong> sw<strong>in</strong>e. J Anim Sci 24, 413–417.Rath D, To¨ pfer-Petersen E, Michelmann H-W, Schwartz P,von Witzendorff D, Ebel<strong>in</strong>g S, Ekhlasi-Hundrieser M,Piehler E, Petrunk<strong>in</strong>a A, Romar R, 2006: Structural,biochemical and functional aspects of sperm–oocyte <strong>in</strong>teractions<strong>in</strong> pigs. In: Ashworth CJ, Krael<strong>in</strong>g RR (eds),Control of Pig <strong>Reproduction</strong> VII, SRF Suppl. 62. Nott<strong>in</strong>ghamUniversity Press, Nott<strong>in</strong>gham, pp. 317–330.Razdan P, Mwanza AM, K<strong>in</strong>dahl H, Rodriguez-Mart<strong>in</strong>ez H,Hultén F, E<strong>in</strong>arsson S, 2002: Effect of repeated ACTHstimulationon early embryonic development and hormonalprofiles <strong>in</strong> sows. Anim Reprod Sci 70, 127–137.Rodriguez-Mart<strong>in</strong>ez H, 2007: Role of the oviduct <strong>in</strong> spermcapacitation. Theriogenology 68S, S138–S146.Rodriguez-Mart<strong>in</strong>ez H, E<strong>in</strong>arsson S, Larsson B, 1982: Spontaneousmotility of the oviduct <strong>in</strong> the anaesthetized pig. JReprod Fertil 66, 615–624.Rodriguez-Mart<strong>in</strong>ez H, Larsson B, E<strong>in</strong>arsson S, 1985: Ovatransport and fertility after resection of the oviductalisthmus <strong>in</strong> pigs. Acta Vet Scand 26, 218–230.Rodriguez-Mart<strong>in</strong>ez H, Nicander L, Vir<strong>in</strong>g S, E<strong>in</strong>arsson S,Larsson K, 1990: Ultrastructure of the uterotubal junction<strong>in</strong> preovulatory pigs. Anat Histol Embryol 19, 16–36.Rodriguez-Mart<strong>in</strong>ez H, Ekstedt E, Ridderstra˚ le Y, 1991:Histochemical localization of carbonic anhydrase <strong>in</strong> thefemale genitalia of pigs dur<strong>in</strong>g the oestrous cycle. Acta Anat140, 41–47.Rodriguez-Mart<strong>in</strong>ez H, Larsson B, Pertoft H, Kjelle´n L, 1998:GAG’s and spermatozoon competence <strong>in</strong> vivo and <strong>in</strong> vitro.In: Lauria A, Gandolfi F, Enne G, Gianaroli L (eds),Gametes: Development and Function. Serono Symposium,Serono, pp. 239–272.Rodriguez-Mart<strong>in</strong>ez H, Tienthai P, Suzuki K, Funahashi H,Ekwall H, Johannisson A 2001: Oviduct <strong>in</strong>volvement <strong>in</strong>sperm capacitation and oocyte development. <strong>Reproduction</strong>(Suppl. 58), 129–145.Rodriguez-Mart<strong>in</strong>ez H, Saravia F, Wallgren M, Tienthai P,Johannisson A, Vázquez JM, Martı´nez E, Roca J, Sanz L,Calvete JJ, 2005: Boar spermatozoa <strong>in</strong> the oviduct. Theriogenology63, 514–535.Spald<strong>in</strong>g JF, Berry RO, Moffit JG, 1955: The maturationprocess of the ovum of sw<strong>in</strong>e dur<strong>in</strong>g normal and <strong>in</strong>ducedovulations. J Anim Sci 14, 609–620.Talbot P, Geiske C, Knoll M, 1999: Oocyte pickup by themammalian oviduct. Mol Biol Cell 10, 5–8.Tanabe TY, Warnick AC, Casida LE, Grummer RH, 1949:The effects of gonadotroph<strong>in</strong>s adm<strong>in</strong>istered to sows and giltsdur<strong>in</strong>g different stages of the estrual cycle. J Anim Sci 8,550–557.Tienthai P, Kjellen L, Pertoft H, Suzuki K, Rodriguez-Mart<strong>in</strong>ez H, 2000a: Localization and quantification ofhyaluronan and sulfated glycosam<strong>in</strong>oglycans <strong>in</strong> the tissueand <strong>in</strong>tralum<strong>in</strong>al fluid of the pig oviduct. Reprod Fertil Dev12, 173–182.Tienthai P, Suzuki K, Pertoft H, Kjellen L, Rodriguez-Mart<strong>in</strong>ez H, 2000b: Production of glycosam<strong>in</strong>oglycans bythe porc<strong>in</strong>e oviduct <strong>in</strong> relation to sperm storage. ReprodDomest Anim 35, 167–170.Tienthai P, Kjellen L, Pertoft H, Suzuki K, Rodriguez-Mart<strong>in</strong>ez H, 2001: Localization and quantitation of hyaluronanand sulfated glycosam<strong>in</strong>oglycans <strong>in</strong> the tissues and<strong>in</strong>tralum<strong>in</strong>al fluid of the pig oviduct. Reprod Fertil Dev 12,173–182.Tienthai P, Kimura N, Held<strong>in</strong> P, Sato E, Rodrı´guez-Martı´nezH, 2003a: Expression of hyaluronan synthase-3 <strong>in</strong> porc<strong>in</strong>eoviductal epithelium dur<strong>in</strong>g oestrus. Reprod Fertil Dev 15,99–105.Tienthai P, Yokoo M, Kimura N, Held<strong>in</strong> P, Sato E,Rodriguez-Mart<strong>in</strong>ez H, 2003b: Immunohistochemical localizationand expression of the hyaluronan receptor CD44 <strong>in</strong>the porc<strong>in</strong>e oviductal epithelium dur<strong>in</strong>g oestrus. <strong>Reproduction</strong>125, 119–132.Tienthai P, Johannisson A, Rodriguez-Mart<strong>in</strong>ez H, 2004:Sperm capacitation <strong>in</strong> the porc<strong>in</strong>e oviduct. Anim Reprod Sci80, 131–146.Wollenhaupt K, Bru¨ ssow K-P, 1995: Isolation of a 97 kdporc<strong>in</strong>e oviductal secretory prote<strong>in</strong> us<strong>in</strong>g a high-performanceelectrophoresis-chromatograph (HPEC) system. ReprodDomest Anim 30, 1–7.Wollenhaupt K, Alm H, Tomek W, Bru¨ ssow K-P, 1997:Studies of the <strong>in</strong>fluence of a specific 97-kd porc<strong>in</strong>e oviductalsecretory prote<strong>in</strong> on the de novo prote<strong>in</strong> synthesis of preimplantationembryos. Reprod Domest Anim 32, 213–219.Yaniz JL, Lopez-Gatius F, Hunter RHF, 2006: Scann<strong>in</strong>gelectron microscopic study of the functional anatomy of theporc<strong>in</strong>e oviductal mucosa. Anat Histol Embryol 35, 28–34.Yokoo M, Miyahayashi Y, Naganuma T, Kimura N, SasadaH, Sato E, 2002: Identification of hyaluronic acid-b<strong>in</strong>d<strong>in</strong>gprote<strong>in</strong>s and their expressions <strong>in</strong> porc<strong>in</strong>e cumulus-oocytecomplexes dur<strong>in</strong>g <strong>in</strong> vitro maturation. Biol Reprod 67, 1165–1171.Yokoo M, Shimizu T, Kimura N, Tunjung WA, MatsumotoH, Abe H, Sasada H, Rodriguez-Mart<strong>in</strong>ez H, Sato E, 2007:Role of the Hyaluronan receptor DC44 dur<strong>in</strong>g porc<strong>in</strong>eoocyte maturation. J Reprod Dev 53, 263–270.Author’s address (for correspondence): K-P Bru¨ ssow, FBN ResearchInstitute for the Biology of Farm <strong>Animals</strong>, Wilhelm-Stahl-Allee 2,18196 Dummerstorf, Germany. E-mail: bruessow@fbn-dummerstorf.deConflict of <strong>in</strong>terest: Grants for K-P Bru¨ ssow were provided byGerman Federal M<strong>in</strong>istry of Nourishment, Agriculture and ConsumersProtection (BMELV) Project 2/05; J Rátky declares thatresearch was partly supported by Hungarian OTKA; H R-M declaresno conflict of <strong>in</strong>terest.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 252–259 (2008); doi: 10.1111/j.1439-0531.2008.01170.xISSN 0936-6768Mastitis <strong>in</strong> Post-Partum Dairy CowsS Pyo¨räläDepartment of Production Animal Medic<strong>in</strong>e, Faculty of Veter<strong>in</strong>ary Medic<strong>in</strong>e, University of Hels<strong>in</strong>ki, Saarentaus, F<strong>in</strong>landContentsTransition from the dry period to lactation is a high riskperiod for the modern dairy cow. The biggest challenge atthat time is mastitis. Environmental bacteria are the mostproblematic pathogens around parturition. Coliforms areable to cause severe <strong>in</strong>fections <strong>in</strong> multiparous cows, andheifers are likely to be <strong>in</strong>fected with coagulase-negativestaphylococci. Dur<strong>in</strong>g the periparturient period, hormonaland other factors make the dairy cows more or lessimmunocompromised. A successful mastitis control programmeis focused on the management of dry and calv<strong>in</strong>gcows and heifers. Clean and comfortable environment, properfeed<strong>in</strong>g and adequate supplementation of the diet withvitam<strong>in</strong>s and trace elements are essential for ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>ggood udder health. Strategies which would enhance closure ofthe teat canal <strong>in</strong> the beg<strong>in</strong>n<strong>in</strong>g of the dry period and wouldprotect teat end from bacteria until the kerat<strong>in</strong> plug hasformed decrease the risk for mastitis after calv<strong>in</strong>g. Dry cowtherapy has been used with considerable success. Yet, aselective approach could be recommended rather than blankettherapy. Non-antibiotic approaches can be useful tools toprevent new <strong>in</strong>fections dur<strong>in</strong>g the dry period, <strong>in</strong> herds wherethe risk for environmental mastitis is high. Vacc<strong>in</strong>ation hasbeen suggested as a means to support the immune defence ofthe dairy cow around parturition. In some countries, implementationof Escherichia coli core antigen vacc<strong>in</strong>e hasreduced the <strong>in</strong>cidence of severe coliform mastitis aftercalv<strong>in</strong>g.IntroductionRate of bov<strong>in</strong>e <strong>in</strong>tramammary <strong>in</strong>fections (IMI) is at thehighest level around parturition (Fig. 1) (Smith et al.1985; Bradley and Green 2004; Valde et al. 2004;McDougall et al. 2007). The factors <strong>in</strong>fluenc<strong>in</strong>g thesusceptibility of the mammary gland to <strong>in</strong>fections arepresence of bacteria at the teat end, level of efficacy ofthe protective characteristics of the teat canal anddefence mechanisms <strong>in</strong> the udder (Sordillo 2005).Dur<strong>in</strong>g the dry period, the mammary gland is consideredto be very resistant aga<strong>in</strong>st <strong>in</strong>fections. The streakcanal is closed and sealed by a kerat<strong>in</strong> plug. In the fully<strong>in</strong>voluted gland, the concentrations of many solublefactors are at a high level and effective <strong>in</strong> prevent<strong>in</strong>g new<strong>in</strong>fections. Concentration of leucocytes is high <strong>in</strong> the drygland, and the environment is more favourable for theirfunction than <strong>in</strong> the lactat<strong>in</strong>g gland (Burvenich et al.2007). The recent decades have shown considerableprogress <strong>in</strong> the understand<strong>in</strong>g of the function of thedefence system of the bov<strong>in</strong>e mammary gland. Despitethis, the <strong>in</strong>efficient host defence and <strong>in</strong>creased susceptibilityto mastitis dur<strong>in</strong>g the transition period cont<strong>in</strong>ue tobe the major problems <strong>in</strong> the dairy cows (Leslie andD<strong>in</strong>gwell 2002). In this article, different factors affect<strong>in</strong>gthe susceptibility of the cow to mastitis aroundparturition, as well as measures for prevention ofmastitis, are reviewed.The Effect of the Dry PeriodDur<strong>in</strong>g <strong>in</strong>volution and aga<strong>in</strong> towards the end of thedry period, the risk for mastitis is at the highest (Oliverand Sordillo 1988). After dry<strong>in</strong>g-off, milk is no longerremoved from the udder, and <strong>in</strong>tramammary pressuremay cause leakage of milk from the teats. Leucocytesstart enter<strong>in</strong>g the gland with<strong>in</strong> 1 week after dry-off, butdo not immediately protect the gland. The kerat<strong>in</strong>plug, which also conta<strong>in</strong>s <strong>in</strong>hibitory substances aga<strong>in</strong>stbacteria, is formed with<strong>in</strong> 1–2 weeks after dry-off andshould naturally seal the teat (D<strong>in</strong>gwell et al. 2004).Quarters that form a kerat<strong>in</strong> plug, which completelycloses the teat soon after dry-off, have significantly lessrisk to develop an IMI. Yet, as many as 23% of thequarters have been found to be still open 6 weeks afterdry<strong>in</strong>g-off (D<strong>in</strong>gwell et al. 2004). In an earlier study,5% of the teats were found to rema<strong>in</strong> completely open(Williamson et al. 1995). Increas<strong>in</strong>g milk yield at thedry-off has been recognized as a significant risk factor:every 5 kg <strong>in</strong>crease <strong>in</strong> milk yield at dry-off above12.5 kg <strong>in</strong>creased the odds of the cow by 77% to havean IMI caused by environmental bacteria at calv<strong>in</strong>g(Rajala-Schultz et al. 2005). The <strong>in</strong>creased susceptibilityto mastitis with <strong>in</strong>creas<strong>in</strong>g milk yield at dry<strong>in</strong>g-off isprobably related to the <strong>in</strong>complete clos<strong>in</strong>g of the teatcanal (D<strong>in</strong>gwell et al. 2004). Teat end condition alsoaffects mastitis susceptibility: teats with cracked endshave higher odds of develop<strong>in</strong>g IMI around calv<strong>in</strong>g(D<strong>in</strong>gwell et al. 2004).Mammary Gland Immunity Around ParturitionFrom 2 weeks prior to calv<strong>in</strong>g, until about 2–3 weeksafter calv<strong>in</strong>g, is the most critical period for the health ofthe mammary gland (Oliver and Sordillo 1988). The<strong>in</strong>nate immune system of the periparturient cows iscompromised. Dur<strong>in</strong>g colostrogenesis, the susceptibilityof the mammary gland to <strong>in</strong>fections <strong>in</strong>creases as the teatcanal starts to open and leaks mammary secretion(Oliver and Sordillo 1988). At the same time, theprotective effect of dry cow therapy (DCT), if used,has disappeared (Oliver et al. 1990). Hormonal changes<strong>in</strong>clude steep rise of the concentration of 17b-oestradiol<strong>in</strong> the plasma dur<strong>in</strong>g the last week of gestation, peak<strong>in</strong>gdur<strong>in</strong>g the last days before parturition and a simultaneousdrop of progesterone <strong>in</strong> circulation. Blood cortisol<strong>in</strong>creases about fivefold at the day of parturition(Burton et al. 2005).Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Mastitis <strong>in</strong> Post-Partum Dairy Cows 253No of cows700600500400300200100Fig. 1. Distribution of the timefrom calv<strong>in</strong>g (days <strong>in</strong> milk) to thediagnosis of cl<strong>in</strong>ical mastitis, basedon data from 28 dairy herds <strong>in</strong>New Zealand (McDougall et al.2007)0–7 7 21 35 49 63 77 91 105 119 133 147 161 175 189 203 217 231 246Time from calv<strong>in</strong>gCalv<strong>in</strong>gCellular FactorsPolymorphonuclear neutrophils (PMN) belong to the<strong>in</strong>nate immune response of the mammary gland to<strong>in</strong>fections (Paape et al. 2003; Burvenich et al. 2007).Around parturition, many functions of the PMN areimpaired. The number of immature neutrophils <strong>in</strong>circulation <strong>in</strong>creases and the number of mature neutrophils<strong>in</strong> the blood and milk are at the lowest. Theproduction of reactive oxygen species (ROS) to killbacteria is reduced from 1 week before parturition overthe first 2 weeks after calv<strong>in</strong>g (Hoeben et al. 2000;Mehrzad et al. 2002). The change <strong>in</strong> the respiratoryburst activity has been found to be parallel withperipartum <strong>in</strong>crease of concentrations of 3b-hydroxybutyricacid (3-BHB), bov<strong>in</strong>e pregnancy-associated glycoprote<strong>in</strong>and bilirub<strong>in</strong> (Hoeben et al. 2000). The rapidrise of the concentration of blood cortisol <strong>in</strong>duceschanges <strong>in</strong> the function of PMN, support<strong>in</strong>g theirextended life span <strong>in</strong> the blood and <strong>in</strong>creas<strong>in</strong>g releasefrom the bone marrow. At that time, neutrophils favourtissue remodell<strong>in</strong>g over defence aga<strong>in</strong>st <strong>in</strong>fections astheir primary task (Burton et al. 2005). At parturition,large numbers of leucocytes are recruited to the reproductivetract and placenta. The ability of the cells tomarg<strong>in</strong>ate on and migrate through endothelium to<strong>in</strong>fected peripheral tissue <strong>in</strong> other sites than uterusdecreases. After parturition, cortisol down-regulates itsown receptors <strong>in</strong> neutrophils and the system returns tonormal function (Burton et al. 2005). The effect ofsteroid hormones on bov<strong>in</strong>e PMN function was studiedby Lamote et al. (2004), who showed that 17b-oestradioltreatment decreased the number of viable cells butprogesterone had no effect. In periparturient cows, a lossof expression of critical neutrophil adhesion moleculeshas been seen (Monfard<strong>in</strong>i et al. 2002), and this loss hasbeen associated with the elevated cortisol levels (Weberet al. 2004). The same was demonstrated by externalglucocorticoid adm<strong>in</strong>istration (Burton et al. 2005). Theproportion of PMN express<strong>in</strong>g, for example, the adhesionreceptor L-select<strong>in</strong>, which is necessary for penetrationto the sites of <strong>in</strong>fection, is dim<strong>in</strong>ished (Diez-Fraileet al. 2004). Neutrophil extracellular traps have recentlyshown to have a role <strong>in</strong> kill<strong>in</strong>g of bacteria and also to befully capable to function <strong>in</strong> the milk environment. Theimpaired efficiency of this system dur<strong>in</strong>g the periparturientperiod may be one more explanation for theimmunosuppression of the dairy cows at that time(Lippolis et al. 2006).Lymphocytes can recognize antigens through specificreceptors and are divided to T and B lymphocytes.CD4+ T lymphocytes cells activate lymphocytes ormacrophages to secrete cytok<strong>in</strong>es, which then canfacilitate either cell-mediated or humoral immuneresponse (Sordillo 2005; Ra<strong>in</strong>ard and Riollet 2006).The proportion of CD4+ cells <strong>in</strong> blood and mammarygland decl<strong>in</strong>es post-partum, and their cytok<strong>in</strong>e productionis different from that <strong>in</strong> mid-lactat<strong>in</strong>g cows. Inperiparturient cows, the percentage of T cells has beenshown to substantially decl<strong>in</strong>e from that <strong>in</strong> mid-lactat<strong>in</strong>gcows (Shafer-Weaver et al. 1996). Macrophages are thedom<strong>in</strong>ant cell type <strong>in</strong> milk of healthy, lactat<strong>in</strong>g gland.Dur<strong>in</strong>g <strong>in</strong>fection, macrophages <strong>in</strong>itiate the immuneresponse by releas<strong>in</strong>g cytok<strong>in</strong>es and other substancesaugment<strong>in</strong>g local <strong>in</strong>flammatory process (Ra<strong>in</strong>ard andRiollet 2006). Bov<strong>in</strong>e macrophage numbers are highest<strong>in</strong> the mammary gland dur<strong>in</strong>g the last week of gestation,but their phagocytic capacity is decreased (Sordillo2005).Humoral FactorsInnate and specific soluble factors represent an importantpart of the defence <strong>in</strong> the mammary gland;complement, lactoferr<strong>in</strong>, lysozyme and antimicrobialpeptides are the most common (Ra<strong>in</strong>ard and Riollet2006). Lactoferr<strong>in</strong> is most active dur<strong>in</strong>g the steady stateof <strong>in</strong>volution (Smith and Schanbacher 1977). Dur<strong>in</strong>gthat time, lactoferr<strong>in</strong> efficiently prevents growth ofbacteria with a high demand of iron such as coliforms(Todhunter et al. 1991). Complement is present <strong>in</strong> highconcentrations <strong>in</strong> colostrum, and seems not to be alimit<strong>in</strong>g factor <strong>in</strong> the defence of the mammary gland atthat time (Ra<strong>in</strong>ard 2003). The most important factors ofthe specific immune response are opsoniz<strong>in</strong>g immunoglobul<strong>in</strong>s(Ig) produced by antigen-activated B lymphocytes.IgG 1 is the primary isotype present <strong>in</strong> the healthymammary gland, but IgG 2 <strong>in</strong>creases dur<strong>in</strong>g <strong>in</strong>flammation.The concentration of IgGs <strong>in</strong> the bov<strong>in</strong>e serum islower around parturition and <strong>in</strong> particular, the lack ofÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


254 S Pyo¨ra¨läthe IgG 2 isotype is associated with the <strong>in</strong>creased<strong>in</strong>dicence of mastitis (Mallard et al. 1998).Dry Cow TherapyUse of DCT has an impact on the <strong>in</strong>cidence of puerperalmastitis <strong>in</strong> two ways: first DCT should elim<strong>in</strong>ate<strong>in</strong>fections present <strong>in</strong> the mammary gland at dry-offand thus prevent their flare-ups at calv<strong>in</strong>g; the secondaim is to protect the mammary gland for new IMIsdur<strong>in</strong>g the dry period (Robert et al. 2006). Antimicrobialsubstances used for DCT do not generally persist <strong>in</strong>the udder until calv<strong>in</strong>g and thus do not offer protectionat that time (Oliver et al. 1990). Yet, <strong>in</strong> an UK study,reduction of cl<strong>in</strong>ical mastitis caused by Gram-negativeagents <strong>in</strong> the subsequent lactation was demonstratedwith a long-act<strong>in</strong>g <strong>in</strong>tramammary preparation withGram-negative spectrum (Bradley and Green 2001).These are the f<strong>in</strong>d<strong>in</strong>gs to date to demonstrate thatselection of dry cow treatment can <strong>in</strong>fluence the<strong>in</strong>cidence of cl<strong>in</strong>ical mastitis <strong>in</strong> the subsequent lactation.Dur<strong>in</strong>g the dry period, 8–12% of previously healthyquarters develop IMI which can be detected at calv<strong>in</strong>g ifno DCT is adm<strong>in</strong>istered (Leslie and D<strong>in</strong>gwell 2003).New IMIs which occur around parturition may greatlyimpact production <strong>in</strong> the subsequent lactation (Oliverand Sordillo 1988; Whist et al. 2006). Blanket DCTcont<strong>in</strong>ues to be the standard procedure <strong>in</strong> most countries(Leslie and D<strong>in</strong>gwell 2003). Rout<strong>in</strong>e treatment ofall cows has recently been questioned, s<strong>in</strong>ce bulk milksomatic cell counts (SCC) have markedly decreased andmastitis has changed from contagious to environmental(Leslie and D<strong>in</strong>gwell 2003). In some countries, especially<strong>in</strong> Scand<strong>in</strong>avia, blanket DCT has never been adaptedbut selective DCT therapy recommended (Osteras et al.1999; Robert et al. 2006). Dry cow therapy does notnecessarily protect cows from mastitis as accord<strong>in</strong>g to aCanadian study, as much as 11% of the treated cowshad new IMI after the dry period (D<strong>in</strong>gwell et al. 2004).Pre-partum <strong>in</strong>tramammary antibiotic therapy for heifershas been suggested to reduce CNS mastitis dur<strong>in</strong>g firstlactation (Oliver et al. 2003; Middleton et al. 2005). In arecent study (Borm et al. 2006), no advantage from thispractice could be shown. Non-antibiotic <strong>in</strong>ternal teatsealants have become widely used, and have proven tobe effective <strong>in</strong> prevention of new <strong>in</strong>fections dur<strong>in</strong>g thedry period (Huxley et al. 2002).Effect of the Metabolic StateThe most important metabolic disturbances occurr<strong>in</strong>gshortly after calv<strong>in</strong>g are milk fever, ketosis and abomasaldisplacement. Hypocalcaemia affects the digestivesystem and pre-disposes the cow to concomitant diseases.It may affect the teat end sph<strong>in</strong>cter and thus<strong>in</strong>crease the risk for mastitis. Cows with periparturienthypocalcaemia are reported to have greater chance ofdevelop<strong>in</strong>g coliform mastitis (Curtis et al. 1983). Negativeenergy balance and perhaps prote<strong>in</strong> imbalances <strong>in</strong>early lactation contribute to the impaired immunedefence (Spa<strong>in</strong> and Scheer 2006). Disturbance <strong>in</strong> fatmetabolism and severe negative energy balance may leadto fatty liver and ketosis. Accumulation of fat <strong>in</strong> theliver disturbs production of humoral immune factorsand is also associated with decreased functional capacityof PMN (Zerbe et al. 2000). There is evidence for adecreased capacity for phagocytosis and kill<strong>in</strong>g ofbacteria <strong>in</strong> cows suffer<strong>in</strong>g from ketosis and fatty liver(Leslie et al. 2001). High concentrations of ketonebodies such as BHB and acetoacetate found at parturitionhave been shown to <strong>in</strong>hibit the proliferation ofhaematopoietic cells (Hoeben et al. 2000). Elevatedlevels of BHB were associated with <strong>in</strong>creased <strong>in</strong>cidenceof cl<strong>in</strong>ical mastitis dur<strong>in</strong>g early lactation (Smith et al.1985; Huszenicza et al. 2004). The course of mastitis wassevere <strong>in</strong> all ketotic cows regardless of the chemotacticresponse before <strong>in</strong>fection (Kremer et al. 1993b).Role of the Caus<strong>in</strong>g AgentBacteriological aetiology of mastitis dur<strong>in</strong>g the puerperalperiod differs between countries, as well asbetween heifers and older cows. The biggest bacterialchallenge for the bov<strong>in</strong>e udder at parturition comesfrom the environment of the cow (Oliver and Sordillo1988). Coliform bacteria appear to be a major problemfor the periparturient cow (Burvenich et al. 2007). In theUK, a high proportion of puerperal mastitis is caused bycoliform bacteria, ma<strong>in</strong>ly Escherichia coli (Bradley andGreen 2004). In a Canadian study, the prevalence ofIMIs at freshen<strong>in</strong>g was 34% and most new IMIs werecaused by environmental streptococci and coliformbacteria (D<strong>in</strong>gwell et al. 2004). Mastitis caused byStreptococcus uberis is most common dur<strong>in</strong>g the dryperiod and <strong>in</strong> early lactation, and <strong>in</strong> some countries thepathogen is most frequently isolated after calv<strong>in</strong>g (Smithet al. 1985; McDougall et al. 2007). On the contrary toenvironmental pathogens, the <strong>in</strong>cidence of mastitiscaused by the contagious pathogen Staphylococcusaureus has been found to <strong>in</strong>crease towards later lactation(Sol et al. 2000; McDougall et al. 2007). In Norwegiansmall herds mostly kept <strong>in</strong> tie stalls, the pathogen mostcommonly isolated at calv<strong>in</strong>g from cows <strong>in</strong> the firstlactation was S. aureus (Waage et al. 1999). In manycountries, the most commonly isolated organism <strong>in</strong>primiparous cows around parturition is CNS (Matthewset al. 1992; Myllys 1995; Taponen et al. 2006, 2007;Parker et al. 2007). CNS mastitis is usually mild(Taponen et al. 2006). Inflammatory reaction <strong>in</strong> theudder <strong>in</strong> mastitis caused by CNS was found to bestronger <strong>in</strong> early lactat<strong>in</strong>g cows as compared with laterlactation (Pyo¨ra¨la¨ and Pyo¨ra¨la¨ 1998).The Special Problem of Puerperal E. coliMastitisMuch research has been carried out on E. coli mastitis,which <strong>in</strong> early lactat<strong>in</strong>g cow is often associated withsevere cl<strong>in</strong>ical signs (Vandeputte-Van Messom et al.1993; Burvenich et al. 2003). Pathophysiology of coliformand staphylococcal mastitis has been shown to bedifferent, and these pathogens elicit a different type ofimmune response (Bannerman et al. 2004). A strongcytok<strong>in</strong>e response and acute or peracute course of thedisease is typical for E. coli mastitis <strong>in</strong> early lactation(Burvenich et al. 2007). Inflammatory reaction of theÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Mastitis <strong>in</strong> Post-Partum Dairy Cows 255mammary gland to endotox<strong>in</strong> was shown to be significantlymore severe <strong>in</strong> cows <strong>in</strong> early lactation than <strong>in</strong> thesame cows close to dry<strong>in</strong>g-off (Lehtola<strong>in</strong>en et al. 2003).The greater severity of coliform mastitis after calv<strong>in</strong>ghas been expla<strong>in</strong>ed by the dysfunction of PMN (Vandeputte-VanMessom et al. 1993). The number of circulat<strong>in</strong>gPMN has been shown to correlate with theseverity of E. coli mastitis (Kremer et al. 1993a). Acutephase mediators produced dur<strong>in</strong>g E. coli mastitis cantrigger an extensive ROS production which damageshost tissues; cells from periparturient dairy cattle havebeen shown to produce significantly more TNF-a thancells from mid-lactat<strong>in</strong>g cows (Sordillo et al. 1995).Multiparous cows have shown to develop more severeE. coli mastitis (Vangroenweghe et al. 2004) as comparedwith young cows. Blood PMN function seems tobe more efficient <strong>in</strong> young cows than <strong>in</strong> older cows(Burvenich et al. 2003). The viability and production ofROS of the milk neutrophils to kill bacteria was foundto be depressed <strong>in</strong> multiparous cows (Mehrzad et al.2002).In the UK, studies have shown that enterobacteria areable to <strong>in</strong>fect the udder dur<strong>in</strong>g the dry period and persistthere until parturition (Bradley and Green 2000). Thishas not been confirmed <strong>in</strong> other countries. For example,<strong>in</strong> a recent US study, the proportion of coliforms asmastitis caus<strong>in</strong>g agents was less than 1% at dry<strong>in</strong>g-offand after calv<strong>in</strong>g (Pantoja and Ruegg 2007). In the studyby Smith et al. (1985), a relatively high proportion ofudder <strong>in</strong>fections at the beg<strong>in</strong>n<strong>in</strong>g and at the end of thedry period was found to be due to Gram-negativebacteria. Dried manure with high counts of coliformbacteria was used as bedd<strong>in</strong>g, which may have affectedthe results.Diagnosis of Mastitis After Calv<strong>in</strong>gUdder health of a dairy cow should be assessed as soonas possible after calv<strong>in</strong>g. Diagnosis of cl<strong>in</strong>ical mastitis <strong>in</strong>early lactation must be adjusted accord<strong>in</strong>g to thephysical properties of milk dur<strong>in</strong>g that period. Somaticcell count is <strong>in</strong>creased at parturition, but decreases tonormal levels with<strong>in</strong> 3–4 days. The difference between<strong>in</strong>fected and healthy quarters is significant at both times(Barkema et al. 1999a). California mastitis test (CMT)was found to have sensitivity and specificity high enoughfor detect<strong>in</strong>g IMI caused by major pathogens at day 3post-partum (Sargeant et al. 2001). Regular monitor<strong>in</strong>gof the udder of the cows around calv<strong>in</strong>g is mostimportant, and milk should be exam<strong>in</strong>ed as soon aspossible after calv<strong>in</strong>g (Green et al. 2007).Effect of Dry Cow Management on theOccurrence of Mastitis Post-PartumDry cow management, such as environment, feed<strong>in</strong>gand tim<strong>in</strong>g of transfer to the calv<strong>in</strong>g unit, significantlyaffect the susceptibility of heifers or dairy cows topuerperal mastitis. Generally, the <strong>in</strong>cidence of cl<strong>in</strong>icalmastitis after calv<strong>in</strong>g <strong>in</strong>creases with parity (Whist et al.2006; Green et al. 2007). Dry and transition dietsshould conta<strong>in</strong> the recommended levels of vitam<strong>in</strong>s andtrace elements (Spa<strong>in</strong> and Scheer 2006). The mostcritical are vitam<strong>in</strong> E and selenium, but also vitam<strong>in</strong> A,copper and z<strong>in</strong>c have a role <strong>in</strong> the defence of the hostaga<strong>in</strong>st <strong>in</strong>fections (Godden et al. 2006). Decades ago,supplementation of dairy cows with selenium andvitam<strong>in</strong> E was shown to have a positive effect onudder health (Smith et al. 1984; Hogan et al. 1993).The effect has mostly been seen <strong>in</strong> herds fed withdeficient or low levels of these elements. In herds fedwith normal diets, daily vitam<strong>in</strong> E supplementationdur<strong>in</strong>g the puerperal period did not affect the <strong>in</strong>cidenceof cl<strong>in</strong>ical mastitis or other puerperal diseases (Perssonet al. 2007).Dry matter <strong>in</strong>take should not be restricted dur<strong>in</strong>g thedry and transition period, but overfeed<strong>in</strong>g of dry cowsshould be avoided. Regular body condition scor<strong>in</strong>g is agood tool <strong>in</strong> monitor<strong>in</strong>g the efficiency of feed<strong>in</strong>g. In arecent study, it was associated with less risk of cl<strong>in</strong>icalmastitis (Green et al. 2007). In a Swedish study, factorswhich <strong>in</strong>creased the risk of elevated SCC after calv<strong>in</strong>gwere <strong>in</strong>tensive concentrate feed<strong>in</strong>g to heifers, andmov<strong>in</strong>g to conf<strong>in</strong>ed hous<strong>in</strong>g on the day of calv<strong>in</strong>g<strong>in</strong>stead of earlier (Svensson et al. 2006). The resultsfrom an Estonian study agreed with the Swedish results,as mov<strong>in</strong>g heifers from separate hous<strong>in</strong>g to the tie stallless than 2 weeks before parturition significantly<strong>in</strong>creased the risk for cl<strong>in</strong>ical mastitis around parturition(Kalmus et al. 2007). An <strong>in</strong>creas<strong>in</strong>g risk of cl<strong>in</strong>icalmastitis was found <strong>in</strong> herds which housed heifers witholder cows (Barkema et al. 1999b). In pasture-grazedconditions, risk factors for peripartum mastitis maydiffer from those <strong>in</strong> housed herds (Compton et al. 2007).Significant risk factors for cl<strong>in</strong>ical and subcl<strong>in</strong>icalmastitis post-calv<strong>in</strong>g were pre-calv<strong>in</strong>g subcl<strong>in</strong>ical mastitis,low teat height above the ground, Friesian breed andudder edema.Provid<strong>in</strong>g a clean, dry environment, good cowcomfort and ventilation are extremely important <strong>in</strong>prevention of mastitis <strong>in</strong> dry and calv<strong>in</strong>g cows andheifers. A recent study from the UK demonstrated theimportance of these factors <strong>in</strong> protect<strong>in</strong>g cows fromcl<strong>in</strong>ical mastitis after calv<strong>in</strong>g (Green et al. 2007). Gooddra<strong>in</strong>age of the dry cow accommodation, use ofmattresses on dry cow cubicle surfaces and dis<strong>in</strong>fectionof cubicle beds were some of the factors found to beprotective aga<strong>in</strong>st mastitis (Fig. 2). Thickness of bedd<strong>in</strong>gof the calv<strong>in</strong>g box was found to be negativelycorrelated with <strong>in</strong>cidence rate of cl<strong>in</strong>ical mastitis(Barkema et al. 1999b). In a Belgian study (DeVliegher et al. 2004) <strong>in</strong> herds with heifers calv<strong>in</strong>g onslatted floors, heifers had lower SCC, probably becausethose calv<strong>in</strong>g places were cleaner than premises withnon-slatted floors. If possible, dry cows and pregnantheifers should not be housed <strong>in</strong> the same barn, nor thetransition cows together with the milk<strong>in</strong>g cows (Barkemaet al. 1999b; Green et al. 2007).Development of vacc<strong>in</strong>es aga<strong>in</strong>st mastitis has beendifficult, because even natural <strong>in</strong>tramammary <strong>in</strong>fectiondoes not provide protection aga<strong>in</strong>st subsequent <strong>in</strong>fections(Talbot and Lacasse 2005). The high number ofmastitis pathogens further complicates the task. Commercialcommon core antigen vacc<strong>in</strong>es aga<strong>in</strong>st coliformmastitis have been available <strong>in</strong> some countries for years.These so-called J5 vacc<strong>in</strong>es are based on the wholeÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


256 S Pyo¨ra¨läFly control used <strong>in</strong> all summermonths for heifersCows of lower paritySCC <strong>in</strong> last 90 days


Mastitis <strong>in</strong> Post-Partum Dairy Cows 257associated with the <strong>in</strong>cidence rate of cl<strong>in</strong>ical mastitis. J DairySci 82, 1643–1654.Borm AA, Fox LK, Leslie KE, Hogan JS, Andrew SM, MoyesKM, Oliver SP, Schukken YH, Hancock DD, Gask<strong>in</strong>s CT,Owens WE, Norman C, 2006: Effects of prepartum <strong>in</strong>tramammaryantibiotic therapy on udder health, milk production,and reproductive performance <strong>in</strong> dairy heifers. J DairySci 89, 2090–2098.Bradley AJ, Green MJ, 2000: A study of the <strong>in</strong>cidence andsignificance of <strong>in</strong>tramammary enterobacterial <strong>in</strong>fectionsacquired dur<strong>in</strong>g the dry period. J Dairy Sci 83, 1957–1965.Bradley AJ, Green MJ, 2001: An <strong>in</strong>vestigation of the impact of<strong>in</strong>tramammary antibiotic dry cow therapy on cl<strong>in</strong>icalcoliform mastitis. J Dairy Sci 84, 1632–1639.Bradley AJ, Green M, 2004: The importance of the nonlactat<strong>in</strong>gperiod <strong>in</strong> the epidemiology of <strong>in</strong>tramammary <strong>in</strong>fectionand strategies for prevention. Vet Cl<strong>in</strong> North Am FoodAnim Pract 20, 547–568.Burton JL, Madsen SA, Chang LC, Weber PS, Buckham KR,van DR, Hickey MC, Earley B, 2005: Gene expressionsignatures <strong>in</strong> neutrophils exposed to glucocorticoids: a newparadigm to help expla<strong>in</strong> ‘‘neutrophil dysfunction’’ <strong>in</strong>parturient dairy cows. Vet Immunol Immunopathol 105,197–219.Burvenich C, Van MV, Mehrzad J, ez-Fraile A, Duchateau L,2003: Severity of E. coli mastitis is ma<strong>in</strong>ly determ<strong>in</strong>ed bycow factors. Vet Res 34, 521–564.Burvenich C, Bannerman DD, Lippolis JD, Peelman L,Nonnecke BJ, Kehrli ME Jr, Paape MJ, 2007: Cumulativephysiological events <strong>in</strong>fluence the <strong>in</strong>flammatory response ofthe bov<strong>in</strong>e udder to Escherichia coli <strong>in</strong>fections dur<strong>in</strong>g thetransition period. J Dairy Sci 90 (Suppl. 1), E39–E54.Compton CW, Heuer C, Parker K, McDougall S, 2007: Riskfactors for peripartum mastitis <strong>in</strong> pasture-grazed dairyheifers. J Dairy Sci 90, 4171–4180.Curtis CR, Erb HN, Sniffen CJ, Smith RD, Powers PA,Smith MC, White ME, Hillman RB, Pearson EJ, 1983:Association of parturient hypocalcemia with eight periparturientdisorders <strong>in</strong> Holste<strong>in</strong> cows. J Am Vet Med Assoc183, 559–561.De Vliegher S, Laevens H, Barkema HW, Dohoo IR, StryhnH, Opsomer G, de Kruif A., 2004: Management practicesand heifer characteristics associated with early lactationsomatic cell count of Belgian dairy heifers. J Dairy Sci 87,937–947.Diez-Fraile A, Meyer E, Duchateau L, Burvenich C, 2004:Effect of pro<strong>in</strong>flammatory mediators and glucocorticoids onL-select<strong>in</strong> expression <strong>in</strong> peripheral blood neutrophils fromdairy cows <strong>in</strong> various stages of lactation. Am J Vet Res 65,1421–1426.D<strong>in</strong>gwell RT, Leslie KE, Schukken YH, Sargeant JM, TimmsLL, Duffield TF, Keefe GP, Kelton DF, Lissemore KD,Conkl<strong>in</strong> J, 2004: Association of cow and quarter-levelfactors at dry<strong>in</strong>g-off with new <strong>in</strong>tramammary <strong>in</strong>fectionsdur<strong>in</strong>g the dry period. Prev Vet Med 63, 75–89.Dosogne H, Vangroenweghe F, Burvenich C, 2002: Potentialmechanism of action of J5 vacc<strong>in</strong>e <strong>in</strong> protection aga<strong>in</strong>stsevere bov<strong>in</strong>e coliform mastitis. Vet Res 33, 1–12.Godden S, Leslie KE, D<strong>in</strong>gwell R, Sanford CJ, 2006: Mastitiscontrol and the dry period: what have we learned. NationalMastitis Council Regional Meet<strong>in</strong>g. Charlottetown, Pr<strong>in</strong>ceEdward Island, Canada. Proceed<strong>in</strong>gs, pp. 56–70.Green MJ, Bradley AJ, Medley GF, Browne WJ, 2007: Cow,farm, and management factors dur<strong>in</strong>g the dry period thatdeterm<strong>in</strong>e the rate of cl<strong>in</strong>ical mastitis after calv<strong>in</strong>g. J DairySci 90, 3764–3776.Hoeben D, Monfard<strong>in</strong>i E, Opsomer G, Burvenich C,Dosogne H, De KA, Beckers JF, 2000: Chemilum<strong>in</strong>escenceof bov<strong>in</strong>e polymorphonuclear leucocytes dur<strong>in</strong>g theperiparturient period and relation with metabolic markersand bov<strong>in</strong>e pregnancy-associated glycoprote<strong>in</strong>. J DairyRes 67, 249–259.Hogan JS, Weiss WP, Smith KL, 1993: Role of vitam<strong>in</strong> E andselenium <strong>in</strong> host defense aga<strong>in</strong>st mastitis. J Dairy Sci 76,2795–2803.Huszenicza G, Janosi S, Gaspardy A, Kulcsar M, 2004:Endocr<strong>in</strong>e aspects <strong>in</strong> pathogenesis of mastitis <strong>in</strong> postpartumdairy cows. Anim Reprod Sci 82–83, 389–400.Huxley JN, Greent MJ, Green LE, Bradley AJ, 2002:Evaluation of the efficacy of an <strong>in</strong>ternal teat sealer dur<strong>in</strong>gthe dry period. J Dairy Sci 85, 551–561.Kalmus P, Viltrop A, Aasma¨e B, Kask K, 2007: Occurrence ofcl<strong>in</strong>ical mastitis <strong>in</strong> primiparous Estonian dairy cows <strong>in</strong>different hous<strong>in</strong>g conditions. Acta Vet Scand 48, 21.Kremer WD, Noordhuizen-Stassen EN, Grommers FJ,Daemen AJ, Henricks PA, Brand A, Burvenich C, 1993a:Pre<strong>in</strong>fection chemotactic response of blood polymorphonuclearleukocytes to predict severity of Escherichia colimastitis. J Dairy Sci 76, 1568–1574.Kremer WD, Noordhuizen-Stassen EN, Grommers FJ, SchukkenYH, Heer<strong>in</strong>ga R, Brand A, Burvenich C, 1993b:Severity of experimental Escherichia coli mastitis <strong>in</strong> ketonemicand nonketonemic dairy cows. J Dairy Sci 76, 3428–3436.Lamote I, Meyer E, Duchateau L, Burvenich C, 2004:Influence of 17beta-estradiol, progesterone, and dexamethasoneon diapedesis and viability of bov<strong>in</strong>e blood polymorphonuclearleukocytes. J Dairy Sci 87, 3340–3349.Lehtola<strong>in</strong>en T, Suom<strong>in</strong>en S, Kutila T, Pyorala S, 2003: Effectof <strong>in</strong>tramammary Escherichia coli endotox<strong>in</strong> <strong>in</strong> early- vs.late-lactat<strong>in</strong>g dairy cows. J Dairy Sci 86, 2327–2333.Leslie KE, D<strong>in</strong>gwell R, 2002: Mastitis control: where are weand where are we go<strong>in</strong>g? In: Kaske M, Scholz H,Ho¨ltersh<strong>in</strong>ken M (eds), Recent developments and perspectives<strong>in</strong> bov<strong>in</strong>e medic<strong>in</strong>e. Hildesheimer Druck und Verlag-GmbH, Hildesheim, pp. 370–382.Leslie KE, D<strong>in</strong>gwell RT, 2003: Background to dry cowtherapy: what, where, why – is it still relevant? NMCAnnual Meet<strong>in</strong>g Proceed<strong>in</strong>gs, Forth Worth, Texas, USA,pp. 5–17.Leslie KE, Duffield TF, Sandals D, Rob<strong>in</strong>son E, 2001: The<strong>in</strong>fluence of negative energy balance on udder health.Proceed<strong>in</strong>gs of the 2nd International Symposium on Mastitisand Milk Quality, Vancouver, Canada, pp. 195–199.Lippolis JD, Re<strong>in</strong>hardt TA, Goff JP, Horst RL, 2006:Neutrophil extracellular trap formation by bov<strong>in</strong>e neutrophilsis not <strong>in</strong>hibited by milk. Vet Immunol Immunopathol113, 248–255.Mallard BA, Dekkers JC, Ireland MJ, Leslie KE, Sharif S,Vankampen CL, Wagter L, Wilkie BN, 1998: Alteration <strong>in</strong>immune responsiveness dur<strong>in</strong>g the peripartum period and itsramification on dairy cow and calf health. J Dairy Sci 81,585–595.Matthews KR, Harmon RJ, Langlois BE, 1992: Prevalence ofStaphylococcus species dur<strong>in</strong>g the periparturient period <strong>in</strong>primiparous and multiparous cows. J Dairy Sci 75, 1835–1839.McDougall S, Arthur DG, Bryan MA, Vermunt JJ, Weir AM,2007: Cl<strong>in</strong>ical and bacteriological response to treatment ofcl<strong>in</strong>ical mastitis with one of three <strong>in</strong>tramammary antibiotics.NZ Vet J 55, 161–170.Mehrzad J, Duchateau L, Pyorala S, Burvenich C, 2002: Bloodand milk neutrophil chemilum<strong>in</strong>escence and viability <strong>in</strong>primiparous and pluriparous dairy cows dur<strong>in</strong>g late pregnancy,around parturition and early lactation. J Dairy Sci85, 3268–3276.Middleton JR, Timms LL, Bader GR, Lakritz J, Luby CD,Steevens BJ, 2005: Effect of prepartum <strong>in</strong>tramammaryÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


258 S Pyo¨ra¨lätreatment with pirlimyc<strong>in</strong> hydrochloride on prevalence ofearly first-lactation mastitis <strong>in</strong> dairy heifers. J Am Vet MedAssoc 227, 1969–1974.Monfard<strong>in</strong>i E, Paape MJ, Wang Y, Capuco AV, Husheem M,Wood L, Burvenich C, 2002: Evaluation of L-select<strong>in</strong>expression and assessment of prote<strong>in</strong> tyros<strong>in</strong>ephosphorylation <strong>in</strong> bov<strong>in</strong>e polymorphonuclear neutrophilleukocytes around parturition. Vet Res 33, 271–281.Myllys V, 1995: Staphylococci <strong>in</strong> heifer mastitis before andafter parturition. J Dairy Res 62, 51–60.Oliver SP, Sordillo LM, 1988: Udder health <strong>in</strong> the periparturientperiod. J Dairy Sci 71, 2584–2606.Oliver SP, Lewis TM, Lewis MJ, Dowlen HH, Maki JL, 1990:Persistence of antibiotics <strong>in</strong> bov<strong>in</strong>e mammary secretionsfollow<strong>in</strong>g <strong>in</strong>tramammary <strong>in</strong>fusion at cessation of milk<strong>in</strong>g.Prev Vet Med 9, 301–311.Oliver SP, Lewis MJ, Gillespie BE, Dowlen HH, Jaenicke EC,Roberts RK, 2003: Prepartum antibiotic treatment ofheifers: milk production, milk quality and economic benefit.J Dairy Sci 86, 1187–1193.Osteras O, Edge VL, Mart<strong>in</strong> SW, 1999: Determ<strong>in</strong>ants of successor failure <strong>in</strong> the elim<strong>in</strong>ation of major mastitis pathogens <strong>in</strong>selective dry cow therapy. J Dairy Sci 82, 1221–1231.Paape MJ, Bannerman DD, Zhao X, Lee JW, 2003: Thebov<strong>in</strong>e neutrophil: structure and function <strong>in</strong> blood and milk.Vet Res 34, 597–627.Pantoja J, Ruegg P, 2007: Somatic cell count and <strong>in</strong>cidence ofnew <strong>in</strong>tramammary <strong>in</strong>fection across lactation. NMC AnnualMeet<strong>in</strong>g, San Antonio, TX, USA, p. 226.Parker KI, Compton C, Anniss FM, Weir A, Heuer C,McDougall S, 2007: Subcl<strong>in</strong>ical and cl<strong>in</strong>ical mastitis <strong>in</strong>heifers follow<strong>in</strong>g the use of a teat sealant precalv<strong>in</strong>g. J DairySci 90, 207–218.Persson WK, Hallen SC, Emanuelson U, Jensen SK, 2007:Supplementation of RRR-alpha-tocopheryl acetate to periparturientdairy cows <strong>in</strong> commercial herds with high mastitis<strong>in</strong>cidence. J Dairy Sci 90, 3640–3646.Pyo¨rälä SH, Pyo¨ra¨la¨ EO, 1998: Efficacy of parenteral adm<strong>in</strong>istrationof three antimicrobial agents <strong>in</strong> treatment ofcl<strong>in</strong>ical mastitis <strong>in</strong> lactat<strong>in</strong>g cows: 487 cases (1989–1995). JAm Vet Med Assoc 212, 407–412.Ra<strong>in</strong>ard P, 2003: The complement <strong>in</strong> milk and defense of thebov<strong>in</strong>e mammary gland aga<strong>in</strong>st <strong>in</strong>fections. Vet Res 34, 647–670.Ra<strong>in</strong>ard P, Riollet C, 2006: Innate immunity of the bov<strong>in</strong>emammary gland. Vet Res 37, 369–400.Rajala-Schultz PJ, Hogan JS, Smith KL, 2005: Short communication:association between milk yield at dry-off andprobability of <strong>in</strong>tramammary <strong>in</strong>fections at calv<strong>in</strong>g. J DairySci 88, 577–579.Robert A, Seegers H, Bareille N, 2006: Incidence of <strong>in</strong>tramammary<strong>in</strong>fections dur<strong>in</strong>g the dry period without or withantibiotic treatment <strong>in</strong> dairy cows – a quantitative analysisof published data. Vet Res 37, 25–48.Sargeant JM, Leslie KE, Shirley JE, Pulkrabek BJ, Lim GH,2001: Sensitivity and specificity of somatic cell count andCalifornia Mastitis Test for identify<strong>in</strong>g <strong>in</strong>tramammary<strong>in</strong>fection <strong>in</strong> early lactation. J Dairy Sci 84, 2018–2024.Shafer-Weaver KA, Pighetti GM, Sordillo LM, 1996: Dim<strong>in</strong>ishedmammary gland lymphocyte functions parallel shifts<strong>in</strong> traffick<strong>in</strong>g patterns dur<strong>in</strong>g the postpartum period. ProcSoc Exp Biol Med 212, 271–280.Smith KL, Schanbacher FL, 1977: Lactoferr<strong>in</strong> as a factor ofresistance to <strong>in</strong>fection of the bov<strong>in</strong>e mammary gland. J AmVet Med Assoc 170, 1224–1227.Smith KL, Harrison JH, Hancock DD, Todhunter DA,Conrad HR, 1984: Effect of vitam<strong>in</strong> E and seleniumsupplementation on <strong>in</strong>cidence of cl<strong>in</strong>ical mastitis andduration of cl<strong>in</strong>ical symptoms. J Dairy Sci 67, 1293–1300.Smith KL, Todhunter DA, Schoenberger PS, 1985: Environmentalpathogens and <strong>in</strong>tramammary <strong>in</strong>fection dur<strong>in</strong>g thedry period. J Dairy Sci 68, 402–417.Sol J, Sampimon OC, Barkema HW, Schukken YH, 2000:Factors associated with cure after therapy of cl<strong>in</strong>ical mastitiscaused by Staphylococcus aureus. J Dairy Sci 83, 278–284.Sordillo LM, 2005: Factors affect<strong>in</strong>g mammary gland immunityand mastitis susceptibility. Livestock Prod Sci 98,89–99.Sordillo LM, Pighetti GM, Davis MR, 1995: Enhancedproduction of bov<strong>in</strong>e tumor necrosis factor-alpha dur<strong>in</strong>gthe periparturient period. Vet Immunol Immunopathol 49,263–270.Spa<strong>in</strong> JN, Scheer WA, 2006: Transition cow nutrition andmastitis – balanc<strong>in</strong>g all the factors. NMC Annual Meet<strong>in</strong>g,Tampa, FL, USA, pp. 172–174.Svensson C, Nyman AK, Persson WK, Emanuelson U, 2006:Effects of hous<strong>in</strong>g, management, and health of dairy heiferson first-lactation udder health <strong>in</strong> southwest Sweden. J DairySci 89, 1990–1999.Talbot BG, Lacasse P, 2005: Progress <strong>in</strong> the development ofmastitis vacc<strong>in</strong>es. Livestock Prod Sci 98, 101–113.Taponen S, Simojoki H, Haveri M, Larsen HD, Pyorala S,2006: Cl<strong>in</strong>ical characteristics and persistence of bov<strong>in</strong>emastitis caused by different species of coagulase-negativestaphylococci identified with API or AFLP. Vet Microbiol115, 199–207.Taponen S, Koort J, Bjo¨rkroth J, Saloniemi H, Pyo¨rälä S,2007: Bov<strong>in</strong>e <strong>in</strong>tramammary <strong>in</strong>fections caused by coagulasenegativestaphylococci may persist throughout lactationaccord<strong>in</strong>g to amplified fragment length polymorphismbasedanalysis. J Dairy Sci 90, 3301–3307.Todhunter DA, Smith KL, Hogan JS, Schoenberger PS, 1991:Gram-negative bacterial <strong>in</strong>fections of the mammary gland <strong>in</strong>cows. Am J Vet Res 52, 184–188.Tomita GM, Ray CH, Nickerson SC, Owens WE, Gallo GF,2000: A comparison of two commercially available Escherichiacoli J5 vacc<strong>in</strong>es aga<strong>in</strong>st E. coli <strong>in</strong>tramammary challenge.J Dairy Sci 83, 2276–2281.Valde JP, Lawson LG, L<strong>in</strong>dberg A, Agger JF, Saloniemi H,Osteras O, 2004: Cumulative risk of bov<strong>in</strong>e mastitis treatments<strong>in</strong> Denmark, F<strong>in</strong>land, Norway and Sweden. Acta VetScand 45, 201–210.Vandeputte-Van Messom G, Burvenich C, Roets E, Massart-Leen AM, Heyneman R, Kremer WD, Brand A, 1993:Classification of newly calved cows <strong>in</strong>to moderate and severeresponders to experimentally <strong>in</strong>duced Escherichia coli mastitis.J Dairy Res 60, 19–29.Vangroenweghe F, Duchateau L, Burvenich C, 2004: Moderate<strong>in</strong>flammatory reaction dur<strong>in</strong>g experimental Escherichiacoli mastitis <strong>in</strong> primiparous cows. J Dairy Sci 87, 886–895.Waage S, Mork T, Roros A, Aasland D, Hunshamar A,Odegaard SA, 1999: Bacteria associated with cl<strong>in</strong>ical mastitis<strong>in</strong> dairy heifers. J Dairy Sci 82, 712–719.Weber PS, Toelboell T, Chang LC, Tirrell JD, Saama PM,Smith GW, Burton JL, 2004: Mechanisms of glucocorticoid-<strong>in</strong>duceddown-regulation of neutrophil L-select<strong>in</strong> <strong>in</strong>cattle: evidence for effects at the gene-expression level andprimarily on blood neutrophils. J Leukoc Biol 75, 815–827.Whist AC, Osteras O, Solverod L, 2006: Cl<strong>in</strong>ical mastitis <strong>in</strong>Norwegian herds after a comb<strong>in</strong>ed selective dry-cow therapyand teat-dipp<strong>in</strong>g trial. J Dairy Sci 89, 4649–4659.Williamson JH, Woolford MW, Day AM, 1995: The prophylacticeffect of a dry-cow antibiotic aga<strong>in</strong>st Streptococcusuberis. NZ Vet J 43, 228–234.Wilson DJ, Grohn YT, Bennett GJ, Gonzalez RN, SchukkenYH, Spatz J, 2007a: Comparison of J5 vacc<strong>in</strong>ates andcontrols for <strong>in</strong>cidence, etiologic agent, cl<strong>in</strong>ical severity, andÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Mastitis <strong>in</strong> Post-Partum Dairy Cows 259survival <strong>in</strong> the herd follow<strong>in</strong>g naturally occurr<strong>in</strong>g cases ofcl<strong>in</strong>ical mastitis. J Dairy Sci 90, 4282–4288.Wilson DJ, Mallard BA, Burton JL, Schukken YH, GrohnYT, 2007b: Milk and serum J5-specific antibodyresponses, milk production change, and cl<strong>in</strong>ical effectsfollow<strong>in</strong>g <strong>in</strong>tramammary Escherichia coli challenge for J5vacc<strong>in</strong>ate and control cows. Cl<strong>in</strong> Vacc<strong>in</strong>e Immunol 14,693–699.Zerbe H, Schneider N, Leibold W, Wens<strong>in</strong>g T, Kruip TA,Schuberth HJ, 2000: Altered functional and immunophenotypicalproperties of neutrophilic granulocytes <strong>in</strong> postpartumcows associated with fatty liver. Theriogenology 54,771–786.Author’s address (for correspondence): S Pyörälä, Department ofProduction Animal Medic<strong>in</strong>e, Faculty of Veter<strong>in</strong>ary Medic<strong>in</strong>e, Universityof Hels<strong>in</strong>ki, Saarentaus, FI-07920, F<strong>in</strong>land. E-mail: satu.pyorala@hels<strong>in</strong>ki.fiConflict of <strong>in</strong>terest: The author declares no conflict of <strong>in</strong>terest.Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 260–267 (2008); doi: 10.1111/j.1439-0531.2008.01171.xISSN 0936-6768Embryonic and Early Foetal Losses <strong>in</strong> Cattle and Other Rum<strong>in</strong>antsMG Disk<strong>in</strong> and DG MorrisTeagasc, Animal Production Research Centre, Mellows Campus, Athenry, Co. Galway, IrelandContentsEmbryo survival is a major factor affect<strong>in</strong>g production andeconomic efficiency <strong>in</strong> all systems of rum<strong>in</strong>ant milk and meatproduction. For heifers, beef and moderate yield<strong>in</strong>g dairycows, does and camelids it appears that fertilization generallylies between 90% and 100%. In high-produc<strong>in</strong>g dairy cowsthere is a less substantive body of literature, but it wouldappear that it is somewhat lower and perhaps more variable.In cattle, the major component of embryo loss occurs beforeday 16 follow<strong>in</strong>g breed<strong>in</strong>g with some evidence of greater lossesbefore day 8 <strong>in</strong> high-produc<strong>in</strong>g dairy cows. In cattle lateembryo loss, while numerically much smaller than earlyembryo mortality loss, nevertheless, causes serious economiclosses to producers because it is often too late to rebreedfemales when they repeat. In multiple ovulat<strong>in</strong>g small rum<strong>in</strong>ants,the loss rate is positively related to ovulation rate.Systemic concentrations of progesterone, dur<strong>in</strong>g both the cyclepreced<strong>in</strong>g and follow<strong>in</strong>g <strong>in</strong>sem<strong>in</strong>ation, affect embryo survivalrate with evidence that too high or <strong>in</strong>deed too low aconcentration be<strong>in</strong>g negatively associated with survival rate.Uter<strong>in</strong>e expression of mRNA for progesterone receptor,oestradiol receptor and ret<strong>in</strong>ol-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> appears to besensitive to changes <strong>in</strong> peripheral concentrations of progesteronedur<strong>in</strong>g the first week after artificial <strong>in</strong>sem<strong>in</strong>ation. Energybalance and dry matter <strong>in</strong>take dur<strong>in</strong>g 4 weeks after calv<strong>in</strong>g arecritically important <strong>in</strong> determ<strong>in</strong><strong>in</strong>g conception rate when cowsare <strong>in</strong>sem<strong>in</strong>ated at 70–100 days post-calv<strong>in</strong>g. Concentratesupplementation of cows at pasture dur<strong>in</strong>g the breed<strong>in</strong>g periodhas m<strong>in</strong>imal effects on conception rates though suddenreductions <strong>in</strong> dietary <strong>in</strong>take should be avoided. For all systemsof milk production, more balanced breed<strong>in</strong>g strategies withgreater emphasis on fertility and feed <strong>in</strong>take and ⁄ or energybalance must be developed. There is sufficient genetic variabilitywith<strong>in</strong> the Holste<strong>in</strong> breed for fertility traits. Alternativedairy breeds such as the Jersey or Norwegian Red could alsobe utilized. Genomic technology will not only provide scientistswith an improved understand<strong>in</strong>g of the underly<strong>in</strong>gbiological processes <strong>in</strong>volved <strong>in</strong> fertilization and the establishmentof pregnancy, but also, <strong>in</strong> the future, identify genesresponsible for improved embryo survival. Its <strong>in</strong>corporation<strong>in</strong>to breed<strong>in</strong>g objectives would <strong>in</strong>crease the rate of geneticprogress for embryo survival.IntroductionEmbryo mortality is a major cause of economic loss <strong>in</strong>all systems of rum<strong>in</strong>ant production. Direct effects ofembryonic mortality are reflected <strong>in</strong> reduced conceptionrates to a service and reduced litter size <strong>in</strong> litter-bear<strong>in</strong>gspecies. In dairy<strong>in</strong>g, the <strong>in</strong>crease <strong>in</strong> milk yield observedover the past 40 years has been accompanied by adecl<strong>in</strong>e <strong>in</strong> cow fertility both <strong>in</strong> high <strong>in</strong>put maize-baseddiets and <strong>in</strong> less <strong>in</strong>tensive, pasture-based systems of milkproduction such as those practised <strong>in</strong> Ireland (see reviewby Disk<strong>in</strong> et al. 2006). In litter-bear<strong>in</strong>g species such asthe ewe, death of one or more embryos dur<strong>in</strong>g theimplantation stage results <strong>in</strong> the birth of smaller lambs(Rh<strong>in</strong>d et al. 1980) with subsequent adverse consequencesfor their post-natal survival and growth rates.The objective of this article is to review current<strong>in</strong>formation on embryonic and early foetal losses <strong>in</strong>cattle and other rum<strong>in</strong>ants and <strong>in</strong> particular on thefactors that affect their <strong>in</strong>cidence and to proposepotential avenues to embryo and foetal survival rates.Fertilization Rate <strong>in</strong> CattleThere are a large number of published estimates offertilization rate <strong>in</strong> heifers and <strong>in</strong> moderate-yield<strong>in</strong>gdairy cows (see review by Sreenan and Disk<strong>in</strong> 1986).Where semen of known high fertility is used <strong>in</strong> artificial<strong>in</strong>sem<strong>in</strong>ation (AI), fertilization rates are of the order of90–100%. In contrast, for higher produc<strong>in</strong>g dairy cowsthere is little quantitative <strong>in</strong>formation on fertilizationrate with only three published reports (Wiebold 1988;Ryan et al. 1993; Sartori et al. 2002). Wiebold (1988),us<strong>in</strong>g a non-surgical embryo recovery technique on day7 follow<strong>in</strong>g oestrus, recovered 25 ova ⁄ embryos from 23lactat<strong>in</strong>g cows with all recovered ova hav<strong>in</strong>g beenfertilized. Ryan et al. (1993) <strong>in</strong> a study on the effectsof ambient temperature on fertilization rate reported noeffect of temperature and quoted fertilization rates of82.4% and 79.5% for high and low temperatures,respectively. Sartori et al. (2002) recorded a low fertilizationrate of 55.6% <strong>in</strong> lactat<strong>in</strong>g dairy cows comparedto 100% for heifers under high ambient temperatures,while <strong>in</strong> a subsequent study dur<strong>in</strong>g the cool seasonfertilization rates were 87.8% and 89.5% for lactat<strong>in</strong>gand non-lactat<strong>in</strong>g dairy cows, respectively. It appearsthat fertilization rate is similar <strong>in</strong> high- and moderateproduc<strong>in</strong>gdairy cows, at least dur<strong>in</strong>g the cool season.Fertilization Rates <strong>in</strong> Small Rum<strong>in</strong>antsThe available evidence is that, like cattle, a fertilizationrate of 90–95% appears to be normal <strong>in</strong> ewes (Restallet al. 1976; Mitchell et al. 1999), does (see review byNancarrow 1994) and South American camelids (Fernandez-Bacaet al. 1970) under natural mat<strong>in</strong>g conditions.There is some suggestion that fertilization is anall-or-none phenomenon <strong>in</strong> multiple ovulat<strong>in</strong>g ewes(Restall et al. 1976), though there is some evidence(Restall et al. 1976; Kleeman et al. 1990) that partialfertilization failure may occur at a low frequency. Thereis some evidence that fertilization rates may be reduced<strong>in</strong> ewes dur<strong>in</strong>g both the early (Hulet et al. 1956) andlatter parts of the breed<strong>in</strong>g season, perhaps as aconsequence of suboptimal mat<strong>in</strong>g activity <strong>in</strong> somerams (Colas 1983) or adverse environmental and nutritionalconditions (Mitchell et al. 1996). Yet, when ewesÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Embryo ⁄ Foetal Losses <strong>in</strong> Rum<strong>in</strong>ants 261are neither environmentally nor nutritionally challengedand mated to fertile rams differences <strong>in</strong> fertilizationfailure rate are likely to be small or non-existent(Mitchell et al. 1999).Extent and Tim<strong>in</strong>g of Early Embryo Loss <strong>in</strong>CattleWhile fertilization rate is apparently similar <strong>in</strong> high- andmoderate-produc<strong>in</strong>g cows and unlikely to be affected bywhether cows are on pasture or high <strong>in</strong>put total mixedration (TMR) diets, nevertheless the average calv<strong>in</strong>g rateto a s<strong>in</strong>gle service is significantly lower <strong>in</strong> high- thaneither <strong>in</strong> low-produc<strong>in</strong>g cows or <strong>in</strong> heifers. Sreenan andDisk<strong>in</strong> (1986) calculated an embryonic and foetalmortality rate (exclud<strong>in</strong>g fertilization failure) of about40% for moderate-produc<strong>in</strong>g cows based on a fertilizationrate of 90% and an average calv<strong>in</strong>g rate of about55% with an estimated 70–80% of it susta<strong>in</strong>ed betweendays 8 and 16 after <strong>in</strong>sem<strong>in</strong>ation. The comparativefigure for high-produc<strong>in</strong>g dairy cows based on afertilization rate of 90% and a calv<strong>in</strong>g rate of 40% is56%.There is some evidence that the pattern of earlyembryo death <strong>in</strong> the modern high-produc<strong>in</strong>g cow maybe different to that observed <strong>in</strong> heifers and loweryield<strong>in</strong>g dairy cows. For example, <strong>in</strong> the study of Sartoriet al. (2002) conducted dur<strong>in</strong>g the summer <strong>in</strong> Wiscons<strong>in</strong>,a significantly higher proportion of the embryos recoveredon days 6–7 were of higher quality grade fromheifers (72%) compared with lactat<strong>in</strong>g cows (33%).Similarly, dur<strong>in</strong>g the w<strong>in</strong>ter component of the samestudy, Sartori et al. (2002) also recorded a higherproportion of higher quality grade embryos from dry(83%) compared to lactat<strong>in</strong>g (53%) cows. This trend ofa comparatively higher proportion of low-quality gradeor abnormal or retarded embryos from high-yield<strong>in</strong>gdairy cows is also evident <strong>in</strong> the study of Wiebold(1988), who found that 52% of the embryos from highyield<strong>in</strong>gdairy cows on days 6–7 were abnormal, basedon morphological criteria. Ryan et al. (1993) reportedthat the proportions of high-quality grade embryosrecovered under the conditions of high (58.5%) and low(51.6%) ambient temperature were similar. A feature ofthese recent studies is the consistent evidence that a high(41–67%) proportion of embryos recovered on day 7post-oestrus from moderate- or high-produc<strong>in</strong>g dairycows were classified as abnormal <strong>in</strong> contrast to a muchlower <strong>in</strong>cidence (17–28%) <strong>in</strong> heifers and non-lactat<strong>in</strong>gcows <strong>in</strong> the same studies or the 6% (average of fourstudies) <strong>in</strong> the report of Sreenan and Disk<strong>in</strong> (1986). Itwould appear that early embryo loss is greater <strong>in</strong> themodern high-produc<strong>in</strong>g dairy cow and that a muchhigher proportion of the embryos die before day 7follow<strong>in</strong>g <strong>in</strong>sem<strong>in</strong>ation. Based on the published evidence,the expected outcome of 100 <strong>in</strong>sem<strong>in</strong>ations ofBritish Friesian and Holste<strong>in</strong>–Friesian cows is summarized<strong>in</strong> Fig. 1.Late Embryo and Foetal Loss <strong>in</strong> CattleOver the past 10 years, there has been significant<strong>in</strong>terest <strong>in</strong> the problem of late embryo and early foetalBritish Friesian 1980 Holste<strong>in</strong> Friesian 2006Late embryo mortality7% Early7%embryodeath 28%EarlyCalv<strong>in</strong>gCalv<strong>in</strong>gembryo40%55%10%death 43%Fertilisation failure10%Fig. 1. Reproductive outcomes <strong>in</strong> British–Friesian versus Holste<strong>in</strong>–Friesian cows (Source: Disk<strong>in</strong> et al. 2006)mortality, which has generally been def<strong>in</strong>ed as the deathof the embryo after about day 24 of gestation. With theadvent of ultrasound scann<strong>in</strong>g, it has been comparativelyeasy to accurately establish the extent and tim<strong>in</strong>gof late embryo ⁄ foetal mortality. Silke et al. (2001)quantified the extent and pattern of embryo ⁄ foetal lossfrom days 28 to 84 of gestation <strong>in</strong> 1046 lactat<strong>in</strong>g dairycows and 162 dairy heifers managed on pasture-basedsystems of milk production. The overall loss ratesbetween days 28 and 84 of gestation and the pattern ofloss over this period are similar for cows (7.2%)produc<strong>in</strong>g on average 7247 kg of milk and heifers(6.1%). Almost half (47.5%) of the total recorded lossoccurred between days 28 and 42 of gestation. Thereare no significant association between level of milkproduction or milk energy output measured on day 120of lactation, milk fat concentration, milk prote<strong>in</strong>concentration or milk lactose concentration andembryo ⁄ foetal loss rate. The extent and pattern ofembryo ⁄ foetal was not related to either cow or cow siregenetic merit. A more recent study by Horan et al.(2004) recorded a similar overall late embryo ⁄ foetal lossrate of 7.5% between days 30 and 67 of gestation <strong>in</strong>dairy cows managed under pasture-based systems ofproduction. The extent of late embryo ⁄ foetal mortalityrecorded <strong>in</strong> these two pasture-based studies is muchlower than that reported for some US-based studies(Silke et al. 2001), though the causes for the apparentdifference are not clear. While the extent of late embryoloss is numerically much smaller than early embryoloss, it nevertheless causes serious economic losses toproducers because it is too late to rebreed cows whenthe loss occurs result<strong>in</strong>g <strong>in</strong> <strong>in</strong>creased cull<strong>in</strong>g particularly<strong>in</strong> seasonal calv<strong>in</strong>g herds.Extent and Tim<strong>in</strong>g of Embryo Loss <strong>in</strong> SmallRum<strong>in</strong>antsUnlike with cattle, there are a much smaller number ofreports of the serial slaughter of other rum<strong>in</strong>ants atspecific time po<strong>in</strong>ts follow<strong>in</strong>g mat<strong>in</strong>g to determ<strong>in</strong>e thepattern of embryo loss. Most studies have measuredembryo survival at about the time of implantation andor at term. Furthermore, most studies have <strong>in</strong>cludedfertilization failure <strong>in</strong> their estimate of embryo loss.This is not unreasonable given that fertilization rate isusually close to 100%. In ewes and does, it is clearthat embryo survival rate is a function of ovulationÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


262 MG Disk<strong>in</strong> and DG MorrisTable 1. Probability of embryo survival as a function of ovulationrate a <strong>in</strong> naturally mated ewes and does (Source: Hanrahan 1994)No. ofcorpora luteaEwesProbability of embryo survival bDoes2 0.82 (5069) c 0.82 (306)3 0.74 (884) 0.60 d (58)4 0.65 (270) –5 0.55 (91) –6 0.45 (38) –a Based on summary of published and unpublished <strong>in</strong>formation on ovulation rateand the number of foetuses ⁄ off-spr<strong>in</strong>gs.b About ‡250 and 0.06 to 0.08 otherwise.c No. of females <strong>in</strong>volved (n).d Includes observations on some does with >3 corpora lutea.rate (Hanrahan 1994) and data are summarized <strong>in</strong>Table 1. While the <strong>in</strong>formation on goats is limitedrelative to sheep, both sets of data show that theprobability of embryo survival decl<strong>in</strong>es as ovulationrate <strong>in</strong>creases. From a comparison of the publishedliterature on embryo survival <strong>in</strong> mated ewes or <strong>in</strong> ewesfollow<strong>in</strong>g embryo transfer, Hanrahan (1994) concludedthat the change <strong>in</strong> pattern of embryo survival with<strong>in</strong>creas<strong>in</strong>g number of ova shed or embryos transferredwas similar, and the clear negative relationshipbetween the number of ovulations and subsequentembryo survival is a direct consequence of the numberof embryos enter<strong>in</strong>g the uterus rather than anyproblems with embryo quality, the time of ovulationor uter<strong>in</strong>e environment. He also concluded that therewas no evidence of an <strong>in</strong>herent physiological or geneticrelationship between ovulation rate and embryo survival.A direct consequence of these conclusions is that<strong>in</strong>formation on embryo survival <strong>in</strong> sheep and goatsshould be exam<strong>in</strong>ed <strong>in</strong> relation to ovulation rate andthe <strong>in</strong>terpretation of differences due to management orenvironmental factors should allow for this directeffect of ovulation rate. Extrapolation of the dataconta<strong>in</strong>ed <strong>in</strong> Table 1 provides estimates ofembryo ⁄ foetal loss of 12%, 18% and 26% for eweswith one, two or three ovulation, respectively. Incomparison with cattle, which are predom<strong>in</strong>atelymono-ovular, the embryo loss rate of 12% formono-ovular sheep is much lower than comparablefigures of about 40% and 60% for heifers and highproduc<strong>in</strong>gdairy cows, respectively.In camelids the proportion pregnant at about60 days post-mat<strong>in</strong>g is about 65% of those thatovulate (Adams et al. 1991), although much lowerfigures have been recorded. Given that fertilizationrates are similar <strong>in</strong> all species, it seems that embryo lossrates are much higher <strong>in</strong> camelids than <strong>in</strong> sheep orgoats. While tw<strong>in</strong> ovulations occur quite frequently <strong>in</strong>camelids, tw<strong>in</strong> births are rare <strong>in</strong>dicat<strong>in</strong>g that themechanisms that control embryo survival are probablydifferent <strong>in</strong> camelids to those <strong>in</strong> cattle, sheep and goats.One of the factors which may contribute to the higherembryo loss rate is the fact that <strong>in</strong> all camelids, over98% of the foetuses implant <strong>in</strong> the left uter<strong>in</strong>e horndespite the fact both ovaries contribute almost equallyto ovulation. It appears that the right uter<strong>in</strong>e horn isunable to susta<strong>in</strong> a pregnancy after about 50 days ofgestation (Arthur et al. 1982).Causes of Embryo or Foetal LossSeveral factors have been implicated <strong>in</strong> embryo andfoetal loss, usually with a lack of support<strong>in</strong>g experimentaldata. The ma<strong>in</strong> factors implicated are normallycategorized as those of genetic, physiological, endocr<strong>in</strong>eand environmental orig<strong>in</strong> (see reviews by Ashworth1994; Bazer 1994; Nancarrow 1994; Zavy 1994). Onlysome possible genetic and environmental factors areconsidered here.Genetic causesGenetic causes of embryo death <strong>in</strong>clude chromosomaldefects, <strong>in</strong>dividual genes and genetic <strong>in</strong>teractions (Van-Raden and Miller 2006). The 1 ⁄ 29 Robertsonian chromosomaltranslocation (Gustavsson 1979) present <strong>in</strong>several beef breeds and <strong>in</strong> the Scand<strong>in</strong>avian Red breeds,but not <strong>in</strong> the Holste<strong>in</strong> breed and has been suggested asa cause of reduced fertility of males and femalesheterozygous for 1 ⁄ 29 translocation. Sweden has successfullyimplemented a test<strong>in</strong>g programme to elim<strong>in</strong>atefrom AI bulls that are carriers for this conditionresult<strong>in</strong>g <strong>in</strong> a significant improvement <strong>in</strong> fertility (Gustavsson1979).In the Holste<strong>in</strong> breed, two major recessive defectsaffect<strong>in</strong>g embryo ⁄ foetal survival have been detected.Deficiency of urid<strong>in</strong>e monophosphate synthase(DUMPS) (Rob<strong>in</strong>son et al. 1984), a homozygous recessivecondition, causes foetal death a 40–50 days ofgestation (Shanks and Rob<strong>in</strong>son 1989). Test<strong>in</strong>g of AIsires for DUMPS has significantly reduced the frequencyof heterozygous sires and of homozygousrecessive embryos and has now almost elim<strong>in</strong>ated thisas a cause of fertility losses (VanRaden and Miller2006). Complex vertebral malformation is a lethalrecessive condition that causes late foetal death <strong>in</strong> cattle.This defect was dissem<strong>in</strong>ated through the widespreaduse of the Holste<strong>in</strong> Carl<strong>in</strong>-M Ivanhoe Bell as a maternalsire of sires. In a Swedish study, Persson (2003) recordedthat carrier bulls had significantly lower breed<strong>in</strong>g valuesfor 168-day non-return rate than non-carriers at168 days follow<strong>in</strong>g <strong>in</strong>sem<strong>in</strong>ation but were not differentat 28 and 56 days <strong>in</strong>dicat<strong>in</strong>g that it primarily affects laterather than early foetal mortality. Aga<strong>in</strong> it is possible togreatly reduce the <strong>in</strong>cidence if carriers for this conditionby test<strong>in</strong>g sires.Several reproductive traits are adversely affected by<strong>in</strong>breed<strong>in</strong>g. Maternal <strong>in</strong>breed<strong>in</strong>g has been reported todecrease the 56–70-day non-return rates by between 1%(Wall et al. 2003) and 2% (Cassell et al. 2003) per 10%<strong>in</strong>breed<strong>in</strong>g of the dam. On the other hand, <strong>in</strong>breed<strong>in</strong>g ofthe embryo has bee reported to reduce the 70-day nonreturnrate by 1% for each 10% <strong>in</strong>crease <strong>in</strong> the level of<strong>in</strong>breed<strong>in</strong>g of the embryo (Cassell et al. 2003; VanRadenand Miller 2006). Genetic variation <strong>in</strong> embryo survivalmay be attributable to genetic constitution of theembryo itself and or the genetic differences among damswith respect to their ability to provide an appropriate<strong>in</strong>traovarian and uter<strong>in</strong>e environment. In sheep, Bod<strong>in</strong>Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Embryo ⁄ Foetal Losses <strong>in</strong> Rum<strong>in</strong>ants 263et al. (1992) concluded, based on results from embryotransfer and breed-cross<strong>in</strong>g studies, that genetic differencesamong embryos were not of any consequence forvariation <strong>in</strong> embryo survival rate, aside from factorssuch as chromosomal abnormalities, specific lethal genesand genetic <strong>in</strong>teraction. Breed differences <strong>in</strong> embryosurvival as a trait of the ewe have been exam<strong>in</strong>ed <strong>in</strong>many studies based on employ<strong>in</strong>g embryo transferprocedures and on estimation of embryo survival us<strong>in</strong>g<strong>in</strong>formation on ovulation rate and the consequent littersize. The general conclusion from the embryo transferstudies is that breed of recipient ewe did not play animportant role <strong>in</strong> determ<strong>in</strong><strong>in</strong>g embryo survival despitethe fact that these studies often <strong>in</strong>volved breeds withvery different levels of natural prolificacy (Hanrahan1994), though there is some evidence (Hanrahan andPiper 1992) that embryo survival may be better when theF<strong>in</strong>n breed was used as an embryo recipient. There isquite a substantial literature on estimation of embryosurvival based on the <strong>in</strong>formation on ovulation andassociated litter size and provides a good <strong>in</strong>dication ofbreed variation <strong>in</strong> embryo survival. Hanrahan (1994),follow<strong>in</strong>g an extensive review of the literature, suggestedthat the likely breed differences for the probability ofembryo survival are 0.15, 0.13 and 0.11 for tw<strong>in</strong>, tripletand quadruplet ovulations, respectively. If the essentiallyl<strong>in</strong>ear relationship between ovulation rate andlitter size is the same <strong>in</strong> all breeds, then a global figure of0.12 has been suggested by Hanrahan (1994) as thelikely range of breed means for embryo survivaladjusted for ovulation rate. Variation among <strong>in</strong>dividualswith<strong>in</strong> a population, measured as repeatability ofdifferences <strong>in</strong> embryo survival, is also quite low (»0.05)(Hanrahan 1982; Ricordeau et al. 1986), and consequentlythe heritability of these differences is alsoextremely low (Ricordeau et al. 1986).Maternal ageWhile the overall embryo survival rates would appearto be largely similar <strong>in</strong> heifers, beef cows and low–moderate-produc<strong>in</strong>g dairy cows, embryo survival rate islower <strong>in</strong> high-produc<strong>in</strong>g dairy cows. Yet, it is improbablethat this is an age- or parity-related phenomenonand is more probable due to the direct and <strong>in</strong>directconsequences of milk production (see later). Recentdata from the US (Kuhn et al. 2006) would suggestboth that heifer conception rate is at a maximum at 15–16 month of age. Breed<strong>in</strong>g heifers at 26 months of ageor older resulted <strong>in</strong> a 13% lower conception ratepresumably due to a lower embryo survival rate. Thereis evidence, at least <strong>in</strong> some breeds of sheep (Galway:Quirke and Hanrahan 1977; Romney: McMillan andMcDonald 1985), that embryo survival is lower <strong>in</strong> ewelambs than <strong>in</strong> adult ewes. These later studies and the asubsequent study of Quirke and Hanrahan (1983) allemploy<strong>in</strong>g embryo transfer <strong>in</strong>dicate that the impairedembryo survival associated with ewe lambs is attributableto the <strong>in</strong>herent quality of embryo rather than anydeficiency of uter<strong>in</strong>e environment. Ricordeau et al.(1982, 1986) found no significant differences <strong>in</strong> embryosurvival between ewe lambs and adults of the Romanovbreed. This may be dependent on the degree of sexualmaturity atta<strong>in</strong>ed at breed<strong>in</strong>g as there is evidence thatconception rate <strong>in</strong>creases between first and later oestrusperiods <strong>in</strong> ewe lambs. Among age groups other thanewe lambs, there is little evidence of differences <strong>in</strong>embryo survival rate (Hanrahan 1990; Kleeman et al.1990).Progesterone dur<strong>in</strong>g the cycle immediately prior to<strong>in</strong>sem<strong>in</strong>ation and embryo survival rateThere is now good evidence l<strong>in</strong>k<strong>in</strong>g circulat<strong>in</strong>g concentrationof progesterone dur<strong>in</strong>g the cycle immediatelyprior to <strong>in</strong>sem<strong>in</strong>ation as well as dur<strong>in</strong>g the early lutealphase of the cycle follow<strong>in</strong>g <strong>in</strong>sem<strong>in</strong>ation with lowembryo survival ⁄ conception rate (see review by Disk<strong>in</strong>et al. 2006). Recent data from this laboratory (Disk<strong>in</strong>et al. 2006) clearly show that there is a positive l<strong>in</strong>earassociation between the concentrations of progesteroneon the day of PGF-2a-<strong>in</strong>duced luteolysis and subsequentembryo survival rate. Potential mechanisms by whichlow concentrations of progesterone dur<strong>in</strong>g the preced<strong>in</strong>goestrous cycle might reduce fertilization and or embryosurvival rates <strong>in</strong>clude the production of oocytes that areat a more advanced stage of maturation at time ofovulation from persistent dom<strong>in</strong>ant follicles; <strong>in</strong>creasedfrequency of pulses of LH which <strong>in</strong> turn <strong>in</strong>duces<strong>in</strong>creased secretion of oestradiol-17b or an alteration<strong>in</strong> endometrial morphology (see review by Disk<strong>in</strong> et al.2006). The more probable effect of low concentrationsof progesterone <strong>in</strong> the cycle preced<strong>in</strong>g oestrus onsubsequent embryo survival rate is to result <strong>in</strong>pre-mature oocyte maturation, which subsequentlycompromises its ability to cont<strong>in</strong>ue normal embryodevelopment after its fertilization.Post-<strong>in</strong>sem<strong>in</strong>ation progesterone and embryo survival rateRecent studies (Stronge et al. 2005; Disk<strong>in</strong> et al. 2006)that exam<strong>in</strong>ed the relationship between early and midlutealphase concentrations of progesterone and subsequentembryo survival ⁄ conception rate have employedlogistic regression techniques to model the relationshipbetween the b<strong>in</strong>omially distributed dependent variable(conception ⁄ embryo survival rate) and the cont<strong>in</strong>uouslydistributed <strong>in</strong>dependent variable (progesterone). In thestudy of Stronge et al. (2005) (Fig. 2), there was apositive l<strong>in</strong>ear and quadratic relationship between milkconcentrations of progesterone on days 5, 6, 7 post<strong>in</strong>sem<strong>in</strong>ationand between the rate of change <strong>in</strong> concentrationsof progesterone between days 4 and 7 andembryo survival rate. Further analysis of this data setreveals that 75%, 72% and 56% of dairy cows haveconcentrations of progesterone that are optimal forconception on days 5, 6 and 7 post-<strong>in</strong>sem<strong>in</strong>ation,respectively. In beef heifers, Disk<strong>in</strong> et al. (2006) haveshown that a similar l<strong>in</strong>ear and quadratic associationbetween peripheral concentrations of progesterone andembryo survival also exists. These recent data for cattleare <strong>in</strong>l<strong>in</strong>e with results from earlier sheep studies of Parret al. (1987) of an <strong>in</strong>verse relationship between circulat<strong>in</strong>gconcentrations of progesterone and embryo survival<strong>in</strong> these species. Furthermore, there is evidence for sheep(Parr et al. 1987) and dairy cows (Starbuck et al. 2001)Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


264 MG Disk<strong>in</strong> and DG Morris10090807060504030201001009080706050403020100Days 5 0.7 70 Days 60.6 600.5 500.4 400.3 300.2 200.1 100 00–12–34–56–78–910–1112–1314–151009080


Embryo ⁄ Foetal Losses <strong>in</strong> Rum<strong>in</strong>ants 265Energy balance at around the time of <strong>in</strong>sem<strong>in</strong>ation andsubsequent conception rateDry matter <strong>in</strong>take is lower for cows graz<strong>in</strong>g pastures thanfor cows fed maize-based TMRs. High-genetic merithigh-produc<strong>in</strong>g cows experience greater NEB <strong>in</strong> earlylactation under graz<strong>in</strong>g conditions relative to lowergenetic merit cows, notwithstand<strong>in</strong>g the somewhat higherDMI capacity of the former cow type (Horan et al.2004). Meta-analysis of the comb<strong>in</strong>ed Kennedy et al.(2003) and Horan et al. (2004) data showed that therewas no service · study · supplementation rate (p >0.10), service · study rate (p > 0.10), study · supplementationrate (p > 0.10) <strong>in</strong>teraction effects on conceptionrate (see Disk<strong>in</strong> et al. 2006). Yet, there was aservice · supplementation rate effect on conception rate(p < 0.05). The rate of supplementation had no effect onfirst service conception rate (56% vs 53%) but cows onthe low level of supplementation had a lower (p < 0.05)second service conception rate (39% vs 58%) comparedwith cows on the high level of supplementation. Interest<strong>in</strong>gly,the withdrawal of the supplementation commencedat about the onset of the breed<strong>in</strong>g period <strong>in</strong> bothstudies. From these studies, there is no clear evidence thatconcentrate supplementation of dairy cows at pastureimproves first service conception rate, but it may bebeneficial <strong>in</strong> ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g second service conception rates.This clearly highlights the difficulty that improv<strong>in</strong>g theEB of the modern dairy cows presents at this stage oflactation where grazed grass is the predom<strong>in</strong>ant componentof the diet. Based on the studies of Sangsritavonget al. (2002), the <strong>in</strong>creased milk production as a result ofthe concentrate supplementation may well be associatedwith <strong>in</strong>creased hepatic blood flow result<strong>in</strong>g <strong>in</strong> <strong>in</strong>creasedmetabolism of progesterone and consequently lower<strong>in</strong>gof peripheral concentrations of progesterone andpre-dispos<strong>in</strong>g to greater risk of embryo death.Sudden reductions <strong>in</strong> feed <strong>in</strong>take and conception rateStudies from this laboratory (Dunne et al. 1999) showthat sudden reductions <strong>in</strong> DMI at around the time of<strong>in</strong>sem<strong>in</strong>ation adversely affect embryo survival <strong>in</strong> heifers.When energy <strong>in</strong>take was reduced from a high level oftwice their ma<strong>in</strong>tenance requirement to 0.8 times ma<strong>in</strong>tenancefor 2 weeks immediately after AI, embryosurvival rate <strong>in</strong> heifers was consistently less than 40%.When heifers were provided with a constant level of feed<strong>in</strong>take or when they were changed from a low to ahigher level feed <strong>in</strong>take, embryo survival was consistentlyhigh at 65–71%. In that study, where heifers wereused, there was no <strong>in</strong>dication of any association betweenenergy <strong>in</strong>take and systemic progesterone concentration.Unlike the situation <strong>in</strong> sheep and pigs, there was nochange <strong>in</strong> systemic progesterone follow<strong>in</strong>g either an<strong>in</strong>crease or reduction <strong>in</strong> energy <strong>in</strong>take. Changes <strong>in</strong>progesterone metabolism may have been balanced bychanges <strong>in</strong> progesterone production.Prote<strong>in</strong> nutrition and conception rateDairy cows at pasture frequently <strong>in</strong>gest high quantitiesof prote<strong>in</strong>, often with a high proportion of the<strong>in</strong>gested prote<strong>in</strong> be<strong>in</strong>g rapidly degradable <strong>in</strong> therumen. The effects of high <strong>in</strong>takes of crude prote<strong>in</strong>on conception rate are equivocal (Disk<strong>in</strong> et al. 2006).They concluded, based on the published evidence, thatelevated concentrations of urea per se are not detrimentalto embryo survival. Yet, it needs to be clarifiedwhether the observed adverse effects of urea onembryo survival are dependent on the energy statusof the animal.Future PossibilitiesIt is well established that embryo survival is criticallyimportant <strong>in</strong> all rum<strong>in</strong>ant species but particularly <strong>in</strong>seasonally bred herds. The extent of late embryomortality is numerically much smaller than earlyembryo mortality loss but nevertheless causes seriouseconomic losses to producers, because it is often too lateto rebreed females when they repeat particularly <strong>in</strong>seasonally bred herds. The major challenge will be toimprove embryonic survival rate and therefore herdreproductive efficiency particularly given the antagonisticrelationship between production and embryo survivalrate. It is clear that genetic variability exists with<strong>in</strong> theHolste<strong>in</strong> breed for important fertility (Berry et al. 2003)and DMI and EB (Berry et al. 2007) traits. Because ofthe difficulty <strong>in</strong> directly measur<strong>in</strong>g DMI and ⁄ or EB <strong>in</strong>large numbers of dairy cows, it is not currently directly<strong>in</strong>cluded <strong>in</strong> <strong>in</strong>ternational breed<strong>in</strong>g objectives. Apply<strong>in</strong>ga positive weight<strong>in</strong>g to traits correlated with DMIand ⁄ or EB or the identification and application ofsuitable molecular markers would be expected toimprove DMI and EB and subsequent reproductiveperformance. Alternatively, stra<strong>in</strong>s of cows derived frommore balanced breed<strong>in</strong>g objectives, such as the NewZealand Friesian (Harris and Kolver 2001) or alternativedairy breeds such as the Jersey or Norwegian Red,could be utilized <strong>in</strong> such production systems. Advances<strong>in</strong> genomic technology have the potential not only toprovide scientists with an improved understand<strong>in</strong>g ofthe underly<strong>in</strong>g biological processes <strong>in</strong>volved <strong>in</strong> fertilizationand the establishment of pregnancy but also toexplore gene profiles and potentially identify genesresponsible for improved embryo survival. Markerassistedselection will <strong>in</strong>crease the rate of geneticprogress for reproductive traits such as embryo survivalby <strong>in</strong>creas<strong>in</strong>g the accuracy of selection and permitt<strong>in</strong>gselection <strong>in</strong> both genders and at a young age (Rohrer2004). <strong>Reproduction</strong>-enhanc<strong>in</strong>g technologies such as AI,embryo transfer and perhaps clon<strong>in</strong>g will have animportant role <strong>in</strong> fully exploit<strong>in</strong>g their potential (Georgeand Massey 1991).ReferencesAdams GP, Sumar J, G<strong>in</strong>ther OJ, 1991: Form and function ofthe corpus luteum <strong>in</strong> llamas. Anim Reprod Sci 24, 127–138.Arthur GH, Noakes DE, Pearson H, 1982: <strong>Reproduction</strong> <strong>in</strong>the camel. In: Arthur GH, Noakes DE, Pearson H (ed.),Veter<strong>in</strong>ary <strong>Reproduction</strong> and Obstetrics. Bailli-ere T<strong>in</strong>dall,London, 482–487.Ashworth JC, 1994: Nutritional factors related to embryonicmortality <strong>in</strong> domestic species. In: Zavy MT, Geisert RDÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


266 MG Disk<strong>in</strong> and DG Morris(eds), Embryonic Mortality <strong>in</strong> <strong>Domestic</strong> Species. CRCPress, Boca Raton, FL, pp. 179–194.Ashworth CJ, Bazer FW, 1989: Changes <strong>in</strong> ov<strong>in</strong>e conceptusand endometrial function follow<strong>in</strong>g asynchronous embryotransfer or adm<strong>in</strong>istration of progesterone. Biol Reprod 40,425–433.Bazer FW, 1994: New frontiers and approaches <strong>in</strong> the study ofembryonic mortality <strong>in</strong> domestic species. In: Geisert RD,Zavy MT (eds), Embryonic Mortality <strong>in</strong> <strong>Domestic</strong> Species.CRC Press, Boca Raton, FL, pp. 195–210.Beam SW, Butler WR, 1997: Energy balance and ovarianfollicle development prior to first ovulation postpartum <strong>in</strong>dairy cows receiv<strong>in</strong>g three levels of dietary fat. Biol Reprod56, 133–142.Berry DP, Buckley F, Dillon P, Evans RD, Veerkamp RF,2003: Genetic parameters for body condition score, bodyweight, milk yield and fertility estimated us<strong>in</strong>g randomregression models. J Dairy Sci 86, 3704–3717.Berry DP, Horan B, O’Donovan M, Buckley F, Kennedy E,McEvoy M, Dillon P, 2007: Genetics of grass dry matter<strong>in</strong>take, energy balance, and digestibility <strong>in</strong> graz<strong>in</strong>g Irishdairy cows. J Dairy Sci 90, 4835–4845.Bod<strong>in</strong> L, Hanrahan JP, Poivey JP, 1992: Variation <strong>in</strong> embryosurvival <strong>in</strong> sheep and goats. Paper Presented to 43rd AnnualMeet<strong>in</strong>g E.A.A.P., Madrid, 24 pp.Britt JH, 1994: Follicular development and fertility, potentialimpacts of negative energy balance. In: Proceed<strong>in</strong>gs ofNational Reproductive Symposium, Pittsburgh, PA,pp.103–112.Cassell BG, Adamec V, Pearson RE, 2003: Maternal and fetal<strong>in</strong>breed<strong>in</strong>g depression for 70-day nonreturn and calv<strong>in</strong>g rate<strong>in</strong> Holste<strong>in</strong>s and Jerseys. J Dairy Sci 86, 2977–2983.Colas G, 1983: Factors affect<strong>in</strong>g the quality of ram semen. In:Haresign W (ed), Sheep Production. Butterworths, London,pp. 453–465.Disk<strong>in</strong> MG, Murphy JJ, Sreenan JM, 2006: Embryo survival<strong>in</strong> dairy cows managed under pastoral conditions. AnimReprod Sci 96, 297–311.Dunne LD, Disk<strong>in</strong> MG, Boland MP, O’Farrell KJ, SreenanJM, 1999: The effects of pre- and post-<strong>in</strong>sem<strong>in</strong>ation plane ofnutrition on embryo survival <strong>in</strong> beef heifers. Anim Sci 69,411–417.Fernandez-Baca S, Hansel W, Novoa C, 1970: Embryonicmortality <strong>in</strong> the Alpaca. Biol Reprod 3, 243–251.Garrett JE, Geisert RD, Zavy MT, Morgan GL, 1988:Evidence for maternal regulation of early conceptus growthand development <strong>in</strong> beef cattle. J Reprod Fertil 84, 437–446.George M, Massey JM, 1991: Velogenetics, or the synergisticuse of marker assisted selection and germ-l<strong>in</strong>e manipulation.Theriogenology 35, 151–159.Gustavsson I, 1979: Distribution and effects of the 1.29Robertsonian translocation <strong>in</strong> cattle. J Dairy Sci 62, 825–835.Hanrahan JP, 1982: Selection for <strong>in</strong>creased ovulation rate,litter size and embryo survival. In: Proceed<strong>in</strong>gs of 2ndWorld Congress Genetics Applied to Livestone Production,Vol. V, pp. 294–309.Hanrahan JP, 1990: Effect of selection for <strong>in</strong>creased litter sizeon ovulation rate and embryo survival <strong>in</strong> sheep. In:Proceed<strong>in</strong>gs of British Society of Animal Production,W<strong>in</strong>ter Meet<strong>in</strong>g, Scarborough, UK, Paper No. 32.Hanrahan JP, 1994: Embryo survival <strong>in</strong> small rum<strong>in</strong>ants:<strong>in</strong>cidence and sources of variation. In: 45th AnnualMeet<strong>in</strong>g European Association of Animal Production,Ed<strong>in</strong>burgh, September, Sheep and Goat Commission,Session 2.Hanrahan JP, Piper LR, 1992: Nature of the genetic control ofovulation rate and its relationship with litter size. In:Proceed<strong>in</strong>gs of 30th Annual Meet<strong>in</strong>g E.A.A.P., Len<strong>in</strong>grad,GS4.2, 12 pp.Harris BL, Kolver ES, 2001: Review of Holste<strong>in</strong>ization on<strong>in</strong>tensive pastoral dairy farm<strong>in</strong>g <strong>in</strong> New Zealand. J Dairy Sci84E(Suppl.), E56–E61.Horan B, Mee JF, Rath M, O’Connor P, Dillon P, 2004: Theeffect of stra<strong>in</strong> of Holste<strong>in</strong>–Friesian cow and feed<strong>in</strong>g systemon reproductive performance <strong>in</strong> seasonal-calv<strong>in</strong>g milk productionsystems. Anim Sci 79, 453–467.Hulet CV, Voigtlander HP Jr, Pope AL, Casida LE, 1956: Thenature of early-season <strong>in</strong>fertility <strong>in</strong> sheep. J Anim Sci 15,607–616.Kennedy J, Dillon P, O’Sullivan K, Buckley F, Rath M, 2003:The effect of genetic merit for milk production andconcentrate feed<strong>in</strong>g level on reproductive performance ofHolste<strong>in</strong>–Friesian cows <strong>in</strong> a grass-based system. Anim Sci76, 297–308.Kerbler TL, Buhr MM, Jordan LT, Leslie KE, Walton JS,1997: Relationship between maternal plasma progesteroneconcentration and <strong>in</strong>terferon-tau synthesis by the conceptus<strong>in</strong> cattle. Theriogenology 47, 703–714.Kleeman DO, Walker SK, Walkley JRW, Smith DH, GrimsonRJ, Seamark RF, 1990: Fertilization and embryo loss <strong>in</strong>Boorolla Mer<strong>in</strong>o · South Australian Mer<strong>in</strong>o ewes: effect ofthe F gene. Theriogenology 33, 487–498.Kuhn MT, Hutchison JL, Wiggans GR, 2006: Characterizationof Holste<strong>in</strong> Heifer fertility <strong>in</strong> the United States. J DairySci 89, 4907–4920.McMillan WH, McDonald MF, 1985: Survival of fertilizedova from ewe lambs and adult ewes <strong>in</strong> the uteri of ewelambs. Anim Reprod Sci 8, 235–240.McNeill RE, Sreenan JM, Disk<strong>in</strong> MG, Cairns MT, FitzpatrickR, Smith TJ, Morris DM, 2006: Effect of progesteroneconcentration on the expression of progesterone-responsivegenes <strong>in</strong> the bov<strong>in</strong>e endometrium dur<strong>in</strong>g the early lutealphase. Reprod Fertil Dev 18, 573–583.Mitchell LM, K<strong>in</strong>g ME, Aitken RP, Wallace JM, 1996: Effectof mat<strong>in</strong>g season and body condition on ovulation, fertilizationand pregnancy rates <strong>in</strong> crossbred ewes. Theriogenology45, 293.Mitchell LM, K<strong>in</strong>g ME, Aitken RP, Gebbie FE, Wallace JM,1999: Ovulation, fertilization and lamb<strong>in</strong>g rates, andperipheral progesterone concentrations, <strong>in</strong> ewes <strong>in</strong>sem<strong>in</strong>atedat a natural oestrus dur<strong>in</strong>g November or February.J Reprod Fertil 115, 133–140.Nancarrow CD, 1994: Embryonic mortality <strong>in</strong> the ewe anddoe. In: Zavy MT, Geisert RD (eds), Embryonic Mortality<strong>in</strong> <strong>Domestic</strong> Species. CRC Press, London, UK, pp. 79–98.Nebel R, McGilliard M, 1993: Interactions of high milk yieldand reproductive performance <strong>in</strong> dairy cows. J Dairy Sci 76,3257–3268.Parr RA, Davis IF, Fairclough RJ, Miles MA, 1987:Overfeed<strong>in</strong>g dur<strong>in</strong>g early pregnancy reduces peripheralprogesterone concentration and pregnancy rate <strong>in</strong> sheep.J Reprod Fertil 80, 317–320.Parr RA, Davis IF, Miles MA, Squires TJ, 1993: Liver bloodflow and metabolic clearance rate of progesterone <strong>in</strong> sheep.Res Vet Sci 55, 311–316.Patton J, Kenny DA, McNamara S, Mee JF, O’Mara FP,Disk<strong>in</strong> MG, Murphy JJ, 2006: Relationships between milkproduction, energy balance, plasma analytes and reproduction<strong>in</strong> Holste<strong>in</strong>–Friesian cows. J Dairy Sci 90, 649–658.Persson A, 2003: Effect of the genetic defect complex vertebralmalformation on fertility <strong>in</strong> Swedish Holste<strong>in</strong> cattle.Undergraduate Thesis, Swedish Agricultural University,Uppsala.Quirke JF, Hanrahan JP, 1977: Comparison of the survival <strong>in</strong>the uteri of adult ewes of cleaved ova from adult ewes andewe lambs. J Reprod Fertil 51, 487–489.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Embryo ⁄ Foetal Losses <strong>in</strong> Rum<strong>in</strong>ants 267Quirke JF, Hanrahan JP, 1983: Comparison of the survival offertilized eggs from adult ewes <strong>in</strong> the uteri of adult ewes andewe lambs. J Reprod Fertil 68, 289–294.Restall BJ, Brown GH, Blickey MdeB, Cahill L, Kearns R,1976: Assessment of reproductive wastage <strong>in</strong> sheep. 1.Fertilization failure and early embryonic survival. Aust JExp Agric Anim Husb 16, 329–335.Rh<strong>in</strong>d SM, Rob<strong>in</strong>son JJ, McDonald I, 1980: Relationshipamong uter<strong>in</strong>e and placental factors <strong>in</strong> prolific ewes andtheir relevance to variation <strong>in</strong> foetal weight. Anim Prod 30,115–124.Ricordeau G, Fazungles J, Lajous D, 1982: Heritability ofovulation rate and level of embryonic losses <strong>in</strong> Romanovbreed. 2nd Wld Congr Appl Livest Prod 7, 591–597.Ricordeau G, Poivey JP, Lajous D, Eychenne R, 1986: Geneticaspects of ovulation rate and embryo mortality <strong>in</strong> Romanovewes 3rd Wld Congr Appl Livest Prod 11, 90–95.Rob<strong>in</strong>son JL, Dombrowski DB, Harpestad GW, Shanks RD,1984: Detection and prevalence of UMP synthase deficiencyamong dairy cattle. J Hered 75, 277–280.Rohrer GA, 2004: An overview of genomics research and itsimpact on livestock reproduction. Reprod Fertil Dev 16, 47–54.Ryan DP, Prichard JF, Kopel E, Godke RA, 1993: Compar<strong>in</strong>gearly embryo mortality <strong>in</strong> dairy cows dur<strong>in</strong>g hot and coolseasons of the year. Theriogenology 39, 719–737.Sangsritavong S, Combs DK, Sartori RF, Armentano LE,Wiltbank MC, 2002: High feed <strong>in</strong>take <strong>in</strong>creases liver bloodflow and metabolism of progesterone and estradiol 17b <strong>in</strong>dairy cattle. J Dairy Sci 85, 2831–2842.Sartori R, Sartori-Bergfelt R, Mertens SA, Guenther JN,Parish JJ, Wiltbank MC, 2002: Fertilization and earlyembryonic development <strong>in</strong> heifers and lactat<strong>in</strong>g cows <strong>in</strong>summer and lactat<strong>in</strong>g and dry cows <strong>in</strong> w<strong>in</strong>ter. J Dairy Sci 85,2803–2812.Shanks RD, Rob<strong>in</strong>son JL, 1989: Embryonic mortality attributedto <strong>in</strong>herited deficiency of urid<strong>in</strong>e monophosphatesynthase. J Dairy Sci 72, 3035–3039.Silke V, Disk<strong>in</strong> MG, Kenny DA, Boland MP, Dillon P, MeeJF, Sreenan JM, 2001: Extent, pattern and factors associatedwith late embryonic loss <strong>in</strong> dairy cows. Anim ReprodSci 15, 1–12.Sreenan JM, Disk<strong>in</strong> MG, 1986: The extent and tim<strong>in</strong>g ofembryonic mortality <strong>in</strong> cattle. <strong>in</strong>: Sreenan JM, Disk<strong>in</strong> MG(eds), Embryonic Mortality <strong>in</strong> Farm <strong>Animals</strong>. Mart<strong>in</strong>usNijhoff, CEC, Brussels, Belgium, pp. 142–158.Sreenan JM, Disk<strong>in</strong> MG, Morris DG, 2001: Embryo survival<strong>in</strong> cattle, a major limitation to the achievement of highfertility. BSAS Occas Publ, 26, 93–104.Starbuck GR, Darwash AO, Mann GE, Lamm<strong>in</strong>g GE, 2001:The detection and treatment of post <strong>in</strong>sem<strong>in</strong>ation progesterone<strong>in</strong>sufficiency <strong>in</strong> dairy cows. BSAS Occas Publ 26,447–450.Stronge AJH, Sreenan JM, Disk<strong>in</strong> MG, Mee JF, Kenny DA,Morris DG, 2005: Post-<strong>in</strong>sem<strong>in</strong>ation milk progesteroneconcentration and embryo survival <strong>in</strong> dairy cows. Theriogenology64, 1212–1224.VanRaden PM, Miller RH, 2006: Effects of nonadditivegenetic <strong>in</strong>teractions, <strong>in</strong>breed<strong>in</strong>g and recessive defects onembryo and fetal loss by seventy days. J Dairy Sci 89, 2716–2721.Wall E, Brotherstone S, Kearney JF, Wolliams JA, CoffeyMP, 2003: Effect of <strong>in</strong>clud<strong>in</strong>g <strong>in</strong>breed<strong>in</strong>g, heterosis andrecomb<strong>in</strong>ation loss <strong>in</strong> prediction of breed<strong>in</strong>g values forfertility traits. Interbull Bull 31, 117–121.Wiebold JL, 1988: Embryonic mortality and the uter<strong>in</strong>eenvironment <strong>in</strong> first service lactat<strong>in</strong>g dairy cows. J ReprodFertil 84, 393–399.Zavy MT, 1994: Embryonic mortality <strong>in</strong> cattle. In: GeisertRD, Zavy MT (eds), Embryonic Mortality <strong>in</strong> <strong>Domestic</strong>Species. CRC Press, Boca Raton, FL, pp. 99–140.Author’s address (for correspondence): MG Disk<strong>in</strong>, Teagasc, AnimalProduction Research Centre, Mellows Campus, Athenry, Co. Galway,Ireland. E-mail: michael@disk<strong>in</strong>@teagasc.ieConflict of <strong>in</strong>terest: MG Disk<strong>in</strong> is a member of a luster that hasrecently been awarded A Science Foundation Ireland Grant; DGMorris declares no conflict of <strong>in</strong>terest.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 268–272 (2008); doi: 10.1111/j.1439-0531.2008.01172.xISSN 0936-6768Role of Cell Death Ligand and Receptor System on Regulation of Follicular Atresia<strong>in</strong> Pig OvariesN Manabe, F Matsuda-M<strong>in</strong>ehata, Y Goto, A Maeda, Y Cheng, S Nakagawa, N Inoue, K Wongpanit, H J<strong>in</strong>, H Gonda and J LiAnimal Resource Science Center, University of Tokyo, Kasama, JapanContentsSeveral hundred thousand primordial follicles are present <strong>in</strong>the mammalian ovary, however, only a limited numberdevelop to the pre-ovulatory stage, and then f<strong>in</strong>ally ovulate.The others, more than 99%, will be elim<strong>in</strong>ated through adegenerative process called ‘atresia’. The endocr<strong>in</strong>ologicalregulatory mechanisms <strong>in</strong>volved <strong>in</strong> follicular developmentand atresia have been characterized to a large extent, but theprecise temporal and molecular mechanisms <strong>in</strong>volved <strong>in</strong> theregulation of these events have rema<strong>in</strong>ed unknown. Frommany recent studies, it is suggested that the apoptosis <strong>in</strong>ovarian granulosa cells plays a crucial role <strong>in</strong> follicular atresia.Notably, death ligand–receptor <strong>in</strong>teraction and subsequent<strong>in</strong>tracellular signall<strong>in</strong>g have been demonstrated to be the keymechanisms regulat<strong>in</strong>g granulosa cell apoptosis. In this review,we provide an overview of granulosa cell apoptosis regulatedby death ligand–receptor signall<strong>in</strong>g. The roles of death ligandsand receptors [Fas ligand (FasL)–Fas, tumour necrosis factor(TNF)a–TNF receptor (TNFR), and TNFa-related apoptosis<strong>in</strong>duc<strong>in</strong>gligand (TRAIL)–TRAIL receptor (TRAILR)] and<strong>in</strong>tracellular death-signal mediators [Fas-associated deathdoma<strong>in</strong> prote<strong>in</strong> (FADD), TNF receptor 1-associated deathdoma<strong>in</strong> prote<strong>in</strong> (TRADD), caspases, apoptotic protease-activat<strong>in</strong>gfactor 1 (Apaf1), TNFR-associated factor 2 (TRAF2),and cellular FLICE-like <strong>in</strong>hibitory prote<strong>in</strong> (cFLIP), etc.] <strong>in</strong>granulosa cells will be discussed.IntroductionIn cells of both vertebrate and <strong>in</strong>vertebrate animalspecies, apoptosis plays a significant role <strong>in</strong> almost allphysiological functions. Apoptosis is a form of celldeath essential for the elim<strong>in</strong>ation of cells that aredamaged, senescent, potentially harmful, or no longeruseful. Apoptosis is characterized by <strong>in</strong>ternucleosomalDNA fragmentation, cell shr<strong>in</strong>kage, plasma membraneblebb<strong>in</strong>g, and the formation of apoptotic bodies (Schwartzmanand Cidlowski 1993). Stimulation by deathligands or deprivation of key survival-promot<strong>in</strong>g growthfactors is the ma<strong>in</strong> contributor to apoptosis, while stress<strong>in</strong>ducers, <strong>in</strong>clud<strong>in</strong>g drugs, toxicants, oxidative stress,and radiation, are also known to cause apoptosis(Hengartner 2000). Recent studies have revealed thatapoptosis also plays a crucial role <strong>in</strong> ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>govarian homeostasis <strong>in</strong> mammals (Hughes and Gorospe1991; Tilly et al. 1991; Manabe et al. 1996; Kaipia andHsueh 1997). Dur<strong>in</strong>g follicular growth and development,more than 99% of follicles disappear primarilydue to apoptosis of granulosa cells as the biochemicaland morphological characteristics of apoptosis havebeen observed <strong>in</strong> the granulosa cells of atretic folliclesgrowth (Grant et al. 1989; Guthrie et al. 1995). Apoptoticstimuli and <strong>in</strong>tracellular signal transduction pathways<strong>in</strong>volved <strong>in</strong> the apoptosis of granulosa cells rema<strong>in</strong>to be determ<strong>in</strong>ed, and <strong>in</strong>vestigators are study<strong>in</strong>gpotential triggers of apoptosis and how <strong>in</strong>tracellularapoptotic signals are transmitted <strong>in</strong> granulosa cells(Manabe et al. 2003, 2004). Many apoptosis-relatedfactors are implicated <strong>in</strong> follicular atresia, <strong>in</strong>clud<strong>in</strong>gdeath ligands and receptors, <strong>in</strong>tracellular pro- and antiapoptoticmolecules, cytok<strong>in</strong>es, and growth factors(Matsuda-M<strong>in</strong>ehata et al. 2006). In particular, cell deathligand–receptor signall<strong>in</strong>g has been revealed to be themajor regulatory system for apoptosis <strong>in</strong> granulosa cells(Nakayama et al. 2003; Matsuda-M<strong>in</strong>ehata et al. 2008).Follicular Growth, Development and Atresia <strong>in</strong>Pig OvaryAfter puberty, a number of primordial follicles start togrow dur<strong>in</strong>g each oestrus cycle <strong>in</strong> adult females.Initiation of follicular growth <strong>in</strong>volves endocr<strong>in</strong>ologicalfactors, ma<strong>in</strong>ly FSH, and local modulat<strong>in</strong>g factors fromgranulosa cells, theca cells, stromal-<strong>in</strong>terstitial cells, andoocytes (Hirshfield 1991; Guthrie et al. 1995). Primaryfollicles (follicles with a monolayer of follicular epithelial⁄ granulosa cells) develop <strong>in</strong>to secondary follicles(follicles with stratified granulosa cells but without anantrum) and subsequently <strong>in</strong>to tertiary follicles (follicleswith a follicular antrum) (Grant et al. 1989). Due to alarge <strong>in</strong>crease <strong>in</strong> the proliferation of granulosa cells andan <strong>in</strong>crease <strong>in</strong> the size of the antrum, tertiary folliclesshow an exponential rate of growth. In the oocyte,meiosis then restarts and the first polar body divides.F<strong>in</strong>ally, selected follicles burst, and the oocytes ovulate.With <strong>in</strong>creas<strong>in</strong>g serum FSH concentrations at the startof the oestrous cycle, follicles produce <strong>in</strong>creas<strong>in</strong>gamounts of oestrogen and <strong>in</strong>hib<strong>in</strong>, produced by granulosacells. As a feedback mechanism by <strong>in</strong>hib<strong>in</strong>, FSHsecretion falls, and the rema<strong>in</strong><strong>in</strong>g small follicles undergoatresia. Although atresia can occur at any time dur<strong>in</strong>gfollicular development, the majority of follicles becomeatretic dur<strong>in</strong>g the early antral stage of development. Thetransition from pre-antral to antral follicular developmentoccurs after exposure of the granulosa cells togonadotrop<strong>in</strong>. Then, the differentiation of granulosacells is <strong>in</strong>itiated, which renders them susceptible toapoptosis. The endonuclease, DNase-I was demonstratedto exist <strong>in</strong> granulosa cells from antral, but notpre-antral, follicles (Manabe et al. 1996a,b). However,the presence of the endonuclease is not sufficient tocause apoptosis; a signal to activate DNase-I and <strong>in</strong>ducecell death is required. Death ligand–receptor <strong>in</strong>teractionis thought to be a major trigger of apoptosis <strong>in</strong>granulosa cells (Manabe et al. 2003, 2004).Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Death Ligand and Receptor Pig Ovarian Follicle 269Death Ligand and Receptor System RegulatesFollicular Atresia <strong>in</strong> Pig OvaryDeath ligands, which are grouped the tumour necrosisfactor (TNF) family, are synthesized as type-II membraneprote<strong>in</strong>s (Ashkenazi and Dixit 1998). Most deathligands can be cleaved from the cell membrane tobecome a soluble form and act as trimers. Deathreceptors, which are classified as type-I membraneprote<strong>in</strong>s, constitute a subfamily with<strong>in</strong> the TNF receptorsuperfamily (TNFRsf) that has a cytoplasmic deathdoma<strong>in</strong> (DD) necessary for the activation of apoptosis(Hengartner 2000). These receptors are trimerized andthen b<strong>in</strong>d to death ligands, which are a trigger forapoptosis. Cell death ligand–receptor systems known <strong>in</strong>mammals <strong>in</strong>clude the Fas ligand (FasL) and Fas (CD95,APO-1 or TNFRsf6), TNFa and TNFa receptors(TNFRs), and TNFa-related apoptosis-<strong>in</strong>duc<strong>in</strong>g ligand(TRAIL, Apo2L) and TRAIL receptors (Nagata 1997;Ashkenazi and Dixit 1998; Wallach et al. 1999). In mostcases, the cell death receptor-mediated apoptotic signall<strong>in</strong>gpathway is as follows: (1) Cell death ligands,b<strong>in</strong>d<strong>in</strong>g with cell membranes or exist<strong>in</strong>g as a solubleform, b<strong>in</strong>d to the extracellular doma<strong>in</strong> of trimerized celldeath receptors, each of which conta<strong>in</strong>s an <strong>in</strong>tracellularDD. (2) The DD of the death receptor b<strong>in</strong>ds with theDD of the adaptor prote<strong>in</strong>s (TNF receptor 1-associateddeath doma<strong>in</strong> prote<strong>in</strong>: TRADD and Fas-associateddeath doma<strong>in</strong> prote<strong>in</strong>: FADD) through homophilic<strong>in</strong>teraction (Inoue et al. 2007). (3) An <strong>in</strong>itiator caspase(procaspase-8) b<strong>in</strong>ds to FADD through homophilic<strong>in</strong>teraction with the death effector doma<strong>in</strong> (DED; theresult<strong>in</strong>g complex is called the ‘death-<strong>in</strong>duc<strong>in</strong>g signall<strong>in</strong>gcomplex’: DISC) (Medema et al. 1997). (4) Dimerizationof procaspase-8 <strong>in</strong>duces auto-proteolytic cleavageand activation (Bold<strong>in</strong> et al. 1995). (5) The activatedcaspase-8 subsequently activates downstream caspaseseither directly (‘type I’) or via mitochondrial perturbation(‘type II; mitochondrion-dependent’) (Matsui et al.2003). (6-a) In type I apoptotic cells, caspase-8 directlyactivates the effector enzyme, caspase-3. (7-a) Trancatedcaspase-3, active form, activates endogenous endonucleasesthat results <strong>in</strong> apoptosis. (6-b) In type II apoptoticcells, caspase-8’s activation leads to the release ofcytochrome c from mitochondrion, which results <strong>in</strong> the<strong>in</strong>teraction of procaspase-9 with apoptotic proteaseactivat<strong>in</strong>gfactor 1 (Apaf1) [formation of cytochromec-Apaf1-caspase-9 complex (apoptosome)]. (7-b) Activatedcaspase-9 cleaves procaspase-3. (8-b) Active caspase-3activates the endonucleases that results <strong>in</strong>apoptosis (Nagata 1997; Ashkenazi and Dixit 1998;Matsui et al. 2003) (Fig. 1).FasL–Fas and other cell death ligand–receptor systems <strong>in</strong>granulosa cellThe FasL–Fas system is one of the most studiedparadigms of <strong>in</strong>structive apoptosis, with strong apoptosis-<strong>in</strong>duc<strong>in</strong>gactivity (Wallach et al. 1999). The apoptosissignal triggered and mediated by FasL and Fas isas follows: (1) FasL b<strong>in</strong>ds to the extracellular doma<strong>in</strong>of Fas. (2) The <strong>in</strong>tracellular DD of Fas <strong>in</strong>teracts withthe adaptor prote<strong>in</strong> (FADD) through DD. (3) FADDDiscFasLFasFADDActivated caspase-8ApoptosisProcaspase-8: Death doma<strong>in</strong> (DD): Death effector doma<strong>in</strong> (DED): Enzymatic doma<strong>in</strong>Cell membraneFig. 1. Intracellular signall<strong>in</strong>g <strong>in</strong> TNF family. When cell death ligandb<strong>in</strong>ds to the extracellular region of cell death receptor, adoptor (Fasassociateddeath doma<strong>in</strong> prote<strong>in</strong>: FADD) b<strong>in</strong>ds to the <strong>in</strong>tracellularregion of receptor through the homophilic <strong>in</strong>teraction of the deathdoma<strong>in</strong>s (DD). Then, <strong>in</strong>itiator caspase (procaspase-8) b<strong>in</strong>ds to FADDthrough the homophilic <strong>in</strong>teraction of the death effector doma<strong>in</strong>(DED). Dimerized procaspase-8 is auto-cleaved, and then truncatedcaspase-8 (active caspase-8) <strong>in</strong>duces activation of down-stream caspases(ex. caspase-3), which results <strong>in</strong> apoptosis<strong>in</strong>teracts with procaspase-8 through their DED and theactive caspase-8 is formed. (4) A caspase cascade isactivated and eventually apoptosis is <strong>in</strong>duced. TheFasL–Fas system is the most characterized apoptosissignall<strong>in</strong>g mach<strong>in</strong>ery <strong>in</strong> granulosa cells (Inoue et al.2006). In many species, both FasL and Fas areexpressed <strong>in</strong> granulosa cells. In mur<strong>in</strong>e ovaries, FasLand Fas mRNA and prote<strong>in</strong> are expressed <strong>in</strong> granulosacells of healthy and atretic follicles (Sakamaki et al.1997). Moreover, treatment of female mice with Fasactivat<strong>in</strong>gantibody promoted granulosa cell apoptosisand follicular atresia, suggest<strong>in</strong>g a pro-apoptotic functionof FasL–Fas signall<strong>in</strong>g <strong>in</strong> vivo. In rat ovaries, FasLand Fas prote<strong>in</strong> occur <strong>in</strong> granulosa cells, which tend tobe more abundant <strong>in</strong> atretic follicles than healthyfollicles (Hakuno et al. 1996). In human females, thegranulosa cells of antral follicles express Fas dur<strong>in</strong>g theearly stages of atresia, the levels of Fas expression<strong>in</strong>crease as atresia progresses, and granulosa FasLprote<strong>in</strong> also <strong>in</strong>creases with atresia <strong>in</strong> antral follicles(Cataldo et al. 2000). In bov<strong>in</strong>e ovaries, FasL mRNAlevels are higher <strong>in</strong> granulosa cells from atretic folliclesthan those from healthy follicles, and Fas mRNAexpression is stronger <strong>in</strong> granulosa cells of atretic thanhealthy follicles (Porter et al. 2000, 2001). Moreover,CD45, a pan-leucocyte marker, was not detected <strong>in</strong>bov<strong>in</strong>e granulosa cells, <strong>in</strong>dicat<strong>in</strong>g that immune cellswere not a source of FasL or Fas <strong>in</strong> the granulosa celllayer (Hu et al. 2001). Trace levels of FasL and FasmRNA and prote<strong>in</strong> were detected <strong>in</strong> granulosa cells ofhealthy follicles but the levels <strong>in</strong>creased dur<strong>in</strong>g atresia<strong>in</strong> porc<strong>in</strong>e ovaries (Inoue et al. 2006). These expressionpatterns of FasL and Fas strongly <strong>in</strong>dicate the importantrole of FasL–Fas signall<strong>in</strong>g <strong>in</strong> follicular atresia.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


270 N Manabe et al.Both FasL and Fas exist on the granulosa cellmembrane and their <strong>in</strong>teraction may <strong>in</strong>duce apoptosis.The pro-apoptotic effect of the FasL–Fas system <strong>in</strong>granulosa cells was also demonstrated by experiments<strong>in</strong> vitro. Stimulation of Fas-signall<strong>in</strong>g can <strong>in</strong>duceapoptosis <strong>in</strong> primary cultured granulosa cells of manyspecies <strong>in</strong>clud<strong>in</strong>g pigs (Matsuda-M<strong>in</strong>ehata et al. 2006).However, pre- or co-treatment with <strong>in</strong>terferon-c orcycloheximide (CHX) is necessary for <strong>in</strong>duc<strong>in</strong>g theapoptosis <strong>in</strong> vitro, <strong>in</strong>dicat<strong>in</strong>g the existence of otheressential factor(s) <strong>in</strong> the FasL–Fas signall<strong>in</strong>g pathwayof granulosa cell apoptosis. Moreover, we found notonly FasL–Fas system, but also TRAIL–TRAILRs(Wada et al. 2002; Inoue et al. 2003), TNFa–TNFRs(Nakayama et al. 2003), and unknown ligand–PFGreceptors (Manabe et al. 2000) systems <strong>in</strong> porc<strong>in</strong>egranulosa cells. However, we have no confirmed<strong>in</strong>formation which system plays dom<strong>in</strong>ant role <strong>in</strong>regulation of granulosa cell apoptosis (Matsuda-M<strong>in</strong>ehataet al. 2008).Intracellular regulator (apoptosis-<strong>in</strong>hibitory factor) <strong>in</strong>porc<strong>in</strong>e granulosa cellDeath ligand–receptor <strong>in</strong>teraction is <strong>in</strong>deed essentialfor trigger<strong>in</strong>g apoptotic signall<strong>in</strong>g, however, it does notnecessarily result <strong>in</strong> apoptotic cell death, <strong>in</strong>dicat<strong>in</strong>g theimportance of <strong>in</strong>tracellular <strong>in</strong>hibitors of the apoptoticsignall<strong>in</strong>g pathway. Recently, we found that cellularFLICE-like <strong>in</strong>hibitory prote<strong>in</strong> (cFLIP: also calledCASH, Casper, CLARP, FLAME, I-FLICE, MRIT,or usurp<strong>in</strong>) (Goltsev et al. 1997; Han et al. 1997; Huet al. 1997; Krueger et al. 2001), which is a homologueof procaspase-8 (also called FLICE) (Muzio et al.1996) and one of the <strong>in</strong>tracellular prote<strong>in</strong>s that<strong>in</strong>terferes with the apoptotic effects of death ligands,plays crucial role <strong>in</strong> regulation of granulosa cellapoptosis <strong>in</strong> porc<strong>in</strong>e ovaries (Goto et al. 2004).FLICE-like <strong>in</strong>hibitory prote<strong>in</strong> was firstly identified <strong>in</strong>several viruses as viral FLIP (vFLIP), which conta<strong>in</strong>stwo DEDs that <strong>in</strong>teract with FADD to avoid thehost’s apoptotic response (Thome et al. 1997). Inmammalian cell, homologue of vFLIP was found andnamed cFLIP (Irmler et al. 1997). There are twosplic<strong>in</strong>g variants of cFLIP, short and long forms(cFLIP S and cFLIP L , respectively). cFLIP S is verysimilar <strong>in</strong> structure to vFLIP, conta<strong>in</strong><strong>in</strong>g two DEDs,while cFLIP L conta<strong>in</strong>s an additional pseudo-enzymaticdoma<strong>in</strong> that is similar to the enzymatic doma<strong>in</strong> ofprocaspase-8 but lacks enzymatic activity. Recently, wefound that cFLIP S and cFLIP L are expressed <strong>in</strong>porc<strong>in</strong>e granulosa cells and both of them to beimportant regulators of apoptosis that block deathligand-<strong>in</strong>ducible apoptosis, by compet<strong>in</strong>g with procaspase-8and <strong>in</strong>hibit<strong>in</strong>g the activation of caspase-8(Thome and Tschopp 2001). The homology of porc<strong>in</strong>ecFLIP with human and mur<strong>in</strong>e cFLIP is very high(more than 75% for both the mRNA and am<strong>in</strong>o acidlevels) (Goto et al. 2004), and we have proposed thatcFLIP also has cell survival-promot<strong>in</strong>g effects <strong>in</strong> thepig. As described above, the FasL–Fas system is wellcharacterizedas a pro-apoptotic signal <strong>in</strong> granulosacells <strong>in</strong> sows, and the expression of FasL and Fas <strong>in</strong>granulosa cells <strong>in</strong>creases dur<strong>in</strong>g atresia, however, bothprote<strong>in</strong>s are also expressed <strong>in</strong> granulosa cells of healthypre-antral and antral follicles, which rapidly grow andhave many proliferat<strong>in</strong>g granulosa cells. Moreover, Fascannot <strong>in</strong>duce apoptosis <strong>in</strong> primarily cultured porc<strong>in</strong>egranulosa cells without either IFN-c or CHX. It hasbeen suggested that the factor(s) that blocks FasL–Fasmediatedapoptotic signall<strong>in</strong>g is essential for ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>ggranulosa cells and keep<strong>in</strong>g follicles healthy. Byreverse-transcription-polymerase cha<strong>in</strong> reaction (RT-PCR) and Western blott<strong>in</strong>g, the mRNA and prote<strong>in</strong> ofcFLIP L were found to be highly expressed <strong>in</strong> thegranulosa cells of healthy follicles and decreased dur<strong>in</strong>gatresia (Goto et al. 2004). The mRNA levels of cFLIP S<strong>in</strong> granulosa cells are low and showed no changesamong the stages of follicular development (Matsuda-M<strong>in</strong>ehata et al. 2005, 2007). Furthermore, the prote<strong>in</strong>level of cFLIP S is extremely low. By <strong>in</strong> situ hybridization,cFLIP L was found to be abundant <strong>in</strong> thegranulosa cells of healthy follicles <strong>in</strong> comparison withthose of atretic follicles. Immunohistochemical analysesshowed that cFLIP prote<strong>in</strong> was found to be highlyexpressed <strong>in</strong> the granulosa cells of healthy follicles butweakly expressed <strong>in</strong> those of atretic follicles. Wepresume that cFLIP, especially cFLIP L , plays ananti-apoptotic role <strong>in</strong> the granulosa cells of healthyfollicles from pig ovaries (Fig. 2). S<strong>in</strong>ce the antiapoptoticactivity of porc<strong>in</strong>e cFLIP (pcFLIP) had notbeen confirmed <strong>in</strong> granulosa cells, we exam<strong>in</strong>ed theeffect of pcFLIP on survival us<strong>in</strong>g granulosa-derivedcells (Matsuda-M<strong>in</strong>ehata et al. 2007). Human ovariangranulosa tumour cell derived KGN cells (Nishi et al.2001) transfected with pcFLIP S or pcFLIP L vectorssurvive the <strong>in</strong>duction of ant-Fas antibody-Fas-mediatedapoptosis, while almost all cells transfected withempty vector die, <strong>in</strong>dicat<strong>in</strong>g the anti-apoptotic activityof pcFLIP <strong>in</strong> granulosa cells. When both cFLIP S andcFLIP L , or cFLIP L only, were suppressed by small<strong>in</strong>terfer<strong>in</strong>g RNA (siRNA), the viability of KGN and Jporc<strong>in</strong>e granulosa-derived JC-410 cells (Chedrese et al.1998) decreased significantly. Thus, we conclude thatporc<strong>in</strong>e cFLIP functions as an anti-apoptotic factor <strong>in</strong>granulosa-derived cells. These our f<strong>in</strong>d<strong>in</strong>gs stronglysuggest that cFLIP acts as a survival-promot<strong>in</strong>g factor<strong>in</strong> granulosa cells and determ<strong>in</strong>es whether porc<strong>in</strong>eovarian follicles survive or undergo atresia. To date,<strong>in</strong>vestigations have revealed that cFLIP can also <strong>in</strong>hibitTNFa- and TRAIL-signall<strong>in</strong>g (Cheng et al. 2007), notonly Fas-signall<strong>in</strong>g (Manabe et al. 2003). Further<strong>in</strong>vestigations are needed to determ<strong>in</strong>e whether cFLIPaffects <strong>in</strong>tracellular signall<strong>in</strong>g transduction <strong>in</strong> other celldeath ligand and receptor systems (e.g. TNFa–TNFRs,TRAIL–TRAILRs, etc.) <strong>in</strong> porc<strong>in</strong>e granulosa cells ornot. More researches are also needed how to regulatethe expression of cFLIP <strong>in</strong> granulosa cells dur<strong>in</strong>gfollicular growth, development, and atresia. Our previousf<strong>in</strong>d<strong>in</strong>gs have shown that <strong>in</strong>terleuk<strong>in</strong>-6 up-regulatesTNFa expression <strong>in</strong> porc<strong>in</strong>e granulosa cells(Maeda et al. 2007a,b), and that TNFa up-regulatescFLIP L expression and acts a survival factor (Nakayamaet al. 2003). However, there is no confirmed<strong>in</strong>formation on what is the <strong>in</strong>itial trigger to controlcFLIP expression.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Death Ligand and Receptor Pig Ovarian Follicle 271Fig. 2. Work<strong>in</strong>g hypothesis on regulation of apoptosis <strong>in</strong> porc<strong>in</strong>e granulosa cells of healthy (left) and atretic follicles (right). Although the celldeath ligands and receptors are expressed and <strong>in</strong>teract on granulosa cells of healthy follicles, subsequent apoptotic signall<strong>in</strong>g is blocked bycFLIP L and ⁄ or cFLIP S , dom<strong>in</strong>antly cFLIP L . As a result, the apoptosis of the granulosa cells is avoided and the follicle is kept healthy. When thecFLIP L and cFLIP S are down-regulated, cell death ligand and receptor <strong>in</strong>teraction causes cleavage of procaspase-8, and subsequent apoptoticsignall<strong>in</strong>g. As a result, the apoptosis of the granulosa cells is <strong>in</strong>duced, and the follicle undergoes atresiaConclusionTo date, <strong>in</strong>ducers of apoptosis <strong>in</strong>clud<strong>in</strong>g FasL–Fas,TNFa–TNFRs, TRAIL–TRAILRs have been the ma<strong>in</strong>targets <strong>in</strong> the studies of regulation mechanism ofgranulosa cell apoptosis, and their contributions tofollicular atresia have been clarified. However, it hasbecome apparent that an <strong>in</strong>tracellular anti-apoptotic⁄ <strong>in</strong>hibitory factor(s), like cFLIP, is critical for <strong>in</strong>hibit<strong>in</strong>ggranulosa cell apoptosis from our recent research.Granulosa cell apoptosis is likely to be regulated by asophisticated balance of pro-apoptotic and anti-apoptoticfactors. The mechanisms of death ligand–receptorsignall<strong>in</strong>g should be determ<strong>in</strong>ed to entirely def<strong>in</strong>egranulosa cell apoptosis. Solv<strong>in</strong>g this problem will helpto establish methods (1) of select<strong>in</strong>g healthy oocytes orto improve damaged oocytes that result <strong>in</strong> an <strong>in</strong>creasedrate of gestation for <strong>in</strong> vitro fertilization <strong>in</strong> domesticanimals and humans, and (2) of rescue oocytes <strong>in</strong> theovaries of wild animals and livestock sacrificed at theslaughterhouse.AcknowledgementsOur works were supported by Grant-<strong>in</strong>-Aid for Scientific Research(13GS0008, S16108003, B18380164, E18658105, and S18108004) to N.M. and by Research Fellowship for Young Scientists to F. M-M. fromthe Japan Society for the Promotion of Science.ReferencesAshkenazi A, Dixit VM, 1998: Death receptors: signal<strong>in</strong>g andmodulation. Science 281, 1305–1308.Bold<strong>in</strong> MP, Mett IL, Varfolomeev EE, Chumakov I, Shemer-Avni Y, Camonis JH, Wallach D, 1995: Self-association ofthe ‘‘death doma<strong>in</strong>s’’ of the p55 tumor necrosis factor(TNF) receptor and Fas ⁄ APO1 prompts signal<strong>in</strong>g for TNFand Fas ⁄ APO1 effects. J Biol Chem 270, 387–391.Cataldo NA, Dumesic DA, Goldsmith PC, Jaffe RB, 2000:Immunolocalization of Fas and Fas ligand <strong>in</strong> the ovaries ofwomen with polycystic ovary syndrome: relationship toapoptosis. Hum Reprod 15, 1889–1897.Chedrese PJ, Rodway MR, Swan CL, Gillio-Me<strong>in</strong>a C, 1998:Establishment of a stable steroidogenic porc<strong>in</strong>e granulosacell l<strong>in</strong>e. J Mol Endocr<strong>in</strong>ol 20, 287–292.Cheng Y, Maeda A, Goto Y, Matsuda-M<strong>in</strong>ehata F, ManabeN, 2007: Molecular clon<strong>in</strong>g of porc<strong>in</strong>e (Sus scrofa) tumornecrosis factor receptor 2. J Reprod Dev 53, 1291–1297.Goltsev YV, Kovalenko AV, Arnold E, Varfolomeev EE,Brodianskii VM, Wallach D, 1997: CASH, a novel caspasehomologue with death effector doma<strong>in</strong>s. J Biol Chem 272,19641–19644.Goto Y, Matsuda-M<strong>in</strong>ehata F, Inoue N, Matsui T, Maeda A,Manabe N, 2004: Porc<strong>in</strong>e (Sus scrofa) cellular FLICE-like<strong>in</strong>hibitory prote<strong>in</strong> (cFLIP): molecular clon<strong>in</strong>g and comparisonwith the human and mur<strong>in</strong>e cFLIP. J Reprod Fertil 50,549–555.Grant SA, Hunter MG, Foxcroft GR, 1989: Morphologicaland biochemical characteristics dur<strong>in</strong>g ovarian folliculardevelopment <strong>in</strong> the pig. J Reprod Fertil 86, 171–183.Guthrie HD, Cooper BS, Welch GR, Zakaria AD, JohnsonLA, 1995: Atresia <strong>in</strong> follicle grown after ovulation <strong>in</strong> the pig:Measurement of <strong>in</strong>creased apoptosis <strong>in</strong> granulosa cells andreduced follicular fluid estradiol-17b. Biol Reprod 52, 920–927.Hakuno N, Koji T, Yano T, Kobayashi N, Tsutsumi O,Taketani Y, Nakane PK, 1996: Fas ⁄ APO-1 ⁄ CD95 system asa mediator of granulosa cell apoptosis <strong>in</strong> ovarian follicleatresia. Endocr<strong>in</strong>ology 137, 1938–1948.Han DK, Chaudhary PM, Wright ME, Friedman C, Trask BJ,Riedel RT, Bask<strong>in</strong> DG, Schwartz SM, Hood L, 1997:MRIT, a novel death-effector doma<strong>in</strong>-conta<strong>in</strong><strong>in</strong>g prote<strong>in</strong>,<strong>in</strong>teracts with caspases and BclXL and <strong>in</strong>itiates cell death.Proc Natl Acad Sci U S A 94, 11333–11338.Hengartner MO, 2000: The biochemistry of apoptosis. Nature407, 770–776.Hirshfield AN, 1991: Development of follicles <strong>in</strong> the mammalianovary. Int Rev Cytol 124, 43–101.Hu S, V<strong>in</strong>cenz C, Ni J, Gentz R, Dixit VM, 1997: I-FLICE, anovel <strong>in</strong>hibitor of tumor necrosis factor receptor-1- and CD-95-<strong>in</strong>duced apoptosis. J Biol Chem 272, 17255–17257.Hu CL, Cowan RG, Harman RM, Porter DA, Quirk SM,2001: Apoptosis of bov<strong>in</strong>e granulosa cells after serumwithdrawal is mediated by Fas antigen (CD95) and Fasligand. Biol Reprod 64, 518–526.Hughes FM Jr, Gorospe WC, 1991: Biochemical identificationof apoptosis (programmed cell death) <strong>in</strong> granulosa cells:evidence for a potential mechanism underly<strong>in</strong>g follicularatresia. Endocr<strong>in</strong>ology 129, 2415–2422.Inoue N, Manabe N, Matsui T, Maeda A, Nakagawa S, WadaS, Miyamoto H, 2003: Roles of tumor necrosisÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


272 N Manabe et al.factor-related ligand signal<strong>in</strong>g pathway <strong>in</strong> granulosa cellapoptosis dur<strong>in</strong>g atresia <strong>in</strong> pig ovaries. J Reprod Dev 49,313–321.Inoue N, Maeda A, Matsuda-M<strong>in</strong>ehata F, Fukuta K, ManabeN, 2006: Expression and localization of Fas ligand and Fasdur<strong>in</strong>g atresia <strong>in</strong> porc<strong>in</strong>e ovarian follicles. J Reprod Dev 52,723–730.Inoue N, Matsuda-M<strong>in</strong>ehata F, Manabe N, 2007: Molecularcharacteristics of porc<strong>in</strong>e Fas-associated death doma<strong>in</strong>(FADD) and procaspase-8. J Reprod Dev 53, 427–436.Irmler M, Thome M, Hahne M, Schneider P, Hofmann K,Ste<strong>in</strong>er V, Bodmer JL, Schroter M, Burns K, Mattmann C,Rimoldi D, French LE, Tschopp J, 1997: Inhibition of deathreceptor signals by cellular FLIP. Nature 388, 190–195.Kaipia A, Hsueh AJ, 1997: Regulation of ovarian follicleatresia. Ann Rev Physiol 59, 349–363.Krueger A, Baumann S, Krammer PH, Kirchhoff S, 2001:FLICE-<strong>in</strong>hibitory prote<strong>in</strong>s: regulators of death receptormediatedapoptosis. Mol Cell Biol 21, 8247–8254.Maeda A, Inoue N, Matsuda-M<strong>in</strong>ehata F, Cheng Y, ManabeN, 2007a: The role of <strong>in</strong>terleuk<strong>in</strong>-6 <strong>in</strong> the regulation ofgranulosa cell apoptosis dur<strong>in</strong>g follicular atresia <strong>in</strong> pigovaries. J Reprod Dev 53, 481–490.Maeda A, Goto Y, Matsuda-M<strong>in</strong>ehata F, Cheng Y, Inoue N,Manabe N, 2007b: Changes <strong>in</strong> expression of <strong>in</strong>terleuk<strong>in</strong>-6receptors <strong>in</strong> granulosa cells dur<strong>in</strong>g follicular atresia <strong>in</strong> pigovaries. J Reprod Dev 53, 727–736.Manabe N, Imai Y, Ohno H, Takahagi Y, Sugimoto M,Miyamoto H, 1996a: Apoptosis occurs <strong>in</strong> granulosa cells butnot cumulus cells <strong>in</strong> the atretic antral follicles <strong>in</strong> pig ovaries.Experientia 52, 647–651.Manabe N, Imai Y, Kimura Y, Myoumoto A, Sugimoto M,Miyamoto H, 1996b: Ca 2+ ⁄ Mg 2+ -dependent endonucleasebut not Ca 2+ -dependent, Mg 2+ -dependent or cation-<strong>in</strong>dependentendonuclease is <strong>in</strong>volved <strong>in</strong> granulosa cell apoptosisof pig atretic follicles. J Reprod Dev 42, 247–253.Manabe N, Myoumoto A, Tajima C, Fukumoto M, NakayamaM, Uchio K, Yamaguchi M, Miyamoto H, 2000:Immunochemical characteristics of a novel cell deathreceptor and a decoy receptor on granulosa cells of porc<strong>in</strong>eovarian follicles. Cytotechnology 33, 189–201.Manabe N, Inoue N, Miyano T, Sakamaki K, Sugimoto M,Myamoto H, 2003: Ovarian follicle selection <strong>in</strong> mammalianovaries: regulatory mechanisms of granulosa cell apoptosisdur<strong>in</strong>g follicular atresia. In: Leung PK, Adashi E (eds), TheOvary, 2nd edn. Academic Press and Elsevier, Amsterdam,pp. 369–385.Manabe N, Goto Y, Matsuda-M<strong>in</strong>ehata F, Inoue N, MaedaA, Sugimoto M, Sakamaki K, Miyano T, 2004: Regulationmechanism of selective atresia <strong>in</strong> porc<strong>in</strong>e follicles: regulationof granulosa cell apoptosis dur<strong>in</strong>g atresia. J Reprod Dev 50,493–514.Matsuda-M<strong>in</strong>ehata F, Goto Y, Inoue N, Manabe N, 2005:Changes <strong>in</strong> expression of anti-apoptotic prote<strong>in</strong>, cFLIP, <strong>in</strong>granulosa cell dur<strong>in</strong>g follicular atresia <strong>in</strong> porc<strong>in</strong>e ovaries.Mol Reprod Dev 72, 145–151.Matsuda-M<strong>in</strong>ehata F, Inoue N, Goto Y, Manabe N, 2006:The regulation of ovarian granulosa cell death by pro- andanti-apoptotic molecules. J Reprod Dev 52, 695–705.Matsuda-M<strong>in</strong>ehata F, Goto Y, Inoue N, Sakamaki K, PChedrese PJ, Manabe N, 2007: Anti-apoptotic activity ofporc<strong>in</strong>e cFLIP <strong>in</strong> ovarian granulosa cell l<strong>in</strong>es. Mol ReprodDev 74, 1165–1170.Matsuda-M<strong>in</strong>ehata F, Maeda A, Cheng Y, Sai T, Gonda H,Manabe N, 2008: Regulation of granulosa cell apoptosis bydeath ligand-receptor signal<strong>in</strong>g. Anim Sci J 79, 1–10.Matsui T, Manabe N, Goto Y, Inoue N, Nishihara S,Miyamoto H, 2003: Expression and activity of Apaf1 andcaspase-9 <strong>in</strong> granulosa cells dur<strong>in</strong>g follicular atresia <strong>in</strong> pigovaries. <strong>Reproduction</strong> 126, 113–120.Medema JP, Scaffidi C, Kischkel FC, Shevchenko A, MannM, Krammer PH, Peter ME, 1997: FLICE is activated byassociation with the CD95 death-<strong>in</strong>duc<strong>in</strong>g signal<strong>in</strong>g complex(DISC). EMBO J 16, 2794–2804.Muzio M, Ch<strong>in</strong>naiyan AM, Kischkel FC, O’Rourke K,Shevchenko A, Ni J, Scaffidi C, Bretz JD, Zhang M, GentzR, Mann M, Krammer PH, Peter ME, Dixit VM, 1996:FLICE, a novel FADD-homologous ICE ⁄ CED-3-like protease,is recruited to the CD95 (Fas ⁄ APO-1) death-<strong>in</strong>duc<strong>in</strong>gsignal<strong>in</strong>g complex. Cell 85, 817–827.Nagata S, 1997: Apoptosis by death factor. Cell 88, 355–365.Nakayama M, Manabe N, Inoue N, Matsui T, Miyamoto H,2003: Changes <strong>in</strong> the expression of tumor necrosis factor(TNF)a, TNFa receptor (TNFR) 2, TNFR-associatedfactor 2 <strong>in</strong> granulosa cells dur<strong>in</strong>g atresia <strong>in</strong> pig ovaries. BiolReprod 68, 530–535.Nishi Y, Yanase T, Mu Y, Oba K, Ich<strong>in</strong>o I, Saito M, NomuraM, Mukasa C, Okabe T, Goto K, Takayanagi R, KashimuraY, Haji M, Nawata H, 2001: Establishment andcharacterization of a steroidogenic human granulosa-liketumor cell l<strong>in</strong>e, KGN, that expresses functional folliclestimulat<strong>in</strong>ghormone receptor. Endocr<strong>in</strong>ology 142, 437–445.Porter DA, Vickers SL, Cowan RG, Huber SC, Quirk SM,2000: Expression and function of Fas antigen vary <strong>in</strong> bov<strong>in</strong>egranulosa and theca cells dur<strong>in</strong>g ovarian follicular developmentand atresia. Biol Reprod 62, 62–66.Porter DA, Harman RM, Cowan RG, Quirk SM, 2001:Relationship of Fas ligand expression and atresia dur<strong>in</strong>gbov<strong>in</strong>e follicle development. <strong>Reproduction</strong> 121, 561–566.Sakamaki K, Yoshida H, Nishimura Y, Nishikawa S, ManabeN, Yonehara S, 1997: Involvement of Fas antigen <strong>in</strong> ovarianfollicular atresia and luteolysis. Mol Reprod Dev 47, 11–18.Schwartzman RA, Cidlowski JA, 1993: Apoptosis: the biochemistryand molecular biology of programmed cell death.Endocr Rev 14, 133–151.Thome M, Tschopp J, 2001: Regulation of lymphocyteproliferation and death by FLIP. Nat Rev Immunol 1, 50–58.Thome M, Schneider P, Hofmann K, Fickenscher H, Me<strong>in</strong>l E,Neipel F, Mattmann C, Burns K, Bodmer JL, Schroter M,Scaffidi C, Krammer PH, Peter ME, Tschopp J, 1997: ViralFLICE-<strong>in</strong>hibitory prote<strong>in</strong>s (FLIPs) prevent apoptosis<strong>in</strong>duced by death receptors. Nature 386, 517–521.Tilly JL, Kowalski KI, Johnson AL, Hsueh AJ, 1991:Involvement of apoptosis <strong>in</strong> ovarian follicular atresia andpostovulatory regression. Endocr<strong>in</strong>ology 129, 2799–2801.Wada S, Manabe N, Inoue N, Nakayama M, Matsui T,Miyamoto H, 2002: TRAIL-decoy receptor-1 disappears <strong>in</strong>granulosa cells of atretic follicles <strong>in</strong> porc<strong>in</strong>e ovaries. JReprod Dev 48, 167–173.Wallach D, Varfolomeev EE, Mal<strong>in</strong><strong>in</strong> NL, Goltsev YV,Kovalenko AV, Bold<strong>in</strong> MP, 1999: Tumor necrosis factorreceptor and Fas signal<strong>in</strong>g mechanisms. Ann Rev Immunol17, 331–367.Author’s address (for correspondence): N Manabe, Animal ResourceScience Center, University of Tokyo, Kasama 319-0206, Japan.E-mail: amanabe@mail.ecc.u-tokyo.ac.jpConflict of <strong>in</strong>terest: The authors declare no conflict of <strong>in</strong>terests.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 273–279 (2008); doi: 10.1111/j.1439-0531.2008.01174.xISSN 0936-6768Epigenetic Programm<strong>in</strong>g of Porc<strong>in</strong>e Endometrial Function and the Lactocr<strong>in</strong>eHypothesisFF Bartol 1 , AA Wiley 1 and CA Bagnell 21 Departments of Animal Sciences and Anatomy, Physiology and Pharmacology, Cellular and Molecular Biosciences Program, Auburn University,Auburn, AL, USA; 2 Department of Animal Sciences, Endocr<strong>in</strong>ology and Animal Biosciences Program, Rutgers University, New Brunswick, NJ, USAContentsEpigenetic programs controll<strong>in</strong>g development of the femalereproductive tract (FRT) are <strong>in</strong>fluenced by the effects ofnaturally occurr<strong>in</strong>g bioactive agents on patterns of geneexpression <strong>in</strong> FRT tissues dur<strong>in</strong>g organizationally criticalperiods of foetal and per<strong>in</strong>atal life. Aberrations <strong>in</strong> suchimportant cellular and molecular events, as may occur withexposure to natural or manmade steroid or peptide receptormodulat<strong>in</strong>gagents, disrupt the developmental program andcan change the developmental trajectory of FRT tissues,<strong>in</strong>clud<strong>in</strong>g the endometrium, with last<strong>in</strong>g consequences. In thepig, as <strong>in</strong> other mammals, maternal programm<strong>in</strong>g of FRTdevelopment beg<strong>in</strong>s pre-natally and is completed postnatally,when maternal effects on development can becommunicated via signals transmitted <strong>in</strong> milk. Studies<strong>in</strong>volv<strong>in</strong>g relax<strong>in</strong> (RLX), a prototypic milk-borne morphoregulatoryfactor (MbF), serve as the basis for ongo<strong>in</strong>gefforts to identify maternal programm<strong>in</strong>g events that affectuter<strong>in</strong>e and cervical tissues <strong>in</strong> the neonatal pig. Data supportthe lactocr<strong>in</strong>e hypothesis for delivery of MbFs to neonatesas a specific consequence of nurs<strong>in</strong>g. Components of amaternally driven lactocr<strong>in</strong>e mechanism for RLX-mediatedsignall<strong>in</strong>g <strong>in</strong> neonatal FRT tissues, <strong>in</strong>clud<strong>in</strong>g evidence thatmilk-borne RLX is delivered <strong>in</strong>to the neonatal circulationwhere it can act on RLX receptor (RXFP1) -positiveneonatal tissues to affect their development, are <strong>in</strong> place <strong>in</strong>the pig. The fact that all newborn mammals dr<strong>in</strong>k milkextends the timeframe of maternal <strong>in</strong>fluence on neonataldevelopment across many species. Thus, lactocr<strong>in</strong>e transmissionof milk-borne developmental signals is an element ofthe maternal epigenetic programm<strong>in</strong>g equation that deservesfurther study.IntroductionIn the pig (Sus scrofa domesticus), as <strong>in</strong> other mammals,development of the female reproductive tract (FRT)beg<strong>in</strong>s pre-natally but is completed post-natally (Y<strong>in</strong>and Ma 2005; Bartol et al. 2006). Structural pattern<strong>in</strong>g(morphogenesis) and functional programm<strong>in</strong>g (cytodifferentiation)of epithelial-mesenchymal tissues derivedfrom the Mu¨llerian ducts, <strong>in</strong>clud<strong>in</strong>g the oviducts, uterus,cervix and anterior vag<strong>in</strong>a, are coupled processessupported by the progressive generation of <strong>in</strong>creas<strong>in</strong>glycomplex and specific cellular relationships and <strong>in</strong>teractions(Gray et al. 2001; Y<strong>in</strong> and Ma 2005). Over time,these <strong>in</strong>teractions drive the evolution of organizationallycritical, temporally and spatially unique morphoregulatorygene expression doma<strong>in</strong>s that def<strong>in</strong>e micro-environmentalconditions which, <strong>in</strong> turn, direct and specifycell fate, dictate patterns of development, and determ<strong>in</strong>ecell and tissue identity and functionality. For a giventissue, this complex sequence of events def<strong>in</strong>es thedevelopmental program and, <strong>in</strong> so do<strong>in</strong>g, establishes adevelopmental trajectory for cells and tissues that willeventually dictate phenotype (Burggren 1999).Genetic potential for developmental success is def<strong>in</strong>edat conception. Thereafter, if development proceedsalong a normal course through embryonic, foetal andper<strong>in</strong>atal life, a normal phenotypic trajectory is establishedand an optimal phenotypic outcome is realized(Fig. 1). However, developmental programs can bedisrupted by a host of epigenetic factors of both biotic(physiological, endocr<strong>in</strong>ological, metabolic) and abiotic(anthropogenic ⁄ macro-environmental) orig<strong>in</strong> (Burggren1999; Bartol 2002). Disruption of the developmentalprogram dur<strong>in</strong>g organizationally critical periods, whencells and tissues are uniquely sensitive to aberrantstimuli, alters micro-environmental conditions requiredfor normal development. Divergence from the normaldevelopmental program can be sufficient to alter thedevelopmental trajectory and, ultimately, the phenotypeof a cell, tissue or organ (Fig. 1) (Bartol et al. 1999;Bartol 2002; Nathanielsz 2006). A wealth of evidence<strong>in</strong>dicates that exposure to organizationally disruptiveconditions dur<strong>in</strong>g per<strong>in</strong>atal life can have last<strong>in</strong>g effectson both the form and function of FRT tissues (Y<strong>in</strong> andMa 2005; Bartol et al. 2006). Factors required toestablish a normal developmental program and to<strong>in</strong>sure an optimal developmental trajectory for FRTtissues rema<strong>in</strong> <strong>in</strong>completely def<strong>in</strong>ed.The term epigenetics, co<strong>in</strong>ed by Wadd<strong>in</strong>gton <strong>in</strong> the1940s, was orig<strong>in</strong>ally def<strong>in</strong>ed to describe the <strong>in</strong>teractionsof genes with their environment which give rise tophenotype (Wadd<strong>in</strong>gton 1940). This def<strong>in</strong>ition hasevolved to describe the study of changes <strong>in</strong> geneexpression that occur without a change <strong>in</strong> DNAsequence (Jirtle and Sk<strong>in</strong>ner 2007). The term is nowused broadly <strong>in</strong> reference to the ‘<strong>in</strong>heritance of <strong>in</strong>formationbased on gene expression levels rather than ongene sequence’ (Junien 2006). Thus, factors that affectdevelopmentally critical gene expression events, particularlythose with last<strong>in</strong>g and potentially heritableconsequences, constitute epigenetic elements of thedevelopmental program (Jirtle and Sk<strong>in</strong>ner 2007).‘Maternal programm<strong>in</strong>g’ refers to maternally drivenepigenetic events with the potential to affect both thedevelopmental program and trajectory of embryonic,foetal and ⁄ or per<strong>in</strong>atal tissues (Szyf et al. 2005; Nathanielsz2006; Wells 2007). It is likely that maternaleffects on development do not end with parturition, butextend <strong>in</strong>to the early neonatal period. Dur<strong>in</strong>g this time,colostrum (first milk) serves as the conduit for communicationof organizationally important developmentalsignals from mother to offspr<strong>in</strong>g.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


274 FF Bartol, AA Wiley and CA BagnellCourse of developmentDevelopmentalprogram affectedDCriticalPeriodTimeDevelopmentaltrajectory affectedFig. 1. Potential effects of an organizationally disruptive event on thedevelopmental program and trajectory. Depicted is a scheme <strong>in</strong> whicha disruptive event (D) is encountered dur<strong>in</strong>g a critical organizationalperiod early <strong>in</strong> development. The normal developmental program isdepicted by the solid l<strong>in</strong>e, with a range of developmental eventsassociated with normal outcomes <strong>in</strong>dicated by the hatched area.Consequences of disruption may <strong>in</strong>clude: (1) alteration of the normaldevelopmental program without effects on the developmental trajectory(dashed l<strong>in</strong>e); or (2) alteration of both the developmental programand trajectory with long-term consequences for adult phenotype(dotted l<strong>in</strong>e)Here, data are reviewed to show that the basic elementsof a signall<strong>in</strong>g system activated by a milk-borne morphoregulatorygrowth factor (MbF) exemplified byrelax<strong>in</strong> (RLX) are present <strong>in</strong> the pig at birth. Collectively,data support the ‘lactocr<strong>in</strong>e hypothesis’ for maternalprogramm<strong>in</strong>g of FRT tissues whereby milk-borne RLX,absorbed <strong>in</strong>to the neonatal circulation dur<strong>in</strong>g the firstdays of post-natal life, acts directly through its ownreceptor to <strong>in</strong>duce and ⁄ or support oestrogen receptor-a(ER) expression and activation <strong>in</strong> a manner necessary to<strong>in</strong>sure normal FRT programm<strong>in</strong>g and an optimal developmentaltrajectory for FRT tissues <strong>in</strong> the pig.Reproductive Tract Development andConsequences of Developmental DisruptionClassic data <strong>in</strong>dicat<strong>in</strong>g that pre-natal exposure ofhuman foetuses to the synthetic oestrogen diethylstilbestrolalters the developmental program of FRT tissuesand sets the stage for cervicovag<strong>in</strong>al cancer and othercomplications (Herbst et al. 1979; Iguchi and Sato 2000)drew attention to the fact that disruption of hormonesensitivedevelopmental events can have serious consequencesfor reproductive performance and health.Studies catalysed by these observations provided important<strong>in</strong>sights <strong>in</strong>to the roles of the steroid hormonesuperfamily of receptors and related ligands <strong>in</strong> normaland aberrant FRT programm<strong>in</strong>g. For the uterus, lossof-functionstudies showed that ER expression isrequired for normal growth, while both the progesteronereceptor (PR) and ER are required for normal uter<strong>in</strong>efunction (Conneely et al. 2001; Emmen and Korach2003). Neither ER nor PR expression is necessary tosupport primary uter<strong>in</strong>e pattern<strong>in</strong>g events <strong>in</strong> pre- orper<strong>in</strong>atal life. However, aberrant activation of these andrelated receptor systems dur<strong>in</strong>g critical organizationalperiods can affect the developmental program withsignificant functional consequences <strong>in</strong> both rodents andungulates (Bartol et al. 1999; Gray et al. 2000; Iguchiand Sato 2000; Huang et al. 2005). In laboratoryanimals, per<strong>in</strong>atal oestrogen exposure produced lesions<strong>in</strong> adult uteri that <strong>in</strong>cluded altered steroid receptorconcentrations and responsiveness; changes <strong>in</strong> oestrogenmetabolism and prote<strong>in</strong> synthesis; persistent <strong>in</strong>ductionor deregulation of gene expression; deregulation ofprotooncogene expression affect<strong>in</strong>g epithelial proliferationand apoptosis; a myriad of structural lesions and,classically, general hypoplasia of FRT tissues andorgans (Iguchi and Sato 2000; Huang et al. 2005).Complementary data <strong>in</strong>volv<strong>in</strong>g ungulate models <strong>in</strong>dicateclearly that adult uter<strong>in</strong>e phenotype can beprogrammed by targeted disruption of both steroidand peptide hormone-sensitive post-natal organizationalevents (Bartol et al. 1999, 2006; Carpenter et al. 2003;Tarleton et al. 2003).Events associated with normal and, particularly,oestrogen-<strong>in</strong>duced disruption of neonatal uter<strong>in</strong>e development<strong>in</strong> the pig have been reviewed (Bartol et al.1993; Yan et al. 2006b) and concepts related to maternaland ⁄ or environmental programm<strong>in</strong>g of reproductive⁄ somatic tissues <strong>in</strong> the pig are illustrated <strong>in</strong> Fig. 2.Data for the porc<strong>in</strong>e uterus show that disruption ofFRT development dur<strong>in</strong>g the early post-natal period hasboth morphological and functional consequences. Radialpattern<strong>in</strong>g of the porc<strong>in</strong>e uter<strong>in</strong>e wall is <strong>in</strong>completeat birth (PND 0), when tissues are ER-negative (Bartolet al. 1993; Tarleton et al. 1998). Early post-natal eventsassociated with development of these tissues <strong>in</strong>cludedifferentiation of glandular epithelium (GE) from lum<strong>in</strong>alepithelium (LE), proliferation of nascent endometrialglands, and onset of regular ER expression <strong>in</strong>stroma and GE, but not <strong>in</strong> LE (Spencer et al. 1993a;Yan et al. 2006a; b; Masters et al. 2007). When adm<strong>in</strong>isteredfor 14 days from birth, the ER antagonist ICI182,780 retards development of the uter<strong>in</strong>e wall and<strong>in</strong>hibits gland genesis <strong>in</strong> the neonatal endometrium(Tarleton et al. 1999). Thus, the ER is both a marker ofGE differentiation and a mediator of radial pattern<strong>in</strong>g<strong>in</strong> the neonatal porc<strong>in</strong>e uterus.Aberrant activation of the neonatal ER system byadm<strong>in</strong>istration of oestradiol valerate (EV) to gilts fromPND 0–13, while acutely uterotrophic (Spencer et al.1993b), alters patterns of morphoregulatory geneexpression <strong>in</strong> the develop<strong>in</strong>g endometrium (Bartol et al.2006) and is ultimately both antiuterotrophic andantiembryotrophic (Bartol et al. 1993; Tarleton et al.2001, 2003). The hypoplastic, neonatally EV-exposedadult porc<strong>in</strong>e uterus does not respond normally tosignals associated with the periattachment stage of earlypregnancy. Compared to unexposed controls on gestationalday 12 (GD 12), uter<strong>in</strong>e lum<strong>in</strong>al fluid prote<strong>in</strong>content was reduced, uter<strong>in</strong>e growth responses to earlypregnancy were <strong>in</strong>hibited, and endometrial gene expressionpatterns were altered <strong>in</strong> neonatally EV-exposed,pregnant adult gilts (Tarleton et al. 2003; Chen et al.2007). Treatment effects on the adult endometrialproteome for GD 12 were also pronounced (Bartolet al. 2006), and embryo survival <strong>in</strong> neonatallyEV-exposed gilts was reduced by 22% when assessedon GD 45 (Bartol et al. 1993). Thus, disruption ofoestrogen-sensitive, ER-dependent developmentalevents between birth and PND 14 has marked andlast<strong>in</strong>g effects on uter<strong>in</strong>e form and function <strong>in</strong> the pig. ItÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Lactocr<strong>in</strong>e Programm<strong>in</strong>g of Uter<strong>in</strong>e Development 275Fig. 2. Maternal and environmental programm<strong>in</strong>g of development.Natural and anthropogenic macroenvironmental factors (oval) imp<strong>in</strong>geupon the maternal system throughout pregnancy and lactation(triangle). Pre-natally, maternal effects on foetal development areprofound and factors of macroenvironmental orig<strong>in</strong> with the potentialto affect development are communicated via the maternal system (left).Dur<strong>in</strong>g this period the uterus serves as the conduit for communicationbetween mother and (potential) offspr<strong>in</strong>g. With parturition, communicationbetween mother and offspr<strong>in</strong>g cont<strong>in</strong>ues via the mammarygland and milk (right). Post-natally, macroenvironmental factors mayaffect neonatal development directly and may also be conductedthrough (and modified <strong>in</strong>) the maternal system. The term lactocr<strong>in</strong>ewas co<strong>in</strong>ed to describe a mechanism through which bioactive milkbornefactors are delivered from mother to offspr<strong>in</strong>g as a specificconsequence of nurs<strong>in</strong>gis clear that factors with the potential to affect uter<strong>in</strong>eER expression and activation, either directly or <strong>in</strong>directly,should be expected to have significant effects ondevelopmental programm<strong>in</strong>g of these tissues.Relax<strong>in</strong> Receptor Expression and Functionality<strong>in</strong> the Neonatal Porc<strong>in</strong>e UterusWhile RLX has been studied for over 80 years (Hisaw1926), the cognate receptor for this 6000 Da peptidehormone was identified <strong>in</strong> 2002 (Hsu et al. 2002). A typeC, leuc<strong>in</strong>e-rich, G-prote<strong>in</strong> coupled receptor (LGR)orig<strong>in</strong>ally designated LGR7, the RLX receptor is nowrecognized as a member of the RLX family of peptide(RXFP) receptors and was recently re-designatedRXFP1 (Bathgate et al. 2006).LGR7 ⁄ RXFP1 is expressed by porc<strong>in</strong>e uter<strong>in</strong>e (Yanet al. 2006b) and cervical tissues from birth (Yan et al.2005). Uter<strong>in</strong>e RXFP1 expression <strong>in</strong> the pig, which ispredom<strong>in</strong>antly if not exclusively stromal, <strong>in</strong>creases fromPND 0 through PND 14. Immunohistochemical evidencesupports <strong>in</strong> situ hybridization (ISH) data show<strong>in</strong>gstromal RXFP1 sta<strong>in</strong><strong>in</strong>g similar to that reported foradult primate and human endometrium (Ivell et al.2003). Stromal expression of RXFP1 suggests that directeffects of RLX on epithelial growth and developmentare mediated through a stromally-driven paracr<strong>in</strong>emechanism (Bagnell et al. 2005). However, the fact thattrophic effects of RLX adm<strong>in</strong>istered for 2 days fromPND 12, after onset of uter<strong>in</strong>e ER expression, were<strong>in</strong>hibited by pre-treatment with ICI 182,780 suggeststhat RLX may also act <strong>in</strong>directly via crosstalk with theER system (Pillai et al. 1999; Yan et al. 2006a). Cervicalexpression of RXFP1 <strong>in</strong> the porc<strong>in</strong>e neonate is higherthan that observed for the uterus (Yan et al. 2005).However, RLX adm<strong>in</strong>istration for 2 days from birthdecreased cervical RXFP1 expression on PND 2 (Yanet al. 2005), suggest<strong>in</strong>g elements of a negative autoregulatorymechanism govern<strong>in</strong>g cervical RXFP1 expression.Prelim<strong>in</strong>ary data (Yan and Bagnell 2003; Yanet al. 2008) also <strong>in</strong>dicate that both uter<strong>in</strong>e and cervicalER expression <strong>in</strong>creased on PND 2 follow<strong>in</strong>g adm<strong>in</strong>istrationof RLX for 2 days from birth. S<strong>in</strong>ce RXFP1expression precedes ER expression <strong>in</strong> the neonataluterus, which is stimulated by exogenous RLX, subtlebut developmentally critical uterotrophic effects of RLXexposure from birth are likely to be RXFP1-specific.The more pronounced effects of RLX exposure observeddur<strong>in</strong>g the second week of neonatal life, after the onsetof ER expression, could reflect amplification of the RLXsignal through cross-talk with the ER (Pillai et al. 1999;Yan et al. 2006a). The fact that RLX can <strong>in</strong>creaseoestrogen-stimulated uter<strong>in</strong>e Hoxa10 expression (Guiet al. 1999) <strong>in</strong>dicates that RLX signall<strong>in</strong>g may also affectER-dependent events driv<strong>in</strong>g morphoregulatory geneexpression <strong>in</strong> the neonatal uterus (Bartol et al. 2006) andother RXFP1-positive, E-sensitive FRT tissues.Like oestrogen, uterotrophic effects of RLX <strong>in</strong> neonatalpigs are age-specific (Spencer et al. 1993b; Bagnellet al. 2005). When given for 2 days from birth, prior toonset of endometrial ER expression, RLX <strong>in</strong>creaseduter<strong>in</strong>e LE height, but not uter<strong>in</strong>e weight on PND 2(Yan et al. 2006a). In strik<strong>in</strong>g contrast, RLX <strong>in</strong>creasedcervical weight by PND 2 when compared with E-treated or control gilts (Yan et al. 2005). In gilts treatedfor 2 days from PND 12, after onset of uter<strong>in</strong>e ERexpression, RLX <strong>in</strong>creased both uter<strong>in</strong>e LE height anduter<strong>in</strong>e weight on PND 14 (Yan et al. 2006a). Data areconsistent with other studies show<strong>in</strong>g that RLX<strong>in</strong>creases uter<strong>in</strong>e LE height <strong>in</strong> pre-pubertal gilts (Ryanet al. 2001) and is important for ma<strong>in</strong>tenance ofreproductive epithelia <strong>in</strong> RLX-null mice (Zhao et al.2000). In RLX-deficient rats, proliferation of vag<strong>in</strong>alepithelial and stromal cells decreased while rates ofapoptosis <strong>in</strong> these cell types <strong>in</strong>creased more than 10- and3-fold relative to controls (Lee et al. 2005).Image analysis based evaluation of cell proliferation,<strong>in</strong> which proliferat<strong>in</strong>g cell nuclear antigen (PCNA)positive cells were identified <strong>in</strong> situ and PCNA labell<strong>in</strong>g<strong>in</strong>dices were determ<strong>in</strong>ed, was used to assess effects of ageat 3-day <strong>in</strong>tervals from birth through PND 15 onpatterns of epithelial proliferation <strong>in</strong> the neonatalporc<strong>in</strong>e endometrium (Masters et al. 2007). Overall,PCNA labell<strong>in</strong>g <strong>in</strong>dex was greater <strong>in</strong> GE than <strong>in</strong> LE andwas affected by neonatal age <strong>in</strong> both cell types. Results<strong>in</strong>dicated that the developmental transition from amorphogenetically <strong>in</strong>active ‘<strong>in</strong>fantile’ endometrial conditionat birth to a morphogenetically active ‘proliferative’state (Spencer et al. 1993a) has occurred by PND3 <strong>in</strong> the pig as marked by a dramatic <strong>in</strong>crease <strong>in</strong>epithelial PCNA labell<strong>in</strong>g <strong>in</strong>dex (Masters et al. 2007).This labell<strong>in</strong>g <strong>in</strong>dex rema<strong>in</strong>ed high through PND 6 <strong>in</strong>both LE and GE, decl<strong>in</strong>ed thereafter <strong>in</strong> both cellcompartments, and <strong>in</strong>creased to PND 15 only <strong>in</strong> GE.Furthermore, adm<strong>in</strong>istration of oestrogen or RLX <strong>in</strong>week two <strong>in</strong>duced LE and GE proliferation on PND 14Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Lactocr<strong>in</strong>e Programm<strong>in</strong>g of Uter<strong>in</strong>e Development 277Fig. 3. Schematic of porc<strong>in</strong>e endometrial development between birth (neonatal day 0) and post-natal day 15. At birth (top left) endometrialglands are absent and the porc<strong>in</strong>e endometrium is ERa negative and RXFP1 positive. Stromal RXFP1 expression suggests that effects of RLX ondevelop<strong>in</strong>g epithelium may <strong>in</strong>volve unidentified RLX-<strong>in</strong>duced factors of stromal orig<strong>in</strong> (relaximed<strong>in</strong>s). Stromal–epithelial <strong>in</strong>teractions (doubleheadedarrows) and the local microenvironmental conditions they affect are essential for uter<strong>in</strong>e developmental success. Milk RLX concentrationsare highest with<strong>in</strong> 24h of birth and decl<strong>in</strong>e l<strong>in</strong>early thereafter. RLX is present (200 pg ⁄ ml) <strong>in</strong> the peripheral circulation of nurs<strong>in</strong>g piglets andrema<strong>in</strong>s so through the second day of neonatal life. Other bioactive milk-borne factors (MbFs) may also be presented to these tissues. Onset ofuter<strong>in</strong>e gland genesis is marked by ERa expression <strong>in</strong> nascent glandular epithelium (GE) and <strong>in</strong>creased proliferative activity <strong>in</strong> GE, evident byPND 3. Data suggest a maternally lactocr<strong>in</strong>e-driven system <strong>in</strong> which milk-borne RLX delivered <strong>in</strong>to the neonatal circulation as a consequence ofnurs<strong>in</strong>g, acts through its cognate receptor system (RXFP1) to stimulate endometrial ERa expression, an event required for normal uter<strong>in</strong>e glandgenesis and endometrial development <strong>in</strong> the neonatal pig. The potential <strong>in</strong>fluence of lactocr<strong>in</strong>e-act<strong>in</strong>g factors wanes as the <strong>in</strong>fluence of ERmediatedevents <strong>in</strong>creases dur<strong>in</strong>g the first weeks of neonatal lifemediated programm<strong>in</strong>g of the porc<strong>in</strong>e FRT dur<strong>in</strong>g theearly post-natal period are <strong>in</strong> place. Included is evidencethat: (1) the w<strong>in</strong>dow of opportunity for post-natalmaternal programm<strong>in</strong>g of FRT tissues is open dur<strong>in</strong>gthe first 3 days of neonatal life as a critical morphoregulatorygene expression axis is develop<strong>in</strong>g; (2) neonataluter<strong>in</strong>e and cervical tissues are RXFP1-positive at birth,prior to onset of uter<strong>in</strong>e ER expression; (3) exogenousporc<strong>in</strong>e RLX has trophic effects on uter<strong>in</strong>e and cervicaltissues that are evident by PND 2 when exposure beg<strong>in</strong>sat birth; (4) RLX adm<strong>in</strong>istered from birth <strong>in</strong>duces ERexpression <strong>in</strong> develop<strong>in</strong>g uter<strong>in</strong>e and cervical tissues byPND 2; (5) a natural source of RLX is found <strong>in</strong> sow’smilk, where RLX concentrations are highest with<strong>in</strong> 48 hof parturition; and (6) RLX is not detectable <strong>in</strong> thesystemic circulation of newborn pigs prior to nurs<strong>in</strong>g or<strong>in</strong> neonatal pigs fed hormone-free milk replacer, but isdetectable at serum concentrations approach<strong>in</strong>g200 pg ⁄ ml on PND 0–1 <strong>in</strong> neonatal pigs allowed tonurse. Us<strong>in</strong>g uter<strong>in</strong>e data as a reference, observationssuggest that RLX, delivered via a lactocr<strong>in</strong>e mechanism,is available to support levels of ER expression requiredto <strong>in</strong>sure that critical oestrogen-sensitive, ER-dependentelements of the post-natal organizational program areoptimized (Bartol et al. 2006). These relationships aresummarized <strong>in</strong> Fig. 3.Relax<strong>in</strong> also has diverse actions <strong>in</strong> non-reproductivetissues (Sherwood 2004). RXFP1 transcripts have beenidentified <strong>in</strong> immature rat (Hsu et al. 2000), adult mouse(Kamat et al. 2004) and human (Sudo et al. 2003)tissues <strong>in</strong> males and females <strong>in</strong>clud<strong>in</strong>g the bra<strong>in</strong>, heart,kidney, lung and liver. Thus, milk-borne RLX maysupport development of other vital tissues and organsdur<strong>in</strong>g neonatal life <strong>in</strong> animals of both sexes. In thislight, the lactocr<strong>in</strong>e-driven, RLX-dependent neonataltissue programm<strong>in</strong>g mechanism envisioned here for theporc<strong>in</strong>e FRT may have broader developmental andsomatotrophic implications. Given that RLX is not theonly bioactive peptide found <strong>in</strong> milk (Donovan andOdle 1994; Meisel 2005), it is important to consider thatother MbFs may be delivered to the neonatal system viaa lactocr<strong>in</strong>e route. Nevertheless, the extent to whichmaternally-derived MbFs contribute to establishment ofan optimal organizational program and affect thedevelopmental trajectory of porc<strong>in</strong>e FRT tissues rema<strong>in</strong>sto be determ<strong>in</strong>ed. This is a central topic ofongo<strong>in</strong>g research <strong>in</strong> our laboratories.AcknowledgementsContributions of Dr Wenbo Yan, Ms Bethany Crean-Harris and MrJoseph Chen are gratefully acknowledged. This work was supported byNational Research Initiative Competitive Grants 2003-35203-13572and 2007-35203-18098 from the USDA-CSREES to FFB and CAB,NSF EPS-0447675, and the NJ and AL Agricultural ExperimentStations.ReferencesArmitage JA, Taylor PD, Poston L, 2005: Experimentalmodels of developmental programm<strong>in</strong>g: consequences ofexposure to an energy rich diet dur<strong>in</strong>g development. JPhysiol 565, 3–8.Baer C, Bilkei G, 2005: Ultrasonographic and gross pathologicalf<strong>in</strong>d<strong>in</strong>gs <strong>in</strong> the mammary glands of weaned sowshav<strong>in</strong>g suffered recidiv<strong>in</strong>g mastitis metritis agalactia. ReprodDomest Anim 40, 544–547.Bagnell CA, Yan W, Wiley AA, Bartol FF, 2005: Effects ofrelax<strong>in</strong> on neonatal porc<strong>in</strong>e uter<strong>in</strong>e growth and development.Ann N Y Acad Sci 1041, 248–255.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


278 FF Bartol, AA Wiley and CA BagnellBartol FF, 2002: The female reproductive system: targets fortoxicants. Proc Soc Theriol Ann Conf Sympos 427–436.Bartol FF, Wiley AA, Spencer TE, Vallet JL, Christenson RK,1993: Early uter<strong>in</strong>e development <strong>in</strong> pigs. J Reprod Fertil48(Suppl), 99–116.Bartol FF, Wiley AA, Floyd JG, Ott TL, Bazer FW, GrayCA, Spencer TE, 1999: Uter<strong>in</strong>e differentiation as afoundation for subsequent fertility. J Reprod Fertil 54(Suppl),287–302.Bartol FF, Wiley AA, Bagnell CA2006: Uter<strong>in</strong>e developmentand endometrial programm<strong>in</strong>g. In: Asworth CJ, Krael<strong>in</strong>gRR(eds), Control of Pig <strong>Reproduction</strong> VII. Nott<strong>in</strong>ghamUniversity Press, UK, pp. 113–130.Bathgate RA, Ivell R, Sanborn BM, Sherwood OD, SummersRJ, 2006: International union of pharmacology vii: recommendationsfor the nomenclature of receptors for relax<strong>in</strong>family peptides. Pharmacol Rev 58, 7–31.Ben-Shlomo Y, Holly J, McCarthy A, Savage P, Davies D,Davey Smith G, 2005: Prenatal and postnatal milk supplementationand adult <strong>in</strong>sul<strong>in</strong>-like growth factor I: Long-termfollow-up of a randomized controlled trial. Cancer EpidemiolBiomarkers Prev 14, 1336–1339.Burggren W, 1999: Genetic, environmental and maternal<strong>in</strong>fluences on embryonic cardiac rhythms. Comp BiochemPhys A 124, 423–427.Burr<strong>in</strong> DG, Davis TA, Ebner S, Schoknecht PA, Fiorotto ML,Reeds PJ, 1997: Colostrum enhances the nutritional stimulationof vital organ prote<strong>in</strong> synthesis <strong>in</strong> neonatal pigs.J Nutr 127, 1284–1289.Carpenter KD, Gray CA, Noel S, Gertler A, Bazer FW,Spencer TE, 2003: Prolact<strong>in</strong> regulation of neonatal ov<strong>in</strong>euter<strong>in</strong>e gland morphogenesis. Endocr<strong>in</strong>ology 144, 110–120.Chen J, Frankshun A-L, Wiley A, Ho TY, Bartol FF, BagnellCA2007: Estrogen exposure from birth affects uter<strong>in</strong>e geneexpression <strong>in</strong> neonatal gilts that persists <strong>in</strong> pregnant adults.Biol Reprod Special Issue, 152 .Coalson JA, Lecce JG, 1973: Influence of nurs<strong>in</strong>g <strong>in</strong>tervals onchanges <strong>in</strong> serum prote<strong>in</strong>s (immunoglobul<strong>in</strong>s) <strong>in</strong> neonatalpigs. J Anim Sci 36, 381–385.Conneely OM, Mulac-Jericevic B, Lydon JP, De Mayo FJ,2001: Reproductive functions of the progesterone receptorisoforms: lessons from knock-out mice. Mol Cell Endocr<strong>in</strong>ol179, 97–103.Donovan SM, Odle J, 1994: Growth factors <strong>in</strong> milk asmediators of <strong>in</strong>fant development. Annu Rev Nutr 14, 147–167.Eddie LW, Sutton B, Fitzgerald S, Bell RJ, Johnston PD,Tregear GW, 1989: Relax<strong>in</strong> <strong>in</strong> paired samples of serum andmilk from women after term and preterm delivery. Am JObstet Gynecol 161, 970–973.Emmen JM, Korach KS, 2003: Estrogen receptor knockoutmice: phenotypes <strong>in</strong> the female reproductive tract. GynecolEndocr<strong>in</strong>ol 17, 169–176.Goldsmith LT, Lust G, Ste<strong>in</strong>etz BG, 1994: Transmission ofrelax<strong>in</strong> from lactat<strong>in</strong>g bitches to their offspr<strong>in</strong>g via suckl<strong>in</strong>g.Biol Reprod 50, 258–265.Gray CA, Taylor KM, Bazer FW, Spencer TE, 2000:Mechanisms regulat<strong>in</strong>g norgestomet <strong>in</strong>hibition of endometrialgland morphogenesis <strong>in</strong> the neonatal ov<strong>in</strong>e uterus. MolReprod Devel 57, 67–78.Gray CA, Bartol FF, Tarleton BJ, Wiley AA, Johnson GA,Bazer FW, Spencer TE, 2001: Developmental biology ofuter<strong>in</strong>e glands. Biol Reprod 65, 1311–1323.Grosvenor CE, Picciano MF, Baumrucker CR, 1993: Hormonesand growth factors <strong>in</strong> milk. Endocr<strong>in</strong>ol Rev 14, 710–728.Gui Y, Zhang J, Yuan L, Lessey BA, 1999: Regulation ofhoxa-10 and its expression <strong>in</strong> normal and abnormal endometrium.Mol Hum Reprod 5, 866–873.Herbst AL, Scully RE, Robboy SJ1979: Prenatal diethylstilbestrolexposure and human genital tract abnormalities.Natl Cancer I Monogr 25–35.Hisaw FL, 1926: Experimental relaxation of the pubicligament of the gu<strong>in</strong>ea pig. PSEBM 23, 661–663.Hsu SY, Kudo M, Chen T, Nakabayashi K, Bhalla A, van derSpek PJ, van Du<strong>in</strong> M, Hsueh AJ, 2000: The three subfamiliesof leuc<strong>in</strong>e-rich repeat-conta<strong>in</strong><strong>in</strong>g g prote<strong>in</strong>-coupledreceptors (lgr): identification of LGR6 and LGR7 and thesignal<strong>in</strong>g mechanism for LGR7. Mol Endocr<strong>in</strong>ol 14, 1257–1271.Hsu SY, Nakabayashi K, Nishi S, Kumagai J, Kudo M,Sherwood OD, Hsueh AJW, 2002: Activation of orphanreceptors by the hormone relax<strong>in</strong>. Science 295, 671–674.Huang W-W, Y<strong>in</strong> Y, Bi Q, Chiang T-C, Garner N, Vuoristo J,McLachlan JA, Ma L, 2005: Developmental diethylstilbestrolexposure alters genetic pathways of uter<strong>in</strong>e cytodifferentiation.Mol Endocr<strong>in</strong>ol 19, 669–682.Iguchi T, Sato T, 2000: Endocr<strong>in</strong>e disruption and developmentalabnormalities of female reproduction. Am Zool 40,402–411.Ivell R, Balvers M, Pohnke Y, Telgmann R, Bartsch O, Milde-Langosch K, Bamberger AM, E<strong>in</strong>spanier A, 2003: Immunoexpressionof the relax<strong>in</strong> receptor LGR7 <strong>in</strong> breast anduter<strong>in</strong>e tissues of humans and primates. Reprod BiolEndocr<strong>in</strong>ol 1, 114.Jirtle RL, Sk<strong>in</strong>ner MK, 2007: Environmental epigenomics anddisease susceptibility. Nat Rev Genet 8, 253–262.Junien C, 2006: Impact of diets and nutrients ⁄ drugs on earlyepigenetic programm<strong>in</strong>g. J Inherit Metab Dis 29, 359–365.Kamat AA, Feng S, Bogatcheva NV, Truong A, Bishop CE,Agoulnik AI, 2004: Genetic target<strong>in</strong>g of relax<strong>in</strong> and <strong>in</strong>sul<strong>in</strong>likefactor 3 receptors <strong>in</strong> mice. Endocr<strong>in</strong>ology 145, 4712–4720.Kl<strong>in</strong>g PJ, 2002: Roles of erythropoiet<strong>in</strong> <strong>in</strong> human milk. ActaPaediatr Suppl 91, 31–35.Koldovsky O, 1996: The potential physiological significance ofmilk-borne hormonally active substances for the neonate.J Mammary Gland Biol 1, 317–323.Lee H-Y, Zhao S, Fields PA, Sherwood OD, 2005: The extentto which relax<strong>in</strong> promotes proliferation and <strong>in</strong>hibits apoptosisof cervical epithelial and stromal cells is greatest dur<strong>in</strong>glate pregnancy <strong>in</strong> rats. Endocr<strong>in</strong>ology 146, 511–518.Lecce JG, 1973: Effect of dietary regimen on cessation ofuptake of macromolecules by piglet <strong>in</strong>test<strong>in</strong>al epithelium(closure) and transport to the blood. J Nutr 103, 751–756.Masters RA, Crean BD, Yan W, Moss AG, Ryan PL, WileyAA, Bagnell CA, Bartol FF, 2007: Neonatal porc<strong>in</strong>eendometrial development and epithelial proliferation affectedby age and exposure to estrogen and relax<strong>in</strong>. DomestAnim Endocr<strong>in</strong>ol 33, 335–346.Meisel H, 2005: Biochemical properties of peptides encrypted<strong>in</strong> bov<strong>in</strong>e milk prote<strong>in</strong>s. Curr Med Chem 12, 1905–1919.Nathanielsz PW, 2006: Animal models that elucidate basicpr<strong>in</strong>ciples of the developmental orig<strong>in</strong>s of adult diseases.ILAR J 47, 73–82.Nusser KD, Frawley S, 1997: Depriv<strong>in</strong>g neonatal rats of milkfrom early lactation has long-term consequences on mammotropedevelopment. Endocr 7, 319–323.O’Byrne EM, Ste<strong>in</strong>etz BG, 1976: Radioimmunoassay (RIA) ofrelax<strong>in</strong> <strong>in</strong> sera of various species us<strong>in</strong>g an antiserum toporc<strong>in</strong>e relax<strong>in</strong>. PSEBM 152, 272–276.Pacha J, 2000: Development of <strong>in</strong>test<strong>in</strong>al transport function <strong>in</strong>mammals. Physiol Rev 80, 1633–1667.Pierce AE, Risdall PC, Shaw B, 1964: Absorption of orallyadm<strong>in</strong>istered <strong>in</strong>sul<strong>in</strong> by the newly born calf. J Physiol 171,203–215.Pillai SB, Rockwell LC, Sherwood OD, Koos RD, 1999: Relax<strong>in</strong>stimulates uter<strong>in</strong>e edema via activation of estrogen receptors:Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Lactocr<strong>in</strong>e Programm<strong>in</strong>g of Uter<strong>in</strong>e Development 279blockade of its effects us<strong>in</strong>g ici 182,780, a specific estrogenreceptor antagonist. Endocr<strong>in</strong>ology 140, 2426–2429.Ryan PL, Baum DL, Lenhart JA, Ohleth KM, Bagnell CA,2001: Expression of uter<strong>in</strong>e and cervical epithelial cadher<strong>in</strong>dur<strong>in</strong>g relax<strong>in</strong>-<strong>in</strong>duced growth <strong>in</strong> pigs. <strong>Reproduction</strong> 122,929–937.Sherwood OD, 2004: Relax<strong>in</strong>’s physiological roles and otherdiverse actions. Endocr<strong>in</strong>ol Rev 25, 205–234.Spencer TE, Bartol FF, Wiley AA, Coleman DA, Wolfe DF,1993a: Neonatal porc<strong>in</strong>e endometrial development <strong>in</strong>volvescoord<strong>in</strong>ated changes <strong>in</strong> DNA synthesis, glycosam<strong>in</strong>oglycandistribution, and 3h-glucosam<strong>in</strong>e label<strong>in</strong>g. Biol Reprod 48,729–740.Spencer TE, Wiley AA, Bartol FF, 1993b: Neonatal age andperiod of estrogen exposure affect porc<strong>in</strong>e uter<strong>in</strong>e growth,morphogenesis, and prote<strong>in</strong>-synthesis. Biol Reprod 48, 741–751.Sudo S, Kumagai J, Nishi S, Layfield S, Ferraro T, BathgateRA, Hsueh AJ, 2003: H3 relax<strong>in</strong> is a specific ligand for LGR7and activates the receptor by <strong>in</strong>teract<strong>in</strong>g with both theectodoma<strong>in</strong> and the exoloop 2. J Biol Chem 278, 7855–7862.Szyf M, Weaver ICG, Champagne FA, Diorio J, Meaney MJ,2005: Maternal programm<strong>in</strong>g of steroid receptor expressionand phenotype through DNA methylation <strong>in</strong> the rat. FrontNeuroendocr<strong>in</strong>ol 26, 139–162.Tarleton BJ, Wiley AA, Spencer TE, Moss AG, Bartol FF,1998: Ovary-<strong>in</strong>dependent estrogen receptor expression <strong>in</strong>neonatal porc<strong>in</strong>e endometrium. Biol Reprod 58, 1009–1019.Tarleton BJ, Wiley AA, Bartol FF, 1999: Endometrialdevelopment and adenogenesis <strong>in</strong> the neonatal pig: effectsof estradiol valerate and the antiestrogen ici 182,780. BiolReprod 61, 253–263.Tarleton BJ, Wiley AA, Bartol FF, 2001: Neonatal estradiolexposure alters uter<strong>in</strong>e morphology and endometrial transcriptionalactivity <strong>in</strong> prepubertal gilts. Domest AnimEndocr<strong>in</strong>ol 21, 111–125.Tarleton BJ, Braden TD, Wiley AA, Bartol FF, 2003:Estrogen-<strong>in</strong>duced disruption of neonatal porc<strong>in</strong>e uter<strong>in</strong>edevelopment alters adult uter<strong>in</strong>e function. Biol Reprod 68,1387–1393.Tashima LS, Mazoujian G, Bryant-Greenwood GD, 1994:Human relax<strong>in</strong>s <strong>in</strong> normal, benign and neoplastic breasttissue. J Mol Endocr<strong>in</strong>ol 12, 351–364.Wadd<strong>in</strong>gton CH1940: Organisers and Genes. CambridgeUniversity Press, Cambridge, UK.Wan Y, Saghatelian A, Chong L-W, Zhang C-L, Cravatt BF,Evans RM, 2007: Maternal PPAR gamma protects nurs<strong>in</strong>gneonates by suppress<strong>in</strong>g the production of <strong>in</strong>flammatorymilk. Gene Dev 21, 1895–1908.Wells JCK, 2007: The thrifty phenotype as an adaptivematernal effect. Biol Rev Camb Phil Soc 82, 143–172.Yan W, Bagnell CA, 2003: Effects of relax<strong>in</strong> on estrogenreceptor and vascular endothelial growth factor expression<strong>in</strong> the cervix and vag<strong>in</strong>a of neonatal pigs. Proc Soc Theriol23.Yan W, Wiley AA, Bartol FF, Bagnell CA, 2005: Tissuespecificeffects of relax<strong>in</strong> on the reproductive tract ofneonatal gilts. Ann N Y Acad Sci 1041, 132–135.Yan W, Ryan PL, Bartol FF, Bagnell CA, 2006a: Uterotrophiceffects of relax<strong>in</strong> related to age and estrogen receptoractivation <strong>in</strong> neonatal pigs. <strong>Reproduction</strong> 131, 943–950.Yan W, Wiley AA, Bathgate RAD, Frankshun A-L, Lasano S,Crean BD, Ste<strong>in</strong>etz BG, Bagnell CA, Bartol FF, 2006b:Expression of LGR7 and LGR8 by neonatal porc<strong>in</strong>e uter<strong>in</strong>etissues and transmission of milk-borne relax<strong>in</strong> <strong>in</strong>to theneonatal circulation by suckl<strong>in</strong>g. Endocr<strong>in</strong>ology 147, 4303–4310.Yan W, Chen J, Wiley AA, Crean-Harris BD, Bartol FF,Bagnell CA, 2008: Relax<strong>in</strong> (RLX) and estrogen affectestrogen receptor a, vascular endothelial growth factor,and RLX receptor expression <strong>in</strong> the neonatal porc<strong>in</strong>e uterusand cervix. <strong>Reproduction</strong> 135, 705–712.Y<strong>in</strong> Y, Ma L, 2005: Development of the mammalian femalereproductive tract. J Biochem 137, 677–683.Zhao L, Samuel CS, Tregear GW, Beck F, W<strong>in</strong>tour EM, 2000:Collagen studies <strong>in</strong> late pregnant relax<strong>in</strong> null mice. BiolReprod 63, 697–703.Author’s address (for correspondence): FF Bartol, Departments ofAnimal Sciences and Anatomy, Physiology and Pharmacology,Cellular and Molecular Biosciences Program, Auburn University,Auburn, AL 36849, USA. E-mail: bartoff@auburn.eduConflict of <strong>in</strong>terest: The authors declare no conflict of <strong>in</strong>terests.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 280–287 (2008); doi: 10.1111/j.1439-0531.2008.01175.xISSN 0936-6768Endocr<strong>in</strong>e Regulation of the Establishment of Spermatogenesis <strong>in</strong> PigsKC Caires, JA Schmidt, AP Oliver, J de Avila and DJ McLeanWash<strong>in</strong>gton State University, Pullman, Wash<strong>in</strong>gton, USAContentsSomatic and germ cell maturation precedes the start ofspermatogenesis and is coord<strong>in</strong>ated, so efficient spermatogenesiswill occur <strong>in</strong> the adults. The present study was conductedto evaluate endocr<strong>in</strong>e regulation of germ and somatic cellhomeostasis <strong>in</strong> the neonatal boar testis associated with theestablishment of spermatogenesis. Testis tissue obta<strong>in</strong>ed from3-, 5-, 7- and 14-day-old piglets were ectopically xenograftedonto castrated, immunodeficient nude mice. Grafts wereremoved 22 weeks later and evaluated for growth and theestablishment of spermatogenesis. Recipient mouse testosteronebiosynthesis and follicle-stimulat<strong>in</strong>g hormone (FSH)concentrations were also assayed. Testis tissue graft growthwas significantly greater <strong>in</strong> testis grafts from 3-day donortissue when compared to all other ages; 5-, 7- and 14-day-olddonor tissue weights were not significantly different atremoval. Follicle-stimulat<strong>in</strong>g hormone concentrations <strong>in</strong>recipient mice support<strong>in</strong>g testis grafts from 5-, 7- and14-day-old donor tissues did not differ and were similar tonormal physiological levels <strong>in</strong> age-matched, <strong>in</strong>tact nude mice.Serum FSH levels were significantly lower <strong>in</strong> recipient micesupport<strong>in</strong>g testis grafts from 3-day-old donor tissue. Radioimmunoassayand biological assay <strong>in</strong>dicated no differences <strong>in</strong>testosterone production by testis tissue grafts of vary<strong>in</strong>gdonor age. Porc<strong>in</strong>e testis tissue obta<strong>in</strong>ed from 3-, 5-, 7- and14-day-old neonatal boars were all capable of produc<strong>in</strong>ground and elongate spermatids after 22 weeks of graft<strong>in</strong>g, buttestis grafts from 14-day-old donors had a significantlygreater (eightfold) percentage of sem<strong>in</strong>iferous tubules withspermatids compared to all other donor ages (p < 0.05).Cryopreservation did not affect the ability of testis tissuegrafts to grow, produce testosterone or establish spermatogenesiswhen compared to controls (p < 0.05). Collectively,these data demonstrate <strong>in</strong>tr<strong>in</strong>sic differences <strong>in</strong> the biologicalactivity of germ and somatic cell populations dur<strong>in</strong>g neonatalboar testis development associated with the establishment ofspermatogenesis.IntroductionSpermatogenesis is an organized process requir<strong>in</strong>g manysystemic and local factors (Zirk<strong>in</strong> 1998; Parv<strong>in</strong>en andVentela 1999) and <strong>in</strong>cludes all cellular transformationsthat occur with<strong>in</strong> the sem<strong>in</strong>iferous epithelium lead<strong>in</strong>g tothe production of mature spermatozoa <strong>in</strong> the sem<strong>in</strong>iferoustubule of the testis. Yet, germ cell <strong>in</strong>teractionswith nearby Sertoli cells are necessary for germ cells toundergo mitosis, meiosis and spermiogenesis (Russellet al. 1990). Furthermore, it is believed that Sertoli cellscontribute to the process of spermatogonial stem cell(SSC) renewal (McLean 2005), allow<strong>in</strong>g for cont<strong>in</strong>ualproduction of sperm dur<strong>in</strong>g a male’s adult life span.Leydig and Sertoli cells also account for the majority oftestosterone and oestrogen synthesis <strong>in</strong> the male (Waiteset al. 1985), and secrete factors exert<strong>in</strong>g negative feedbackon the pituitary gland; both processes promot<strong>in</strong>gnormal male fertility (Padmanabhan and Sharma 2001;Plant and Marshall 2001).The majority of <strong>in</strong>formation about the cell biologyassociated with spermatogenesis has been ga<strong>in</strong>ed withthe use of rodent models and as a result, there rema<strong>in</strong>s ageneral lack of knowledge regard<strong>in</strong>g testis developmentand sperm production <strong>in</strong> livestock. Further, due to thecomplexity <strong>in</strong>volved with germ cell differentiation, mostresearch <strong>in</strong>vestigat<strong>in</strong>g testis biology is often restricted to<strong>in</strong> vitro studies. Although <strong>in</strong>sightful, the <strong>in</strong>timate relationshipbetween Sertoli cells and germ cells are oftenlost, and cell l<strong>in</strong>es can mutate dur<strong>in</strong>g culture. Ectopictestis tissue xenograft<strong>in</strong>g is a procedure whereby smallpieces of testicular parenchyma from a donor animal aregrafted under the sk<strong>in</strong> of an immunodeficient nudemouse. This technique ma<strong>in</strong>ta<strong>in</strong>s <strong>in</strong>teractions betweengerm and somatic cells <strong>in</strong> the grafted tissue and thusprovides a biological assay for the establishment ofspermatogenesis <strong>in</strong> a variety of species (Honaramoozet al. 2002; Oatley et al. 2004). Additionally, multiplestudies have demonstrated that testis tissue xenograftsare representative of <strong>in</strong> vivo testis development (Schmidtet al. 2006; Zeng et al. 2006).The onset of puberty is commonly associated with thecompletion of the first wave of spermatogenesis <strong>in</strong> avariety of species (Amann 1970). It is known that Sertolicell number established before puberty determ<strong>in</strong>es adulttestis size (Russell 1993; Sharpe et al. 2000) and maturespermatogenic capacity (Orth et al. 1988) <strong>in</strong> variousmammalian species. Puberty <strong>in</strong> most domestic pigbreeds occurs around 20 weeks of age (Lunstra et al.1986) and researchers have <strong>in</strong>vestigated the establishmentof the spermatogenesis <strong>in</strong> the post-natal boar testisfrom 1 month of age to adulthood (Lunstra et al. 2003;McCoard et al. 2003; Almeida et al. 2006), but little isknown about relationships between somatic and germcells <strong>in</strong> the neonatal testis. Although porc<strong>in</strong>e Sertoli cellsstill express markers of proliferat<strong>in</strong>g cells around4 months of age (Klobucar et al. 2003), several reportssuggest the importance of Sertoli cell proliferation <strong>in</strong> theboar testis dur<strong>in</strong>g the first 2 weeks of life and neonatally(McCoard et al. 2001, 2003). This represents severalcandidate physiological stages <strong>in</strong> development that maygovern the spermatogenic capacity of the mature boartestis.Porc<strong>in</strong>e Sertoli cell numbers <strong>in</strong>crease fourfold dur<strong>in</strong>gthe first 2 weeks of neonatal life, and this represents animportant time for Sertoli cell homeostasis, thus wehypothesized that testicular tissue from 14-day-oldboars would have the greatest ability to establish andsupport spermatogenesis follow<strong>in</strong>g graft<strong>in</strong>g when comparedto other donor ages. We also predicted that nodifferences <strong>in</strong> testis graft androgen biosynthesis orÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Endocr<strong>in</strong>e Regulation of Porc<strong>in</strong>e Spermatogenesis 281recipient mouse serum follicle-stimulat<strong>in</strong>g hormone(FSH) would exist between the donor ages. Completionof these objectives will provide a better understand<strong>in</strong>g ofthe mechanisms regulat<strong>in</strong>g neonatal germ and somaticcell development associated with establish<strong>in</strong>g the firstwave of spermatogenesis and future spermatogeniccapacity <strong>in</strong> the boar testis.Materials and MethodsChemicals and reagents were all purchased from Sigma(http://www.sigmaaldrich.com) unless otherwise stated.Donor animals and tissue collectionThe Wash<strong>in</strong>gton State University Animal Care and UseCommittee approved all animal procedures. Neonataltestis was obta<strong>in</strong>ed from 3-, 5-, 7- and 14-day-old whitel<strong>in</strong>e composite boars at the Wash<strong>in</strong>gton State UniversitySw<strong>in</strong>e Facility us<strong>in</strong>g standard castration protocols. Am<strong>in</strong>imum of three boars were randomly chosen torepresent each donor age. To assess age-related differences<strong>in</strong> testicular characteristics of porc<strong>in</strong>e donormaterial prior to graft<strong>in</strong>g, <strong>in</strong>dividual testis weights,Sertoli cell number and markers of Sertoli cell maturitystatus were measured (n = 6 neonatal boars per age).We also evaluated these traits <strong>in</strong> 21-day-old boars.Ectopic testis xenograft<strong>in</strong>gTesticular parenchyma tissue was cut <strong>in</strong>to 5 mg piecesand ectopically xenografted onto castrated, recipientimmunodeficient NCr nude mice (Taconic, http://www.taconic.com; CrTac:NCR-Fox1 ) or placed<strong>in</strong> Bou<strong>in</strong>’s fixative for 4 h at 4°C followed by dehydrationand storage <strong>in</strong> 70% ethanol for eventual morphologicalevaluation. Briefly, mice were anesthetized withketam<strong>in</strong>e (0.1 mg ⁄ kg body weight, BW) and xylaz<strong>in</strong>e(0.5 mg ⁄ kg BW) castrated us<strong>in</strong>g surgical procedures,and managed as previously described (Oatley et al.2004).Analysis of donor grafts and recipient miceRecipient mice were sacrificed 22 weeks after graft<strong>in</strong>g.Testis grafts were removed, weighed and processed forhistological analysis of establishment of spermatogenesisas previously described (Schmidt et al. 2006). Atsacrifice, mouse vesicular gland weights were recordedas a bioassay for androgen biosynthesis, and bloodwas also collected from recipient mice by cardiacpuncture and serum evaluated for FSH and testosteroneby radioimmunoassay (RIA). The extent of germcell differentiation <strong>in</strong> testis tissue grafts was visuallyevaluated by compar<strong>in</strong>g the average number ofsem<strong>in</strong>iferous tubules cross-sections conta<strong>in</strong><strong>in</strong>g spermatogonia,meiotic germ cells, differentiat<strong>in</strong>g spermatidsor only Sertoli cells with<strong>in</strong> the largest centre section ofeach tissue sample. Digital images were captured us<strong>in</strong>ga Leica DFC 280 camera and a Leica DMEcompound microscope (Leica Microsystems Imag<strong>in</strong>gSolutions Ltd, http://www.leica-microsystems.com) at400 · magnification.ImmunohistochemistryAll antisera were obta<strong>in</strong>ed from Santa Cruz Biotechnology(Santa Cruz, CA, USA). Tissues were fixed <strong>in</strong>Bou<strong>in</strong>’s solution and embedded <strong>in</strong> paraff<strong>in</strong> accord<strong>in</strong>gto standard procedures. The tissues were sectioned(thickness, 5 lm), deparaff<strong>in</strong>ized, rehydrated and microwavedon high for 15 m<strong>in</strong> while submerged <strong>in</strong>0.01 M of sodium citrate (pH 6.0) to retrieve antigens.Tissues were submerged for 20 m<strong>in</strong> <strong>in</strong> 3% hydrogenperoxide (<strong>in</strong> methanol) to quench endogenous peroxidaseactivity. Sections were then blocked with 10%normal serum for 30 m<strong>in</strong> at room temperature, and<strong>in</strong>cubated overnight at 4°C with an aff<strong>in</strong>ity-purified,rabbit anti-mouse GATA4 polyclonal antiserum(C-20), mouse cytokerat<strong>in</strong> 18 monoclonal antiserum(C-04), rabbit anti-human GATA1 polyclonal antiserum(H-200), each diluted 1 : 200; or rabbit antihumanandrogen receptor (AR) polyclonal antiserum(1 : 100). As a negative control, serial sections wereprocessed without any primary antibody. At roomtemperature, sections were then washed <strong>in</strong> phosphatebufferedsal<strong>in</strong>e (PBS) (2 · 5 m<strong>in</strong>) and <strong>in</strong>cubated withbiot<strong>in</strong>ylated secondary antibody diluted 1 : 300 for 1 h,r<strong>in</strong>sed <strong>in</strong> PBS (2 · 5 m<strong>in</strong>) and exposed to streptavid<strong>in</strong>⁄ horseradish peroxidase (HRP) for 30 m<strong>in</strong>. Immunoreactivitywas detected follow<strong>in</strong>g a 5-m<strong>in</strong> <strong>in</strong>cubation<strong>in</strong> diam<strong>in</strong>obenzid<strong>in</strong>e (DAB) and sections were countersta<strong>in</strong>edlightly with haematoxyl<strong>in</strong>. Slides were storedat room temperature until morphological analysis.Photomicrograph cross-sections represent<strong>in</strong>g immunolocalizationof GATA-4, cytokerat<strong>in</strong> 18, GATA-1 andAR can be found <strong>in</strong> Fig. 3.CryopreservationOn the day of collection, four pieces of testis tissue from5-day-old boars (n = 3) were ectopically grafted ontocastrated NCr nude mice as a pre-cryopreservationcontrol. The rema<strong>in</strong>der of the samples that was notgrafted immediately was frozen <strong>in</strong> a dimethyl sulfoxide(DMSO)-based cryopreservation medium us<strong>in</strong>g a programmableembryo freezer. Briefly, freez<strong>in</strong>g media wasprepared by mix<strong>in</strong>g 0.68 g sucrose (0.1 M) and 0.2 gbov<strong>in</strong>e serum album<strong>in</strong> (1%) <strong>in</strong>to Dulbecco’s modifiedeagle medium (DMEM) to a f<strong>in</strong>al volume of 17.87 ml.Freez<strong>in</strong>g media was then filtered with a 0.4 mm syr<strong>in</strong>gefilter, and 2.13 ml of DMSO (1.5 M) was added. About4–8 pieces of testis tissue were added to 1 ml of freez<strong>in</strong>gmedia <strong>in</strong> cryovials. The cryovials were placed <strong>in</strong> aCrylogic (Model CL856) freezer at 20°C. The vials werecooled at 2°C ⁄ m<strong>in</strong> to )7°C and seeded with liquidnitrogen cooled tongs. The freez<strong>in</strong>g procedure cont<strong>in</strong>uedat 0.3°C ⁄ m<strong>in</strong> to )30°C and then at 0.1°C ⁄ m<strong>in</strong> to )35°C.Once the tissue reached )30°C, it was transferred to an)80°C freezer for 1 h and then submersed <strong>in</strong> liquidnitrogen. Tissue was stored for 1 week <strong>in</strong> liquid nitrogen,removed and subsequently thawed. After removalfrom liquid nitrogen, the cryovials were placed at roomtemperature for 1 m<strong>in</strong> and then <strong>in</strong>cubated for 1 m<strong>in</strong> <strong>in</strong> a37°C water bath. Samples were washed <strong>in</strong> mediaconta<strong>in</strong><strong>in</strong>g decreas<strong>in</strong>g concentrations of DMSO (1.0and 0.5 M) <strong>in</strong> DMEM for 3 m<strong>in</strong>, each followed byÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


282 KC Caires, JA Schmidt, AP Oliver, J de Avila and DJ McLeanwash<strong>in</strong>g <strong>in</strong> Hank’s balanced salt solutions (HBSS)(0.0 M DMSO). After the f<strong>in</strong>al wash, donor tissues(n = 3) were grafted onto n<strong>in</strong>e mice (three mice perdonor) <strong>in</strong> the same manner as the controls. To determ<strong>in</strong>ethe immediate affects of cryopreservation on tissuemorphology, samples of control and cryopreserved pregrafttestis tissue were also processed for evaluation bylight microscopy.Statistical analysisGerm cell proportions were averaged per mouse toprevent unequal weight<strong>in</strong>g due to variation <strong>in</strong> thesetraits. All statistical comparisons were performed us<strong>in</strong>gthe Proc GLM function of Statistical Analysis System(SAS) software, version 9.13 (SAS Institute, Cary, NC,USA), and differences were considered significant at thep £ 0.05 level. Pair-wise comparisons were evaluatedbetween donor ages us<strong>in</strong>g the Duncan’s multiple rangetest for significance. Data are presented as the mean ± -SEM and differences between donor ages were consideredsignificant at the p £ 0.05 level.ResultsNeonatal donor testicular characteristicsTesticular growth and Sertoli cell numberTestis weight <strong>in</strong>creased with advanc<strong>in</strong>g age(p < 0.001). The average <strong>in</strong>crease <strong>in</strong> testis weight after3 days post-partum (dpp) <strong>in</strong> neonatal boars was about1.5-, 2-, 6- and 12-fold at ages 5, 7, 14 and 21 dpp,respectively (Fig. 1). The number of Gata4-positiveSertoli cells per sem<strong>in</strong>iferous cord cross-section also(a)(b)Fig. 1. Porc<strong>in</strong>e testis tissue weight and Sertoli cell number from prepubertalboars at 3, 5, 7, 14 and 21 days post-partum (dpp). (a)Testicular growth and (b) number of Gata4-positive Sertoli cell nucleiper sem<strong>in</strong>iferous tubule (cross-section). Bars with different letters<strong>in</strong>dicate significant differences between means (p < 0.05)<strong>in</strong>creased with advanc<strong>in</strong>g age (p < 0.05, Fig. 1b).Sertoli cell nuclei reacted strongly with the Gata4antibody, but a low level of expression was alsolocalized to Leydig cell nuclei <strong>in</strong> the <strong>in</strong>terstitial compartment(Fig. 2b), as previously described (McCoardet al. 2003).Prote<strong>in</strong> markers of Sertoli cell maturationTo determ<strong>in</strong>e the maturation status of Sertoli cells theexpression of cytokerat<strong>in</strong> 18 and Gata1, prote<strong>in</strong>markers for immaturity and differentiation wereimmunolocalized. Strong cytoplasmic sta<strong>in</strong><strong>in</strong>g forcytokerat<strong>in</strong> 18, a foetal Sertoli cell marker, wasvisualized <strong>in</strong> Sertoli cells with<strong>in</strong> 3, 5, 7, 14 and 21dpp boar testis tissues (Fig. 2c,f). Immunolocalizationof GATA1, a l<strong>in</strong>eage specific differentiation marker,revealed weak but specific cytoplasmic sta<strong>in</strong><strong>in</strong>g nearthe apical region of Sertoli cell nuclei of all neonatalages evaluated (Fig. 2d) as is consistent with theexpression pattern of precursor Sertoli cells <strong>in</strong> the prenataland early neonatal mouse testis (Yomogida et al.1994). Nuclear immunoreactivity for AR was visualized<strong>in</strong> Sertoli, peritubular myoid, Leydig cells andgonocytes with<strong>in</strong> 3, 5, 7, 14 and 21 dpp boar testistissues (Fig. 2e).Porc<strong>in</strong>e testis tissue graft growth, androgen biosynthesisand recipient mouse serum follicle-stimulat<strong>in</strong>g hormoneconcentrationTo assess age-related differences <strong>in</strong> growth potential ofporc<strong>in</strong>e donor tissue at removal, testis graft weightswere measured and the numbers of sem<strong>in</strong>iferoustubule cross-sections were determ<strong>in</strong>ed. Analysis ofgrowth potential <strong>in</strong>dicated donor-age-related variation<strong>in</strong> testis tissue graft weight and tubule number. Bothparameters for graft growth were significantly higher(p < 0.05) <strong>in</strong> testis grafts from 3-day donor tissue,when compared to all other ages (Fig. 3). In contrast,donor testis tissue from 5-, 7- and 14-day-old neonatalboars had similar growth rates as no differences weredetected <strong>in</strong> graft weight or sem<strong>in</strong>iferous tubule number.To assess the ability for testis tissue grafts tosynthesize biologically active androgen, recipient mouseserum testosterone levels and vesicular gland weightswere compared between donor age groups. Despitevariation <strong>in</strong> donor age, RIA for serum testosterone andbiological assay of vesicular gland weights <strong>in</strong>dicated nodifferences <strong>in</strong> the ability of testis tissue grafts to producetestosterone (Table 1). We also <strong>in</strong>vestigated the abilityof porc<strong>in</strong>e testis tissue xenografts to produce factors thatexert negative feedback on pituitary FSH synthesiswhen compared to normal, <strong>in</strong>tact mice of similar ageand stra<strong>in</strong> by RIA. Follicle-stimulat<strong>in</strong>g hormone concentrations<strong>in</strong> recipient mice support<strong>in</strong>g testis graftsfrom 5-, 7- and 14-day-old donor tissue did not differand were similar to normal physiological levels <strong>in</strong> agematched,<strong>in</strong>tact nude mice (Table 1). Yet, serum FSHlevels were significantly lower (p < 0.05) than normal <strong>in</strong>recipient mice support<strong>in</strong>g testis grafts from 3-day-olddonor tissue.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Endocr<strong>in</strong>e Regulation of Porc<strong>in</strong>e Spermatogenesis 283(a)(b)(c)(d)(e)(f)Fig. 2. Immunolocalization ofSertoli cell prote<strong>in</strong> markers <strong>in</strong>pre-pubertal porc<strong>in</strong>e testis tissue:(a) negative control (no primaryIgG), (b) GATA4, (c, f) cytokerat<strong>in</strong>18, (d) GATA1 and (e) androgenreceptor. Scale bars = 50 lmDegree of germ cell differentiation <strong>in</strong> porc<strong>in</strong>e testis tissuegraftsThe extent of spermatogenesis <strong>in</strong> testis tissue fromneonatal boars of different ages was determ<strong>in</strong>ed after22 weeks of graft<strong>in</strong>g. Porc<strong>in</strong>e testis tissue obta<strong>in</strong>ed from3-, 5-, 7- and 14-day-old neonatal boars were all capableof produc<strong>in</strong>g round and elongate spermatids aftergraft<strong>in</strong>g (Fig. 4). Testis grafts from 14-day-old donorsconta<strong>in</strong>ed a significantly greater (p < 0.05) percentageof sem<strong>in</strong>iferous tubules with spermatids compared to allother donor ages (Fig. 4). In contrast, spermatogenesiswas established at similar rates <strong>in</strong> donor testis tissuefrom 3-, 5- and 7-day-old neonatal boars (Fig. 4). Theaverage percentage of tubules conta<strong>in</strong><strong>in</strong>g spermatids <strong>in</strong>testis tissue from 14-day-old donors was about eightfoldhigher (45.4%) when compared to other donor agesaverag<strong>in</strong>g 5.4%. Photomicrograph cross-sections represent<strong>in</strong>gthe degree of germ cell differentiation with<strong>in</strong>porc<strong>in</strong>e testis grafts can be found <strong>in</strong> Fig. 5.Effect of cryopreservation on porc<strong>in</strong>e testis tissuedevelopment post-graft<strong>in</strong>gWe evaluated the affect of pre-graft cryopreservationon the ability of porc<strong>in</strong>e testis tissue grafts to grow,produce testosterone and support complete germ celldifferentiation by utiliz<strong>in</strong>g 5-day-old donor testis tissue(n = 3). Morphological evaluation prior to graft<strong>in</strong>grevealed that cryopreservation adversely affected sem<strong>in</strong>iferouscord organization when compared to controls(Fig. 6a,b). Follow<strong>in</strong>g the graft<strong>in</strong>g period, no differenceswere observed between control and cryopreserved testistissue weight, sem<strong>in</strong>iferous tubule number and diameteror testosterone production (data not shown). Establishmentof spermatogenesis <strong>in</strong> cryopreserved and controlporc<strong>in</strong>e testis tissue grafts was similar as no differenceswere observed <strong>in</strong> the distribution of sem<strong>in</strong>iferous tubulecross-sections conta<strong>in</strong><strong>in</strong>g spermatogonia, meiotic germcells and spermatids, respectively (Fig. 6c). Photomicrographcross-sections represent<strong>in</strong>g complete germ celldifferentiation <strong>in</strong> control and cryopreserved porc<strong>in</strong>etestis tissue grafts can be found <strong>in</strong> Fig. 6d,e.DiscussionIn the present study, porc<strong>in</strong>e testis tissue grafts from alldonor ages <strong>in</strong>itiated the first round of spermatogenesisassociated with pubertal age <strong>in</strong> a similar developmentaltimel<strong>in</strong>e to the boar testis <strong>in</strong> situ, and were capable ofcomplete germ cell differentiation after 22 weeks ofgraft<strong>in</strong>g. Yet, we observed a significant <strong>in</strong>crease (eightfold)<strong>in</strong> the extent of germ cell differentiation <strong>in</strong> testisÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


284 KC Caires, JA Schmidt, AP Oliver, J de Avila and DJ McLean(a)(b)Fig. 4. Percentage of sem<strong>in</strong>iferous tubules conta<strong>in</strong><strong>in</strong>g round andelongat<strong>in</strong>g spermatids with<strong>in</strong> porc<strong>in</strong>e testis tissue grafts of vary<strong>in</strong>gdonor age. Different letters show significant differences (p < 0.05)between meansFig. 3. Weight of porc<strong>in</strong>e testis tissue grafts obta<strong>in</strong>ed from prepubertalboars of 3, 5, 7 and 14 days of age follow<strong>in</strong>g the ectopicgraft<strong>in</strong>g period. (a) Testis tissue graft weight and (b) number ofsem<strong>in</strong>iferous tubule cross-sections per testis graft. Different letters<strong>in</strong>dicate significant differences (p < 0.05) between meanstissue grafts from 14-day-old donors, as demonstratedby about 40% more sem<strong>in</strong>iferous tubules conta<strong>in</strong><strong>in</strong>gspermatids, when compared to other donor ages. Thus,<strong>in</strong>tr<strong>in</strong>sic differences exist <strong>in</strong> neonatal boar testis developmentassociated with the support<strong>in</strong>g the first wave ofspermatogenesis <strong>in</strong> porc<strong>in</strong>e testis tissue xenografts.Differences also existed <strong>in</strong> the ability for testis graftsto grow and exert negative feedback on pituitary FSHproduction. Testis tissue grafts from 3-day donor tissuewere on average twofold larger when compared to testistissue obta<strong>in</strong>ed from other donor ages, while serum FSHlevels were significantly lower than controls (p < 0.05)<strong>in</strong> recipient mice support<strong>in</strong>g testis grafts from 3-day-olddonor tissue. In contrast, testis grafts from 5-, 7- and14-day-old donor tissue were of similar size andrecipient mice support<strong>in</strong>g these testis grafts conta<strong>in</strong>edserum concentrations of FSH <strong>in</strong> similar to normalphysiological levels <strong>in</strong> age-matched, <strong>in</strong>tact nude mice.This f<strong>in</strong>d<strong>in</strong>g is <strong>in</strong>terest<strong>in</strong>g, as no differences wereobserved <strong>in</strong> testosterone production by RIA or biologicalassay, and suggests <strong>in</strong>creased potential for testisgrafts from 3-day donors to negatively regulate pituitaryproduction of FSH, likely due to <strong>in</strong>creased <strong>in</strong>hib<strong>in</strong>production from a larger population of Sertoli cells.This effect may also be due to germ<strong>in</strong>al aplasia (Zarateet al. 1974) or failure of germ cells to develop, butthis appears unlikely based on the data demonstrat<strong>in</strong>gthe establishment of spermatogenesis was similar <strong>in</strong>testis tissue grafts from 3-, 5- and 7-day neonataldonors.The timel<strong>in</strong>e for Sertoli cell proliferation and differentiationevents are highly species-dependent, occurbefore puberty and are essential for spermatogenesis <strong>in</strong>the adult. It is well established that dur<strong>in</strong>g pre-natal andpost-natal testis development, FSH stimulates Sertolicells to proliferate and <strong>in</strong> response to thyroid hormones,ret<strong>in</strong>oic acid and testosterone, Sertoli cells cease proliferationand <strong>in</strong>itiate term<strong>in</strong>al differentiation (Orth 1984;Buzzard et al. 2003; Holsberger and Cooke 2005).Although androgens may limit Sertoli cell expansion<strong>in</strong> rodents (Buzzard et al. 2003), elevated testosteronelevels dur<strong>in</strong>g pre-pubertal development <strong>in</strong> the rhesusmonkey testis <strong>in</strong>creases Sertoli proliferation and thepopulation of germ cells <strong>in</strong>dependent of FSH (Arslanet al. 1993; Ramaswamy et al. 2000).The first 3 weeks of neonatal life <strong>in</strong> boars correspondswith significant <strong>in</strong>creases <strong>in</strong> testis weight and Sertoli cellnumber (Fig. 1), as previously reported (McCoard et al.2003). Our morphologic evaluation of testicular characteristics<strong>in</strong>dicates that Sertoli cells <strong>in</strong> the post-natalboar testis ma<strong>in</strong>ta<strong>in</strong> a primitive phenotype until at least21 days of age, as def<strong>in</strong>ed by the expression patterns oftwo well-characterized prote<strong>in</strong> markers of Sertoli cellmaturation: cytokerat<strong>in</strong> 18 (Stosiek et al. 1990) andGATA1 (Yomogida et al. 1994; Bartu˚ nek et al. 2003).Dur<strong>in</strong>g this same timel<strong>in</strong>e, the germ cell populationconsists of mitotically dormant gonocytes, locatedpericentrally <strong>in</strong> the sem<strong>in</strong>iferous tubule, that do notmigrate to the basement membrane of until after14 days of age.Non-graftedGrafted: listed by donor age (dpp)Intact Castrate 3 5 7 14VG wt. (mg) 220.4 ± 14.8* 14.7 ± 2.5* 139.0 ± 21.3 84.0 ± 29.3 63.5 ± 36.4 92.5 ± 22.3Test. (ng ⁄ ml) 0.88 ± 0.4 0.09 ± 0.4* 1.26 ± 0.2 0.94 ± 0.2 0.54 ± 0.2 0.65 ± 0.3FSH (ng ⁄ ml) 36.7 ± 4.5 NA 22.4 ± 2.8* 32.4 ± 4.3 36.7 ± 5.2 29.5 ± 3.8Table 1. Vesicular gland weights,serum concentrations of testosterone(test.) and follicle stimulat<strong>in</strong>ghormone (FSH) of <strong>in</strong>tact (positivecontrol), castrated (negativecontrol) and castrated micesupport<strong>in</strong>g testis tissue grafts*Significant differences (p < 0.05) between means.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Endocr<strong>in</strong>e Regulation of Porc<strong>in</strong>e Spermatogenesis 285(a)(b)(c)(d)Fig. 5. Morphological analysis ofgerm cell populations <strong>in</strong> porc<strong>in</strong>etestis tissue grafts. Tubule crosssectionsrepresent<strong>in</strong>g mostadvanced germ cell type: (a) spermatocytesa, (b) round spermatidsb, (c, d) elongat<strong>in</strong>g spermatids c.Scale bars = 50 lm(a) (b) (c)Fig. 6. Morphological analysis of(a) non-frozen control and (b)cryopreserved porc<strong>in</strong>e testis tissueprior to graft<strong>in</strong>g. (c) Morphometricevaluation of germ cell differentiation<strong>in</strong> control and frozen porc<strong>in</strong>etestis tissue after graft<strong>in</strong>g(20 weeks) represented by percentageof sem<strong>in</strong>iferous tubule crosssectionsconta<strong>in</strong><strong>in</strong>g spermatogonia[Gonia], spermatocytes [Cytes]and spermatids [Tids] as mostadvanced cell type. There were nosignificant differences betweenmeans (p > 0.05). Micrographs of(d) control and (e). Cryopreservedporc<strong>in</strong>e testis tissue follow<strong>in</strong>g thegraft<strong>in</strong>g period Bar = 50 lm(d)(e)In vivo studies have suggested the potential tomanipulate establishment of Sertoli cell populations <strong>in</strong>non-human primates (Sharpe et al. 2000), rodents (Orthet al. 1988), bulls (Majdic et al. 1998) and boars(Lunstra et al. 2003; Nonneman et al. 2005). In anattempt to <strong>in</strong>crease Sertoli cell number and pubertaltestis size similar to rodents, researchers used theapproach of <strong>in</strong>duc<strong>in</strong>g post-natal hypothyroidism <strong>in</strong> 21-day-old, pre-pubertal boars (Klobucar et al. 2003). Yet,it was concluded that post-natal hypothyroidism doesnot affect testis development, Sertoli cell number orsperm production <strong>in</strong> boars, despite observations ofporc<strong>in</strong>e Sertoli cells express<strong>in</strong>g markers of proliferat<strong>in</strong>gcells until 4 months of age (Klobucar et al. 2003).Interest<strong>in</strong>gly, Sertoli cells and gonocytes were immunopositivefor nuclear AR prote<strong>in</strong> <strong>in</strong> the testes of allÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


286 KC Caires, JA Schmidt, AP Oliver, J de Avila and DJ McLeanneonatal boar ages evaluated (Fig. 2e). This suggeststhat testosterone may directly or <strong>in</strong>directly regulate thehomeostasis of these cell types <strong>in</strong> the neonatal boartestis, potentially contribut<strong>in</strong>g to the observed donor agevariation <strong>in</strong> the establishment of spermatogenesis <strong>in</strong> thegraft<strong>in</strong>g study, and it is known that post-natal testisdevelopment is conserved between grafted and <strong>in</strong> vivotestis tissue (Oatley et al. 2004; Zeng et al. 2006).Our results demonstrate age-related differences <strong>in</strong> thepotential of neonatal testis tissue to grow and supportcomplete germ cell differentiation when grafted onimmunodeficient mice. More importantly, these f<strong>in</strong>d<strong>in</strong>gs<strong>in</strong>dicate important physiological changes <strong>in</strong> germ andSertoli cell homeostasis dur<strong>in</strong>g neonatal testis development<strong>in</strong> the boar, and may expla<strong>in</strong> why previousattempts to <strong>in</strong>crease testis size and sperm production<strong>in</strong> boars have been unsuccessful. This conclusion issupported by <strong>in</strong> vivo evidence (At-Taras et al. 2006,2008), and as a result we suggest future efforts to impactlifetime fertility <strong>in</strong> boars should occur dur<strong>in</strong>g the first2 weeks of neonatal life.From an application standpo<strong>in</strong>t, tissue orig<strong>in</strong>at<strong>in</strong>gfrom 14-day boar testis resulted <strong>in</strong> the greatest degree ofgraft germ cell differentiation; this stage <strong>in</strong> developmentmay be ideally suited for germ l<strong>in</strong>e genetic manipulationof cells, potentially facilitat<strong>in</strong>g a novel means to producetransgenic boar spermatozoa. Further, as Sertoli cellsprovide an immunoprotective barrier for develop<strong>in</strong>ggerm cells, neonatal porc<strong>in</strong>e Sertoli cells have seen<strong>in</strong>creas<strong>in</strong>g use to encapsulate porc<strong>in</strong>e islet cells prior totransplantation <strong>in</strong> human patients with diabetes mellitus(Dufour et al. 2003; Valdes-Gonzalez et al. 2007), andthus a better understand<strong>in</strong>g of the basic mechanismsregulat<strong>in</strong>g their homeostasis, and survival after transplantationis necessitated. We also demonstrated nodifferences <strong>in</strong> the ability of cryopreserved and freshlygrafted donor porc<strong>in</strong>e testis tissue to grow, producetestosterone and establish spermatogenesis. Thus, assomatic cell function is restored after freez<strong>in</strong>g, cryopreservationof pre-pubertal testis tissue prior to graft<strong>in</strong>gpresents a novel means for male germ l<strong>in</strong>epreservation. The factors regulat<strong>in</strong>g the biologicalactivity of Sertoli and germ cell homeostasis <strong>in</strong> theneonatal boar testis will be the focus of future experimentsand may provide <strong>in</strong>sights for both agriculturaland biomedical applications.AcknowledgementThis research was supported <strong>in</strong> part by a graduate research fellowshipprovided by the Seattle Chapter of the Achievement Rewards forCollege Scientists Foundation on behalf of K.C.C.ReferencesAlmeida FF, Leal MC, Franca LR, 2006: Testis morphometry,duration of spermatogenesis, and spermatogenic efficiency<strong>in</strong> the wild boar (Sus scrofa). Biol Reprod 75, 792–799.Amann RP, 1970: Sperm production rates. In: Johnson AD,Gomes WR, VanDemark NL (eds), The Testis, Vol. 1.Academic Press, New York, pp. 433–482.Arslan M, We<strong>in</strong>bauer GF, Schlatt S, Shahab M, Nieschlag E,1993: FSH and testosterone, alone or <strong>in</strong> comb<strong>in</strong>ation,<strong>in</strong>itiate testicular growth and <strong>in</strong>crease the number ofspermatogonia and Sertoli cells <strong>in</strong> a juvenile non-humanprimate (Macaca mulatta). J Endocr<strong>in</strong>ol 136, 235–243.At-Taras EE, Berger T, McCarthy MJ, Conley AJ, Nitta-OdaBJ, Roser JF, 2006: Reduc<strong>in</strong>g estrogen synthesis <strong>in</strong> develop<strong>in</strong>gboars <strong>in</strong>creases testis size and total sperm production.J Androl 27, 552–559.At-Taras EE, Kim IC, Berger T, Conley A, Roser JF, 2008:Reduc<strong>in</strong>g endogenous estrogen dur<strong>in</strong>g development altershormone production by porc<strong>in</strong>e Leydig cells and sem<strong>in</strong>iferoustubules. Domest Anim Endocr<strong>in</strong>ol 34, 100–108.Bartu˚ nek P, Králova´ J, Blend<strong>in</strong>ger G, Dvora´k M, Zenke M,2003: GATA-1 and c-myb crosstalk dur<strong>in</strong>g red blood celldifferentiation through GATA-1 b<strong>in</strong>d<strong>in</strong>g sites <strong>in</strong> the c-mybpromoter. Oncogene 22, 1927–1935.Buzzard JJ, Wreford NG, Morrison JR, 2003: Thyroidhormone, ret<strong>in</strong>oic acid, and testosterone suppress proliferationand <strong>in</strong>duce markers of differentiation <strong>in</strong> cultured ratsertoli cells. Endocr<strong>in</strong>ology 144, 3722–3731.Dufour JM, Rajotte RV, Korbutt GS, Emerich DF, 2003:Harness<strong>in</strong>g the immunomodulatory properties of Sertolicells to enable xenotransplantation <strong>in</strong> type I diabetes.Immunol Invest 32, 275–297.Holsberger DR, Cooke PS, 2005: Understand<strong>in</strong>g the role ofthyroid hormone <strong>in</strong> Sertoli cell development: a mechanistichypothesis. Cell Tissue Res 322, 133–140.Honaramooz A, Snedaker A, Boiani M, Scholer H, Dobr<strong>in</strong>skiI, Schlatt S, 2002: Sperm from neonatal mammalian testesgrafted <strong>in</strong>to mice. Nature 418, 778–781.Klobucar I, Kosec M, Cebulj-Kadunc N, Majdic G, 2003:Postnatal hypothyroidism does not affect prepubertaltestis development <strong>in</strong> boars. Reprod Domest Anim 38,193–198.Lunstra DD, Ford JJ, Christenson RK, Allrich RD, 1986:Changes <strong>in</strong> Leydig cell ultrastructure and function dur<strong>in</strong>gpubertal development <strong>in</strong> the boar. Biol Reprod 34, 145–158.Lunstra DD, Wise TH, Ford JJ, 2003: Sertoli cells <strong>in</strong> the boartestis: changes dur<strong>in</strong>g development and compensatoryhypertrophy after hemicastration at different ages. BiolReprod 68, 140–150.Majdic G, Snoj T, Horvat A, Mrkun J, Kosec M, Cestnik V,1998: Higher thyroid hormone levels <strong>in</strong> neonatal life result <strong>in</strong>reduced testis volume <strong>in</strong> postpubertal bulls. Int J Androl 21,352–357.McCoard SA, Lunstra DD, Wise TH, Ford JJ, 2001: Specificsta<strong>in</strong><strong>in</strong>g of Sertoli cell nuclei and evaluation of Sertoli cellnumber and proliferative activity <strong>in</strong> Meishan and WhiteComposite boars dur<strong>in</strong>g the neonatal period. Biol Reprod64, 689–695.McCoard SA, Wise TH, Ford JJ, 2003: Endocr<strong>in</strong>e andmolecular <strong>in</strong>fluences on testicular development <strong>in</strong> Meishanand White Composite boars. J Endocr<strong>in</strong>ol 178, 405–416.McLean DJ, 2005: Spermatogonial stem cell transplantationand testicular function. Cell Tissue Res 322, 21–31.Nonneman D, Rohrer GA, Wise TH, Lunstra DD, Ford JJ,2005: A variant of porc<strong>in</strong>e thyrox<strong>in</strong>e-b<strong>in</strong>d<strong>in</strong>g globul<strong>in</strong> hasreduced aff<strong>in</strong>ity for thyrox<strong>in</strong>e and is associated with testissize. Biol Reprod 72, 214–220.Oatley JM, de Avila DM, Reeves JJ, McLean DJ, 2004:Establishment of spermatogenesis and the sem<strong>in</strong>iferousepithelium <strong>in</strong> ectopically xenografted bov<strong>in</strong>e testiculartissue. Biol Reprod 71, 494–501.Orth JM, 1984: The role of follicle-stimulat<strong>in</strong>g hormone <strong>in</strong>controll<strong>in</strong>g Sertoli cell proliferation <strong>in</strong> testes of fetal rats.Endocr<strong>in</strong>ology 115, 1248–1255.Orth JM, Gunsalus GL, Lamperti AA, 1988: Evidence fromSertoli cell-depleted rats <strong>in</strong>dicates that spermatid number<strong>in</strong> adults depends on numbers of Sertoli cells produceddur<strong>in</strong>g per<strong>in</strong>atal development. Endocr<strong>in</strong>ology 122, 787–794.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Endocr<strong>in</strong>e Regulation of Porc<strong>in</strong>e Spermatogenesis 287Padmanabhan V, Sharma TP, 2001: Neuroendocr<strong>in</strong>e vs.paracr<strong>in</strong>e control of follicle-stimulat<strong>in</strong>g hormone. ArchMed Res 32, 533–543.Parv<strong>in</strong>en M, Ventela S, 1999: Local regulation of spermatogenesis:a liv<strong>in</strong>g cell approach. Hum Fertil (Camb) 2, 138–142.Plant TM, Marshall GR, 2001: The functional significance ofFSH <strong>in</strong> spermatogenesis and the control of its secretion <strong>in</strong>male primates. Endocr Rev 22, 764–786.Ramaswamy S, Plant TM, Marshall GR, 2000: Pulsatilestimulation with recomb<strong>in</strong>ant s<strong>in</strong>gle cha<strong>in</strong> human lute<strong>in</strong>iz<strong>in</strong>ghormone elicits precocious sertoli cell proliferation <strong>in</strong> thejuvenile male rhesus monkey (Macaca mulatta). Biol Reprod63, 82–88.Russell LD, 1993: Form, dimensions and cytology of mammalianSertoli cells. In: Russell LD, Griswold MD (eds),The Sertoli Cell. Cache River Press, Clearwater, FL, pp.1–37.Russell LD, Ettl<strong>in</strong> RA, S<strong>in</strong>ha Hikim AP, Clegg ED, 1990:Mammalian spermatogenesis. <strong>in</strong>: Russell LD, Ettl<strong>in</strong> RA,S<strong>in</strong>ha Hikim AP, Clegg ED (eds), Histological and HistopathologicalEvaluation of the Testis. Cache River Press,Clearwater, FL, pp. 1–40.Schmidt JA, de Avila JM, McLean DJ, 2006: Graft<strong>in</strong>gperiod and donor age affect the potential for spermatogenesis<strong>in</strong> bov<strong>in</strong>e ectopic testis xenografts. Biol Reprod 75,160–166.Sharpe RM, Walker M, Millar MR, Atanassova N, Morris K,McK<strong>in</strong>nell C, Saunders PT, Fraser HM, 2000: Effect ofneonatal gonadotrop<strong>in</strong>-releas<strong>in</strong>g hormone antagonistadm<strong>in</strong>istration on Sertoli cell number and testicular development<strong>in</strong> the marmoset: comparison with the rat. BiolReprod 62, 1685–1693.Stosiek P, Kasper M, Karsten U, 1990: Expression of cytokerat<strong>in</strong>8 and 18 <strong>in</strong> human Sertoli cells of immature and atrophicsem<strong>in</strong>iferous tubules. Differentiation 43, 66–70.Valdes-Gonzalez RA, White DJ, Dorantes LM, Teran L,Garibay-Nieto GN, Bracho-Blanchet E, Davila-Perez R,Evia-Viscarra L, Ormsby CE, Ayala-Sumuano JT, Silva-Torres ML, Ramirez-Gonzalez B, 2007: Three-yr follow-upof a type 1 diabetes mellitus patient with an islet xenotransplant.Cl<strong>in</strong> Transplant 21, 352–357.Waites GM, Speight AC, Jenk<strong>in</strong>s N, 1985: The functionalmaturation of the Sertoli cell and Leydig cell <strong>in</strong> themammalian testis. J Reprod Fertil 75, 317–326.Yomogida K, Ohtani H, Harigae H, Ito E, Nishimune Y,Engel JD, Yamamoto M, 1994: Developmental stage- andspermatogenic cycle-specific expression of transcriptionfactor GATA-1 <strong>in</strong> mouse Sertoli cells. Development 120,1759–1766.Zarate A, Garrido J, Canales ES, Soria J, Schally AV, 1974:Disparity <strong>in</strong> the negative gonadal feedback control for LHand FSH secretion <strong>in</strong> cases of germ<strong>in</strong>al aplasia or Sertolicell-onlysyndrome. J Cl<strong>in</strong> Endocr<strong>in</strong>ol Metab 38, 1125–1127.Zeng W, Avelar GF, Rathi R, Franca LR, Dobr<strong>in</strong>ski I, 2006:The length of the spermatogenic cycle is conserved <strong>in</strong>porc<strong>in</strong>e and ov<strong>in</strong>e testis xenografts. J Androl 27, 527–533.Zirk<strong>in</strong> BR, 1998: Spermatogenesis: its regulation by testosteroneand FSH. Sem<strong>in</strong> Cell Dev Biol 9, 417–421.Author’s address (for correspondence): DJ McLean, Wash<strong>in</strong>gton StateUniversity, Pullman, Wash<strong>in</strong>gton, 99164–6353, USA. E-mail:dmclean@wsu.eduConflict of <strong>in</strong>terest: All authors declare no conflict of <strong>in</strong>terests.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 288–294 (2008); doi: 10.1111/j.1439-0531.2008.01176.xISSN 0936-6768Male Germ Cell TransplantationI Dobr<strong>in</strong>skiSchool of Veter<strong>in</strong>ary Medic<strong>in</strong>e, Center for Animal Transgenesis and Germ Cell Research, University of Pennsylvania, Kennett Square, PA, USAContentsTransplantation of male germ l<strong>in</strong>e stem cells from a donoranimal to the testes of an <strong>in</strong>fertile recipient was first described<strong>in</strong> 1994. Donor germ cells colonize the recipient’s testis andproduce donor-derived sperm, such that the recipient male candistribute the genetic material of the germ cell donor. Germcell transplantation represents a functional reconstitutionassay for male germ l<strong>in</strong>e stem cells and as such has vastly<strong>in</strong>creased our ability to study the biology of stem cells <strong>in</strong> thetestis and def<strong>in</strong>e phenotypes of <strong>in</strong>fertility. First developed <strong>in</strong>rodents, the technique has now been used <strong>in</strong> a number ofanimal species, <strong>in</strong>clud<strong>in</strong>g domestic mammals, chicken and fish.There are three major applications for this technology <strong>in</strong>animals: first, to study fundamental aspects of male germ l<strong>in</strong>estem cell biology and male fertility; second, to preserve thereproductive potential of genetically valuable <strong>in</strong>dividuals bymale germ cell transplantation with<strong>in</strong> or between species;third, to produce transgenic sperm by genetic manipulation ofisolated germ l<strong>in</strong>e stem cells and subsequent transplantation.Transgenesis through the male germ l<strong>in</strong>e has tremendouspotential <strong>in</strong> species <strong>in</strong> which embryonic stem cells are notavailable and somatic cell nuclear transfer has limited success.Therefore, transplantation of male germ cells is a uniquelyvaluable approach for the study, preservation and manipulationof male fertility <strong>in</strong> animals.IntroductionMale fertility requires efficient production of spermatozoathroughout the adult life of the male. Spermatogenesisis characterized by sequential steps of cellproliferation and differentiation result<strong>in</strong>g <strong>in</strong> the productionof virtually unlimited numbers of spermatozoa(Russell et al. 1990). The foundation of this system is themale germ l<strong>in</strong>e stem cell, which has the unique potentialfor both self-renewal and production of differentiateddaughter cells that ultimately form spermatozoa (Huck<strong>in</strong>s1971; Clermont 1972; Meistrich and van Beek1993a). The male germ l<strong>in</strong>e stem cell is the only cell <strong>in</strong> anadult body that divides and can contribute genes tosubsequent generations, mak<strong>in</strong>g it an obvious target forgenetic manipulations (see below). Because stem cellsare ultimately def<strong>in</strong>ed by function, unequivocal identificationdepends on an assay to demonstrate thepotential to reconstitute the appropriate body system.For male germ l<strong>in</strong>e stem cells, this assay wasestablished <strong>in</strong> 1994, when it was demonstrated thattransplantation of germ cells from fertile donor mice tothe testes of <strong>in</strong>fertile recipient mice resulted <strong>in</strong> donorderivedspermatogenesis and sperm production by therecipient animal (Br<strong>in</strong>ster and Zimmermann 1994). Theuse of donor males carry<strong>in</strong>g a marker gene allowed foridentification of donor-derived spermatogenesis <strong>in</strong> therecipient mouse testis and proved that the donorhaplotype is passed on to the offspr<strong>in</strong>g by recipientanimals (Br<strong>in</strong>ster and Avarbock 1994). In 13 years s<strong>in</strong>cethe <strong>in</strong>itial report, the technique has found widespreaduse <strong>in</strong> rodents, and more recently, also <strong>in</strong> non-rodentanimals (see below). Some of the most importantmilestones are summarized <strong>in</strong> Table 1.Cross-Species Transplantation of Male GermCellsIn 1996, production of rat sperm <strong>in</strong> mouse testes wasachieved follow<strong>in</strong>g cross-species (xenogeneic) spermatogonialtransplantation from rats to mice (Clouthieret al. 1996) and was subsequently successful from miceto rats (Ogawa et al. 1999b; Zhang et al. 2003). Thiswork illustrated that the cell cycle dur<strong>in</strong>g spermatogenesisis controlled by the germ cell, not the Sertolicell (Franca et al. 1998). Recently, it was confirmedthat rat sperm produced <strong>in</strong> a host mouse testis arecapable of support<strong>in</strong>g normal development when<strong>in</strong>troduced <strong>in</strong>to rat oocytes by ICSI (Sh<strong>in</strong>ohara et al.2006). Hamster spermatogenesis also occurred successfully<strong>in</strong> the mouse host (Ogawa et al. 1999a); yet, with<strong>in</strong>creas<strong>in</strong>g phylogenetic distance between donor andrecipient species, meiotic differentiation could no longerbe achieved <strong>in</strong> the mouse testis. Transplantation ofgerm cells from donors rang<strong>in</strong>g from rabbits and dogs,to pigs and bulls, and ultimately non-human primatesand humans, resulted <strong>in</strong> colonization of the mousetestis, but spermatogenesis became arrested at the stageof spermatogonial expansion (Dobr<strong>in</strong>ski et al. 1999,2000; Nagano et al. 2001b, 2002a). It appears that the<strong>in</strong>itial steps of germ cell recognition by the Sertoli cells,localization to the basement membrane and <strong>in</strong>itiationof spermatogonial proliferation are conserved betweenevolutionary divergent species. Yet, the testicularenvironment (Sertoli cells and paracr<strong>in</strong>e factors) ofthe recipient mouse appears to be unable to supportspermatogenic differentiation and meiosis from donorspecies other than rodents. This <strong>in</strong>compatibility couldtheoretically be addressed by co-transplantation ofgerm cells and donor Sertoli cells to the mouse testis(Sh<strong>in</strong>ohara et al. 2003), and complete spermatogenesisfrom different mammalian species <strong>in</strong> a mouse host wasachieved by testis tissue transplantation (Honaramoozet al. 2002b). Although xenogeneic spermatogonialtransplantation to rodent testes did not result <strong>in</strong>spermatogenesis from donor species other thanrodents, it nonetheless provides a bioassay for stemcell potential of germ cells isolated from other species(Dobr<strong>in</strong>ski et al. 1999, 2000; Izadyar et al. 2003a).Recently, an elegant study demonstrated spermatogenesisand fertility after transplantation of primordialgerm cells between ra<strong>in</strong>bow trout and salmon,Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Male Germ Cell Transplantation 289Table 1. Male germ cell transplantation – a time l<strong>in</strong>e of progressYear of first report Major f<strong>in</strong>d<strong>in</strong>g References1994 Spermatogenesis and transmission of donor haplotype after germ cell transplantation <strong>in</strong> mice Br<strong>in</strong>ster and Avarbock (1994),Br<strong>in</strong>ster and Zimmermann (1994)1995 Male germ cell transplantation <strong>in</strong> rats Jiang and Short (1995)1996 Reconstitution of spermatogenesis after transplantation of frozen spermatogonial stem cells Avarbock et al. (1996)Rat spermatogenesis <strong>in</strong> mouse testis Clouthier et al. (1996)1998 Culture of mouse spermatogonial stem cells Nagano et al. (1998)1999 Transplantation of germ cells from domestic animals <strong>in</strong>to mouse testis Dobr<strong>in</strong>ski et al. (1999)2000 Restoration of fertility by germ cell transplantation <strong>in</strong> mice Ogawa et al. (2000)2001 Transgenic mice produced by retroviral transduction of male germ l<strong>in</strong>e stem cells Nagano et al. (2001a)2002 Production of transgenic rats by lentiviral transduction of male germ l<strong>in</strong>e stem cells Hamra et al. (2002)Germ cell transplantation <strong>in</strong>to X-irradiated monkey testes Schlatt et al. (2002)Germ cell transplantation <strong>in</strong> pigsHonaramooz et al. (2002a)Generation of a spermatogonial cell l<strong>in</strong>e <strong>in</strong> mice Feng et al. (2002)2003 Fertility and transmission of donor haplotype after germ cell transplantation <strong>in</strong> goats Honaramooz et al. (2003b)Transplantation of bov<strong>in</strong>e spermatogonial stem cellsIzadyar et al. (2003b)2004 Generation of pluripotent stem cells from neonatal mouse testis Kanatsu-Sh<strong>in</strong>ohara et al. (2004a)2006 Testicular germ cells can colonize sexually undifferentiated embryonic gonad and produceOkutsu et al. (2006)functional eggs <strong>in</strong> fishKnock-out mice produced after transplantation of cultured germ cells Kanatsu-Sh<strong>in</strong>ohara et al. (2006)2008 Adeno-associated virus-mediated transgenesis after germ cell transplantation <strong>in</strong> mice and goats Honaramooz et al. (2008)highlight<strong>in</strong>g the plasticity of the approach <strong>in</strong> fish(Okutsu et al. 2007).Male Germ Cell Transplantation <strong>in</strong> Non-RodentSpeciesWhile the majority of studies are performed <strong>in</strong> rodentmodels, germ cell transplantation is also applied to nonrodentspecies and has so far been reported <strong>in</strong> pigs,goats, cattle, monkeys and recently fish and chickens(Honaramooz et al. 2002a, 2003a; b; Schlatt et al. 2002;Izadyar et al. 2003b; Takeuchi et al. 2003; Yoshizakiet al. 2005; Lee et al. 2006; Mikkola et al. 2006; Okutsuet al. 2006; Trefil et al. 2006).The application of germ cell transplantation to nonrodentmammalian species was <strong>in</strong>itially difficult due todifferences <strong>in</strong> testicular anatomy and physiology. Whiledirect <strong>in</strong>jection of donor cells <strong>in</strong>to rodent sem<strong>in</strong>iferoustubules is possible via the efferent ducts, this is notfeasible <strong>in</strong> larger mammalian species. Instead, a comb<strong>in</strong>ationof ultrasound-guided cannulation of the centrallylocated rete testis with delivery of germ cells by gravityflow (Honaramooz et al. 2002a, 2003a) was shown to bea successful approach <strong>in</strong> large animals. When germ cellsfrom transgenic donor goats were transplanted <strong>in</strong>to thetestes of immunocompetent, pre-pubertal recipient animals,the recipients produced sperm carry<strong>in</strong>g the donorhaplotype and transmitted the donor genetic makeup tothe offspr<strong>in</strong>g. This provided proof-of-pr<strong>in</strong>ciple thatgerm cell transplantation results <strong>in</strong> donor-derived spermproduction and fertility also <strong>in</strong> a non-rodent species(Honaramooz et al. 2003b).In cattle, Izadyar et al. (2003b) first reported thetechnique of germ cell transfer and showed that transplantedautologous germ cells can <strong>in</strong>itiate spermatogenesis<strong>in</strong> the recipient testis. Subsequently, we have shownthat heterologous transplantation of bov<strong>in</strong>e germ cellscan succeed between breeds of cattle (Herrid et al. 2006).The success of germ cell transplantation requires theavailability of a stem cell niche <strong>in</strong> the recipient testis. Inrodents, the use of young mice and treat<strong>in</strong>g recipientswith GnRH agonists to suppress high <strong>in</strong>tratesticulartestosterone levels improved donor cell colonization(Ogawa et al. 1998, 1999b; Dobr<strong>in</strong>ski et al. 2001;Sh<strong>in</strong>ohara et al. 2001). Similarly, us<strong>in</strong>g pre-pubertalmales as recipients has proven a successful strategy <strong>in</strong>germ cell transplantation <strong>in</strong> pigs, goats and cattle(Honaramooz et al. 2003a; b; Herrid et al. 2006). Theefficiency of colonization of sem<strong>in</strong>iferous tubules by thetransplanted germ cells can be further improved ifthe recipient testes have little or no endogenousspermatogonia. Busulfan, a DNA-alkylat<strong>in</strong>g agent thatdestroys proliferat<strong>in</strong>g cells, is frequently used <strong>in</strong> rodentsto deplete recipient germ cells prior to germ celltransplantation. However, the steriliz<strong>in</strong>g dose of busulfanis species- and stra<strong>in</strong>-specific and treatment can belethal due to severe bone marrow depression (Ogawaet al. 1999b; Br<strong>in</strong>ster et al. 2003). While its use <strong>in</strong> adultboars has been described <strong>in</strong> comb<strong>in</strong>ation with methylprednisoloneto prevent thrombocytopenia (Mikkolaet al. 2006), <strong>in</strong> utero treatment of pigs dur<strong>in</strong>g a period ofhigh proliferation of foetal gonocytes provides a morepractical approach that resulted <strong>in</strong> depletion of germcells with no observed adverse effects on the piglets oron sow health or fertility (Honaramooz et al. 2005). Asan alternative to cytotoxic treatment, irradiation of thetestes will also result <strong>in</strong> depletion of endogenous germcells (Creemers et al. 2002; Schlatt et al. 2002). Provideda suitable radiation source is available, local testicularirradiation appears to be the method of choice <strong>in</strong> specieswhere the anatomical position of the testes facilitatesshield<strong>in</strong>g of the body from irradiation, thereby m<strong>in</strong>imiz<strong>in</strong>gsystemic effects. Fractionated testicular irradiationresulted <strong>in</strong> depletion of germ cells <strong>in</strong> goats, ramsand cats (Honaramooz et al. 2005; Oatley et al. 2005;Kim et al. 2006). Interest<strong>in</strong>gly, germ cell transplantation<strong>in</strong> rodents and perhaps also <strong>in</strong> older pigs and cattlerequires that donor and recipients are closely related orthat recipient animals are immunosuppressed (Izadyaret al. 2003b; Kanatsu-Sh<strong>in</strong>ohara et al. 2003b; Zhanget al. 2003; Mikkola et al. 2006), whereas germ celltransplantation <strong>in</strong> young pigs, goats and bulls wasÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


290 I Dobr<strong>in</strong>skisuccessful also between unrelated <strong>in</strong>dividuals (Honaramoozet al. 2002a, 2003a; b; Herrid et al. 2006). Thetestis is considered to be an immune-privileged site, butit is unclear why transplantation between unrelated,immunocompetent animals is possible <strong>in</strong> domesticanimal species but not <strong>in</strong> rodents. Nonetheless, thismakes the technique <strong>in</strong>f<strong>in</strong>itely more applicable <strong>in</strong> nonrodentspecies.Applications of Male Germ CellTransplantationTo study fundamental aspects of male germ l<strong>in</strong>e stem cellbiology and male fertilityGerm cell transplantation <strong>in</strong> rodents made it possible tostudy the stem cell niche <strong>in</strong> the testis and to characterizeputative spermatogonial stem cells (Parreira et al. 1998;Nagano et al. 1999; Ventela et al. 2002; Kubota et al.2003; Nagano 2003; Hamra et al. 2004). It was alsoconfirmed that sperm aris<strong>in</strong>g from transplanted donorgerm cells are capable of fertilization <strong>in</strong> vivo and <strong>in</strong> vitro(Br<strong>in</strong>ster and Avarbock 1994; Goossens et al. 2003,2006; Honaramooz et al. 2003b).As there are only few stem cells <strong>in</strong> the testis (Meistrichand van Beek 1993b; Tegelenbosch and de Rooij 1993),a great deal of work has been directed towards enrichmentof stem cells from the testis and their expansion <strong>in</strong>culture. Initially it was shown that selection of mousegerm cells for expression of a 6 - and b 1 -<strong>in</strong>tegr<strong>in</strong> <strong>in</strong> theabsence of c-kit receptor, as well as collection of cellsfrom experimentally <strong>in</strong>duced cryptorchid testes resulted<strong>in</strong> a significant enrichment for spermatogonial stem cells(Sh<strong>in</strong>ohara et al. 1999, 2000a; b). Expression of Thy-1,CD9 or Egr3 was also utilized as markers for enrichmentof mouse germ l<strong>in</strong>e stem cell populations (Kubotaet al. 2003; Hamra et al. 2004; Kanatsu-Sh<strong>in</strong>ohara et al.2004b). In contrast, germ cell isolation and enrichment<strong>in</strong> pigs and cattle relies on expression of different markerprote<strong>in</strong>s (Luo et al. 2006; Herrid et al. 2007).Nagano et al. (1998) showed first that stem cells couldbe ma<strong>in</strong>ta<strong>in</strong>ed <strong>in</strong> culture for a long period of time.Co-culture with embryonic fibroblast or bone marrowstromal cells, but not Sertoli cell l<strong>in</strong>es, and addition ofseveral growth factors known to be beneficial for cultureof other stem cell types or primordial germ cells, such asglial cell l<strong>in</strong>e-derived neurotrophic factor (GDNF),leukemia <strong>in</strong>hibitory factor (LIF), epidermal growthfactor (EGF) and basic fibroblast growth factor(bFGF), successfully ma<strong>in</strong>ta<strong>in</strong>ed mouse germ l<strong>in</strong>e stemcells <strong>in</strong> culture for vary<strong>in</strong>g periods of time (Naganoet al. 1998, 2003; Kanatsu-Sh<strong>in</strong>ohara et al. 2003a; Yehet al. 2007). Efficient long-term culture systems formouse and rat spermatogonial stem cells have now beendescribed (Kubota et al. 2004; Hamra et al. 2005;Kanatsu-Sh<strong>in</strong>ohara et al. 2005; Tenenhaus Dann et al.2006). The majority of the work to date was performedwith primary cultures of putative male germ l<strong>in</strong>e stemcells; yet, immortalized germ cell l<strong>in</strong>es have beendescribed <strong>in</strong> rat and mouse (Feng et al. 2000; van Peltet al. 2002; Hofmann et al. 2005).Recently, the developmental plasticity of culturedmale germ cells was highlighted by reports that pluripotentstem cells could be isolated from cultures ofneonatal and adult mouse testis cells (Kanatsu-Sh<strong>in</strong>oharaet al. 2004a; Guan et al. 2006; Seandel et al. 2007)and by the demonstration that adult male germ l<strong>in</strong>e stemcells can even give rise to fertilization competent eggswhen transplanted <strong>in</strong>to an undifferentiated gonad <strong>in</strong> fish(Okutsu et al. 2006, 2007).When presented with a phenotype of male <strong>in</strong>fertilitywith a defect <strong>in</strong> spermatogenesis, it is often difficult todeterm<strong>in</strong>e whether the defect orig<strong>in</strong>ates <strong>in</strong> the germ cells,or <strong>in</strong> the somatic components of the testis (Ogawa et al.2000). To characterize an unknown defect, standardexperimental design now <strong>in</strong>cludes reciprocal transplantationof germ cells from affected donors to wild-typetestes and vice versa. Us<strong>in</strong>g this approach, male germcell transplantation was successfully applied to characterizethe role of the c-kit receptor and its ligand stemcell factor <strong>in</strong> regulation of germ cell proliferation (Ohtaet al. 2000), to show that the defect associated with thejuvenile spermatogonial depletion (jsd) mutation was<strong>in</strong>herent to the germ cells (Boettger-Tong et al. 2000;Ohta et al. 2001), that germ cells do not requireoestrogen receptors (Mahato et al. 2000) or androgenreceptors (Johnston et al. 2001) for development, andthat germ cell differentiation is regulated by GDNF(Creemers et al. 2002; Yomogida et al. 2003), plzf(Buaas et al. 2004; Costoya et al. 2004) and CREMfunction (Wistuba et al. 2002). Transplantation of wildtypegerm cells <strong>in</strong>to the testes of Dazl null miceestablished that the somatic compartment of the Dazlnull testes rema<strong>in</strong>s functional (Rilianawati et al. 2003).In the rat, germ cell transplantation experiments elucidatedthe defect underly<strong>in</strong>g the as-mutation (Noguchiet al. 2002).To preserve the reproductive potential of geneticallyvaluable <strong>in</strong>dividuals by male germ cell transplantationMouse spermatogonial stem cells can be cryopreservedfor prolonged periods of time before transplantation,can still establish spermatogenesis <strong>in</strong> the recipient testis(Avarbock et al. 1996) and live off spr<strong>in</strong>g resulted fromspermatozoa produced after transplantation of frozenthawedmouse germ cells (Kanatsu-Sh<strong>in</strong>ohara et al.2003c; Frederickx et al. 2004). Therefore, germ celltransplantation could serve to restore male fertilityafter an <strong>in</strong>sult to the testis, such as irradiation orcytotoxic treatment <strong>in</strong> cancer patients (Orwig andSchlatt 2005). Germ cell transplantation has an advantageover the cryopreservation of sperm prior totreatment, <strong>in</strong> that it could be applied to pre-pubertalmales where sperm cannot be obta<strong>in</strong>ed or to adultmales rendered azoospermic or teratozoospermic by thedisease. The technique is also of great <strong>in</strong>terest <strong>in</strong>domestic or endangered animals for its potential topreserve genetic material from immature males that arelost before they reach puberty (Pukazhenthi et al. 2006;Dobr<strong>in</strong>ski and Travis 2007). Even when cryopreservationof sperm is possible from adult <strong>in</strong>dividuals, thepreserved sperm will provide a f<strong>in</strong>ite resource. Incontrast, cryopreserved germ cells will undergo geneticrecomb<strong>in</strong>ation after transplantation, thereby virtuallypreserv<strong>in</strong>g the entire genetic potential of the donormale. This will provide an <strong>in</strong>valuable advantage forÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Male Germ Cell Transplantation 291application of germ cell transplantation to the conservationof genetic diversity.To produce transgenic sperm by genetic manipulation ofthe male germ l<strong>in</strong>eTransgenic mice and rats have been generated by viraltransduction of germ cells prior to transplantation(Nagano et al. 2001a, 2002b; Hamra et al. 2002; Orwiget al. 2002; Honaramooz et al. 2008). Recently, theadvances <strong>in</strong> mouse germ cell culture even allowed forgene target<strong>in</strong>g and production of knock-out mice us<strong>in</strong>ggerm cell transplantation (Kanatsu-Sh<strong>in</strong>ohara et al.2006). This approach has tremendous potential <strong>in</strong>species where embryonic stem cell technology is notavailable and options to generate genetically modifiedanimals are <strong>in</strong>efficient. Current strategies to generategenetically modified large animals <strong>in</strong>clude pronuclearmicro<strong>in</strong>jection of DNA (Hammer et al. 1985) andnuclear transfer technology us<strong>in</strong>g modified donor cells(Schnieke et al. 1997; Cibelli et al. 1998; Chen et al.2002; Lai et al. 2002; Park et al. 2002), lentiviraltransduction of oocytes (Hofmann et al. 2003, 2004;McGrew et al. 2004; Whitelaw et al. 2004), as well assperm-mediated DNA transfer (Lazzereschi et al. 2000;Lavitrano et al. 2002). Yet, except for lentiviral transductionof oocytes, currently available technology isfrequently fraught with low efficiency and developmentalabnormalities <strong>in</strong> the few result<strong>in</strong>g off-spr<strong>in</strong>g, mak<strong>in</strong>gthe approach of us<strong>in</strong>g germ cell transplantation a veryvaluable alternative. The approach is schematicallyillustrated <strong>in</strong> Fig. 1. Introduction of a genetic modificationprior to fertilization will circumvent problemsassociated with manipulation of gametes and earlyembryos and developmental abnormalities associatedwith nuclear reprogramm<strong>in</strong>g. We recently providedproof-of-pr<strong>in</strong>ciple for transgenesis through the malegerm l<strong>in</strong>e <strong>in</strong> a non-rodent animal by demonstrat<strong>in</strong>gtransgene transmission after transduction of goat germcells prior to transplantation with a GFP marker genecarried by an adeno-associated viral vector (Honaramoozet al. 2008). While still experimental, transgenesisthrough the male germ l<strong>in</strong>e is expected to provide anefficient alternative to exist<strong>in</strong>g technology for the <strong>in</strong>troductionof genetic modifications <strong>in</strong> domestic animals.Fig. 1. Schematic representation of transgene <strong>in</strong>troduction throughmale germ cell transplantationConclusionsGerm cell transplantation was <strong>in</strong>itially developed <strong>in</strong>rodents <strong>in</strong> 1994. Application to a domestic animalspecies was first reported <strong>in</strong> the pig and subsequently <strong>in</strong>goats and cattle, chickens and fish. Male germ celltransplantation allows us to explore basic biologicalaspects of male germ l<strong>in</strong>e stem cells and testis function aswell as potential causes of male <strong>in</strong>fertility. Practicalapplications <strong>in</strong>clude the <strong>in</strong>troduction of genetic modifications<strong>in</strong>to the germ l<strong>in</strong>e of domestic animals, and thepreservation of fertility <strong>in</strong> rare and endangered animalsand <strong>in</strong> patients undergo<strong>in</strong>g potentially steriliz<strong>in</strong>g treatmentsfor cancer therapy. Transplantation of germ cellswill cont<strong>in</strong>ue to significantly enhance our understand<strong>in</strong>gof testis function and our ability to control and preservemale fertility.AcknowledgementWork from the author’s laboratory presented here was supported by5R01RR017359-05 (NCRR ⁄ NIH) and 2R42-HD044780-02 (NIH ⁄ -NICHD).ReferencesAvarbock MR, Br<strong>in</strong>ster CJ, Br<strong>in</strong>ster RL, 1996: Reconstitutionof spermatogenesis from frozen spermatogonial stem cells.Nat Med 2, 693–696.Boettger-Tong H, Johnston DS, Russell LD, Griswold MD,Bishop CE, 2000: Sem<strong>in</strong>iferous tubules from mutant jsd(juvenile spermatogonial depletion) mice are capable ofsupport<strong>in</strong>g transplanted spermatogenesis. Abstract. BiolReprod 62(Suppl. 1), 108.Br<strong>in</strong>ster RL, Avarbock MR, 1994: Germl<strong>in</strong>e transmission ofdonor haplotype follow<strong>in</strong>g spermatogonial transplantation.Proc Natl Acad Sci U S A 91, 11303–11307.Br<strong>in</strong>ster RL, Zimmermann JW, 1994: Spermatogenesis follow<strong>in</strong>gmale germ-cell transplantation. Proc Natl Acad SciUSA91, 11298–11302.Br<strong>in</strong>ster CJ, Ryu B-Y, Avarbock MR, Karagenc L, Br<strong>in</strong>sterRL, Orwig KE, 2003: Restoration of fertility by germ celltransplantation requires effective recipient preparation. BiolReprod 69, 412–420.Buaas FW, Kirsh AL, Sharma M, McLean DJ, Morris JL,Griswold MD, de Rooij DG, Braun RE, 2004: Plzf isrequired <strong>in</strong> adult male germ cells for stem cell self-renewal.Nat Genet 36, 647–652.Chen S-H, Vaught TD, Monahan JA, Boone J, Emslie E,Jobst PM, Lamborn AE, Schnieke A, Robertson L, ColmanA, Dai Y, Polejaeva IA, Ayares DL, 2002: Efficientproduction of transgenic cloned calves us<strong>in</strong>g preimplantationscreen<strong>in</strong>g. Biol Reprod 67, 1488–1492.Cibelli JB, Stice SL, Golueke PG, Kane JJ, Jerry J, BlackwellC, Ponce de Leon FA, Robl JM, 1998: Cloned transgenicclaves produced from nonquiescent fetal fibroblasts. Science280, 1256–1258.Clermont Y, 1972: K<strong>in</strong>etics of spermatogenesis <strong>in</strong> mammals:sem<strong>in</strong>iferous epithelium cycle and spermatogonial renewal.Physiol Rev 52, 198–236.Clouthier DE, Avarbock MR, Maika SD, Hammer RE,Br<strong>in</strong>ster RL, 1996: Rat spermatogenesis <strong>in</strong> mouse testis.Nature 381, 418–421.Costoya JA, Hobbs RM, Barna M, Cattoretti G, Manova K,Sukhwani M, Orwig KE, Wolgemuth DJ, Pandolfi PP, 2004:Essential role of Plzf <strong>in</strong> ma<strong>in</strong>tenance of spermatogonial stemcells. Nat Genet 36, 653–659.Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


292 I Dobr<strong>in</strong>skiCreemers LB, Meng X, den Ouden K, van Pelt AMM, IzadyarF, Santoro M, de Rooij DG, Sariola H, 2002: Transplantationof germ cells from glial cell l<strong>in</strong>e-derived neurotrophicfactor-overexpress<strong>in</strong>g mice to host testes depleted of endogenousspermatogenesis by fractionated irradiation. BiolReprod 66, 1579–1584.Dobr<strong>in</strong>ski I, Travis AJ, 2007: Germ cell transplantation for thepropagation of companion animals, non-domestic andendangered species. Reprod Fertil Dev 19, 732–739.Dobr<strong>in</strong>ski I, Avarbock MR, Br<strong>in</strong>ster RL, 1999: Transplantationof germ cells from rabbits and dogs <strong>in</strong>to mouse testes.Biol Reprod 61, 1331–1339.Dobr<strong>in</strong>ski I, Avarbock MR, Br<strong>in</strong>ster RL, 2000: Germ celltransplantation from large domestic animals <strong>in</strong>to mousetestes. Mol Rep Dev 57, 270–279.Dobr<strong>in</strong>ski I, Ogawa T, Avarbock MR, Br<strong>in</strong>ster RL, 2001:Effect of the GnRH-agonist leuprolide on colonization ofrecipient testes by donor spermatogonial stem cells aftertransplantation <strong>in</strong> mice. Tissue Cell 33, 200–207.Feng B, Inaba M, Lian Z, Cui Y, Toki J, Ito T, J<strong>in</strong> T, Fan T,Yang G, Yu C, Kushida T, Ikehara S, 2000: Development ofmouse dendritic cells from l<strong>in</strong>eage-negative c-kit


Male Germ Cell Transplantation 293Kanatsu-Sh<strong>in</strong>ohara M, Inoue K, Lee J, Yoshimoto M,Ogonuki N, Miki H, Baba S, Kato T, Kazuki Y, ToyokuniS, Toyoshima M, Niwa O, Oshimura M, Heike T, NakahataT, Ish<strong>in</strong>o F, Ogura A, Sh<strong>in</strong>ohara T, 2004a: Generation ofpluripotent stem cells from neonatal mouse testis. Cell 119,1001–1012.Kanatsu-Sh<strong>in</strong>ohara M, Toyokuni S, Sh<strong>in</strong>ohara T, 2004b: CD9is a surface marker on mouse and rat male germl<strong>in</strong>e stemcells. Biol Reprod 70, 70–75.Kanatsu-Sh<strong>in</strong>ohara M, Miki H, Inoue K, Ogonuki N,Toyokuni S, Ogura A, Sh<strong>in</strong>ohara T, 2005: Long-termculture of mouse male germl<strong>in</strong>e stem cells under serum- orfeeder-free conditions. Biol Reprod 72, 985–991.Kanatsu-Sh<strong>in</strong>ohara M, Ikawa M, Takehashi M, Ogonuki N,Miki H, Inoue K, Kazuki Y, Lee J, Toyokuni S, OshimuraM, Ogura A, Sh<strong>in</strong>ohara T, 2006: Production of knockoutmice by random or targeted mutagenesis <strong>in</strong> spermatogonialstem cells. Proc Natl Acad Sci U S A 103, 8018–8023.Kim Y, Selvaraj V, Dobr<strong>in</strong>ski I, Lee H, McEntee MC, TravisAJ, 2006: Recipient preparation and mixed germ cellisolation for spermatogonial stem cell transplantation <strong>in</strong>domestic cats. J Androl 27, 248–256.Kubota H, Avarbock MR, Br<strong>in</strong>ster RL, 2003: Spermatogonialstem cells share some, but not all, phenotypic and functionalcharacteristics with other stem cells. Proc Natl Acad Sci U SA 100, 6487–6492.Kubota H, Avarbock MR, Br<strong>in</strong>ster RL, 2004: Cultureconditions and s<strong>in</strong>gle growth factors affect fate determ<strong>in</strong>ationof mouse spermatogonial stem cells. Biol Reprod 71,722–731.Lai L, Kolber-Simonds D, Park K-W, Cheong H-T, Greenste<strong>in</strong>JL, Im G-S, Samuel M, Bonk A, Rieke A, Day BN,Murphy CN, Carter DB, Hawley RJ, Prather RS, 2002:Production of a-1,3-galactosyltransferase knockout pigs bynuclear transfer clon<strong>in</strong>g. Science 295, 1089–1092.Lavitrano M, Bacci ML, Forni M, Lazzereschi D, Di StefanoC, Fioretti D, Giancotti P, Marfe G, Pucci L, Renzi L,Wang H, Stoppacciaro A, Stassi G, Sargiacomo M,S<strong>in</strong>ibaldi P, Turchi V, Giovannoni R, Della Casa G, SerenE, Rossi G, 2002: Efficient production by sperm-mediatedgene transfer of human decay accelerat<strong>in</strong>g factor (hDAF)transgenic pigs for xenotransplantation. Proc Natl Acad SciUSA99, 14230–14235.Lazzereschi D, Forni M, Cappello F, Bacci ML, Di Stefano C,Marfe G, Giancotti P, Renzi L, Wang HJ, Rossi M, DellaCasa G, Pretagost<strong>in</strong>i R, Frati G, Bruzzone P, Stassi G,Stoppacciaro A, Turchi V, Cortes<strong>in</strong>i R, S<strong>in</strong>ibaldi P, Frati L,Lavitrano M, 2000: Efficiency of transgenesis us<strong>in</strong>g spermmediatedgene transfer: generation of hDAF transgenic pigs.Transplant Proc 32, 892–894.Lee YM, Jung JG, Kim JN, Park TS, Kim TM, Sh<strong>in</strong> SS, KangDK, Lim JM, Han JY, 2006: A testis-mediated germl<strong>in</strong>echimera production based on transfer of chicken testicularcells directly <strong>in</strong>to heterologous testes. Biol Reprod 75, 380–386.Luo J, Megee SO, Rathi R, Dobr<strong>in</strong>ski I, 2006: Prote<strong>in</strong> geneproduct 9.5 is a spermatogonia-specific marker <strong>in</strong> the pigtestis: application to enrichment and culture of porc<strong>in</strong>espermatogonia. Mol Reprod Dev 73, 1531–1540.Mahato D, Gould<strong>in</strong>g EH, Korach KS, Eddy EM, 2000:Spermatogenic cells do not require estrogen receptor-a fordevelopment or function. Endocr<strong>in</strong>ology 141, 1273–1276.McGrew MJ, Sherman A, Ellard FM, Lillico SG, GilhooleyHJ, K<strong>in</strong>gsman AJ, Mitrophanous KA, Sang H, 2004:Efficient production of germl<strong>in</strong>e transgenic chickens us<strong>in</strong>glentiviral vectors. EMBO Rep 5, 728–733.Meistrich ML, van Beek MEAB, 1993a: Spermatogonial stemcells. In: Desjard<strong>in</strong>s C, Ew<strong>in</strong>g LL (eds), Cell and MolecularBiology of the Testis. Oxford University Press, New York,pp. 266–295.Meistrich ML, van Beek MEAB, 1993b: Spermatogonial stemcells: assess<strong>in</strong>g their survival and ability to produce differentiatedcells. Meth Toxicol 3A, 106–123.Mikkola M, Sironen A, Kopp C, Taponen J, Sukura A, VilkkiJ, Katila T, Andersson M, 2006: Transplantation of normalboar testicular cells resulted <strong>in</strong> complete focal spermatogenesis<strong>in</strong> a boar affected by the immotile short-tail spermdefect. Reprod Domest Anim 41, 124–128.Nagano M, 2003: Hom<strong>in</strong>g efficiency and proliferation k<strong>in</strong>eticsof male germ l<strong>in</strong>e stem cells follow<strong>in</strong>g transplantation <strong>in</strong>mice. Biol Reprod 69, 701–707.Nagano M, Avarbock MR, Leonida EB, Br<strong>in</strong>ster CJ, Br<strong>in</strong>sterRL, 1998: Culture of mouse spermatogonial stem cells.Tissue Cell 30, 389–397.Nagano M, Avarbock MR, Br<strong>in</strong>ster RL, 1999: Pattern andk<strong>in</strong>etics of mouse donor spermatogonial stem cell colonization<strong>in</strong> recipient testes. Biol Reprod 60, 1429–1436.Nagano M, Br<strong>in</strong>ster CJ, Orwig KE, Ryu B-Y, Avarbock MR,Br<strong>in</strong>ster RL, 2001a: Transgenic mice produced by retroviraltransduction of male germ-l<strong>in</strong>e stem cells. Proc Natl AcadSci U S A 98, 13090–13095.Nagano M, McCarrey JR, Br<strong>in</strong>ster RL, 2001b: Primatespermatogonial stem cells colonize mouse testes. BiolReprod 64, 1409–1416.Nagano M, Patrizio P, Br<strong>in</strong>ster RL, 2002a: Long-term survivalof human spermatogonial stem cells <strong>in</strong> mouse testes. FertilSteril 78, 1225–1233.Nagano M, Watson DJ, Ryu B-Y, Wolfe JH, Br<strong>in</strong>ster RL,2002b: Lentiviral vector transduction of male germ l<strong>in</strong>e stemcells <strong>in</strong> mice. FEBS Lett 524, 111–115.Nagano M, Ryu B-Y, Br<strong>in</strong>ster CJ, Avarbock MR, Br<strong>in</strong>sterRL, 2003: Ma<strong>in</strong>tenance of mouse male germ l<strong>in</strong>e stem cells<strong>in</strong> vitro. Biol Reprod 68, 2207–2214.Noguchi J, Toyama Y, Yuasa S, Kikuchi K, Kaneko M, 2002:Hereditary defects <strong>in</strong> both germ cells and the blood–testisbarrier system <strong>in</strong> as-mutant rats: evidence from spermatogonialtransplantation and tracer-permeability analysis. BiolReprod 67, 880–888.Oatley JM, Tibary A, de Avila DM, Wheaton JE, McLean DJ,Reeves JJ, 2005: Changes <strong>in</strong> spermatogenesis and endocr<strong>in</strong>efunction <strong>in</strong> the ram testis due to irradiation and activeimmunization aga<strong>in</strong>st lute<strong>in</strong>iz<strong>in</strong>g hormone-releas<strong>in</strong>g hormone.J Anim Sci 83, 604–612.Ogawa T, Dobr<strong>in</strong>ski I, Avarbock MR, Br<strong>in</strong>ster RL, 1998:Leuprolide, a gonadotrop<strong>in</strong>-releas<strong>in</strong>g hormone agonist,enhances colonization after spermatogonial transplantation<strong>in</strong>to mouse testes. Tissue Cell 30, 583–588.Ogawa T, Dobr<strong>in</strong>ski I, Avarbock MR, Br<strong>in</strong>ster RL, 1999a:Xenogeneic spermatogenesis follow<strong>in</strong>g transplantation ofhamster germ cells to mouse testes. Biol Reprod 60, 515–521.Ogawa T, Dobr<strong>in</strong>ski I, Br<strong>in</strong>ster RL, 1999b: Recipient preparationis critical for spermatogonial transplantation <strong>in</strong> therat. Tissue Cell 31, 461–472.Ogawa T, Dobr<strong>in</strong>ski I, Avarbock MR, Br<strong>in</strong>ster RL, 2000:Transplantation of male germ l<strong>in</strong>e stem cells restores fertility<strong>in</strong> <strong>in</strong>fertile mice. Nat Med 6, 29–34.Ohta H, Yomogida K, Dohmae K, Nishimune Y, 2000:Regulation of proliferation and differentiation <strong>in</strong> spermatogonialstem cells: the role of c-kit and its ligand SCF.Development 127, 2125–2131.Ohta H, Yomogida K, Tadokoro Y, Tohda A, Dohmae K,Nishimune Y, 2001: Defect <strong>in</strong> germ cells, not support<strong>in</strong>gcells, is the cause of male <strong>in</strong>fertility <strong>in</strong> the jsd mutant mouse:proliferation of spermatogonial stem cells without differentiation.Int J Androl 24, 15–23.Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


294 I Dobr<strong>in</strong>skiOkutsu T, Suzuki K, Takeuchi Y, Takeuchi T, Yoshizaki G,2006: Testicular germ cells can colonize sexually undifferentiatedembryonic gonad and produce functional eggs <strong>in</strong>fish. Proc Natl Acad Sci U S A 103, 2725–2729.Okutsu T, Shik<strong>in</strong>a S, Kanno M, Takeuchi Y, Yoshizaki G,2007: Production of trout offspr<strong>in</strong>g from triploid salmonparents. Science 317, 1517.Orwig KE, Schlatt S, 2005: Cryopreservation and transplantationof spermatogonia and testis tissue for preservation ofmale fertility. J Natl Cancer Inst Monogr 34, 56.Orwig KE, Avarbock MR, Br<strong>in</strong>ster RL, 2002: Retrovirusmediatedmodification of male germl<strong>in</strong>e stem cells <strong>in</strong> rats.Biol Reprod 67, 874–879.Park K-W, Lai L, Cheong H-T, Cabot R, Sun Q-Y, Wu G,Rucker EB, Durtschi D, Bonk A, Samuel M, Rieke A, DayBN, Murphy CN, Carter DB, Prather RS, 2002: Mosaicgene expression <strong>in</strong> nuclear transfer-derived embryos and theproduction of cloned transgenic pigs from ear-derivedfibroblasts. Biol Reprod 66, 1001–1005.Parreira GC, Ogawa T, Avarbock MR, Franca LR, Br<strong>in</strong>sterRL, Russell LD, 1998: Development of germ cell transplants<strong>in</strong> mice. Biol Reprod 59, 1360–1370.van Pelt aMM, Roepers-Gajadien HL, Gademan IS, CreemersLB, de Rooij DG, van Dissel-Emiliani FMF, 2002: Establishmentof cell l<strong>in</strong>es with rat spermatogonial stem cellcharacteristics. Endocr<strong>in</strong>ology 143, 1845–1850.Pukazhenthi BS, Comizzoli P, Travis AJ, Wildt DE, 2006:Applications of emerg<strong>in</strong>g technologies to the study andconservation of threatened and endangered species. ReprodFertil Dev 18, 77–90.Rilianawati R, Speed R, Taggart M, Cooke HJ, 2003:Spermatogenesis <strong>in</strong> testes of Dazl null mice after transplantationof wild-type germ cells. <strong>Reproduction</strong> 126, 599–604.Russell LD, Ettl<strong>in</strong> RA, S<strong>in</strong>haHikim AP, Clegg ED, 1990:Mammalian spermatogenesis. In: Russell LD, Ettl<strong>in</strong> RA,S<strong>in</strong>haHikim AP, Clegg ED (eds), Histological and HistopathologicalEvaluation of the Testis. Cache River Press,Clearwater, pp. 1–40.Schlatt S, Foppiani L, Rolf C, We<strong>in</strong>bauer GF, Nieschlag E,2002: Germ cell transplantation <strong>in</strong>to X-irradiated monkeytestes. Hum Reprod 17, 55–62.Schnieke AE, K<strong>in</strong>d AJ, Ritchie WA, Mycock K, Scott AR,Ritchie M, Wilmut I, Colman A, Campbell KHS, 1997:Human factor IX transgenic sheep produced by transfer ofnuclei from transfected fetal fibroblasts. Science 278, 2130–2133.Seandel M, James D, Shmelkov SV, Falciatori I, Kim J,Chavala S, Scherr DS, Zhang F, Torres R, Gale NW,Yancopoulos GD, Murphy A, Valenzuela DM, Hobbs RM,Pandolfi PP, Rafi S, 2007: Generation of functional multipotentadult stem cells from GPR125+ germl<strong>in</strong>e progenitors.Nature 449, 346–352.Sh<strong>in</strong>ohara T, Avarbock MR, Br<strong>in</strong>ster RL, 1999: b 1 - and a 6 -<strong>in</strong>tegr<strong>in</strong> are surface markers on mouse spermatogonial stemcells. Proc Natl Acad Sci U S A 96, 5504–5509.Sh<strong>in</strong>ohara T, Avarbock MR, Br<strong>in</strong>ster RL, 2000a: Functionalanalysis of spermatogonial stem cells <strong>in</strong> Steel and cryptorchid<strong>in</strong>fertile mouse models. Dev Biol 220, 401–411.Sh<strong>in</strong>ohara T, Orwig KE, Avarbock MR, Br<strong>in</strong>ster RL, 2000b:Spermatogonial stem cell enrichment by multiparameterselection of mouse testis cells. Proc Natl Acad Sci U S A 97,8346–8351.Sh<strong>in</strong>ohara T, Orwig KE, Avarbock MR, Br<strong>in</strong>ster RL, 2001:Remodel<strong>in</strong>g of the postnatal mouse testis is accompanied bydramatic changes <strong>in</strong> stem cell number and niche accessibility.Proc Natl Acad Sci U S A 98, 6186–6191.Sh<strong>in</strong>ohara T, Orwig KE, Avarbock MR, Br<strong>in</strong>ster RL, 2003:Restoration of spermatogenesis <strong>in</strong> <strong>in</strong>fertile mice by Sertolicell transplantation. Biol Reprod 68, 1064–1071.Sh<strong>in</strong>ohara T, Kato M, Takehashi M, Lee J, Chuma S,Nakatsuji N, Kanatsu-Sh<strong>in</strong>ohara M, Hirabayashi M, 2006:Rats produced by <strong>in</strong>traspecies spermatogonial transplantation<strong>in</strong> mice and <strong>in</strong> vitro micro<strong>in</strong>sem<strong>in</strong>ation. Proc Natl AcadSci U S A 103, 13624–13628.Takeuchi Y, Yoshizaki G, Takeuchi T, 2003: Generation oflive fry from <strong>in</strong>traperitoneally transplanted primordial germcells <strong>in</strong> ra<strong>in</strong>bow trout. Biol Reprod 69, 1142–1149.Tegelenbosch RAJ, de Rooij DG, 1993: A quantitative studyof spermatogonial multiplication and stem cell renewal <strong>in</strong>the C3H ⁄ 101 F 1 hybrid mouse. Mut Res 290, 193–200.Tenenhaus Dann C, Alvadro AL, Hammer RE, Garbers DL,2006: Heritable and stable gene knockdown <strong>in</strong> rats. ProcNatl Acad Sci U S A 103, 11246–11251.Trefil P, Micakova A, Mucksova J, Hejnar J, Poplste<strong>in</strong> M,Bakst MR, Kal<strong>in</strong>a J, Brillard J-P, 2006: Restoration ofspermatogenesis and male fertility by transplantation ofdispersed testicular cells <strong>in</strong> the chicken. Biol Reprod 75,575–581.Ventela S, Ohta H, Parv<strong>in</strong>en M, Nishimune Y, 2002: Developmentof stages of the cycle <strong>in</strong> mouse sem<strong>in</strong>iferousepithelium after transplantation of green fluorescent prote<strong>in</strong>-labeledspermatogonial stem cells. Biol Reprod 66,1422–1429.Whitelaw CBA, Radcliffe PA, Ritchie WA, Carlisle A, EllardFM, Pena RN, Rowe J, Clark AJ, K<strong>in</strong>g TJ, MitrophanousKA, 2004: Efficient generation of transgenic pigs us<strong>in</strong>gequ<strong>in</strong>e <strong>in</strong>fectious anaemia virus (EIAV) derived vector.FEBS Lett 571, 233–236.Wistuba J, Schlatt S, Cantauw C, von Schonfeldt V, NieschlagE, Behr R, 2002: Transplantation of wild-type spermatogonialeads to complete spermatogenesis <strong>in</strong> testes of cyclic3’,5’-adenos<strong>in</strong>e monophosphate response element modulator-deficientmice. Biol Reprod 67, 1052–1057.Yeh JR, Zhang X, Nagano M, 2007: Establishment of a shortterm<strong>in</strong> vitro assay for mouse spermatogonial stem cells. BiolReprod 77, 904.Yomogida K, Yagura Y, Tadokoro Y, Nishimune Y, 2003:Dramatic expansion of germ<strong>in</strong>al stem cells by ectopicallyexpressed human glial cell l<strong>in</strong>e-derived neurotrophic factor<strong>in</strong> mouse Sertoli cells. Biol Reprod 69, 1303–1307.Yoshizaki G, Tago Y, Takeuchi Y, Sawatari E, Kobayashi T,Takeuchi T, 2005: Green fluorescent prote<strong>in</strong> label<strong>in</strong>g ofprimordial germ cells us<strong>in</strong>g a nontransgenic method and itsapplication for germ cell transplantation <strong>in</strong> salmonidae. BiolReprod 73, 88–93.Zhang Z, Renfree MB, Short RV, 2003: Successful <strong>in</strong>tra- and<strong>in</strong>terspecific male germ cell transplantation <strong>in</strong> the rat. BiolReprod 68, 961–967.Author’s address (for correspondence): I Dobr<strong>in</strong>ski, School ofVeter<strong>in</strong>ary Medic<strong>in</strong>e, Center for Animal Transgenesis and Germ CellResearch, University of Pennsylvania, Kennett Square, PA 19348,USA. E-mail: dobr<strong>in</strong>sk@vet.upenn.eduConflict of <strong>in</strong>terest: The author received grants from NIH whosupported the work described <strong>in</strong> this paper.Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 295–301 (2008); doi: 10.1111/j.1439-0531.2008.01177.xISSN 0936-6768Sexual Maturation <strong>in</strong> the BullN Rawl<strong>in</strong>gs 1 , ACO Evans 2 , RK Chandolia 3 and ET Bagu 41 Department of Veter<strong>in</strong>ary Biomedical Sciences, University of Saskatchewan, Saskatoon, Saskatchewan, Canada; 2 School of Agriculture FoodSciences and Veter<strong>in</strong>ary Medic<strong>in</strong>e, University College Dubl<strong>in</strong>, Dubl<strong>in</strong>, Ireland; 3 Institute fu¨r Reproducktions Mediz<strong>in</strong>, Universitat Mu¨nster, Mu¨nster,Germany; 4 Centre de recherche´ en biologie de la <strong>Reproduction</strong>, Laurier, Quebec, CanadaContentsIn this review, we describe the process of sexual maturation<strong>in</strong> the bull calf. The testes of the bull grow relatively slowlyuntil approximately 25 weeks of age and then a rapid phaseof growth occurs until puberty, at 37–50 weeks of age.Dur<strong>in</strong>g the early postnatal phase of slower growth of thetestis pre-spermatogonia and some spermatogonia are established,adult Leydig cells appear and undifferentiated Sertolicells are produced. The rapid testicular growth, after25 weeks of age, consists of marked <strong>in</strong>creases <strong>in</strong> thediameter and length of the sem<strong>in</strong>iferous tubules, dramaticproliferation and differentiation of germ cells, with maturespermatozoa occurr<strong>in</strong>g between 32 and 40 weeks of age.The adult Leydig cell population is largely <strong>in</strong> place by30 weeks of age and that of Sertoli cells by 30–40 weeksof age. Serum concentrations of LH <strong>in</strong>crease from 4 to5 weeks of age, to an early postnatal peak at 12–16 weeksof age, followed by a decl<strong>in</strong>e to 25 weeks of age.Serum FSH concentrations are high postnatally, decl<strong>in</strong><strong>in</strong>gto approximately 25 weeks of age. Serum testosteroneconcentrations <strong>in</strong>crease dur<strong>in</strong>g the phase of rapid testiculargrowth. Hypothalamic opioidergic <strong>in</strong>hibition may abatetransiently to allow the early postnatal <strong>in</strong>crease <strong>in</strong> LHsecretion, while testicular androgenic negative feedbackprobably contributes to the decl<strong>in</strong>e <strong>in</strong> gonadotrop<strong>in</strong>secretion to 25 weeks of age. Several l<strong>in</strong>es of study haveled us to suggest that early postnatal gonadotrop<strong>in</strong> secretionis pivotal <strong>in</strong> <strong>in</strong>itiat<strong>in</strong>g the process of sexual maturation <strong>in</strong>the bull calf.IntroductionA fairly widely accepted end po<strong>in</strong>t for pubertal development<strong>in</strong> the bull was def<strong>in</strong>ed by Wolf et al. (1965) asan ejaculate that conta<strong>in</strong>ed at least 50 million spermwith no less than 10% progressive motility. In bullsfrom European breeds puberty is reached over a rangeof approximately 37–50 weeks of age, with dairy breedsmatur<strong>in</strong>g somewhat earlier than beef breeds (Lunstraet al. 1978; Amann 1983; Evans et al. 1995). Althoughscrotal circumference at puberty, as def<strong>in</strong>ed by Wolfet al. (1965), varies somewhat between breeds, a scrotalcircumference of 28 cm is often used as the po<strong>in</strong>t of, orage at puberty (Wolf et al. 1965; Lunstra et al. 1978).However, sperm output and quality <strong>in</strong>crease for sometime after the age of puberty (Almquist and Cunn<strong>in</strong>gham1967; Lunstra and Echternkamp 1982). Thispresent review will focus on development of the reproductivesystem <strong>in</strong> the bull, with particular emphasis onregulatory mechanisms. As much as possible we havetried to describe trends and mechanisms for bulls <strong>in</strong>general, regardless of breed, season of birth andnutrition.Testicular Growth and Development of theReproductive TractTesticular growth follows a sigmoid pattern <strong>in</strong> the bullcalf, with more rapid growth after approximately25 weeks of age through puberty, slow<strong>in</strong>g as the bullachieves adult sperm output (Fig. 1; Abdel-Raouf 1960;Macmillan and Hafs 1968; Amann 1983). Sem<strong>in</strong>iferoustubule diameter <strong>in</strong>creases gradually until 20–25 weeks ofage and then more rapidly (Abdel-Raouf 1960; Macmillanand Hafs 1969; Amann 1983; Evans et al. 1996).Increased diameter and length of the tubules accountsfor most of the <strong>in</strong>crease <strong>in</strong> testis size up to 32 weeks ofage, but tubule length predom<strong>in</strong>ates dur<strong>in</strong>g later stagesof development (Curtis and Amann 1981). Lum<strong>in</strong>ationof the tubules occurs at approximately 25 weeks of age(Evans et al. 1996). Fetal Leydig cells degenerate byapproximately 8 weeks after birth and adult Leydig cellnumbers <strong>in</strong>crease rapidly from 4 to 30 weeks of age(Wrobel 1990). Undifferentiated Sertoli cells proliferatefrom 4 weeks of age up until 13–20 weeks of age andthen the number decreases as these cells differentiate<strong>in</strong>to mature, adult Sertoli cells; differentiation of matureSertoli cells appears to be complete somewhere between30 and 40 weeks of age (Abdel-Raouf 1960; Curtis andAmann 1981; S<strong>in</strong>owatz and Amselgruber 1986; Wrobel2000; Bagu et al. 2006a). The number of Sertoli cells is acritical determ<strong>in</strong>ant of daily sperm production <strong>in</strong> thebull (Berndtson et al. 1987). At birth the solid sem<strong>in</strong>iferoustubules or chords conta<strong>in</strong> the primordial germcells or gonocytes; these largely disappear by 30 weeksof age (Curtis and Amann 1981; Wrobel 2000; Baguet al. 2006a). Proliferation of pre-spermatogonia andsome spermatogonia occurs from 4 to 5 weeks of ageonwards (Abdel-Raouf 1960; Curtis and Amann 1981;Wrobel 1990; Evans et al. 1996; Bagu et al. 2006a). Prespermatogonialcell numbers decrease after 24 weeksof age (Curtis and Amann 1981), but numbers ofspermatogonia <strong>in</strong>crease rapidly until 44 weeks of age(Abdel-Raouf 1960). Primary spermatocytes appear atapproximately 20 weeks of age, secondary spermatocytesat 20–30 weeks of age, round spermatids between25 and 30 weeks of age, long spermatids at 25–35 weeksof age and f<strong>in</strong>ally mature spermatozoa are clearly seenbetween 32 and 40 weeks of age (Abdel-Raouf 1960;Macmillan and Hafs 1968; Curtis and Amann 1981;Evans et al. 1996; Bagu et al. 2006a). It is <strong>in</strong>terest<strong>in</strong>gthat rapid testicular growth, differentiation and development,occur after 20–25 weeks of age.As with testicular development, epididymal growth isslow <strong>in</strong>itially, but <strong>in</strong>creases rapidly after 28 weeks of ageÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


296 N Rawl<strong>in</strong>gs, ACO Evans, RK Chandolia and ET BaguFig. 1. Growth and developmentof some segments of the reproductivesystem of the bull calf frombirth to puberty based on Abdel-Raouf (1960), Macmillan and Hafs(1969), Curtis and Amann (1981),S<strong>in</strong>owatz and Amselgruber (1986),Evans et al. (1996), Wrobel (2000)and Bagu et al. (2006a). Horizontall<strong>in</strong>es <strong>in</strong>dicate approximate periodsfor the activities shown; l<strong>in</strong>es witharrow heads <strong>in</strong>dicate start po<strong>in</strong>tsfor cont<strong>in</strong>ued activities(Fig. 1; Abdel-Raouf 1960; Macmillan and Hafs 1969).Rapid development of the vesicular glands occurs afterapproximately 2 months of age (L<strong>in</strong>dner and Mann1960; Macmillan and Hafs 1969; Chandolia et al. 1997c)<strong>in</strong> the bull. Content of the two pr<strong>in</strong>ciple secretoryproducts of the vesicular glands, fructose and citric acid,<strong>in</strong>crease markedly <strong>in</strong> bulls after 20–24 weeks of age(Abdel-Raouf 1960; Macmillan and Hafs 1969) but <strong>in</strong>one report (L<strong>in</strong>dner and Mann 1960) this <strong>in</strong>crease wasseen as early as 8 weeks of age.Endocr<strong>in</strong>e Changes Dur<strong>in</strong>g Sexual Maturation;Alignment with Testicular DevelopmentSerum concentrations of LH are low <strong>in</strong> the first 4–5 weeks after birth but <strong>in</strong>crease thereafter to a peak atapproximately 12–16 weeks of age and then concentrationsdecrease to approximately 25 weeks of age,rema<strong>in</strong><strong>in</strong>g low but variable through the time of puberty(Fig. 2; Rawl<strong>in</strong>gs et al. 1978; Lacroix and Pelletier1979; McCarthy et al. 1979; Amann and Walker 1983;Rodriguez and Wise 1989; Rawl<strong>in</strong>gs and Evans 1995;Fig. 2. The temporal patterns of serum concentrations of reproductivehormones from birth to puberty <strong>in</strong> the bull calf based on Evans et al.(1996), Chandolia et al. (1997c), Arav<strong>in</strong>dakshan et al. (2000a) andWrobel (2000). Horizontal l<strong>in</strong>es <strong>in</strong>dicate approximate periods for theactivities shownArav<strong>in</strong>dakshan et al. 2000a; Bagu et al. 2006a). Thisearly, postnatal <strong>in</strong>crease <strong>in</strong> LH secretion is caused by atransient <strong>in</strong>crease <strong>in</strong> the frequency of peaks or episodes<strong>in</strong> LH secretion (Rawl<strong>in</strong>gs and Evans 1995). The earlypostnatal <strong>in</strong>crease <strong>in</strong> LH pulse frequency is clearlydriven by an <strong>in</strong>crease <strong>in</strong> the frequency of pulses ofGnRH secretion (Rodriguez and Wise 1989) and isaccompanied by an <strong>in</strong>crease <strong>in</strong> pituitary LH stores(Rodriguez and Wise 1991), pituitary GnRH receptors(Amann et al. 1986; Rodriguez and Wise 1991) and LHrelease <strong>in</strong> vitro (McAndrews et al. 1994). The pattern ofserum concentrations of FSH dur<strong>in</strong>g development <strong>in</strong> thebull is less clear rang<strong>in</strong>g from no real trend (McCarthyet al. 1979; Amann and Walker 1983) to elevatedconcentrations postnatally, decreas<strong>in</strong>g to 25 weeks ofage; FSH secretion <strong>in</strong> the bull does not appear to bepulsatile (Miyamoto et al. 1989; Evans et al. 1996; Baguet al. 2006a).Interest<strong>in</strong>gly, serum concentrations of <strong>in</strong>hib<strong>in</strong> arehigh postnatally decl<strong>in</strong><strong>in</strong>g to 7 or 8 months of age(Miyamoto et al. 1989; Matsuzaki et al. 2001). Serumconcentrations of testosterone <strong>in</strong>crease slowly after birthand up to approximately 20 weeks of age; subsequently,concentrations <strong>in</strong>crease rapidly, start<strong>in</strong>g anywhere fromapproximately 20–35 weeks of age (Secchiara et al.1976; Lacroix et al. 1977; Sundby and Velle 1980;Miyamoto et al. 1989; Rawl<strong>in</strong>gs and Evans 1995; Evanset al. 1996). Dur<strong>in</strong>g the early postnatal period, up to20 weeks of age, the testis also secretes significantamounts of androstenedione and dihydrotestosterone,albeit at lower levels than testosterone (Rawl<strong>in</strong>gs et al.1972; McCarthy et al. 1979; Sundby et al. 1984; Rawl<strong>in</strong>gsand Cook 1986).The slow <strong>in</strong>crease <strong>in</strong> testosterone secretion andproduction of androstenedione and dihydrotestosterone,seen prior to 20 weeks of age, occur dur<strong>in</strong>g thephase of the rapid <strong>in</strong>crease <strong>in</strong> number of adult Leydigcells <strong>in</strong> the testes and the early postnatal transientÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Puberty <strong>in</strong> the Bull 297<strong>in</strong>crease <strong>in</strong> LH secretion. The later, rapid <strong>in</strong>crease <strong>in</strong>serum concentrations of testosterone occurs after theearly rise <strong>in</strong> LH secretion and once the production ofadult Leydig cells is virtually complete, but importantly,over a period of time when gonadotrop<strong>in</strong> secretion islow. Rapid growth of the testes and the androgendependentsexual accessory structures occurs after20 weeks of age dur<strong>in</strong>g the rapid <strong>in</strong>crease <strong>in</strong> testosteronesecretion. When LH and FSH receptor concentrationsper milligram of prote<strong>in</strong>, <strong>in</strong> the testes, were studied every4 weeks from 5 to 33 weeks of age and aga<strong>in</strong> at56 weeks of age, the major f<strong>in</strong>d<strong>in</strong>g was a transientdecrease <strong>in</strong> concentrations from 13 to 25 weeks of age(Bagu et al. 2006a). For these bulls (Hereford · Charolais)this transient decrease <strong>in</strong> gonadotrop<strong>in</strong> receptorconcentrations occurred as serum gonadotrop<strong>in</strong> concentrationsdecl<strong>in</strong>ed from their high postnatal levels andas numbers of adult Leydig and Sertoli cells <strong>in</strong>creasedquite rapidly at the onset of rapid testicular growth.Proliferation of immature Sertoli cells occurs dur<strong>in</strong>gthe early postnatal period <strong>in</strong> the bull calf while serumFSH concentrations are high. Maturation of immatureto mature Sertoli cells starts around the time of the earlypostnatal rise <strong>in</strong> LH secretion and cont<strong>in</strong>ues as gonadotrop<strong>in</strong>secretion decl<strong>in</strong>es. It is <strong>in</strong>terest<strong>in</strong>g that thephase of most active production of spermatogonia andforward progression of spermatogenesis, <strong>in</strong> the develop<strong>in</strong>gbull, occurs at the end of the early postnatal<strong>in</strong>crease <strong>in</strong> LH secretion and as serum concentrations ofFSH decl<strong>in</strong>e from their postnatal zenith. Aga<strong>in</strong>, theperiod of rapid testicular growth and development ofspermatogenesis, occur after 20–25 weeks of age whenserum concentrations of LH and FSH are lower than <strong>in</strong>the early postnatal period. However, pulsatile secretionof LH is still important for testis growth <strong>in</strong> this period ofrapid testicular growth, as suppression of LH secretionwith oestradiol slows testicular growth, but treatmentwith LHRH restores it (Schanbacher 1981; Schanbacheret al. 1982).The Critical Role of the Early PostnatalIncrease <strong>in</strong> Gonadotrop<strong>in</strong> SecretionWhen 48 Hereford · Charolais bull calves were split<strong>in</strong>to two groups, based on age at puberty as def<strong>in</strong>ed byWolf et al. (1965), serum LH concentrations were higherdur<strong>in</strong>g the postnatal <strong>in</strong>crease <strong>in</strong> LH secretion, <strong>in</strong> earlymatur<strong>in</strong>g (42 weeks age) compared to late matur<strong>in</strong>gbulls (48 weeks of age; Evans et al. 1995; Arav<strong>in</strong>dakshanet al. 2000b). Serum FSH concentrations did notdiffer between groups. We were surprised to see subsequently,that late matur<strong>in</strong>g bull calves had higher serumconcentrations of LH <strong>in</strong> response to LHRH at 4 and20 months of age (at the onset and end of the earlypostnatal <strong>in</strong>crease <strong>in</strong> LH secretion) than early matur<strong>in</strong>gbull calves (Bagu et al. 2006b). This was perhapsbecause early matur<strong>in</strong>g bull calves experienced an earlierand greater <strong>in</strong>crease <strong>in</strong> GnRH secretion postnatally, thatcaused depletion of LH stores <strong>in</strong> the early develop<strong>in</strong>gpituitary, giv<strong>in</strong>g a lower response to exogenous LHRH,compared to late matur<strong>in</strong>g bull calves. Treatment ofHereford · Charolais bull calves with a GnRH agonistdesigned to lower LH secretion, from 6 to 18 weeks ofage (Chandolia et al. 1997a), delayed the peak <strong>in</strong> theearly postnatal rise <strong>in</strong> LH secretion from 20 (controlcalves) to 25 weeks of age. Serum concentrations ofFSH were lower <strong>in</strong> treated calves at 14, 16, 18 and26 weeks of age compared to control calves. Serumtestosterone concentrations were significantly suppressedat 14, 16 and 18 weeks of age. Scrotal circumferencewas smaller <strong>in</strong> treated calves from birth to50 weeks of age. At 50 weeks of age spermatid andspermatocyte numbers, but not Sertoli cell numbers,were lower <strong>in</strong> tubule cross sections for tubules <strong>in</strong> stageVI of spermatogenesis <strong>in</strong> treated compared to controlbulls. In another study, similar bull calves were givenLHRH <strong>in</strong>travenously, every 2 h for 14 days, between 4and 6 weeks of age (Chandolia et al. 1997b). Thistreatment <strong>in</strong>creased the frequency of secretory pulsesof LH dur<strong>in</strong>g the period of treatment and resulted <strong>in</strong> agreater scrotal circumference. Spermatogenesis wasenhanced and numbers of Sertoli cells were <strong>in</strong>creasedbased on histology of testicular tubular cross sections <strong>in</strong>stage VI of spermatogenesis, from testes collected at54 weeks of age. Serum concentrations of FSH were notaffected by treatment. In subsequent studies, <strong>in</strong> Hereford· Charolais bull calves, treatments were given lessfrequently; GnRH was given (i.m.) twice every day from4 to 8 weeks of age (Madgwick et al. 2007) and <strong>in</strong>separate sets of bull calves, either bov<strong>in</strong>e LH or bov<strong>in</strong>eFSH were given every 2 days from 4 to 8 weeks of age(Bagu et al. 2004). Based on achievement of a scrotalcircumference of 28 cm (Wolf et al. 1965; Lunstra et al.1978) GnRH treatment advanced age at puberty(42 weeks of age) compared to control bulls (47 weeksof age). The FSH treatment also advanced puberty(39 weeks of age) compared to control bulls (45 weeksof age). Schuenemann et al. (2007) gave FSH at 27 daysof age, when bull calves were immunized aga<strong>in</strong>st <strong>in</strong>hib<strong>in</strong>and saw <strong>in</strong>creased germ cell numbers at 17 weeks of age.It appears that early postnatal secretion of FSH andparticularly LH, prior to 25 weeks of age, are critical to<strong>in</strong>itiate testicular differentiation and development <strong>in</strong> thebull calf, with the process cont<strong>in</strong>u<strong>in</strong>g after this timeaga<strong>in</strong>st a background of lower serum gonadotrop<strong>in</strong>concentrations, but <strong>in</strong>creas<strong>in</strong>g serum concentrations oftestosterone.Regulation of Gonadotrop<strong>in</strong> Secretion Dur<strong>in</strong>gDevelopmentThe regulation of the relatively high levels of FSHsecretions seen postnatally <strong>in</strong> the bull calf and the early,transient, postnatal rise <strong>in</strong> LH secretion are critical asthey appear to be pivotal signals for the onset of sexualmaturation (Fig. 3). When bull calves were castrated atdifferent ages there were only limited <strong>in</strong>creases <strong>in</strong> FSHsecretion but LH secretion rose markedly <strong>in</strong> calves24 weeks old and older (Amann and Walker 1983;Deaver and Peters 1988). In another study, castrationdid not result <strong>in</strong> an <strong>in</strong>crease <strong>in</strong> LH secretion <strong>in</strong> bullcalves at 7, 8 or 9 weeks of age, but a response was seenat 10, 11, and 13 weeks of age (Wise et al. 1987).However, Bass et al. (1978) concluded that castrationresulted <strong>in</strong> <strong>in</strong>creased LH secretion at 1, 7 and 17 weeksof age. Testosterone and dihydrotestosterone suppressÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


298 N Rawl<strong>in</strong>gs, ACO Evans, RK Chandolia and ET BaguFig. 3. Regulation of LH and FSH secretion dur<strong>in</strong>g sexual maturation<strong>in</strong> bull calves based on Amann and Walker (1983), Wise et al. (1987),Deaver and Peters (1988), Rodriguez and Wise (1989), Macdonaldet al. (1990), Evans et al. (1993), Shahab et al. (1993, 1995), Evanset al. (1996), Chandolia et al. (1997c) and Arav<strong>in</strong>dakshan et al.(2000a). Horizontal l<strong>in</strong>es and grey boxes <strong>in</strong>dicate approximate periodsfor the activities shown; l<strong>in</strong>es with arrow heads <strong>in</strong>dicate start po<strong>in</strong>ts forcont<strong>in</strong>ued activities. NMA, N-methyl-D,L-aspartic acidserum concentrations of LH and FSH <strong>in</strong> bull calvesbetween 4 and 20 weeks of age (Kennedy et al. 1985;Godfrey et al. 1992). However, when Hereford · Charolaiscalves were treated with three <strong>in</strong>jections, 12 hoursapart, of the androgen receptor blocker Flutamide, at 8,16 and 24 weeks of age, the authors concluded thatreduced suppression of LH secretion by androgens isnot critical for the onset of the early postnatal <strong>in</strong>crease<strong>in</strong> LH secretion but <strong>in</strong>creased suppression could be afactor <strong>in</strong> the subsequent decrease <strong>in</strong> both LH and FSHsecretion (Rawl<strong>in</strong>gs and Evans 1995). There has beenconsiderable <strong>in</strong>terest <strong>in</strong> the role of oestradiol as afeedback regulator of gonadotrop<strong>in</strong> secretion <strong>in</strong> the bullcalf. Oestradiol is produced by the bov<strong>in</strong>e testes but anadrenal source is also likely (Henricks et al. 1988).Serum concentrations of oestradiol appear to <strong>in</strong>creasefrom 1 to 6 weeks of age (Amann et al. 1986; Evanset al. 1993) and aga<strong>in</strong> after 24 weeks of age, dur<strong>in</strong>grapid testicular growth (Evans et al. 1993). The concentrationsof oestradiol receptors decrease <strong>in</strong> the hypothalamus,but <strong>in</strong>crease <strong>in</strong> the pituitary, from 6 to10 weeks of age (Amann et al. 1986). Treatment of bullcalves with oestradiol as early as 5 weeks of age willsuppress LH and FSH secretion (Schanbacher et al.1982; Wise et al. 1987; Deaver et al. 1988; Wildeus et al.1988; Macdonald et al. 1990; Godfrey et al. 1992).Interest<strong>in</strong>gly, <strong>in</strong> the study of Wildeus et al. (1988),treatment with oestradiol suppressed mean LH secretionat 4, 6, 8 and 10 weeks of age but LH pulse frequencywas suppressed only at 4 and 8 weeks of age.It would appear that negative feedback suppression ofgonadotrop<strong>in</strong> secretion, by products of the testes, mayterm<strong>in</strong>ate the early postnatal <strong>in</strong>crease <strong>in</strong> LH secretionand contribute to decreased FSH secretion at that time(circa 20 to 25 weeks of age). This negative feedbackstrengthens as the calf gets older and probably, pr<strong>in</strong>cipally,<strong>in</strong>volves androgens, but oestradiol is also <strong>in</strong>volved.The role of <strong>in</strong>hib<strong>in</strong> <strong>in</strong> regulat<strong>in</strong>g FSH secretiondur<strong>in</strong>g development is unclear; although immunizationaga<strong>in</strong>st <strong>in</strong>hib<strong>in</strong> <strong>in</strong> the bull calf does cause an <strong>in</strong>crease <strong>in</strong>FSH secretion (Kaneko et al. 2001). Some attemptshave been made to further study the neuroendrocr<strong>in</strong>eregulation of gonadotrop<strong>in</strong> secretion <strong>in</strong> the postnatalbull calf between birth and 25 weeks of age. WhenNaloxone, a l opiate receptor antagonist, was adm<strong>in</strong>isteredto bull calves, some evidence was produced thatsupported opioidergic suppression of LH secretiondur<strong>in</strong>g the early postnatal period (Macdonald et al.1990; Evans et al. 1993). The authors of these studiessuggested that a transient decrease <strong>in</strong> opioidergic suppressionof LH secretion, particularly LH pulse frequency,may allow the early postnatal <strong>in</strong>crease <strong>in</strong> LHsecretion (Macdonald et al. 1990; Evans et al. 1993;Chandolia et al. 1997d). Some evidence for opioidergicsuppression of FSH secretion was seen <strong>in</strong> 24-week-oldbull calves (Evans et al. 1993; Chandolia et al. 1997d).In the study of Chandolia et al. (1997d), the dopam<strong>in</strong>eantagonist pimozide was also used and the authorsconcluded that a dopam<strong>in</strong>ergic drive for LH and FSHsecretion was established <strong>in</strong> 24-week-old bull calves. At24 weeks of age, but not earlier, opioidergic <strong>in</strong>hibitionof gonadotrop<strong>in</strong> secretion largely <strong>in</strong>volved <strong>in</strong>hibition ofthe dopam<strong>in</strong>ergic drive. Adm<strong>in</strong>istration of MK801, anNMDA (N-methyl-D,L-aspartic acid) receptor antagonist,did not affect LH secretion at 1 and 12 weeks ofage, but at 24 weeks of age pulsed secretion of LH wasdecreased (Shahab et al. 1995); NMA caused LH releaseat 24 weeks of age (Shahab et al. 1993).The early postnatal <strong>in</strong>crease <strong>in</strong> LH secretion consistsof an <strong>in</strong>crease <strong>in</strong> the frequency of pulses of LH secretion,driven by <strong>in</strong>creased GnRH pulse frequency (Rodriguezand Wise 1989). Dim<strong>in</strong>ished opioidergic tone may allowthis <strong>in</strong>creased GnRH secretion and the term<strong>in</strong>ation ofthe early postnatal <strong>in</strong>crease <strong>in</strong> LH secretion is perhapspartly due to negative feedback by testicular androgensas well as opioidergic tone. Similar regulatory mechanismsmay cause the decrease <strong>in</strong> FSH secretion fromapproximately 20 to 30 weeks of age. Central drives forgonadotrop<strong>in</strong> secretion, <strong>in</strong>volv<strong>in</strong>g NMA and dopam<strong>in</strong>e,appear to be established after the early postnatal<strong>in</strong>crease <strong>in</strong> LH secretion, but androgenic negativefeedback of gonadotrop<strong>in</strong> secretion also strengthens atand after this time. The role of <strong>in</strong>hib<strong>in</strong> <strong>in</strong> the regulationof the temporal patterns of FSH secretion dur<strong>in</strong>gdevelopment <strong>in</strong> the bull calf is unclear.Effects of Breed, Season and NutritionIn the <strong>in</strong>troduction to this review the effect of breed onage at puberty was briefly alluded to; nutrition (Brattonet al. 1959) and season of birth (Arav<strong>in</strong>dakshan et al.2000a; Tatman et al. 2004) can also <strong>in</strong>fluence reproductivedevelopment. In general, it appears that lownutritional <strong>in</strong>take after wean<strong>in</strong>g h<strong>in</strong>ders sexual development,whereas, the effects of enhanced nutrition,although less clear, appear positive (Mann et al. 1967;Pruitt et al. 1986). It has been suggested that variousmetabolic hormones may provide a reflection of nutritional<strong>in</strong>take and <strong>in</strong>fluence patterns of secretion ofreproductive hormones dur<strong>in</strong>g development (I’Ansonet al. 1991). In the bull calf, serum concentrations of<strong>in</strong>sul<strong>in</strong>, IGF-1 and lept<strong>in</strong> <strong>in</strong>crease with age, but growthhormone concentrations decrease (Breier et al. 1988;Ronge and Blum 1989; McAndrews et al. 1993; Renavilleet al. 1996; Brito et al. 2007a; b). Treatment of bullcalves with recomb<strong>in</strong>ant bov<strong>in</strong>e somatotroph<strong>in</strong> from 4to 32 weeks of age, did not affect sexual developmentÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Puberty <strong>in</strong> the Bull 299(MacDonald and Deaver 1993). When nutritionalrestriction was applied pre-wean<strong>in</strong>g and through theperiod of the early postnatal <strong>in</strong>crease <strong>in</strong> LH secretion,LH secretion was reduced and puberty delayed; testicularweight was reduced (Brito et al. 2007a). Enhancednutrition dur<strong>in</strong>g the same period of time positivelyaffected the early postnatal <strong>in</strong>crease <strong>in</strong> LH secretion and<strong>in</strong>creased testicular growth (Brito et al. 2007b). Dur<strong>in</strong>gthe treatment period, nutritional restriction caused adecrease <strong>in</strong> serum concentrations of IGF-1 and improvednutrition <strong>in</strong>creased serum concentrations ofIGF-1 and <strong>in</strong>sul<strong>in</strong> (Brito et al. 2007a,b). It is <strong>in</strong>trigu<strong>in</strong>gthat nutritional manipulation of the early <strong>in</strong>crease <strong>in</strong> LHsecretion affects testicular growth and age at puberty.Fall born bull calves reached puberty later (Tatmanet al. 2004) or the tim<strong>in</strong>g of puberty was more variable(Arav<strong>in</strong>dakshan et al. 2000a) compared to spr<strong>in</strong>g borncalves. Fall born bull calves experienced a prolongedearly postnatal <strong>in</strong>crease <strong>in</strong> LH secretion, with greater LHpulse amplitude, compared to spr<strong>in</strong>g born calves (Arav<strong>in</strong>dakshanet al. 2000a). Although photoperiod wouldbe the obvious basis for these seasonal differences,treatment of spr<strong>in</strong>g born bulls with implants releas<strong>in</strong>gmelaton<strong>in</strong> at birth, 6 and 11 weeks of age, did not disruptdevelopment <strong>in</strong> any way (Arav<strong>in</strong>dakshan et al. 2000a).ConclusionsThe testes of the bull grow relatively slowly untilapproximately 25 weeks of age and then a rapid phaseof growth occurs until puberty at 37–50 weeks of age.Dur<strong>in</strong>g the early postnatal phase of slower growth of thetestis, pre-spermatogonia and some spermatogonia areestablished, adult Leydig cells appear and undifferentiatedSertoli cells are produced. The rapid testiculargrowth, after 25 weeks of age, consists of marked<strong>in</strong>creases <strong>in</strong> the diameter and length of the sem<strong>in</strong>iferoustubules, dramatic proliferation and differentiation ofgerm cells, with mature spermatozoa occurr<strong>in</strong>g between32 and 40 weeks of age. The adult Leydig cell populationis largely <strong>in</strong> place by 30 weeks of age and that ofSertoli cells by 30–40 weeks of age. Serum concentrationsof LH <strong>in</strong>crease from 4 to 5 weeks of age, to anearly postnatal peak at 12–16 weeks of age, followed bya decl<strong>in</strong>e to 25 weeks of age. Serum FSH concentrationsare high postnatally, decl<strong>in</strong><strong>in</strong>g to approximately25 weeks of age. Serum testosterone concentrations<strong>in</strong>crease dur<strong>in</strong>g the phase of rapid testicular growth.Hypothalamic opioidergic <strong>in</strong>hibition may abate transientlyto allow the early postnatal <strong>in</strong>crease <strong>in</strong> LHsecretion, while testicular androgenic negative feedbackprobably contributes to the decl<strong>in</strong>e <strong>in</strong> gonadotrop<strong>in</strong>secretion to 25 weeks of age. Several l<strong>in</strong>es of study haveled us to suggest that early postnatal gonadotrop<strong>in</strong>secretion is pivotal to <strong>in</strong>itiate the process of sexualmaturation <strong>in</strong> the bull calf.AcknowledgementsWe thank Dr R. Kennedy, Ms S. Cook, Dr J.P. Arav<strong>in</strong>dakshan, Dr S.Madgwick and Mr W. Kerr for help with our studies. F<strong>in</strong>ancialsupport was from the Canadian Natural Sciences and Eng<strong>in</strong>eer<strong>in</strong>gResearch Council and the Saskatchewan Agricultural Research Fund.ReferencesAbdel-Raouf M, 1960: The postnatal development of thereproductive organs <strong>in</strong> bullswith special reference topuberty. (Includ<strong>in</strong>g growth of the hypophysis and theadrenals). Acta Endocr<strong>in</strong>ol 34(Suppl. 49), 1–109.Almquist J, Cunn<strong>in</strong>gham D, 1967: Reproductive capacity ofbeef bulls. I. Postpuberal changes <strong>in</strong> semen production atdifferent ejaculation frequencies. J Anim Sci 26, 174–181.Amann RP, 1983: Endocr<strong>in</strong>e changes associated with onset ofspermatogenesis <strong>in</strong> Holste<strong>in</strong> bulls. J Dairy Sci 66, 2606–2622.Amann R, Walker O, 1983: Changes <strong>in</strong> pituitary-gonadal axisassociated with puberty <strong>in</strong> Holste<strong>in</strong> bulls. J Anim Sci 57,422–433.Amann R, Wise M, Glass J, Nett T, 1986: Prepubertal changes<strong>in</strong> the hypothalamic–pituitary axis of Holste<strong>in</strong> bulls. BiolReprod 34, 71–80.Arav<strong>in</strong>dakshan JP, Honaramooz A, Bartlewski PM, BeardAP, Pierson RR, Rawl<strong>in</strong>gs NC, 2000a: Gonadotroph<strong>in</strong>secretion <strong>in</strong> prepubertal bull calves born <strong>in</strong> spr<strong>in</strong>g andautumn. J Reprod Fertil 120, 159–167.Arav<strong>in</strong>dakshan JP, Honaramooz A, Bartlewski PM, BeardAP, Pierson RA, Rawl<strong>in</strong>gs NC, 2000b: Pattern of gonadotrop<strong>in</strong>secretion and ultrasonographic evaluation of developmentalchanges <strong>in</strong> the testis of early and late matur<strong>in</strong>gbull calves. Theriogenology 54, 339–354.Bagu ET, Madgwick S, Duggavathi R, Bartlewski PM, BarrettDM, Huchkowsky S, Cook SJ, Rawl<strong>in</strong>gs NC, 2004: Effectsof treatment with LH or FSH from 4 to 8 weeks of age onthe atta<strong>in</strong>ment of puberty <strong>in</strong> bull calves. Theriogenology 62,861–873.Bagu ET, Cook SJ, Gratton C, Rawl<strong>in</strong>gs NC, 2006a: Postnatalchanges <strong>in</strong> testicular gonadotrop<strong>in</strong> receptors, serum gonadotrop<strong>in</strong>,and testosterone concentrations and functionaldevelopment of the testes <strong>in</strong> bulls. <strong>Reproduction</strong> 132, 403–411.Bagu ET, Cook SJ, Honaramooz A, Arav<strong>in</strong>dakshan JP,Huchkowsky S, Rawl<strong>in</strong>gs NC, 2006b: Changes <strong>in</strong> serumlute<strong>in</strong>iz<strong>in</strong>g hormone (LH) concentrations <strong>in</strong> response tolute<strong>in</strong>iz<strong>in</strong>g hormone releas<strong>in</strong>g hormone (LHRH) <strong>in</strong> bullcalves that atta<strong>in</strong>ed puberty early or late. Theriogenology66, 937–944.Bass JJ, Peterson AJ, Payne E, 1978: Plasma concentrations oflute<strong>in</strong>iz<strong>in</strong>g hormone, testosterone and androstenedione <strong>in</strong>castrated and adrenalectomized bull calves. J Endocr<strong>in</strong>ol 79,137–138.Berndtson WE, Igboeli G, Parker WG, 1987: The numbers ofSertoli cells <strong>in</strong> mature Holste<strong>in</strong> bulls and their relationshipto quantitative aspects of spermatogenesis. Biol Reprod 37,60–67.Bratton R, Musgrave S, Dunn H, Foote R, 1959: Causes andprevention of reproductive failures <strong>in</strong> dairy cattle. II.Influence of underfeed<strong>in</strong>g and overfeed<strong>in</strong>g from birth to80 weeks of age on growth, sexual development, and semenproduction of Holste<strong>in</strong> bulls. Bull Cornell Agric Exp Sta 94,45.Breier BH, Gluckman PD, Bass JJ, 1988: Influence ofnutritional status and oestradiol-17 beta on plasma growthhormone, <strong>in</strong>sul<strong>in</strong>-like growth factors-I and -II and theresponse to exogenous growth hormone <strong>in</strong> young steers.J Endocr<strong>in</strong>ol 118, 243–250.Brito LF, Barth AD, Rawl<strong>in</strong>gs NC, Wilde RE, Crews DH Jr,Boisclair YR, Ehrhardt RA, Kastelic JP, 2007a: Effect offeed restriction dur<strong>in</strong>g calfhood on serum concentrations ofmetabolic hormones, gonadotrop<strong>in</strong>s, testosterone, and onsexual development <strong>in</strong> bulls. <strong>Reproduction</strong> 134, 171–181.Brito LF, Barth AD, Rawl<strong>in</strong>gs NC, Wilde RE, Crews DH Jr,Mir PS, Kastelic JP, 2007b: Effect of improved nutritionÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


300 N Rawl<strong>in</strong>gs, ACO Evans, RK Chandolia and ET Bagudur<strong>in</strong>g calfhood on serum metabolic hormones, gonadotrop<strong>in</strong>s,and testosterone concentrations, and on testiculardevelopment <strong>in</strong> bulls. Domest Anim Endocr<strong>in</strong>ol 33, 460–469.Chandolia RK, Evans ACO, Rawl<strong>in</strong>gs NC, 1997a: Effect of<strong>in</strong>hibition of <strong>in</strong>creased gonadotroph<strong>in</strong> secretion before20 weeks of age <strong>in</strong> bull calves on testicular development.J Reprod Fertil 109, 65–71.Chandolia RK, Honaramooz A, Bartlewski PM, Beard AP,Rawl<strong>in</strong>gs NC, 1997b: Effects of treatment with LH releas<strong>in</strong>ghormone before the early <strong>in</strong>crease <strong>in</strong> LH secretion onendocr<strong>in</strong>e and reproductive development <strong>in</strong> bull calves.J Reprod Fertil 111, 41–50.Chandolia RK, Honaramooz A, Omeke BC, Pierson R, BeardAP, Rawl<strong>in</strong>gs NC, 1997c: Assessment of development of thetestes and accessory glands by ultrasonography <strong>in</strong> bullcalves and associated endocr<strong>in</strong>e changes. Theriogenology48, 119–132.Chandolia RK, Evans ACO, Rawl<strong>in</strong>gs NC, 1997d: The<strong>in</strong>volvement of dopam<strong>in</strong>ergic and opioidergic neuronalsystems <strong>in</strong> the control of the early rise <strong>in</strong> LH secretion <strong>in</strong>bull calves. J Neuroendocr<strong>in</strong>ol 9, 121–127.Curtis S, Amann R, 1981: Testicular development andestablishment of spermatogenesis <strong>in</strong> Holste<strong>in</strong> bulls. J AnimSci 53, 1645–1647.Deaver DR, Glass GD, Grieger DM, Reeves JJ, 1998: Effectsof estradid on secretion of LH, hypothalamic functionand testical development <strong>in</strong> bull calves. Domest AnimEndocr<strong>in</strong>al 5, 307–316.Evans A-CO, Currie WD, Rawl<strong>in</strong>gs NC, 1993: Opioidergicregulation of gonadotroph<strong>in</strong> secretion <strong>in</strong> the early prepubertalbull calf. J Reprod Fertil 99, 45–51.Evans A-CO, Davies FJ, Nasser LF, Bowman P, Rawl<strong>in</strong>gsNC, 1995: Differences <strong>in</strong> early patterns of gonadotroph<strong>in</strong>secretion between early and late matur<strong>in</strong>g bulls, and changes<strong>in</strong> semen characteristics at puberty. Theriogenology 43, 569–578.Evans A-CO, Pierson RA, Garcia A, McDougall LM, HrudkaF, Rawl<strong>in</strong>gs NC, 1996: Changes <strong>in</strong> circulat<strong>in</strong>g hormoneconcentrations, testes histology and testes ultrasonographydur<strong>in</strong>g sexual maturation <strong>in</strong> beef bulls. Theriogenology 46,345–357.Godfrey RW, Lunstra DD, Schanbacher BD, 1992: Effect ofimplant<strong>in</strong>g bull calves with testosterone propionate, dihydrotestosteronepropionate or oestradiol-17 beta prepubertallyon the pituitary–testicular axis and onpostpubertal social and sexual behaviour. J Reprod Fertil94, 57–69.Henricks D, Hoover J, Gimenez T, Grimes L, 1988: A study ofthe source of estradiol-17 beta <strong>in</strong> the bull. Horm Met Res 20,494–497.I’Anson H, Foster DL, Foxcroft GR, Booth PJ, 1991:Nutrition and reproduction. Oxf Rev Reprod Biol 13,239–311.Kaneko H, Noguchi J, Kikuchi K, Akagi S, Shimada A, TayaK, Watanabe G, Hasegawa Y, 2001: Production andendocr<strong>in</strong>e role of <strong>in</strong>hib<strong>in</strong> dur<strong>in</strong>g the early development ofbull calves. Biol Reprod 65, 209–215.Kennedy R I, Rawl<strong>in</strong>gs N, Cook S, 1985: Effects ofdihydrotestosterone and 5 alpha-androstane-3 alpha,17beta-diol on serum concentrations of LH and FSH <strong>in</strong> theprepubertal bull. Can J Anim Sci 65, 95–99.Lacroix A, Pelletier J, 1979: Short-term variations <strong>in</strong> plasmaLH and testosterone <strong>in</strong> bull calves from birth to 1 year ofage. J Reprod Fertil 55, 81–85.Lacroix A, Garnier DH, Pelletier J, 1977: Temporal fluctuationsof plasma LH and testosterone <strong>in</strong> Charolais bull calvesdur<strong>in</strong>g the first year of life. Ann Biol Anim Biochim Biophys17, 1013–1019.L<strong>in</strong>dner H, Mann T, 1960: Relationship between the contentof androgenic steroids <strong>in</strong> the testes and the secretory activityof the sem<strong>in</strong>al vesicles <strong>in</strong> the bull. J Endocr<strong>in</strong>ol 21, 341–361.Lunstra D, Echternkamp S, 1982: Puberty <strong>in</strong> beef bulls:acrosome morphology and semen quality <strong>in</strong> bulls ofdifferent breeds. J Anim Sci 55, 638–648.Lunstra DD, Ford JJ, Echterkamp SE, 1978: Puberty <strong>in</strong> beefbulls: hormone concentrations, growth, testicular development,sperm production and sexual aggressiveness <strong>in</strong> bulls ofdifferent breeds. J Anim Sci 46, 1054–1062.MacDonald R, Deaver D, 1993: Testicular development <strong>in</strong>bulls treated with recomb<strong>in</strong>ant bov<strong>in</strong>e somatotrop<strong>in</strong>.J Anim Sci 71, 1540–1545.Macdonald RD, Peters JL, Deaver DR, 1990: Effect ofnaloxone on the secretion of LH <strong>in</strong> <strong>in</strong>fantile and prepubertalHolste<strong>in</strong> bull calves. J Reprod Fertil 89, 51–58.Macmillan K, Hafs H, 1968: Gonadal and extra gonadalsperm numbers dur<strong>in</strong>g reproductive development of Holste<strong>in</strong>bulls. J Anim Sci 27, 1614–1620.Macmillan K, Hafs H, 1969: Reproductive tract of Holste<strong>in</strong>bulls from birth through puberty. J Anim Sci 28, 233–239.Madgwick S, Bagu ET, Duggavathi R, Bartlewski PM, BarrettDM, Huchkowsky S, Cook SJ, Beard AP, Rawl<strong>in</strong>gs NC,2007: Effects of treatment with GnRH from 4 to 8 weeks ofage on the atta<strong>in</strong>ment of sexual maturity <strong>in</strong> bull calves.Anim Reprod Sci, <strong>in</strong> press.Mann T, Rowson L, Short RV, Sk<strong>in</strong>ner J, 1967: Therelationship between nutrition and androgenic activity <strong>in</strong>pubescent tw<strong>in</strong> calves, and the effect of orchitis. Endocr<strong>in</strong>ology38, 455–468.Matsuzaki S, Uenoyama Y, Okuda K, Watanabe G, KitamuraN, Taya K, Cruzana MB, Yamada J, 2001: Prepubertalchanges <strong>in</strong> immunoreactive <strong>in</strong>hib<strong>in</strong> concentration <strong>in</strong> bloodserum and testicular tissue <strong>in</strong> Holste<strong>in</strong> bull calves. J Vet MedSci 63, 1303–1307.McAndrews JM, Stroud CM, Macdonald RD, Hymer WC,Deaver DR, 1993: Age-related changes <strong>in</strong> the secretion ofgrowth hormone <strong>in</strong> vivo and <strong>in</strong> vitro <strong>in</strong> <strong>in</strong>fantile andprepubertal Holste<strong>in</strong> bull calves. J Endocr<strong>in</strong>ol 139, 307–315.McAndrews JM, Peters JL, Deaver DR, 1994: Age-relatedchanges <strong>in</strong> the secretion of LH <strong>in</strong> vivo and <strong>in</strong> vitro <strong>in</strong><strong>in</strong>fantile and prepubertal Holste<strong>in</strong> bull calves. J ReprodFertil 101, 453–458.McCarthy MS, Hafs HD, Convey EM, 1979: Serum hormonepatterns associated with growth and sexual development <strong>in</strong>bulls. J Anim Sci 49, 1012–1020.Miyamoto A, Umezu M, Ishii S, Furusawa T, Masaki J,Hasegawa Y, Ohta M, 1989: Serum <strong>in</strong>hib<strong>in</strong>, FSH, LH andtestosterone levels and testicular <strong>in</strong>hib<strong>in</strong> content <strong>in</strong> beef bullsfrom birth to puberty. Anim Reprod Sci 20, 165–178.Pruitt RJ, Corah LR, Stevenson JS, Kiracofe GH, 1986: Effectof energy <strong>in</strong>take after wean<strong>in</strong>g on the sexual development ofbeef bulls. II. Age at first mat<strong>in</strong>g, age at puberty, testosteroneand scrotal circumference. J Anim Sci 63, 579–585.Rawl<strong>in</strong>gs NC, Cook SJ, 1986: Plasma concentrations oftestosterone, androstenedione, dihydrotestosterone, 5alpha-androstane-3alpha,17beta-diol,5alpha-androstane-3beta,17beta-diol,and androsterone <strong>in</strong> bull calves: response tohCG. Can J Anim Sci 66, 975–982.Rawl<strong>in</strong>gs NC, Evans ACO, 1995: Androgen negative feedbackdur<strong>in</strong>g the early rise <strong>in</strong> LH secretion <strong>in</strong> bull calves.J Endocr<strong>in</strong>ol 145, 243–249.Rawl<strong>in</strong>gs N, Hafs H, Swanson LV, 1972: Testicular and bloodplasma androgens <strong>in</strong> Holste<strong>in</strong> bulls from birth throughpuberty. J Anim Sci 34, 435–440.Rawl<strong>in</strong>gs NC, Fletcher PW, Henricks DM, Hill JR, 1978:Plasma lute<strong>in</strong>iz<strong>in</strong>g hormone (LH) and testosterone levelsdur<strong>in</strong>g sexual maturation <strong>in</strong> beef bull calves. Biol Reprod19, 1108–1112.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Puberty <strong>in</strong> the Bull 301Renaville R, Massart S, Sneyers M, Falaki M, Gengler N,Burny A, Portetelle D, 1996: Dissociation of <strong>in</strong>creases <strong>in</strong>plasma <strong>in</strong>sul<strong>in</strong>-like growth factor I and testosterone dur<strong>in</strong>gthe onset of puberty <strong>in</strong> bulls. J Reprod Fertil 106, 79–86.Rodriguez RE, Wise ME, 1989: Ontogeny of pulsatile secretionof gonadotrop<strong>in</strong>-releas<strong>in</strong>g hormone <strong>in</strong> the bull calfdur<strong>in</strong>g <strong>in</strong>fantile and pubertal development. Endocr<strong>in</strong>ology124, 248–256.Rodriguez RE, Wise ME, 1991: Advancement of postnatalpulsatile lute<strong>in</strong>iz<strong>in</strong>g hormone secretion <strong>in</strong> the bull calf bypulsatile adm<strong>in</strong>istration of gonadotrop<strong>in</strong>-releas<strong>in</strong>g hormonedur<strong>in</strong>g <strong>in</strong>fantile development. Biol Reprod 44, 432–439.Ronge H, Blum J, 1989: Insul<strong>in</strong>-like growth factor I dur<strong>in</strong>ggrowth <strong>in</strong> bulls. Reprod Nutr Dev 29, 105–111.Schanbacher BD, 1981: Importance of the episodic nature oflute<strong>in</strong>iz<strong>in</strong>g hormone secretion for normal development ofthe bov<strong>in</strong>e testis dur<strong>in</strong>g puberty: <strong>in</strong>terference with oestradiol-17beta. J Endocr<strong>in</strong>ol 88, 393–400.Schanbacher BD, D’Occhio MJ, K<strong>in</strong>der JE, 1982: Initiation ofspermatogenesis and testicular growth <strong>in</strong> oestradiol-17 beta-implanted bull calves with pulsatile <strong>in</strong>fusion of lute<strong>in</strong>iz<strong>in</strong>ghormone releas<strong>in</strong>g hormone. J Endocr<strong>in</strong>ol 93, 183–192.Schuenemann GM, Mendis-Handagama SM, Hopk<strong>in</strong>s FM,Kania SA, Schrick FN, 2007: Changes <strong>in</strong> the testis sem<strong>in</strong>iferoustubules and <strong>in</strong>terstitium <strong>in</strong> prepubertal bull calvesimmunised aga<strong>in</strong>st <strong>in</strong>hib<strong>in</strong> at the time of gonadotrop<strong>in</strong>adm<strong>in</strong>istration. Reprod Fertil Dev 19, 840–849.Secchiara P, Martorana F, Pellegr<strong>in</strong>i S, Luisi M, 1976:Variation of plasma testosterone <strong>in</strong> develop<strong>in</strong>g Friesianbulls. J Anim Sci 42, 405–409.Shahab M, Nusser KD, Griel LC, Deaver DR, 1993: Effect ofa s<strong>in</strong>gle <strong>in</strong>travenous <strong>in</strong>jection of N-methyl-D,L-aspartic acidon secretion of lute<strong>in</strong>iz<strong>in</strong>g hormone and growth hormone <strong>in</strong>Holste<strong>in</strong> bull calves. J Neuroendocr<strong>in</strong>ol 5, 469–473.Shahab M, Nusser KD, Peters JL, Deaver DR, 1995:Involvement of N-methyl-D-aspartate receptor us<strong>in</strong>g excitatoryam<strong>in</strong>o acid neurotransmitters <strong>in</strong> control of pulsatilesecretion of LH dur<strong>in</strong>g sexual development <strong>in</strong> Holste<strong>in</strong> bullcalves. J Reprod Fertil 105, 77–78.S<strong>in</strong>owatz F, Amselgruber W, 1986: Postnatal development ofbov<strong>in</strong>e Sertoli cells. Anat Embryol 174, 413–423.Sundby A, Velle W, 1980: Plasma concentration of testosterone<strong>in</strong> young bulls <strong>in</strong> relation to age, rate of weight ga<strong>in</strong> andstimulation with human chorionic gonadotroph<strong>in</strong>. J Endocr<strong>in</strong>ol86, 465–469.Sundby A, Andersen D, Purvis K, Hansson V, 1984: Testiculargonadotrop<strong>in</strong> receptors, testicular testosterone, dihydrotestosteroneand androstenedione <strong>in</strong> the develop<strong>in</strong>g bull.Archiv Androl 12, 59–64.Tatman SR, Neuendorff DA, Wilson TW, Randel RD,2004: Influence of season of birth on growth andreproductive development of Brahman bulls. Theriogenology62, 93–102.Wildeus S, Randel R, Humphrey W, 1988: Age-relatedchanges <strong>in</strong> the suppression of serum lute<strong>in</strong>iz<strong>in</strong>g hormoneby estradiol-17b <strong>in</strong> develop<strong>in</strong>g steers. Theriogenology 29,1065–1073.Wise ME, Rodriguez RE, Kelly CM, 1987: Gonadal regulationof LH secretion <strong>in</strong> prepubertal bull calves. DomestAnim Endocr<strong>in</strong>ol 4, 175–181.Wolf F, Almquist J, Hale E, 1965: Prepuberal behavior andpuberal characteristics of beef bulls on high nutrientallowance. J Anim Sci 24, 761–765.Wrobel K, 1990: The postnatal development of the bov<strong>in</strong>eLeydig cell population. Reprod Dom Anim 25, 51–60.Wrobel KH, 2000: Prespermatogenesis and spermatogoniogenesis<strong>in</strong> the bov<strong>in</strong>e testis. Anat Embryol 202, 209–222.Author’s address (for correspondence): NC Rawl<strong>in</strong>gs, The DeansOffice, WCVM, University of Saskatchewan, Saskatoon, SaskatchewanS7N 5B4, Canada. E-mail: norman.rawl<strong>in</strong>gs@usask.caConflict of <strong>in</strong>terest: All authors declare no conflict of <strong>in</strong>terests.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 302–309 (2008); doi: 10.1111/j.1439-0531.2008.01178.xISSN 0936-6768Epigenetic Regulation of Foetal Development <strong>in</strong> Nuclear Transfer Animal ModelsA D<strong>in</strong>nyes 1,2 , XC Tian 3 and X Yang 31 Genetic Reprogramm<strong>in</strong>g Group, Agricultural Biotechnology Centre, Godollo, Hungary; 2 Molecular Animal Biotechnology Laboratory, Szent IstvanUniversity, Godollo, Hungary; 3 Department of Animal Science ⁄ Center for Regenerative Biology, University of Connecticut, Storrs, CT, USAContentsSomatic cell nuclear transfer (SCNT, ‘clon<strong>in</strong>g’) holds greatpotential for agricultural applications, generation of medicalmodel animals, transgenic farm animals or by ‘therapeuticclon<strong>in</strong>g’ for generat<strong>in</strong>g human embryonic stem cells for thetreatment of human diseases. However, the low survival rate ofSCNT-derived pregnancies represents a serious limitation ofthe current technology. In order to overcome this hurdle, adeeper understand<strong>in</strong>g of the epigenetic reprogramm<strong>in</strong>g of thesomatic cell nuclei and its effect on the pregnancy is needed.Here we review the literature on nuclear reprogramm<strong>in</strong>g bySCNT, <strong>in</strong>clud<strong>in</strong>g studies of gene expression, DNA methylation,chromat<strong>in</strong> remodell<strong>in</strong>g, genomic impr<strong>in</strong>t<strong>in</strong>g and Xchromosome <strong>in</strong>activation. Reprogramm<strong>in</strong>g of genes expressed<strong>in</strong> the <strong>in</strong>ner cell mass, from which the body of the foetus isformed, seems to be highly efficient. Defects <strong>in</strong> the extraembryonictissues are probably the major cause of the lowsuccess rate of reproductive clon<strong>in</strong>g. Methods to partiallyovercome such problems exist, yet more future research isneeded to f<strong>in</strong>d practical and efficient methods to remedy thisproblem. Improvement of the survival of foetuses is a centralissue for the future of agricultural SCNT not only for itseconomic viability, but also because <strong>in</strong> lack of improvements<strong>in</strong> animal welfare current regulations can block the use of themethod <strong>in</strong> the EU and several other countries.Importance of Somatic Cell Nuclear TransferSomatic cell nuclear transfer is a promis<strong>in</strong>g technologyfor agricultural applications on its own or comb<strong>in</strong>edwith transgenesis for the generation of transgenic farmanimals or novel medical animal models. Somatic cellnuclear transfer offspr<strong>in</strong>gs have been produced <strong>in</strong> 18mammalian species, among these the largest number ofsomatic cell nuclear transfer (SCNT) progeny have beenborn <strong>in</strong> cattle and pig, with an estimated number of 3000and 600, respectively. Although, most of the ‘clones’(F 0 ) are not alive by now, by add<strong>in</strong>g the number (severalthousands) of healthy progeny derived by naturalbreed<strong>in</strong>g from such ‘clone’ F 0 founders, the impact ofthe technology is much bigger. Even though the numberof F 0 ‘clones’ produced for food purposes is still small,the potential of subsequent generations developed fromthose ‘clones’ by sexual reproduction is already high.The recently published risk assessment report of FDA(Animal Clon<strong>in</strong>g 2006) on the food safety of the milkand meat of SCNT-derived animals concludes that thereare no food safety concerns with such products, thus<strong>in</strong>creases the future chances for more animals producedfor human consumption.Despite all these advances, the technology is still very<strong>in</strong>efficient, mostly due to the major (80–99%) loss ofembryos transferred dur<strong>in</strong>g the pregnancy and withfurther losses <strong>in</strong> the per<strong>in</strong>atal period and dur<strong>in</strong>g the firstyear of life. Such <strong>in</strong>efficiency makes the technologycommercially less valuable and socially hardly acceptable.This later issue is particularly important <strong>in</strong> the EUwhere the legal framework for animal welfare regulationsand the negative public perception can block theuse of such a wasteful method, therefore improvementof the survival of foetuses is a central issue for the futureof agricultural SCNT.Pre- and Post-implantation Development ofSCNT EmbryosBasic steps of nuclear reprogramm<strong>in</strong>gSomatic cell nuclear reprogramm<strong>in</strong>g <strong>in</strong>volves two majorsteps: (i) dedifferentiation of the already differentiateddonor cell to a totipotent ‘embryonic’ state, followed by(ii) redifferentiation of SCNT embryos to differentprecursor and somatic cell types dur<strong>in</strong>g later development.The first step of nuclear reprogramm<strong>in</strong>g <strong>in</strong>volvesthe erasure of the donor cell epigenetic pattern after NTand the re-establishment of embryonic epigenetic characteristicsand gene expression <strong>in</strong> the SCNT embryo.The second step of nuclear reprogramm<strong>in</strong>g refers toredifferentiation of SCNT embryos from a totipotentstatus to various differentiated stages of organogenesisdur<strong>in</strong>g post-implantation development. Dur<strong>in</strong>g thesesteps there is evidence for normal and abnormalreprogramm<strong>in</strong>g, as well. Recent results <strong>in</strong>dicate thatmany of the defects <strong>in</strong> SCNT embryos occur <strong>in</strong> the laterreprogramm<strong>in</strong>g stage, which may be attributable toproblems with placental development and function.Pre- and post-implantation developmentMammalian embryos derived by SCNT are capable ofdevelopment to the blastocyst stage with an efficiency of30–50%, comparable to that of embryos produced by<strong>in</strong> vitro fertilization (IVF) <strong>in</strong> species such as cattle andpigs (30–40%) (Cibelli et al. 2002; Zhu et al. 2002).Unlike embryos derived from fertilization, however,most SCNT embryos die dur<strong>in</strong>g post-implantationdevelopment, and those that survive to term arefrequently defective.In cattle Panarace et al. (2007) reported efficiency ofSCNT up to 20%, but analys<strong>in</strong>g data <strong>in</strong> three countries,Brazil, Argent<strong>in</strong>e and USA over 5 years shows thatamong the 3374 cloned embryos transferred <strong>in</strong>to surrogatefemales, 9% live calves were born, 8% were alive24 h after birth, and 7% survived for 150 days or more.With<strong>in</strong> a given species, success rates can vary extensivelyreflect<strong>in</strong>g a lack of full understand<strong>in</strong>g of the roleof various factors <strong>in</strong>volved <strong>in</strong> the SCNT process such asÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Epigenetics of Foetal Development <strong>in</strong> Clones 303donor cell selection, cell cycle stage, culture conditions.For unknown reasons, approximately one-third of thedonor cell l<strong>in</strong>es lead to a success rate of live calvesobta<strong>in</strong>ed from <strong>in</strong>itiated pregnancy as high as 40%whereas approximately 25% of donor cell l<strong>in</strong>es totallyfailed (Panarace et al. 2007). Those differences <strong>in</strong> therate of live calves even occur when donor cell l<strong>in</strong>ecultures are run simultaneously with<strong>in</strong> the same experimentalprogram. The different cell l<strong>in</strong>es with noevidence of abnormal chromosomal constitution gavethe same high number of blastocysts <strong>in</strong> vitro afterSCNT, irrespectively of the follow<strong>in</strong>g success rate ofdevelopment (Renard et al. 2007).Higher success rates of SCNT <strong>in</strong> cattle are largely dueto the extensive knowledge on the female (and male)reproductive physiology <strong>in</strong> this economically importantspecies. In most mammalian species studied thus far, thesurvival rate to birth for cloned blastocysts is onlyapproximately 1–5%, compared with a 30–60% birthrate for IVF blastocysts. Many pathologies have beendescribed <strong>in</strong> conceptuses and neonates derived fromSCNT (Cibelli et al. 2002). Large offspr<strong>in</strong>g syndrome(LOS), which also occurs <strong>in</strong> cattle and sheep derivedfrom IVF, is common <strong>in</strong> cloned cattle, sheep and mice.LOS refers to a heterogeneous group of symptoms:notably, large size at birth and severe placental deficiency(Young et al.1998). Other symptoms are morevariable but <strong>in</strong>clude prolonged gestation, dystocia(strenuous labour), foetal and placental oedema, abnormalsize of organs, hydrallantois and hydramnios,respiratory problems and per<strong>in</strong>atal death (Young et al.1998; Far<strong>in</strong> et al. 2006). Most of the observed defects<strong>in</strong>volve abnormal placentation (Cibelli et al. 2002).Placentas of cloned calves have fewer but much largerplacentomes, presumably to compensate for the reducednumber of sites of foeto-maternal exchange (Chavatte-Palmer et al. 2002). In SCNT mice placentas are alwaysenlarged (Suemizu et al. 2003). It is possible that manyof the observed abnormalities are consequences ofprimary defects <strong>in</strong> placental function (Constant et al.2006). SCNT embryos from mice and cattle develop tothe blastocyst stage with a high success rate, and NTembryonicstem cells (NT-ESCs) can be derived fromsuch embryos with high efficiency (Wang et al. 2005;Wakayama et al. 2006) . Indeed, we have producednumerous NT-ESC l<strong>in</strong>es <strong>in</strong> mouse from donor cells<strong>in</strong>clud<strong>in</strong>g cumulus, fibroblast and ESCs. Gene expressionanalyses of such cell l<strong>in</strong>es have shown almost nodifferences compared to the ESCs with the same geneticbackground or from each others, regardless of the celltype of donor cells (D<strong>in</strong>nyes et al., unpublished results).The molecular, epigenetic mechanisms beh<strong>in</strong>d the placentalabnormalities are particularly <strong>in</strong>terest<strong>in</strong>g as aunique model for epigenetic regulation of the pregnancy,as discussed <strong>in</strong> details below.Molecular Level Changes <strong>in</strong> SCNT EmbryosGlobal gene expressionMost SCNT embryos have abnormalities at the molecularlevel. Early reports on reprogramm<strong>in</strong>g of variouscandidate genes <strong>in</strong> SCNT embryos are <strong>in</strong>consistent(Daniels et al. 2000; Wrenzycki et al. 2001; Inoue et al.2006). For example, expression of the pluripotencymarker gene Pou5f1 (formerly known as Oct4) afterSCNT has been reported to be both normal (Danielset al. 2000; Jouneau et al. 2006), or abnormal (Bortv<strong>in</strong>et al. 2003). The global gene expression pattern <strong>in</strong>SCNT bov<strong>in</strong>e blastocysts reflects well the extent ofnuclear reprogramm<strong>in</strong>g. In our studies the gene expressionprofiles of SCNT embryos were very different fromthose of somatic donor cells, and they resembled thoseof naturally fertilized embryos with


304 A D<strong>in</strong>nyes, XC Tian and X Yanggrowth and development. Some genes are onlyimpr<strong>in</strong>ted <strong>in</strong> the placenta (Wagschal and Feil 2006).In the mouse dist<strong>in</strong>ct allelic expression patterns ofimpr<strong>in</strong>ted genes can be established as early as the twocellstage, and by the blastocyst stage, monoallelicexpression of many impr<strong>in</strong>ted genes is observed, whichclosely follows the re-establishment of genome-wideDNA methylation <strong>in</strong> early development (Latham1999).Epigenetic perturbations <strong>in</strong> SCNT embryosFor clon<strong>in</strong>g to succeed, the adult pattern of epigeneticmodifications must be erased, and the patternsdescribed above must be established <strong>in</strong> the SCNTembryos. In the development of naturally fertilizedembryos, epigenetic marks are established over longperiods of time dur<strong>in</strong>g gametogenesis and fertilization(Morgan et al. 2005). In SCNT, however, an adultsomatic pattern of epigenetic modification that isnormally very stable must be reversed with<strong>in</strong> a shortperiod of time before zygotic genome activation(Zuccotti et al. 2000). Although no study has beenable to measure the epigenetic status of SCNT embryosand then follow their development after transfer torecipients, several studies have shown aberrant methylationpatterns <strong>in</strong> SCNT bov<strong>in</strong>e embryos whencompared with <strong>in</strong> vivo- or <strong>in</strong> vitro-fertilized controls(Bourc’his et al. 2001; Dean et al. 2001; Kang et al.2002). Interest<strong>in</strong>gly, at the blastocyst stage, the methylationstatus of the ICM is relatively normal, whereasthe trophoblast cells show abnormal hypermethylation(Dean et al. 2001). Methylation of histone H3K9 alsoshows a similar pattern, with abnormally high levels ofmethylation <strong>in</strong> the trophectoderm of cloned bov<strong>in</strong>eblastocysts (Santos et al. 2003). Studies on the acetylationof histones <strong>in</strong> cloned embryos have foundaberrancies, <strong>in</strong>clud<strong>in</strong>g hypoacetylation (Santos et al.2003; Enright et al. 2005; Suteevun et al. 2006). Thehistone deacetylase (HDAC) <strong>in</strong>hibitor trichostat<strong>in</strong> A(TSA) <strong>in</strong>creases clon<strong>in</strong>g efficiency <strong>in</strong>clud<strong>in</strong>g live births(Kishigami et al. 2006). The reprogramm<strong>in</strong>g of impr<strong>in</strong>tedgenes <strong>in</strong> SCNT can be altered and someabnormalities <strong>in</strong> cloned animals resemble those seen <strong>in</strong>human impr<strong>in</strong>t<strong>in</strong>g diseases and <strong>in</strong> mutant mice withexperimentally disrupted impr<strong>in</strong>t<strong>in</strong>g (Debaun et al.2003). SCNT animals with abnormal phenotypes canbe naturally bred, and all their offspr<strong>in</strong>g show normalphenotypes, suggest<strong>in</strong>g that the abnormalities of theclones are due to epigenetic errors rather than geneticmutations (Tamashiro et al. 2002). There is evidencethat the placenta is especially vulnerable to problems ofthis type. In cloned mice several impr<strong>in</strong>ted genes showabnormally low expression <strong>in</strong> the placenta, whereas nodifferences are seen <strong>in</strong> foetal expression (Inoue et al.2002). We also found abnormal allelic expressionpattern of the impr<strong>in</strong>ted IGF2R gene <strong>in</strong> placentasbut not <strong>in</strong> the organs of cloned bov<strong>in</strong>e calves (Yanget al. 2005). In our microarray study (Smith et al.2005), we found that Cd81, a gene that <strong>in</strong> mice showsimpr<strong>in</strong>t<strong>in</strong>g only <strong>in</strong> the placenta, was downregulated <strong>in</strong>SCNT bov<strong>in</strong>e blastocysts compared with <strong>in</strong> vivoembryos.X chromosome <strong>in</strong>activationIn all eutherian mammalian species XCI is used toachieve an equality of expression of X-l<strong>in</strong>ked genesbetween males and females (Lyon 1961). Inactivationof the X chromosome is achieved through epigeneticmechanisms. The Xist gene encodes a non-cod<strong>in</strong>gRNA that is expressed <strong>in</strong> cis from the chromosomethat is to be <strong>in</strong>activated (Borsani et al. 1991). Xisttranscripts coat the X chromosome and recruit chromat<strong>in</strong>-modify<strong>in</strong>gprote<strong>in</strong>s that convert the X chromosome<strong>in</strong>to a heterochromatic, silenced state (Okamotoet al. 2004). X chromosome <strong>in</strong>activation is also subjectto impr<strong>in</strong>t<strong>in</strong>g. In mice impr<strong>in</strong>ted XCI occurs dur<strong>in</strong>gpre-implantation development, with the paternal chromosomebe<strong>in</strong>g preferentially silenced <strong>in</strong> the placenta(Takagi and Sasaki 1975). By preferential paternalXist expression at the time of zygotic genome activation,lead<strong>in</strong>g to Xist transcript accumulation andsilenc<strong>in</strong>g of the paternal X chromosome at the fourcellstage (Okamoto et al. 2005). This pattern persists<strong>in</strong> the trophectoderm l<strong>in</strong>eage, such that only thematernal X chromosome is expressed <strong>in</strong> the placenta.In the ICM, however, the paternal X chromosome isreactivated, after which the paternal and maternalchromosomes are subject to random <strong>in</strong>activation <strong>in</strong>the develop<strong>in</strong>g embryo (Okamoto et al. 2005). There isalso evidence of impr<strong>in</strong>ted XCI <strong>in</strong> cattle placentas(Xue et al. 2002). Although a systematic analysis ofXCI <strong>in</strong> cattle has yet to be done, Xist transcripts havebeen detected as early as the two-cell stage (De LaFuente et al. 1999). The late-replicat<strong>in</strong>g (and presumptive<strong>in</strong>active) X chromosome is first observed at theearly blastocyst stage (day 8), and XCI is complete byday 14.X chromosome <strong>in</strong>activation perturbations dur<strong>in</strong>g SCNTIn SCNT animals that are generated from femaledonor nuclei, XCI appears to be random, which isnormal. This means that the <strong>in</strong>active X chromosomefrom the donor nucleus must have been reactivated,after which the embryo must have reestablishedrandom activation of maternal and paternal chromosomes.This pattern of XCI <strong>in</strong> the ICM of clonedmouse blastocysts was confirmed to be normal. Otherstudies have confirmed that the <strong>in</strong>active X chromosomeis reactivated <strong>in</strong> cloned mouse blastocysts (Eggan et al.2000) but also observed aberrant expression of X-l<strong>in</strong>ked genes at later developmental stages (particularly<strong>in</strong> the mid- to late-gestation placenta) that failed toshow the normal pattern of impr<strong>in</strong>ted (paternal-<strong>in</strong>active)XCI (Nolen et al. 2005). Placental XCI is usuallyskewed <strong>in</strong> these clones, with the orig<strong>in</strong>al <strong>in</strong>active Xchromosome <strong>in</strong>activated <strong>in</strong> the (female) donor nucleus.This suggests that no reprogramm<strong>in</strong>g occurs or thataberrant reprogramm<strong>in</strong>g of XCI occurs later <strong>in</strong> theextraembryonic l<strong>in</strong>eages after NT. We have performedsimilar studies <strong>in</strong> cattle, with similar results. At theblastocyst stage, we did not f<strong>in</strong>d any evidence forabnormal expression of X-l<strong>in</strong>ked genes <strong>in</strong> clonedembryos (Smith et al. 2005). In later development,however, we observed biallelic expression of X-l<strong>in</strong>kedÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Epigenetics of Foetal Development <strong>in</strong> Clones 305genes <strong>in</strong> the placentas of dead clones rather than thematernal-specific expression that is expected if placentalXCI is normal. Interest<strong>in</strong>gly, the placentas ofsurviv<strong>in</strong>g clones, unlike those of dead clones, had onlyone active X chromosome, suggest<strong>in</strong>g that aberrantpatterns of XCI might have contributed to foetal death(Xue et al. 2002). Aberrations <strong>in</strong> X-l<strong>in</strong>ked gene expressioncould have severe consequences for placentaldevelopment (Hemberger 2002).Trophectoderm and placental defects <strong>in</strong> SCNT embryosRecent molecular evidence supports the hypothesis thatthe placental l<strong>in</strong>eage is especially vulnerable to problemsaris<strong>in</strong>g from reprogramm<strong>in</strong>g of the somatic nucleus afterNT. Anomalies <strong>in</strong> the trophectoderm of cloned embryoshave been described as early as the blastocyst stage.SCNT bov<strong>in</strong>e blastocysts have been found to have alower ratio of trophectoderm-to-ICM cells comparedwith fertilized ones (Koo et al. 2002). Furthermore, TP-1 (also known as IFN-tau), the gene specificallyexpressed by the trophoblast for maternal recognitionof pregnancy, was abnormally expressed <strong>in</strong> SCNTbov<strong>in</strong>e blastocysts (Wrenzycki et al. 2001). In theplacentas or placentomes of SCNT bov<strong>in</strong>e foetusesaberrant gene expression has been detected as early asday 25 and throughout gestation to term (Hashizumeet al. 2002; Hill et al. 2002). Furthermore, 60 prote<strong>in</strong>swere differentially expressed <strong>in</strong> term placentas of clonedcalves compared with fertilized controls (Kim et al.2005). The majority of the cloned mouse embryos diebefore a functional placenta can develop (Jouneau et al.2006) show<strong>in</strong>g phenotypic defects dur<strong>in</strong>g gastrulation(E7–E8) characterized by an abnormal ratio between theembryonic and extraembryonic portions of the embryo.The abnormal phenotypes were not fully rescued withthe addition of ESCs or ICM cells from normal embryosbut were rescued by the addition of tetraploid cells.Gene expression <strong>in</strong> cloned mice derived from nuclei ofdifferent cell types shown some cell type-specific effects,but most of the abnormalities were <strong>in</strong>dependent ofdonor cell type and seemed to be a consequence of theNT procedure, with at least 4% of genes expressed <strong>in</strong> theplacenta show<strong>in</strong>g dysregulation (Humpherys et al.2002).Strategies to Increase the Pregnancy Rates andReduce Foetal Abnormalities After SCNTFrom a practical po<strong>in</strong>t of view SCNT pregnanciespresent different level of problems: (i) negligible reprogramm<strong>in</strong>gerrors, without any obvious physiologicalconsequences; (ii) small errors, where favourable environmentalconditions or veter<strong>in</strong>ary care can compensatefor negative effects; (iii) serious reprogramm<strong>in</strong>g errorsresult<strong>in</strong>g <strong>in</strong> placental or foetal failures and loss ofpregnancy.Currently there is a lack of diagnostic methods topredict the expected level of problems <strong>in</strong> <strong>in</strong>dividualembryos and for their pre-selection prior to ET.However, there are several different practical strategieswhich might <strong>in</strong>crease the live birth rates of SCNTpregnancies:Modification of reprogramm<strong>in</strong>g success of both foetal andplacental genesThis strategy <strong>in</strong>cludes efforts to f<strong>in</strong>d the best comb<strong>in</strong>ationof many variables dur<strong>in</strong>g SCNT. The ma<strong>in</strong> effortsare focused on the core elements of the SCNT procedure:the donor cells, the recipient cytoplast, and certa<strong>in</strong>modifiers of the reprogramm<strong>in</strong>g.The ‘best’ donor cellF<strong>in</strong>d<strong>in</strong>g of cell types more amenable for reprogramm<strong>in</strong>gwould improve the SCNT, although the exist<strong>in</strong>g data isstill controversial concern<strong>in</strong>g the effect of cell type onthe reprogramm<strong>in</strong>g – as approximately 200 cell typescould be tested, the task is rather big.Improv<strong>in</strong>g donor cell culture methods and us<strong>in</strong>g lowpassage number of such cells could help to avoid geneticaberrations and mutations accumulated dur<strong>in</strong>g cellculture – although long term cultured cells can be usedas well for SCNT (Kubota et al. 2000) and telomereshorten<strong>in</strong>g seems not to be a limit<strong>in</strong>g factor <strong>in</strong> SCNT(Lanza et al. 2000). The genetic orig<strong>in</strong> of the cells is animportant factor, as it was studied <strong>in</strong> mouse <strong>in</strong> details(Wakayama et al. 1999; 2001; Rideout et al. 2000;Eggan et al. 2001). However, there is almost no dataon the effect of genetic component <strong>in</strong> domestic animals.The cell cycle status of the donor cells plays animportant role, and several studies reported on thepositive effect of serum starvation <strong>in</strong>duced G0 (Wilmutet al. 1997) or cell culture confluency <strong>in</strong>duced G1 stages(Cibelli et al. 1998a, 1998b, 2002; Wells et al. 2003). Thecell cycle must be <strong>in</strong> harmony with the state of therecipient cytoplast (Campbell et al. 1996; 2002; Wilmutet al. 2002) (see below).The ‘best’ recipient cytoplastThe orig<strong>in</strong> of the recipient oocyte can be <strong>in</strong> vivo derivedor <strong>in</strong> vitro matured. The later ones <strong>in</strong> farm animals aremuch less expensive if slaughterhouse ovaries areavailable. In mouse there is evidence, that naturallyovulated oocytes are better quality than superovulatedones (Hiiragi and Solter 2005), but <strong>in</strong> farm animals thischoice would <strong>in</strong>crease the cost substantially. The geneticorig<strong>in</strong> of the recipient oocyte also plays a role, accord<strong>in</strong>gto mouse data (Wakayama et al. 1999; Gao et al. 2004),but there is very limited <strong>in</strong>formation on this issue <strong>in</strong>other species. Genotype <strong>in</strong> that case is probably importantdue to variations <strong>in</strong> the amount of maternal-orig<strong>in</strong>reprogramm<strong>in</strong>g factors available <strong>in</strong> the cytoplast. Furthermore,<strong>in</strong>dividual differences among oocyte donorsprobably exist, but scientific exam<strong>in</strong>ation of such factorsis rather difficult, due to the low number of observationspossible when the oocyte donors are not used repetitively.Usually cytoplasts for SCNT are generated byenucleation of non-activated Metaphase II stage oocytes(Wilmut et al. 1997, 2002; Cibelli et al. 2002), butTelophase II stage can be also used as recipient withsimilar efficacy (Bordignon and Smith 1998; Baguisiet al. 1999), and recently it was demonstrated, that theuse of mitotic zygotes is also a viable option (Gredaet al. 2006; Schurmann et al. 2006; Egli et al. 2007).Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


306 A D<strong>in</strong>nyes, XC Tian and X YangIncreas<strong>in</strong>g the volume of the recipient cytoplast can alsoprovide a better reprogramm<strong>in</strong>g environment (Peuraet al. 1998).In vitro modifiers of reprogramm<strong>in</strong>gA number of <strong>in</strong> vitro approaches have been devised to<strong>in</strong>crease SCNT success. These <strong>in</strong>clude treat<strong>in</strong>g donorcells with pharmacological agents to modify theirepigenetic marks (Enright et al. 2003, 2005; Shi et al.2003) or cell cycle stage (Wells et al. 2003). In mice theHDAC <strong>in</strong>hibitor TSA can significantly improve theefficiency of both reproductive clon<strong>in</strong>g (Enright et al.2005; Kishigami et al. 2006) and NT-ESC derivation(Kishigami et al. 2006). The efficiency of TSA treatmentvaries among species, but prelim<strong>in</strong>ary data <strong>in</strong> rabbit(D<strong>in</strong>nyes et al., unpublished results) suggest that chemicaltreatments may be a promis<strong>in</strong>g alternative. Anotherapproach is fus<strong>in</strong>g transiently permeabilized cells conta<strong>in</strong><strong>in</strong>gartificially condensed chromat<strong>in</strong> (Sullivan et al.2004). Activat<strong>in</strong>g with sperm rather than by artificialmethods (Schurmann et al. 2006) can have a positive<strong>in</strong>fluence on SCNT by provid<strong>in</strong>g a better Ca-signall<strong>in</strong>gimpulse and contribut<strong>in</strong>g some factors (mRNAs, microRNAs) to the cytoplast. Serial NT (Ono et al. 2001) hasbeen shown to improve the reprogramm<strong>in</strong>g process, aswell.Favourable environmental conditionsThe environmental conditions to which the SCNTembryos and foetuses are exposed <strong>in</strong>fluence the epigenomeand can compensate for a certa<strong>in</strong> level of geneticreprogramm<strong>in</strong>g errors. Epigenetics also depend on thatcomplex ‘environment’ of the genes <strong>in</strong> question and IVCconditions can play a major role <strong>in</strong> species, where theIVC period is fairly long prior to embryo transfer.Early-stage embryo transfer can reduce aberrationsaccumulated due to IVC, and strong empirical evidence<strong>in</strong> rabbit support the importance of asynchrony betweenthe SCNT embryo age and the post-ovulatory age of therecipient oviduct or uterus (Chesne et al. 2002).Co-transfer of parthenogenetically activated or <strong>in</strong> vivoembryos together with the SCNT ones can overcomesome of the signall<strong>in</strong>g and ‘communicational’ problems ofthe SCNT embryos with the uter<strong>in</strong>e environment. Thisapproach was beneficial <strong>in</strong> pig (De Sousa et al. 2002) and<strong>in</strong> mouse (D<strong>in</strong>nyes et al., unpublished data).Husbandry, nutritional and veter<strong>in</strong>ary managementof pregnancy and delivery can be particularly important<strong>in</strong> SCNT cases and must be aimed at improved birthrates and animal welfare for both the recipient femalesand the newborns. Measures <strong>in</strong>clude precise monitor<strong>in</strong>gof foetal and placental development us<strong>in</strong>g tools such asrepeated ultrasonography and, if needed, treat<strong>in</strong>g hydropsand other <strong>in</strong>-utero complications, provid<strong>in</strong>g<strong>in</strong>duced deliveries, timed caesarean sections, and <strong>in</strong>tensivecare options <strong>in</strong> the per<strong>in</strong>atal period.Tetraploid complementationThe method of tetraploid placenta complementation isoften used for produc<strong>in</strong>g ESC-derived mice (Nagyet al. 1990) when the ESCs are mixed with atetraploid embryo, which will contribute only to theplacenta and not the foetus itself. In case of SCNTembryos the extra trophectoderm cells contributed bythe tetraploid embryo can rescue the foetus (Jouneauet al. 2006). In mouse NT-ESCs can develop to termwith a much higher efficiency when comb<strong>in</strong>ed withfertilized tetraploid placenta complement, comparedwith direct transfer of SCNT embryos (Eggan et al.2001; Hochedl<strong>in</strong>ger and Jaenisch 2002). These studiesshowed support<strong>in</strong>g evidence that poor clon<strong>in</strong>gefficiency is largely due to placenta abnormality. Incattle chimeric calves have been generated with NT-ESC-like cells (Cibelli et al. 1998a, 1998b; Iwasakiet al. 2000).Embryo aggregationComplet<strong>in</strong>g SCNT embryos with each other by embryoaggregation has been shown to be beneficial as presumablyeach embryo will conta<strong>in</strong> a different set ofreprogramm<strong>in</strong>g errors, potentially compensat<strong>in</strong>g forsome of the <strong>in</strong>dividual errors. Indeed, <strong>in</strong> mouse,aggregation of two or three SCNT embryos led to<strong>in</strong>creased cell numbers, normalized gene expressions andeightfold higher <strong>in</strong> vivo development of the result<strong>in</strong>gs<strong>in</strong>gle blastocysts (Boiani et al. 2003). In bov<strong>in</strong>e, similaraggregation of three one-cell NT embryos dur<strong>in</strong>g <strong>in</strong> vitroculture affected development of embryonic and somaticcloned embryos differently. Aggregates of embryonicclones showed a higher clon<strong>in</strong>g efficiency but there wasno evidence for complementary <strong>in</strong>teractions. In contrast,SCNT aggregates developed normally <strong>in</strong> vitro, butthe <strong>in</strong> vivo development was not improved (Misica-Turner et al. 2007).ConclusionsAfter the first decade of mammalian SCNT it is notclear yet how to overcome the current serious limitationsof the technology, but the overall progress ispromis<strong>in</strong>g, and there are no reasons to doubt thatwith<strong>in</strong> another decade a much more efficient SCNTmethodology will be available for the breeders. Asdescribed above, several l<strong>in</strong>es of evidence po<strong>in</strong>t towardto the hypothesis of relatively normal reprogramm<strong>in</strong>g<strong>in</strong> the embryonic l<strong>in</strong>eage of SCNT embryos butaberrant reprogramm<strong>in</strong>g <strong>in</strong> their trophectoderm (Cibelliet al. 1998a, 1998b; Iwasaki et al. 2000; Jouneauet al. 2006; Wakayama et al. 2006). A better reprogramm<strong>in</strong>gis the key for safety, reproducibility andanimal welfare. Fundamental knowledge on reprogramm<strong>in</strong>gmechanisms, epigenetics and developmentalpathways will provide the base for practical resolutions.Replacement or rescue of faulty SCNT placentaSomatic cell nuclear transfer placental deficiencies couldbe rescued by provid<strong>in</strong>g a well function<strong>in</strong>g placenta.AcknowledgementsThe work was supported by EU FP6 (MEXT-CT-2003-509582,LSHG-CT-2006-518240, MRTN-CT-2006-035468).Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Epigenetics of Foetal Development <strong>in</strong> Clones 307ReferencesAnimal Clon<strong>in</strong>g, 2006. A Draft Risk Assessment. FDA,Rockville, MD, USA. Available at: http://www.fda.gov/cvm/Documents/Clon<strong>in</strong>g_Risk_Assessment.pdf.Baguisi A, Behboodi E, Melican DT, Pollock JS, DestrempesMM, Cammuso C, Williams JL, Nims SD, Porter CA,Midura P, Palacios MJ, Ayres SL, Denniston RS, HayesML, Ziomek CA, Meade HM, Godke RA, Gav<strong>in</strong> WG,Overstrom EW, Echelard Y, 1999: Production of goats bysomatic cell nuclear transfer. Nat Biotechnol 17, 456–461.Boiani M, Eckardt S, Leu NA, Scholer HR, McLaughl<strong>in</strong> KJ,2003: Pluripotency deficit <strong>in</strong> clones overcome by clone-cloneaggregation: epigenetic complementation? EMBO J 22,5304–5312.Bordignon V, Smith LC, 1998: Telophase enucleation: animproved method to prepare recipient cytoplasts for use <strong>in</strong>bov<strong>in</strong>e nuclear transfer. Mol Reprod Dev 49, 29–36.Borsani G, Tonlorenzi R, Simmler Mc, Dandolo L, Arnaud D,Capra V, Grompe M, Pizzuti A, Muzny D, Lawrence C,Willard Hf, Avner P, Ballabio A, 1991: Characterization ofa mur<strong>in</strong>e gene expressed from the <strong>in</strong>active X chromosome.Nature 351, 325–329.Bortv<strong>in</strong> A, Eggan K, Skaletsky H, Akutsu H, Berry DL,Yanagimachi R, Page DC, Jaenisch R, 2003: Incompletereactivation of Oct4-related genes <strong>in</strong> mouse embryos clonedfrom somatic nuclei. Development 130, 1673–1680.Bourc’his D, Le Bourhis D, Pat<strong>in</strong> D, Niveleau A, Comizzoli P,Renard JP, Viegas-Pequignot E, 2001: Delayed and <strong>in</strong>completereprogramm<strong>in</strong>g of chromosome methylation patterns<strong>in</strong> bov<strong>in</strong>e cloned embryos. Curr Biol 11, 1542–1546.Campbell KH, Loi P, Otaegui PJ, Wilmut I., 1996: Cell cycleco-ord<strong>in</strong>ation <strong>in</strong> embryo clon<strong>in</strong>g by nuclear transfer. RevReprod 1, 40–46.Cardoso MC, Leonhardt H, 1999: DNA methyltransferase isactively reta<strong>in</strong>ed <strong>in</strong> the cytoplasm dur<strong>in</strong>g early development.J Cell Biol 147, 25–32.Chavatte-Palmer P, Heyman Y, Richard C, Monget P,LeBourhis D, Kann G, Chilliard Y, Vignon X, Renard JP,2002: Cl<strong>in</strong>ical, hormonal, and hematologic characteristics ofbov<strong>in</strong>e calves derived from nuclei from somatic cells. BiolReprod 66, 1596–1603.Chesne P, Adenot PG, Viglietta C, Baratte M, Boulanger L,Renard JP, 2002: Cloned rabbits produced by nucleartransfer from adult somatic cells. Nat Biotechnol 20, 366–369.Cibelli JB, Stice SL, Golueke PJ, Kane JJ, Jerry J, Blackwell C,de Leon FAP, Robl JM, 1998a: Cloned transgenic calvesproduced from nonquiescent fetal fibroblasts. Science 280,1256–1258.Cibelli JB, Stice SL, Golueke PJ, Kane JJ, Jerry J, Blackwell C,de Leon FAP, Robl JM, 1998b: Transgenic bov<strong>in</strong>e chimericoffspr<strong>in</strong>g produced from somatic cell-derived stem-like cells.Nat Biotechnol 16, 642–646.Cibelli JB, Campbell KH, Seidel GE, West MD, Lanza RP,2002: The health profile of cloned animals. Nat Biotechnol20, 13–14.Constant F, Guillomot M, Heyman Y, Vignon X, Laigre P,Servely JL, Renard JP, Chavatte-Palmer P, 2006: Largeoffspr<strong>in</strong>g or large placenta syndrome? Morphometric analysisof late gestation bov<strong>in</strong>e placentomes from somaticnuclear transfer pregnancies complicated by hydrallantois.Biol Reprod 75, 122–130.Daniels R, Hall V, Trounson AO, 2000: Analysis of genetranscription <strong>in</strong> bov<strong>in</strong>e nuclear transfer embryos reconstructedwith granulosa cell nuclei. Biol Reprod 63, 1034–1040.De La Fuente R, Hahnel A, Basrur PK, K<strong>in</strong>g WA, 1999: X<strong>in</strong>active-specific tran-script (Xist) expression and X chromosome<strong>in</strong>activation <strong>in</strong> the preattachment bov<strong>in</strong>e embryo.Biol Reprod 60, 769–775.De Sousa PA, Dobr<strong>in</strong>sky JR, Zhu J, Archibald AL, A<strong>in</strong>slie A,Bosma W, Bower<strong>in</strong>g J, Bracken J, Ferrier PM, Fletcher J,Gasparr<strong>in</strong>i B, Harkness L, Johnston P, Ritchie M, RitchieWA, Travers A, Albert<strong>in</strong>i D, D<strong>in</strong>nyes A, K<strong>in</strong>g TJ, Wilmut I,2002: Somatic cell nuclear transfer <strong>in</strong> the pig: control ofpronuclear formation and <strong>in</strong>tegration with improved methodsfor activation and ma<strong>in</strong>tenance of pregnancy. BiolReprod 66, 642–650.Dean W, Santos F, Stojkovic M, Zakhartchenko V, Walter J,Wolf E, Reik W, 2001: Conservation of methylationreprogramm<strong>in</strong>g <strong>in</strong> mammalian development: aberrantreprogramm<strong>in</strong>g <strong>in</strong> cloned embryos. Proc Natl Acad Sci USA98, 13734–13738.Dean W, Santos F, Reik W, 2003: Epigenetic reprogramm<strong>in</strong>g<strong>in</strong> early mammalian development and follow<strong>in</strong>g somaticnuclear transfer. Sem<strong>in</strong> Cell Dev Biol 14, 93–100.Debaun MR, Niemitz EL, Fe<strong>in</strong>berg AP, 2003: Association of<strong>in</strong> vitro fertilization with Beckwith-Wiedemann syndromeand epigenetic alterations of Lit1 and H19. Am J HumGenet 72, 156–160.Eggan K, Akutsu H, Hochedl<strong>in</strong>ger K, Rideout W III,Yanagimachi R, Jaenisch R, 2000: X-chromosome <strong>in</strong>activation<strong>in</strong> cloned mouse embryos. Science 290, 1578–1581.Eggan K, Akutsu H, Lor<strong>in</strong>g J, Jackson-Grusby L, Klemm M,Rideout WM, Yanagimachi R, Jaenisch R, 2001: Hybridvigor, fetal overgrowth, and viability of mice derived bynuclear clon<strong>in</strong>g and tetraploid embryo complementation.Proc Natl Acad Sci U S A 98, 6209–6214.Egli D, Rosa<strong>in</strong>s J, Birkhoff G, Eggan K, 2007: Developmentalreprogramm<strong>in</strong>g after chromosome transfer <strong>in</strong>to mitoticmouse zygotes. Nature 447, 679–685.Enright BP, Kubota C, Yang X, Tian XC, 2003: Epigeneticcharacteristics and development of embryos cloned fromdonor cells treated by trichostat<strong>in</strong> A or 5-aza-2’-deoxycytid<strong>in</strong>e.Biol Reprod 69, 896–901.Enright BP, Sung LY, Chang CC, Yang X, Tian XC, 2005:Methylation and acetylation characteristics of cloned bov<strong>in</strong>eembryos from donor cells treated with 5-aza-2¢-deoxycytid<strong>in</strong>e.Biol Reprod 72, 944–948.Far<strong>in</strong> PW, Piedrahita JA, Far<strong>in</strong> CE, 2006: Errors <strong>in</strong> developmentof fetuses and placentas from <strong>in</strong> vitro-produced bov<strong>in</strong>eembryos. Theriogenology 65, 178–191.Gao S, Czirr E, Chung YG, Han Z, Latham KE, 2004:Genetic variation <strong>in</strong> oocyte phenotype revealed throughparthenogenesis and clon<strong>in</strong>g: correlation with differences<strong>in</strong> pronuclear epigenetic modification. Biol Reprod 70,1162–1170.Greda P, Karasiewicz J, Modl<strong>in</strong>ski JA, 2006: Mouse zygotesas recipients <strong>in</strong> embryo clon<strong>in</strong>g. <strong>Reproduction</strong> 132, 741–748.Hashizume K, Ishiwata H, Kizaki K, Yamada O, TakahashiT, Imai K, Patel OV, Akagi S, Shimizu M, Takahashi S,Katsuma S, Shiojima S, Hirasawa A, Tsujimoto G, TodorokiJ, Izaike Y, 2002: Implantation and placental development<strong>in</strong> somatic cell clone recipient cows. Clon<strong>in</strong>g StemCells 4, 197–209.Hemberger M, 2002: The role of the X chromosome <strong>in</strong>mammalian extra embryonic development. Cytogenet GenomeRes 99, 210–217.Hiiragi T, Solter D, 2005: Reprogramm<strong>in</strong>g is essential <strong>in</strong>nuclear transfer. Mol Reprod Dev 70, 417–421.Hill JR, Schlafer DH, Fisher PJ, Davies CJ, 2002: Abnormalexpression of trophoblast major histocompatibility complexclass I antigens <strong>in</strong> cloned bov<strong>in</strong>e pregnancies is associatedwith a pronounced endometrial lymphocytic response. BiolReprod 67, 55–63.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


308 A D<strong>in</strong>nyes, XC Tian and X YangHochedl<strong>in</strong>ger K, Jaenisch R, 2002: Monoclonal mice generatedby nuclear transfer from mature B and T donor cells.Nature 415, 1035–1038.Humpherys D, Eggan K, Akutsu H, Friedman A, Hochedl<strong>in</strong>gerK, Yanagimachi R, Lander ES, Golub TR, Jaenisch R,2002: Abnormal gene expression <strong>in</strong> cloned mice derivedfrom embryonic stem cell and cumulus cell nuclei. Proc NatlAcad Sci U S A 99, 12889–12894.Inoue K, Kohda T, Lee J, Ogonuki N, Mochida K, NoguchiY, Tanemura K, Kaneko-Ish<strong>in</strong>o T, Ish<strong>in</strong>o F, Ogura A,2002: Faithful expression of impr<strong>in</strong>ted genes <strong>in</strong> cloned mice.Science 295, 297.Inoue K, Ogonuki N, Miki H, Hirose M, Noda S, Kim JM,Aoki F, Miyoshi H, Ogura A, 2006: Inefficient reprogramm<strong>in</strong>gof the hematopoietic stem cell genome follow<strong>in</strong>gnuclear transfer. J Cell Sci 119, 1985–1991.Iwasaki S, Campbell KH, Galli C, Akiyama K, 2000:Production of live calves derived from embryonic stem-likecells aggregated with tetraploid embryos. Biol Reprod 62,470–475.Jouneau A, Zhou Q, Camus A, Brochard V, Maulny L,Collignon J, Renard JP, 2006: Developmental abnormalitiesof NT mouse embryos appear early after implantation.Development 133, 1597–1607.Kang YK, Park JS, Koo DB, Choi YH, Kim SU, Lee KK,Han YM, 2002: Limited demethylation leaves mosaic-typemethylation states <strong>in</strong> cloned bov<strong>in</strong>e pre-implantationembryos. EMBO J 21, 1092–1100.Kim HR, Kang JK, Yoon JT, Seong HH, Jung JK, Lee HM,Park CS, J<strong>in</strong> DI, 2005: Prote<strong>in</strong> profiles of bov<strong>in</strong>e placentaderived from somatic cell nuclear transfer. Proteomics 5,4264–4273.Kishigami S, Mizutani E, Ohta H, Hikichi T, Van Thuan N,Wakayama S, Bui HT, Wakayama T, 2006: Significantimprovement of mouse clon<strong>in</strong>g technique by treatment withtrichostat<strong>in</strong> A after somatic nuclear transfer. BiochemBiophys Res Commun 340, 183–189.Koo DB, Kang YK, Choi YH, Park JS, Kim HN, Oh KB, SonDS, Park H, Lee KK, Han YM, 2002: Aberrant allocationsof <strong>in</strong>ner cell mass and trophectoderm cells <strong>in</strong> bov<strong>in</strong>e nucleartransfer blastocysts. Biol Reprod 67, 487–492.Kubota C, Yamakuchi H, Todoroki J, Mizoshita K, TabaraN, Barber M, Yang XZ, 2000: Six cloned calves producedfrom adult fibroblast cells after long-term culture. Proc NatlAcad Sci U S A 97, 990–995.Lanza RP, Cibelli JB, Blackwell C, Cristofalo VJ, FrancisMK, Baerlocher GM, Mak J, Schertzer M, Chavez EA,Sawyer N, Lansdorp PM, West MD, 2000: Extension of celllife-span and telomere length <strong>in</strong> animals cloned fromsenescent somatic cells. Science 288, 665–669.Latham KE, 1999: Epigenetic modification and impr<strong>in</strong>t<strong>in</strong>g ofthe mammalian genome dur<strong>in</strong>g development. Curr Top DevBiol 43, 1–49.Li E, 2002: Chromat<strong>in</strong> modification and epigenetic reprogramm<strong>in</strong>g<strong>in</strong> mammalian development. Nat Rev Genet 3, 662–673.Lyon MF, 1961: Gene action <strong>in</strong> the X-chromosome of themouse (Mus musculus L.). Nature 190, 372–373.Mayer W, Niveleau A, Walter J, Fundele R, Haaf T, 2000:Demethylation of the zygotic paternal genome. Nature 403,501–502.Misica-Turner PM, Oback FC, Eichenlaub M, Wells DN,Oback B, 2007: Aggregat<strong>in</strong>g embryonic but not somaticnuclear transfer embryos <strong>in</strong>creases clon<strong>in</strong>g efficiency <strong>in</strong>cattle. Biol Reprod 76, 268–278.Morgan HD, Santos F, Green K, Dean W, Reik W, 2005:Epigenetic reprogramm<strong>in</strong>g <strong>in</strong> mammals. Hum Mol Genet14, R47–R58.Nagy A, Gocza E, Diaz Em, Prideaux Vr, Ivanyi E, MarkkulaM, Rossant J, 1990: Embryonic stem cells alone are able tosupport fetal development <strong>in</strong> the mouse. Development 110,815–821.Nolen LD, Gao SR, Han ZM, Mann MRW, Chung YG, OtteAP, Bartolomei MS, Latham KE, 2005: X chromosomereactivation and regulation <strong>in</strong> cloned embryos. Dev Biol279, 525–540.Okamoto I, Otte A P, Allis C D, Re<strong>in</strong>berg D, Heard E, 2004:Epigenetic dynamics of impr<strong>in</strong>ted X <strong>in</strong>activation dur<strong>in</strong>gearly mouse development. Science 303, 644–649.Okamoto I, Arnaud D, Le Baccon P, Otte AP, Disteche CM,Avner P, Heard E, 2005: Evidence for de novo impr<strong>in</strong>ted X-chromosome <strong>in</strong>activation <strong>in</strong>dependent of meiotic <strong>in</strong>activation<strong>in</strong> mice. Nature 438, 369–373.Ono Y, Shimozawa N, Ito M, Kono T, 2001: Cloned micefrom fetal fibroblast cells arrested at metaphase by a serialnuclear transfer. Biol Reprod 64, 44–50.Panarace M, Agu¨ero JI, Garrote M, Jauregui G, Segovia A,Canè L, Gutie´rrez J, Marfil M, Rigali F, Pugliese M, YoungS, Lagioia J, Garmil C, Forte Pontes JE, Ereno Junio JC,Mower S, Med<strong>in</strong>a M, 2007: How healthy are clones andtheir progeny: 5 years of field experience. Theriogenology67, 142–151.Peura TT, Lewis IM, Trounson AO., 1998: The effect ofrecipient oocyte volume on nuclear transfer <strong>in</strong> cattle. MolReprod Dev 50, 185–191.Reik W, Santos F, Mitsuya K, Morgan H, Dean W, 2003:Epigenetic Asymmetry In The Mammalian Zygote AndEarly Embryo: Relationship to l<strong>in</strong>eage commitment? PhilosTrans R Soc Lond B Biol Sci 358, 1403–1409.Renard J P, Maruotti J, Jouneau A, Vignon X, 2007: Nuclearreprogramm<strong>in</strong>g and pluripotency of embryonic cells: applicationto the isolation of embryonic stem cells <strong>in</strong> farmanimals. Theriogenology 68, S196–S205.Rideout WM, Wakayama T, Wutz A, Eggan K, Jackson-Grusby L, Dausman J, Yanagimachi R, Jaenisch R, 2000:Generation of mice from wild-type and targeted ES cells bynuclear clon<strong>in</strong>g. Nat Genet 24, 109–110.Santos F, Zakhartchenko V, Stojkovic M, Peters A, Jenuwe<strong>in</strong>T, Wolf E, Reik W, Dean W, 2003: Epigenetic mark<strong>in</strong>gcorrelates with developmental potential <strong>in</strong> cloned bov<strong>in</strong>epreimplantation embryos. Curr Biol 13, 1116–1121.Schurmann A, Wells DN, Oback B, 2006: Early zygotes aresuitable recipients for bov<strong>in</strong>e somatic nuclear transfer andresult <strong>in</strong> cloned offspr<strong>in</strong>g. <strong>Reproduction</strong> 132, 839–848.Shi W, Hoeflich A, Flasw<strong>in</strong>kel H, Stojkovic M, Wolf E,Zakhartchenko V, 2003: Induction of a senescent-likephenotype does not confer the ability of bov<strong>in</strong>e immortalcells to support the development of nuclear transferembryos. Biol Reprod 69, 301–309.Smith SL, Everts RE, Tian XC, Du FL, Sung LY, Rodriguez-Zas SL, Jeong BS, Renard JP, Lew<strong>in</strong> HA, Yang XZ, 2005:Global gene expression profiles reveal significant nuclearreprogramm<strong>in</strong>g by the blastocyst stage after clon<strong>in</strong>g. ProcNatl Acad Sci U S A 102, 17582–17587.Somers J, Smith C, Donnison M, Wells DN, Henderson H,McLeay L, Pfeffer PL, 2006: Gene expression profil<strong>in</strong>g of<strong>in</strong>dividual bov<strong>in</strong>e nuclear transfer blastocysts. <strong>Reproduction</strong>131, 1073–1084.Suemizu H, Aiba K, Yoshikawa T, Sharov AA, Shimozawa N,Tamaoki N, Ko MSH, 2003: Expression profil<strong>in</strong>g ofplacentomegaly associated with nuclear transplantation ofmouse es cells. Dev Biol 253, 36–53.Sullivan EJ, Kas<strong>in</strong>athan S, Kas<strong>in</strong>athan P, Robl JM, Collas P,2004: Cloned calves from chromat<strong>in</strong> remodeled <strong>in</strong> vitro. BiolReprod 70, 146–153.Suteevun T, Parnpai R, Smith SL, Chang CC, MuenthaisongS, Tian XC, 2006: Epigenetic characteristics of cloned and <strong>in</strong>vitro-fertilized swamp buffalo (Bubalus bubalis) embryos.J Anim Sci 84, 2065–2071.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Epigenetics of Foetal Development <strong>in</strong> Clones 309Takagi N, Sasaki M, 1975: Preferential <strong>in</strong>activation of thepaternally derived X chromosome <strong>in</strong> the extraembryonicmembranes of the mouse. Nature 256, 640–642.Tamashiro KLK, Wakayama T, Akutsu H, Yamazaki Y,Lachey JL, Wortman MD, Seeley RJ, D’Alessio DA,Woods SC, Yanagimachi R, Sakai RR, 2002: Cloned micehave an obese phenotype not transmitted to their offspr<strong>in</strong>g.Nat Med 8, 262–267.Wagschal A, Feil R, 2006: Genomic impr<strong>in</strong>t<strong>in</strong>g <strong>in</strong> the placenta.Cytogenet Genome Res 113, 90–98.Wakayama T, Yanagimachi R, 2001: Mouse clon<strong>in</strong>g withnucleus donor cells of different age and type. Mol ReprodDev 58, 376–383.Wakayama T, Rodriguez I, Perry AC, Yanagimachi R,Mombaerts P, 1999: Mice cloned from embryonic stemcells. Proc Natl Acad Sci U S A 96, 14984–14989.Wakayama S, Jakt ML, Suzuki M, Araki R, Hikichi T,Kishigami S, Ohta H, Van Thuan N, Mizutani E, SakaideY, Senda S, Tanaka S, Okada M, Miyake M, Abe M,Nishikawa SI, Shiota K, Wakayama T, 2006: Equivalencyof nuclear transfer-derived embryonic stem cells to thosederived from fertilized mouse blastocyst. Stem Cells 24,2023–2033.Wang L, Duan E, Sung LY, Jeong BS, Yang X, Tian XC,2005: Generation and characterization of pluripotentstem cells from cloned bov<strong>in</strong>e embryos. Biol Reprod 73,149–155.Wells DN, Laible G, Tucker FC, Miller AL, Oliver JE, XiangT, Forsyth JT, Berg MC, Cockrem K, L’Huillier PJ, TervitHR, Oback B, 2003a: Coord<strong>in</strong>ation between donor cell typeand cell cycle stage improves nuclear clon<strong>in</strong>g efficiency <strong>in</strong>cattle. Theriogenology 59, 45–59.Wells DN, Laible G, Tucker FC, Miller AL, Oliver JE, XiangT, Forsyth JT, Berg MC, Cockrem K, L’Huillier PJ, TervitHR, Oback B, 2003b: Coord<strong>in</strong>ation between donor cell typeand cell cycle stage improves nuclear clon<strong>in</strong>g efficiency <strong>in</strong>cattle. Theriogenology 59, 45–59.Wilk<strong>in</strong>s JF, Haig D, 2003: What good is genomic impr<strong>in</strong>t<strong>in</strong>g:the function of parent-specific gene expression. Nat RevGenet 4, 359–368.Wilmut I, Schnieke AE, McWhir J, K<strong>in</strong>d AJ, Campbell KH,1997: Viable offspr<strong>in</strong>g derived from fetal and adult mammaliancells. Nature 385, 810–813.Wilmut I, Beaujean N, de Sousa PA, D<strong>in</strong>nyes A, K<strong>in</strong>g TJ,Paterson LA, Wells DN, Young LE, 2002: Somatic cellnuclear transfer. Nature 419, 583–587.Wrenzycki C, Wells D, Herrmann D, Miller A, Oliver J, TervitR, Niemann H, 2001: Nuclear transfer protocol affectsmessenger RNA expression patterns <strong>in</strong> cloned bov<strong>in</strong>eblastocysts. Biol Reprod 65, 309–317.Xue F, Tian XC, Du FL, Kubota C, Taneja M, D<strong>in</strong>nyes A,Dai YP, Lev<strong>in</strong>e H, Pereira LV, Yang XZ, 2002: Aberrantpatterns of X chromosome <strong>in</strong>activation <strong>in</strong> bov<strong>in</strong>e clones.Nat Genet 31, 216–220.Yang L, Chavatte-Palmer P, Kubota C, O’Neill M, HoaglandT, Renard JP, Taneja M, Yang XZ, Tian XC, 2005:Expression of impr<strong>in</strong>ted genes is aberrant <strong>in</strong> deceasednewborn cloned calves and relatively normal <strong>in</strong> surviv<strong>in</strong>gadult clones. Mol Reprod Dev 71, 431–438.Young LE, S<strong>in</strong>clair KD, Wilmut I, 1998: Large offspr<strong>in</strong>gsyndrome <strong>in</strong> cattle and sheep. Rev Reprod 3, 155–163.Zhu J, Telfer EE, Fletcher J, Spr<strong>in</strong>gbett A, Dobr<strong>in</strong>sky JR, DeSousa PA, Wilmut I, 2002: Improvement of an electrical activationprotocol for porc<strong>in</strong>e oocytes. Biol Reprod 66, 635–641.Zuccotti M, Garagna S, Redi CA, 2000: Nuclear transfer,genome reprogramm<strong>in</strong>g and novel opportunities <strong>in</strong> celltherapy. J Endocr<strong>in</strong>ol Invest 23, 623–629.Author’s address (for correspondence): A D<strong>in</strong>nyes, Genetic Reprogramm<strong>in</strong>gGroup, Agricultural Biotechnology Centre and MolecularAnimal Biotechnology Laboratory, Szent Istvan University, Godollo,H–2100, Hungary. E-mail: d<strong>in</strong>nyes@abc.huConflict of <strong>in</strong>terest: The authors declare no conflict of <strong>in</strong>terests.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 310–316 (2008); doi: 10.1111/j.1439-0531.2008.01179.xISSN 0936-6768A Proposed Role for VEGF Isoforms <strong>in</strong> Sex-Specific Vasculature Development <strong>in</strong> theGonadRC Bott, DT Clopton and AS CuppAnimal Science Department, University of Nebraska-L<strong>in</strong>coln, L<strong>in</strong>coln, NE, USAContentsMany scientists have expended efforts to determ<strong>in</strong>e whatregulates development of an <strong>in</strong>different gonad <strong>in</strong>to either atestis or ovary. Expression of Sry and upregulation of Sox9 arefactors that <strong>in</strong>itiate formation of the testis-specific pathway toallow for both sex-specific vasculature and sem<strong>in</strong>iferous cordformation. Migration of mesonephric precursors of peritubularmyoid cells and endothelial cells <strong>in</strong>to the differentiat<strong>in</strong>gtestis is a critical step <strong>in</strong> formation of both of these structures.Furthermore, these events appear to be <strong>in</strong>itiated downstreamfrom Sry expression. Sertoli cell secretion of growth factorsacts to attract these mesonephric cells. One hypothesis is that agrowth factor specific for these cell l<strong>in</strong>ages act <strong>in</strong> concert tocoord<strong>in</strong>ate migration of both peritubular and endothelial cells.A second hypothesis is that several growth factors stimulatemigration and differentiation of mesonephric ‘stem-like’ cellsto result <strong>in</strong> migration and differentiation <strong>in</strong>to several differentcell l<strong>in</strong>eages. While the specific mechanism is unclear, severalgrowth factors have been implicated <strong>in</strong> the <strong>in</strong>itiation ofmesonephric cell migration. This review will focus on theproposed mechanisms of a growth factor, Vascular EndothelialGrowth Factor, and how different angiogenic and <strong>in</strong>hibitoryisoforms from this s<strong>in</strong>gle gene may aid <strong>in</strong> development oftestis-specific vascular development.IntroductionWhy study gonadal differentiation and sex-specificvascular development?Infertility affects 40–70 million couples; of thoseapproximately 50% of the <strong>in</strong>fertility problems areattributed to male-related factors which <strong>in</strong>clude: lowsperm count, abnormal spermatogenesis, and reducedandrogen production (Skakkebaek 2004). In the past12 years, the <strong>in</strong>cidence of male <strong>in</strong>fertility cases has<strong>in</strong>creased at an alarm<strong>in</strong>g rate result<strong>in</strong>g <strong>in</strong> the developmentof a new syndrome – testicular dysgenesis syndrome.The reasons for the <strong>in</strong>crease <strong>in</strong> testicularabnormalities are not understood but may be a resultof genetic or environmental disruption of cell differentiationdur<strong>in</strong>g testis morphogenesis (Skakkebaek 2004).Formation of testicular cords, and sex-specific vasculature,are the two morphological hallmarks that dist<strong>in</strong>guisha testis from an ovary. Testis development is<strong>in</strong>itiated by Sertoli cell differentiation and expression ofSry, caus<strong>in</strong>g mesonephric cell migration <strong>in</strong>to the differentiat<strong>in</strong>gtestis to form sem<strong>in</strong>iferous cords. Removal ofthe mesonephros or blockage of mesonephric cellmigration prevents cord formation. Thus, the migrationof mesonephric derived cells, which <strong>in</strong>clude pre-endothelialand pre-peritubular cells, are critical to testisdevelopment.What is known about embryonic testis morphogenesis?In the mouse, the Sry gene is expressed <strong>in</strong> embryos at10.5 days post-coitus (dpc) and ceases after 12.5 dpc.Sry <strong>in</strong>duces expression of genes which cause differentiationof the Sertoli cell l<strong>in</strong>eage from precursorsomatic cells <strong>in</strong> the coelomic epithelium (Fig. 1) (Cuppand Sk<strong>in</strong>ner 2005). So even though Sry function isimportant, expression <strong>in</strong> the rodent is brief. The Sertolicell is the first cell of the testis to differentiate (Magreand Jost 1991). After differentiation, Sertoli cells beg<strong>in</strong>to proliferate and simultaneously move <strong>in</strong>to the gonadproper, form<strong>in</strong>g aggregates with primordial germ cells.Proliferation of Sertoli cells <strong>in</strong>creases the size of thetestis, and this proliferation appears to be solelydependent on the expression of Sry (Schmahl et al.2000). Without differentiation and proliferation of theSertoli cells, the <strong>in</strong>different gonad would not develop<strong>in</strong>to a testis.After Sertoli cell differentiation, testicular cords andsex-specific vasculature develop to establish adult testismorphology. In the mouse, these events occur dur<strong>in</strong>g11.5–12 dpc and are complete by 12.5 dpc while <strong>in</strong> therat these events occur between embryonic day 13.5–14(Mart<strong>in</strong>eau et al. 1997). Induction of cord formation is<strong>in</strong>itiated by cell migration from the adjacent mesonephros<strong>in</strong>to the develop<strong>in</strong>g testis to surround theprimordial germ and Sertoli cell aggregates (Buehret al. 1993; Merchant-Larios et al. 1993). Mesonephriccell migration is the result of Sry regulated expressionof paracr<strong>in</strong>e growth factors secreted by Sertoli cells(Mart<strong>in</strong>eau et al. 1997). Paracr<strong>in</strong>e growth factors act aschemo-attractants <strong>in</strong>duc<strong>in</strong>g mesonephric cell migrationand cord formation (Ricci et al. 1999; Cupp et al. 2000,2002, 2003; Colv<strong>in</strong> et al. 2001). If mesonephric cellmigration is blocked, or if the mesonephros is removed,no cords will form. The population of cells that migratefrom the mesonephros is proposed to be pre-endothelialand ⁄ or pre-peritubular cells (Buehr et al. 1993; Merchant-Larioset al. 1993). Therefore, <strong>in</strong> addition tosurround<strong>in</strong>g the Sertoli-primordial germ cell aggregatesto form testis cords, the migrat<strong>in</strong>g mesonephric cellsmay also <strong>in</strong>itiate the formation of vasculature with<strong>in</strong>the develop<strong>in</strong>g testis. Platelet-derived growth factor(Uzumcu et al. 2002a) and VEGF (Bott et al. 2006)have been demonstrated to be necessary for cordformation; however, only VEGF has been demonstratedto be critical for both vascular development andcord formation.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


VEGFA Isoforms as Regulators of Testis Vasculature 311Sex determ<strong>in</strong>ationTestis morphogenesis# Tail somites (ts)Da yspostcoitum(dpc)8 10–15 16–18 19–23 24–3010. 5 10.7–11.2 11.3–11.5 11.6–11.9 12–12.5Fig. 1. Sry-regulated events occurr<strong>in</strong>gdur<strong>in</strong>g sex determ<strong>in</strong>ation andtestis morphogenesis us<strong>in</strong>g dayspost-coitum and tail somites asdevelopmental markers (modifiedfrom Cupp and Sk<strong>in</strong>ner 2005)MesonephrosGenital RidgeSRY mRN APrim ordialGe rm CellSertoli celldifferentiationSertoli cellproliferationMesonephric ce llmigrationCord formatio nWhat is known about formation of sex-specific vasculardevelopment?Very little is known about what regulates sex specificvascular development <strong>in</strong> the gonads. Pre-endothelialcells <strong>in</strong> the <strong>in</strong>different gonad express markers for bothve<strong>in</strong>s (EphB4) and arteries (ephr<strong>in</strong> B2); however thischanges as each gonad develops sex-specific structures.In the testis, endothelial cells migrate from the mesonephrosand form vascular networks with arterialmarkers between cords and the coelomic vessel. Thecoelomic vessel is the major testicular artery that isformed when mesonephric endothelial cells migratefrom the mesonephros <strong>in</strong>to the testis and is unique tothe testis. As endothelial cells differentiate a greaterpercentage develop <strong>in</strong>to arteries and contribute to thecoleomic vessel than those which contribute to ve<strong>in</strong>swith<strong>in</strong> the testis. The development of arteries may benecessary to aid movement of testosterone to thedevelop<strong>in</strong>g male reproductive tract which allows for itsma<strong>in</strong>tenance and differentiation (Brennan et al. 2002).In contrast, the ovary develops similar amounts ofarterial and venous networks from endothelial cells.The orig<strong>in</strong> of vasculature is different <strong>in</strong> the testis andovary with vascular networks develop<strong>in</strong>g throughangiogenesis (branch<strong>in</strong>g from exist<strong>in</strong>g vasculature <strong>in</strong>the mesonephros) <strong>in</strong> the testis and through vasculogenesis(de novo or neovascularization) <strong>in</strong> the ovary(Brennan et al. 2002). It is likely that the testis andovary develop vascular patterns from different endothelialcell orig<strong>in</strong>s due to expression of sex-specifictranscription factors. Furthermore, sex-specific transcriptionfactors stimulate different growth factors tonot only <strong>in</strong>itate blood vessel formation but organspecificsupport structures such as sem<strong>in</strong>iferous cordsor oogonial cysts.Experiments us<strong>in</strong>g knockout mice demonstrate thatdisruption of vascular development also causes sexreversaland abnormalities <strong>in</strong> germ cell development.Mice that are null for Wnt4 (Jeays-Ward et al. 2003)and Follistat<strong>in</strong> (Fst) (Yao et al. 2004) have sex-reversedXX gonads with ectopic expression of a coelomic vesseland vasculature surround<strong>in</strong>g sem<strong>in</strong>iferous cord-likestructures. The Wnt4 gene regulates expression of Fstso it is not surpris<strong>in</strong>g that mice null for both of thesegenes have the same phenotype. Follistat<strong>in</strong> is onlyexpressed <strong>in</strong> the XX gonad and <strong>in</strong> general acts to <strong>in</strong>hibitactiv<strong>in</strong>s. Therefore, there is potential for activ<strong>in</strong>s to be<strong>in</strong>volved <strong>in</strong> formation of the coelomic vessel. In contrastInhib<strong>in</strong> beta b (Inhbb) XY null mice do not formcoelomic vessels 50% of the time. Thus, Inhbb null XYgonads are sometimes sex-reversed to XX vascularphenotypes. Activ<strong>in</strong> A is composed of two <strong>in</strong>hib<strong>in</strong> betaa, Inhba, subunits while Activ<strong>in</strong> B is composed of twoInhbb subunits. In the ovary, only Inhbb is expressedand not Inhba. Furthermore, expression of Inhbb isfourfold higher <strong>in</strong> the testis compared to the ovary at12.5 dpc. Mat<strong>in</strong>g of Wnt4 and Inhbb null mice recapitulatethe normal phenotype <strong>in</strong> XY gonads to form acoelomic vessel. Yet, mat<strong>in</strong>gs between Inhbb and Fstnull mice did not recapitulate the XY vascular phenotype,thus Wnt4 suppression of Inhbb appears to be<strong>in</strong>dependent of Fst <strong>in</strong>hibitory actions on activ<strong>in</strong>s (Jeays-Ward et al. 2003; Yao et al. 2006).Dur<strong>in</strong>g normal testis development, Sry antagonizesexpression of Wnt4 to suppress Fst and relieve <strong>in</strong>hibitionof Inhbb (Fig. 2). We can only speculate that Inhbbtranscriptional regulation is due to expression of Sox-9,or other growth factors downstream of Sry expression.Inhbb stimulates endothelial cell migration and formationof a coelomic vessel. In XX gonads with no Wnt4repression, Fst expression is <strong>in</strong>creased and Inhbbexpression is down regulated to prevent endothelial cellmigration and formation of ovarian-specific vascularpatterns (Yao et al. 2004).AMHXYSrySox9FGF-9?InhbbActiv<strong>in</strong> ABMPsTestis vascular patternsXXWnt4FstOvarian vascular patternsFig. 2. Genes demonstrated to be <strong>in</strong>volved <strong>in</strong> sex-specific vascularpatterns through development of null mice or treatment of <strong>in</strong>differentgonads <strong>in</strong> organ cultureÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


312 RC Bott, DT Clopton and AS CuppIn addition to the potential actions of Activ<strong>in</strong> B, XXgonads treated with bone morphogenic prote<strong>in</strong>s(BMP), anti-mullerian hormone (AMH) and Activ<strong>in</strong>A <strong>in</strong> vitro developed a sex-reversed phenotype withformation of an ectopic coelomic vessel; however,AMH deficient mice did not develop this same phenotype(Ross et al. 2003; Yao et al. 2004). Bone morphogenicprote<strong>in</strong>, AMH, Activ<strong>in</strong> A and Inhbb aremembers of the transform<strong>in</strong>g growth factor-beta(TGF-b) superfamily and directly regulate cell functionthrough <strong>in</strong>hibition or activation of the SMAD signaltransduction pathway. In the zebrafish, SMAD responseelements <strong>in</strong> the promoter region of Vegfa genealso appear to regulate expression of different isoforms(He and Chen 2005). Thus, the Vegfa gene may be adownstream gene regulated by members of the TGFfamily to regulate sex-specific vasculature <strong>in</strong> the develop<strong>in</strong>ggonad (Fig. 2).What is known about the vascular endothelial growthfactor family?The VEGF family is composed of five ligands: VEGF(VEGF-A), VEGF-B, VEGF-C, VEGF-D and placentagrowth factor. VEGF (VEGF-A) is the bestcharacterized and most potent VEGF molecule.VEGF works through both Fms-like tyros<strong>in</strong>e k<strong>in</strong>ase1 (FLT1) and K<strong>in</strong>ase doma<strong>in</strong> region receptor (KDR),to elicit its effects on endothelial cell migration,differentiation, proliferation and survival and apoptosis.The primary receptor <strong>in</strong>volved <strong>in</strong> neovascularizationof organs with VEGF is KDR which conta<strong>in</strong>s atyros<strong>in</strong>e k<strong>in</strong>ase signal transduction doma<strong>in</strong> (Waltenbergeret al. 1994). There are conflict<strong>in</strong>g reports aboutthe role of FLT1 <strong>in</strong> regulation of VEGF’s actions. InFlt1 knockouts, the mice die due to vasculatureovergrowth suggest<strong>in</strong>g that FLT1 acts as a decoyreceptor to <strong>in</strong>hibit VEGF-dependent vascular development.Furthermore, FLT1 can also <strong>in</strong>hibit VEGF’sactions by dimeriz<strong>in</strong>g with KDR to <strong>in</strong>hibit signaltransduction. Thus, FLT1 appears to act as a negativeregulator of VEGF’s actions on endothelial cellmigration, survival and proliferation (Ferrara 2000;Olsson et al. 2006).Both mRNA and prote<strong>in</strong> for VEGF and KDR arepresent dur<strong>in</strong>g testis morphogenesis and are expressed<strong>in</strong> cells, like the Sertoli cell, that direct testis development.However, Flt1 is not expressed until after cordformation <strong>in</strong> the testis (Bott et al. 2006). The absenceof FLT1 dur<strong>in</strong>g mesonephric endothelial cell migrationand establishment of vasculature <strong>in</strong> the testis mayallow for endothelial cell migration through VEGF. Incontrast, the ovary, which does not have mesonephriccell migration, expresses both Flt1 and Kdr dur<strong>in</strong>gdevelopment (Pohlmann et al. 2004). Thus, VEGFangiogenic isoforms may be bound by FLT1 receptor<strong>in</strong> the ovary <strong>in</strong>hibit<strong>in</strong>g the ability of mesonephricendothelial cells to be recruited <strong>in</strong>to the develop<strong>in</strong>govary; thus provid<strong>in</strong>g a different endothelial cell orig<strong>in</strong>for ovarian vasculature.Vascular endothelial growth factor is transcribedfrom a s<strong>in</strong>gle gene that has eight exons and is alternativelyspliced <strong>in</strong>to different isoforms each conta<strong>in</strong><strong>in</strong>g aVEGF 205VEGF 188VEGF 164VEGF 144VEGF 120VEGF 110VEGF 164bExons 1–5 6A 6B 7 8aExons 1–5 6A 7 8aExons 1–5 7 8aExons 1–5 6A 8aExons 1–5Exons 1–37Bdifferent number of am<strong>in</strong>o acids. The most commonangiogenic isoforms are VEGF205, 188, 164, 144 and120 (<strong>in</strong> humans the angiogenic isoforms are one am<strong>in</strong>oacid longer; Fig. 3). The predom<strong>in</strong>ant isoforms expressed<strong>in</strong> tissues are VEGF188, 164 and 120 (Veikkolaand Alitalo 1999).In 2002, an additional isoform, VEGF165b, wasidentified which conta<strong>in</strong>ed part of the 3’ UTR that isnow determ<strong>in</strong>ed to be exon 8b (Fig. 3). Furthermore,recent studies have demonstrated that the humanVEGF165b isoform is anti-angiogenic <strong>in</strong> function and<strong>in</strong>hibits signal transduction through KDR (Bates et al.2002; Woolard et al. 2004). Thus, this isoform is<strong>in</strong>hibitory to the actions of VEGF. Recently severalother <strong>in</strong>hibitory isoforms have been identified <strong>in</strong> thehuman and we have cloned several <strong>in</strong> both the rat andbov<strong>in</strong>e (VEGF165b accession number EU040284;VEGF189b accession number EU040285). Therefore,it appears that for every angiogenic isoform there is asister <strong>in</strong>hibitory isoform that is formed when exon 8ais replaced with exon 8b. (In rodents and cattle thesister <strong>in</strong>hibitory isoform is one am<strong>in</strong>o acid longer.)The primary functions of angiogenic VEGF isoformsare to <strong>in</strong>duce endothelial cell migration, survival,proliferation and differentiation to <strong>in</strong>itiate angiogenesiswith<strong>in</strong> develop<strong>in</strong>g organs and tumors. The <strong>in</strong>hibitoryVEGF isoforms appear to modulate thesefunctions.In addition to the multiple VEGF isoforms, two coreceptors,neuropil<strong>in</strong>1 (NRP1) and neuropil<strong>in</strong>2 (NRP2),that b<strong>in</strong>d to specific VEGF isoforms (i.e. VEGF164,VEGF188) have been identified. The predom<strong>in</strong>antfunction of NRP1 appears to be stabilization ofVEGF164 b<strong>in</strong>d<strong>in</strong>g to KDR which augments signaltransduction. The two co-receptors can also stabilizeb<strong>in</strong>d<strong>in</strong>g of VEGF isoforms to FLT1. In fact, FLT1 mayregulate VEGF actions by ty<strong>in</strong>g up NRP boundisoforms. While both NRP co-receptors can b<strong>in</strong>d toFLT1 it appears that only NRP1 can stabilize signaltransduction through KDR. Interest<strong>in</strong>gly, KDR, butnot FLT1 is expressed dur<strong>in</strong>g sem<strong>in</strong>iferous cord formation(Bott et al. 2006) thus the ability of NRP1 toenhance signal transduction upon b<strong>in</strong>d<strong>in</strong>g of VEGFisoforms to KDR may be critical dur<strong>in</strong>g testis differentiation.8a8aExons 1–5 7 8bFig. 3. Exons <strong>in</strong> VEGF gene that compose each VEGF isoformÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


VEGFA Isoforms as Regulators of Testis Vasculature 313What Signal Transduction Pathways DoesVEGF Activate Through KDR and Which areCritical for Testis Cord Formation?Vascular endothelial growth factor <strong>in</strong>duces the activationof several signal transduction pathways uponb<strong>in</strong>d<strong>in</strong>g to KDR. Of these pathways, Phospholipase Cgamma activates prote<strong>in</strong> k<strong>in</strong>ase C caus<strong>in</strong>g activation ofRas or Raf and ultimately MAPK and ERK phosphorylation.The result of ERK phosphorylation is endothelialcell proliferation (Shu et al. 2002). The PI3-k<strong>in</strong>asepathway has been demonstrated to affect migration andpermeability through Rac and permeability and survivalthrough Akt (Gerber et al. 1998). The p38MAPK andFAK pathways are also postulated to affect act<strong>in</strong>reorganization and focal adhesion turnover to <strong>in</strong>fluencemigration (Goligorsky et al. 1999). Several of theseVEGF signal transduction pathways may <strong>in</strong>teract to<strong>in</strong>itiate endothelial cell differentiation, migration, proliferationand survival. Inhibition of the MAPK pathwayaffects cord formation (Uzumcu et al. 2002b) <strong>in</strong>testis organ cultures; however, PI3-k<strong>in</strong>ase <strong>in</strong>hibits bothsem<strong>in</strong>iferous cord formation (Uzumcu et al. 2002b; Bottet al. 2006) and vascular development (Bott et al. 2006).Thus, both of these sex-specific events, vascular developmentand cord formation may be regulated bymultiple growth factors through a similar signal transductionpathway.What specific roles do the VEGF isoforms have <strong>in</strong>endothelial cell migration?Endothelial cell recruitment and chemoattractionrequires several different VEGF isoforms to establish aVEGF concentration gradient. At least three of theangiogenic VEGF isoforms are associated with specificendothelial cell functions (Veikkola and Alitalo 1999).VEGF120, a highly diffusible isoform, recruits endothelialcells <strong>in</strong>to tissue to <strong>in</strong>itiate the development ofvasculogenesis while VEGF164 can recruit endothelialcells and establish large blood vessels. VEGF188, due toits hepar<strong>in</strong> b<strong>in</strong>d<strong>in</strong>g doma<strong>in</strong>, acts locally to <strong>in</strong>ducebranch<strong>in</strong>g of large blood vessels <strong>in</strong>to smaller capillariesand the development of capillary beds (Grunste<strong>in</strong> et al.2000) (Fig. 3). Furthermore, these three isoforms arethought to establish a VEGF chemoattractant gradientto allow for endothelial cell migration. In the testis,VEGF120, 188 and 164 are present dur<strong>in</strong>g sem<strong>in</strong>iferouscord formation and can develop a viable chemoattractantgradient (Bott et al. 2006). In contrast, <strong>in</strong> the ovaryonly VEGF164 and 120 are present (Pohlmann et al.2004). The absence of the larger VEGF isoforms toanchor the chemoattractant gradient <strong>in</strong>hibits endothelialcell migration and weakens the attraction to VEGFsecret<strong>in</strong>g cells (Ruhrberg 2003). Therefore, <strong>in</strong> the ovarya viable chemoattractant gradient may not be presentcontribut<strong>in</strong>g to the lack of mesonephric cell migration.VEGF165b is anti-angiogenic and appears to <strong>in</strong>hibitthe actions of VEGF164 and to a lesser extent otherVEGF isoforms. The <strong>in</strong>hibitory isoform, VEGF165b isexpressed <strong>in</strong> all tissues but is down-regulated <strong>in</strong> prostatetumors (Woolard et al. 2004), renal tumors (Bates et al.2002) and <strong>in</strong> disorders of the eye such as maculardegeneration (Perr<strong>in</strong> et al. 2005). The mechanism ofdifferential VEGF isoform expression is not known butis likely related to transcriptional and ⁄ or post-transcriptionalregulation of the VEGF gene which results <strong>in</strong>different ratios of angiogenic to <strong>in</strong>hibitory VEGFisoforms expressed.One role of the <strong>in</strong>hibitory VEGF isoforms may be to<strong>in</strong>hibit the chemoattractant gradient and <strong>in</strong>hibit endothelialcell migration (Bates et al. 2002; Woolard et al.2004). Our laboratory has subcloned and sequenced therat VEGF165b and determ<strong>in</strong>ed that there is a sexspecificexpression of VEGF165b (fourfold higher <strong>in</strong>ovary than testis; unpublished data) dur<strong>in</strong>g the timeendothelial cell migration occurs. Therefore, theVEGF165b isoform may be expressed <strong>in</strong> the ovary toprevent establishment of a chemoattractant gradientand migration of endothelial cells from the mesonephros<strong>in</strong>to the develop<strong>in</strong>g ovary.How are specific VEGF isoform actions regulated?Neuropil<strong>in</strong>1, a VEGFA co-receptor, appears to modulateVEGFA signal transduction (Klagsbrun and Eichmann2005). Neuropil<strong>in</strong>s were first identified asreceptors for axon guidance form<strong>in</strong>g complexes withthe plex<strong>in</strong>-A subfamily to modulate semaphor<strong>in</strong> signals(Takahashi et al. 1998). However, recent studies haveidentified a role for neuropil<strong>in</strong>s <strong>in</strong> VEGF-dependentangiogenic processes. Specifically, NRP1 knockout micehave vasculature, heart and neural defects and die atE10.5–12.5 suggest<strong>in</strong>g that NRP1 is necessary fornormal vascular development (Kawasaki et al. 1999).If NRP1 b<strong>in</strong>ds to semaphor<strong>in</strong> <strong>in</strong>stead of VEGF thenendothelial cell migration is <strong>in</strong>hibited (Miao et al. 1999).Likewise, if NRP1 ⁄ VEGF complexes b<strong>in</strong>d to FLT1 thisalso may <strong>in</strong>hibit endothelial cell migration and thedevelopment of vasculature.The predom<strong>in</strong>ant function of NRP1 appears to bestabilization of VEGFA164 b<strong>in</strong>d<strong>in</strong>g to KDR whichaugments signal transduction. The two co-receptors canalso stabilize b<strong>in</strong>d<strong>in</strong>g of angiogenic VEGFA isoforms toFLT1. Infact, FLT1 may regulate angiogenic VEGFactions by entic<strong>in</strong>g NRP bound isoforms to b<strong>in</strong>d to thisdecoy receptor. While both NRP co-receptors can b<strong>in</strong>dto FLT1 it appears that only NRP1 can stabilize signaltransduction through KDR.Initially, VEGFA isoforms conta<strong>in</strong><strong>in</strong>g exon 7 or ahepar<strong>in</strong> b<strong>in</strong>d<strong>in</strong>g doma<strong>in</strong> (also <strong>in</strong> exon 6) were thought tob<strong>in</strong>d to NRP1. However, there have been recent studiesthat suggest NRPs (<strong>in</strong> addition to b<strong>in</strong>d<strong>in</strong>g to VEGF164and 188 that conta<strong>in</strong> hepar<strong>in</strong> b<strong>in</strong>d<strong>in</strong>g doma<strong>in</strong>s) alsob<strong>in</strong>d to VEGFA120 (which does not have a hepar<strong>in</strong>b<strong>in</strong>d<strong>in</strong>g doma<strong>in</strong>). Although they b<strong>in</strong>d VEGFA120, the<strong>in</strong>teraction does not allow for the bridg<strong>in</strong>g of the tworeceptors (NRP1 and KDR) that occur with VEGF164or 188. Therefore, the <strong>in</strong>teraction with VEGF120 doesnot appear to stabilize and amplify signal transductionwhich occurs with VEGF164 (Pan et al. 2007).Exon 8a also appears to be important <strong>in</strong> b<strong>in</strong>d<strong>in</strong>g ofNRPs to VEGF angiogenic isoforms (Jia et al. 2006). Ithas been demonstrated that <strong>in</strong>hibitory VEGF isoformssuch as 165b cannot b<strong>in</strong>d NRPs s<strong>in</strong>ce exon 8b replacesÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


314 RC Bott, DT Clopton and AS Cupp8a. Therefore, NRPs can convey angiogenic vs <strong>in</strong>hibitoryisoform specificity to cells and if NRPs areelim<strong>in</strong>ated from cells then the actions of angiogenicisoforms may be impaired.What regulates VEGF gene expression?Hypoxia (Schweda et al. 2000), oestradiol (Muelleret al. 2000), progesterone (Mueller et al. 2003), andfactors signall<strong>in</strong>g through the SMAD signal transductionpathway (He and Chen 2005) have all beenimplicated <strong>in</strong> <strong>in</strong>creased or decreased expression ofVEGF isoforms. Results from <strong>in</strong> vitro studies (Activ<strong>in</strong>A ⁄ B, BMPs, AMH) (Yao et al. 2004) and null mice(Inhbb, Follistat<strong>in</strong> and Wnt4) (Yao et al. 2004, 2006)suggest that TGF-b family members or prote<strong>in</strong>s that<strong>in</strong>hibit their actions are <strong>in</strong>volved <strong>in</strong> modulation ofvascular development <strong>in</strong> the develop<strong>in</strong>g gonad. Interest<strong>in</strong>gly,<strong>in</strong> the zebrafish SMAD response elementsregulate VEGF gene expression (He and Chen 2005).Deletion of either SMAD1 or SMAD5 response elementson the zebrafish VEGF promoter resulted <strong>in</strong>either <strong>in</strong>hibition or enhanced transcription of the VEGFgene (He and Chen 2005). With<strong>in</strong> the mur<strong>in</strong>e VEGFpromoter there are multiple consensus sequences forSMAD response elements (AS Cupp, unpublished data).Therefore, it is highly possible that growth factorssignall<strong>in</strong>g through the SMAD signal transduction pathwaysalter the expression of VEGF and VEGF-signaltransduction mediators or potentially affect post-translationalmodification of VEGF to allow for differentratios of angiogenic vs <strong>in</strong>hibitory isoforms to beexpressed (Fig. 2).What Mouse Models Can Be Used toUnderstand the Function of VEGF Dur<strong>in</strong>gTestis Morphogenesis?Null mutations of VEGF are embryonic lethal with eventhe loss of one allele result<strong>in</strong>g <strong>in</strong> embryonic death at E11dpc (Carmeliet et al. 1996; Ferrara et al. 1996). Similarly,homozygous knockouts for either receptor, FLT1or KDR, die on 9–10 dpc (Millauer et al. 1994; Hiratsukaet al. 1998). Thus, these ubiquitous knockoutmodels prevent us from determ<strong>in</strong><strong>in</strong>g the role of VEGF<strong>in</strong> testis morphogenesis and ⁄ or vascular pattern<strong>in</strong>g ofthe gonad. Overexpression of VEGF is equally detrimentalto fertility and embryonic viability (Miquerolet al. 2000; Hum<strong>in</strong>iecki et al. 2001).Many conditional mutants have been developedwhich knock out VEGF ubiquitously later <strong>in</strong> development(<strong>in</strong> the neonate; Gerber et al. 1999) or <strong>in</strong> specifictissues (i.e. nerves <strong>in</strong> sk<strong>in</strong> (Mukouyama et al. 2005). Inthese studies VEGF was determ<strong>in</strong>ed to be required forproliferation and survival of endothelial cells. Thedeletion of VEGF <strong>in</strong> newborn mice <strong>in</strong> every cell of thebody resulted <strong>in</strong> smaller mice that did not survive toadulthood. However, if VEGF was knocked out later <strong>in</strong>the fully developed animal there was less of an effect ongrowth.Overexpression of VEGF <strong>in</strong> transgenic mice results <strong>in</strong><strong>in</strong>fertility (Korpela<strong>in</strong>en et al. 1998). Furthermore, overexpressionof human165b through the MMLV promoterresults <strong>in</strong> disruption of mammary gland development,and death postnatally <strong>in</strong> offspr<strong>in</strong>g from over-express<strong>in</strong>gfemales due to malnutrition (Qiu et al. 2007). However,until a testis-specific knockout mouse is developed it isunclear how removal of Sertoli-cell secretion of VEGFwill affect testis development and function. Thus, furtherexperiments need to be conducted <strong>in</strong> vivo to demonstratethe role of VEGF on sex-specific vascular development,sem<strong>in</strong>iferous cord formation and testis function.ConclusionApproximately 2 million couples seek treatment for<strong>in</strong>fertility every year and less than half f<strong>in</strong>d successfultreatments (Carlsen et al. 2005). Infertility problems <strong>in</strong>at least half of these couples are a result of male-relatedfactors that are created by testicular dysgenesis. Manyof the problems associated with testicular dysgenesis areproposed to <strong>in</strong>volve a disruption <strong>in</strong> embryonic differentiationof cells with<strong>in</strong> the <strong>in</strong>different gonad result<strong>in</strong>g <strong>in</strong>altered testicular development. Elucidat<strong>in</strong>g the factors<strong>in</strong>volved <strong>in</strong> sex-specific vascular development will allowfor a better understand<strong>in</strong>g of how transcription factorscoord<strong>in</strong>ate regulation of growth factors to result <strong>in</strong> atestis-specific vascular system. Furthermore, del<strong>in</strong>eat<strong>in</strong>gthe <strong>in</strong>teraction of VEGF angiogenic and <strong>in</strong>hibitoryisoforms <strong>in</strong> sex-specific vascular development promisesto be an <strong>in</strong>terest<strong>in</strong>g piece <strong>in</strong> the puzzle of gonadaldevelopment.AcknowledgementThis research was funded by NIH grants (R03HD41546-01;R03HD045350-01).ReferencesBates D, Cui T-G, Doughty J, W<strong>in</strong>kler M, Sugiono M, ShieldsJ, Peat D, Gillatt D, Harper S., 2002: VEGF 165 b, an<strong>in</strong>hibitory splice variant of vascular endothelial growthfactor, is down-regulated <strong>in</strong> renal cell carc<strong>in</strong>oma. CancerRes 62, 4123–4131.Bott RC, McFee RM, Clopton DT, Toombs C, Cupp AS,2006: Vascular endothelial growth factor and k<strong>in</strong>ase doma<strong>in</strong>region receptor are <strong>in</strong>volved <strong>in</strong> both sem<strong>in</strong>iferous cordformation and vascular development dur<strong>in</strong>g testis morphogenesis<strong>in</strong> the rat. Biol Reprod 75, 56–67.Brennan J, Karl J, Capel B, 2002: Divergent vascularmechanisms downstream of Sry establish the arterial system<strong>in</strong> the XY gonad. Dev Biol 244, 418–428.Buehr M, Gu S, McLaren A, 1993: Mesonephric contributionto testis differentiation <strong>in</strong> the fetal mouse. Development 117,273–281.Carlsen E, Swan SH, Petersen JH, Skakkebaek NE, 2005:Longitud<strong>in</strong>al changes <strong>in</strong> semen parameters <strong>in</strong> young Danishmen from the Copenhagen area. Hum Reprod 20, 942–949.Carmeliet P, Ferreira V, Breier G, Pollefeyt S, Kieckens L,Gertsenste<strong>in</strong> M, Fahrig M, Vandenhoeck A, HarpalK, Eberhardt C, Declercq C, Pawl<strong>in</strong>g J, Moons L, CollenD, Risau W, Nagy A, 1996: Abnormal blood vesseldevelopment and lethality <strong>in</strong> embryos lack<strong>in</strong>g a s<strong>in</strong>gleVEGF allele. Nature 380, 435–439.Colv<strong>in</strong> JS, Green RP, Schmahl J, Capel B, Ornitz DM, 2001:Male-to-female sex reversal <strong>in</strong> mice lack<strong>in</strong>g fibroblastgrowth factor 9. Cell 104, 875–889.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


VEGFA Isoforms as Regulators of Testis Vasculature 315Cupp A, Sk<strong>in</strong>ner M, 2005: Embryonic sertoli cell differentiation.<strong>in</strong>: Griswold M, Sk<strong>in</strong>ner M (eds), Sertoli Cell Biology,Vol. 1. Elsevier Academic Press, San Diego, pp. 43–70.Cupp AS, Kim GH, Sk<strong>in</strong>ner MK, 2000: Expression and actionof neurotrop<strong>in</strong>-3 and nerve growth factor <strong>in</strong> embryonic andearly postnatal rat testis development. Biol Reprod 63,1617–1628.Cupp AS, Tessarollo L, Sk<strong>in</strong>ner MK, 2002: Testis developmentalphenotypes <strong>in</strong> neurotrop<strong>in</strong> receptor trkA and trkCnull mutations: role <strong>in</strong> formation of sem<strong>in</strong>iferous cords andgerm cell survival. Biol Reprod 66, 1838–1845.Cupp AS, Uzumcu M, Sk<strong>in</strong>ner MK, 2003: Chemotactic role ofneurotrop<strong>in</strong> 3 <strong>in</strong> the embryonic testis that facilitates malesex determ<strong>in</strong>ation. Biol Reprod 68, 2033–2037.Ferrara N, 2000: VEGF: an update on biological andtherapeutic aspects. Curr Op<strong>in</strong> Biotechnol 11, 617–624.Ferrara N, Carver-Moore K, Chen H, Dowd M, Lu L, O’SheaKS, Powell-Braxton L, Hillan KJ, Moore MW, 1996:Heterozygous embryonic lethality <strong>in</strong>duced by targeted<strong>in</strong>activation of the VEGF gene. Nature 380, 439–442.Gerber HP, McMurtrey A, Kowalski J, Yan M, Keyt BA,Dixit V, Ferrara N, 1998: Vascular endothelial growthfactor regulates endothelial cell survival through the phosphatidyl<strong>in</strong>ositol3’-k<strong>in</strong>ase ⁄ Akt signal transduction pathway.Requirement for Flk-1 ⁄ KDR activation. J Biol Chem 273,30336–30343.Gerber H.-P, Hilan K, Ryan A, Kowalski J, Keller G.A,Rangell L, Wright B, Radtke F, Aguet M, Ferrara N, 1999:VEGF is required for growth and survival <strong>in</strong> neonatal mice.Development 126, 1149–1159.Goligorsky MS, Abedi H, Noiri E, Takhtajan A, Lense S,Romanov V, Zachary I, 1999: Nitric oxide modulation offocal adhesions <strong>in</strong> endothelial cells. Am J Physiol 276,C1271–C1281.Grunste<strong>in</strong> J, Masbad JJ, Hickey R, Giordano F, Johnson RS,2000: Isoforms of vascular endothelial growth factor act <strong>in</strong> acoord<strong>in</strong>ate fashion to recruit and expand tumor vasculature.Mol Cell Biol 20, 7282–7291.He C, Chen X, 2005: Transcription regulation of the vegf geneby the BMP ⁄ Smad pathway <strong>in</strong> the angioblast of zebrafishembryos. Biochem Biophys Res Commun 329, 324–330.Hiratsuka S, M<strong>in</strong>owa O, Kuno J, Noda T, Shibuya M, 1998:Flt-1 lack<strong>in</strong>g the tyros<strong>in</strong>e k<strong>in</strong>ase doma<strong>in</strong> is sufficient fornormal development and angiogenesis <strong>in</strong> mice. Proc NatlAcad Sci U S A 95, 9349–9354.Hum<strong>in</strong>iecki L, Chan H, Lui S, Poulsom R, Stamp G, Harris A,Bicknell R, 2001: Vascular endothelial growth factor transgenicmice exhibit reduced male fertility and placentalrejection. Mol Human Reprod 7, 255–264.Jeays-Ward K, Hoyle C, Brennan J, Dandonneau M, AlldusG, Capel B, Swa<strong>in</strong> A, 2003: Endothelial and steroidogeniccell migration are regulated by WNT4 <strong>in</strong> the develop<strong>in</strong>gmammalian gonad. Development 130, 3663–3670.Jia H, Bagherzadeh A, Hartzoulakis B, Jarvis A, Lohr M,Shaikh S, Aqil R, Cheng L, Tickner M, Esposito D, HarrisR, Driscoll PC, Selwood DL, Zachary IC, 2006: Characterizationof a bicyclic peptide neuropil<strong>in</strong>-1 (NP-1) antagonist(EG3287) reveals importance of vascular endothelial growthfactor exon 8 for NP-1 b<strong>in</strong>d<strong>in</strong>g and role of NP-1 <strong>in</strong> KDRsignal<strong>in</strong>g. J Biol Chem 281, 13493–13502.Kawasaki T, Kitsukawa T, Bekku Y, Matsuda Y, Sanbo M,Yagi T, Fujisawa H, 1999: A requirement for neuropil<strong>in</strong>-1 <strong>in</strong>embryonic vessel formation. Development 126, 4895–4902.Klagsbrun M, Eichmann A, 2005: A role for axon guidancereceptors and ligands <strong>in</strong> blood vessel development andtumor angiogenesis. Cytok<strong>in</strong>e Growth Factor Rev 16, 535–548.Korpela<strong>in</strong>en E, Karkka<strong>in</strong>en M, Tenhunen A, Lakso M,Rauvala H, Vierula M, Parv<strong>in</strong>en M, Alitalo K, 1998:Overexpression of VEGF <strong>in</strong> testis and epididymis causes<strong>in</strong>fertility <strong>in</strong> transgenic mice: evidence for nonendothelialtargets for VEGF. J Cell Biol 143, 1705–1712.Magre S, Jost A, 1991: Sertoli cells and testicular differentiation<strong>in</strong> the rat fetus. J Electron Microsc Tech 19, 172–188.Mart<strong>in</strong>eau J, Nordqvist K, Tilmann C, Lovell-Badge R, CapelB, 1997: Male-specific cell migration <strong>in</strong>to the develop<strong>in</strong>ggonad. Curr Biol 7, 958–968.Merchant-Larios H, Moreno-Mendoza N, Buehr M, 1993:The role of the mesonephros <strong>in</strong> cell differentiation andmorphogenesis of the mouse fetal testis. Int J Dev Biol 37,407–415.Miao HQ, Soker S, Fe<strong>in</strong>er L, Alonso JL, Raper JA, KlagsbrunM, 1999: Neuropil<strong>in</strong>-1 mediates collaps<strong>in</strong>-1 ⁄ semaphor<strong>in</strong> III<strong>in</strong>hibition of endothelial cell motility: functional competitionof collaps<strong>in</strong>-1 and vascular endothelial growth factor-165. J Cell Biol 146, 233–242.Millauer B, Shauver L, Plate K, Risau W, Ullrich A, 1994:Glioblastoma growth <strong>in</strong>hibited <strong>in</strong> vivo by a dom<strong>in</strong>antnegativeFlk-1 mutant. Nature 367, 576–579.Miquerol L, Langille BL, Nagy A, 2000: Embryonic developmentis disrupted by modest <strong>in</strong>creases <strong>in</strong> vascular endothelialgrowth factor gene expression. Development 127, 3941–3946.Mueller MD, Vigne JL, M<strong>in</strong>chenko A, Lebovic DI, LeitmanDC, Taylor RN, 2000: Regulation of vascular endothelialgrowth factor (VEGF) gene transcription by estrogenreceptors alpha and beta. Proc Natl Acad Sci U S A 97,10972–10977.Mueller MD, Vigne JL, Pritts EA, Chao V, Dreher E, TaylorRN, 2003: Progest<strong>in</strong>s activate vascular endothelial growthfactor gene transcription <strong>in</strong> endometrial adenocarc<strong>in</strong>omacells. Fertil Steril 79, 386–392.Mukouyama YS, Gerber HP, Ferrara N, Gu C, Anderson DJ,2005: Peripheral nerve-derived VEGF promotes arterialdifferentiation via neuropil<strong>in</strong> 1-mediated positive feedback.Development 132, 941–952.Olsson AK, Dimberg A, Kreuger J, Claesson-Welsh L, 2006:VEGF receptor signall<strong>in</strong>g – <strong>in</strong> control of vascular function.Nat Rev 7, 359–371.Pan Q, Chathery Y, Wu Y, Rathore N, Tong RK, Peale F,Bagri A, Tessier-Lavigne M, Koch AW, Watts R. J, 2007:Neuropil<strong>in</strong>-1 b<strong>in</strong>ds to VEGF121 and regulates endothelialcell migration and sprout<strong>in</strong>g. J Biol Chem 282, 24049–24056.Perr<strong>in</strong> RM, Konopatskaya O, Qiu Y, Harper S, Bates DO,Churchill AJ, 2005: Diabetic ret<strong>in</strong>opathy is associated with aswitch <strong>in</strong> splic<strong>in</strong>g from anti- to pro-angiogenic isoforms ofvascular endothelial growth factor. Diabetologia 48, 2422–2427.Pohlmann R, Clopton D, McFee R, Rozell T, Cupp A, 2004:Inhibition of Vascular Endothelial Growth Factor (VEGF)Signal<strong>in</strong>g blocks follicle progression <strong>in</strong> the per<strong>in</strong>atal ratovary. J Anim Sci 82, 233.Qiu YBH, Weeraperuma S, Wratt<strong>in</strong>g D, Murphy D, Neal CR,Bates DO, Harper SJ, 2008: Mammary alveolar developmentdur<strong>in</strong>g lactation is <strong>in</strong>hibited by the endogenousantiangiogenic growth factor isoform, VEGF165b. FASEBJ 22, 1104–1112.Ricci G, Catizone A, Innocenzi A, Galdieri M, 1999: Hepatocytegrowth factor (HGF) receptor expression and role ofHGF dur<strong>in</strong>g embryonic mouse testis development. Dev Biol216, 340–347.Ross AJ, Tilman C, Yao H, MacLaughl<strong>in</strong> D, Capel B, 2003:AMH <strong>in</strong>duces mesonephric cell migration <strong>in</strong> XX gonads.Mol Cell Endocr<strong>in</strong>ol 211, 1–7.Ruhrberg C, 2003: Grow<strong>in</strong>g and shap<strong>in</strong>g the vascular tree:multiple roles for VEGF. BioEssays 25, 1052–1060.Schmahl J, Eicher EM, Washburn LL, Capel B, 2000: Sry<strong>in</strong>duces cell proliferation <strong>in</strong> the mouse gonad. Development127, 65–73.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


316 RC Bott, DT Clopton and AS CuppSchweda F, Blumberg FC, Schweda A, Nabel C, Holmer SR,Riegger GA, Pfeifer M, Kramer BK, 2000: Effects of chronichypoxia on renal PDGF-A, PDGF-B, and VEGF geneexpression <strong>in</strong> rats. Nephron 86, 161–166.Shu X, Wu W, Mosteller RD, Broek D, 2002: Sph<strong>in</strong>gos<strong>in</strong>ek<strong>in</strong>ase mediates vascular endothelial growth factor–<strong>in</strong>ducedactivation of ras and mitogen–activated prote<strong>in</strong> k<strong>in</strong>ases.Mol Cell Biol 22, 7758–7768.Skakkebaek NE, 2004: Testicular dysgenesis syndrome: newepidemiological evidence. Int J Androl 27, 189–191.Takahashi T, Nakamura F, J<strong>in</strong> Z, Kalb RG, Strittmatter SM,1998: Semaphor<strong>in</strong>s A and E act as antagonists of neuropil<strong>in</strong>-1 and agonists of neuropil<strong>in</strong>-2 receptors. Nat Neurosci 1,487–493.Uzumcu M, Dirks KA, Sk<strong>in</strong>ner MK, 2002a: Inhibition ofplatelet-derived growth factor actions <strong>in</strong> the embryonic testis<strong>in</strong>fluences normal cord development and morphology. BiolReprod 66, 745–753.Uzumcu M, Westfall SD, Dirks KA, Sk<strong>in</strong>ner MK, 2002b:Embryonic testis cord formation and mesonephric cellmigration requires the phosphotidyl<strong>in</strong>ositol 3-k<strong>in</strong>ase signal<strong>in</strong>gpathway. Biol Reprod 67, 1927–1935.Veikkola T, Alitalo K, 1999: VEGF’s receptors and angiogenesis.Sem<strong>in</strong> Cancer Biol 9, 211–220.Waltenberger J, Claesson-Welsh L, Siegbahn A, Shibuya M,Held<strong>in</strong> CH, 1994: Different signal transduction properties ofKDR and Flt1, two receptors for vascular endothelialgrowth factor. J Biol Chem 269, 26988–26995.Woolard J, Wang W-Y, Bevan H, Qui Y, MorbidelliL, Pritchard-Jones R, Cui T-G, Sugiono M, Wa<strong>in</strong>eE, Perr<strong>in</strong> R, Foster R, Digby-Bell J, Shields J,Whittles C, Muchens R, Gillatt D, Ziche M,Harper S, Bates D, 2004: VEGF 165 b, an <strong>in</strong>hibitoryvascular endothelial growth factor splice variant:mechanism of action, <strong>in</strong> vivo effect on angiogenesisand endogenous prote<strong>in</strong> expression. Cancer Res 64,7822–7835.Yao HH, Matzuk MM, Jorgez CJ, Menke DB, Page DC,Swa<strong>in</strong> A, Capel B, 2004: Follistat<strong>in</strong> operates downstream ofWnt4 <strong>in</strong> mammalian ovary organogenesis. Dev Dyn 230,210–215.Yao HH, Aardema J, Holthusen K, 2006: Sexually dimorphicregulation of <strong>in</strong>hib<strong>in</strong> beta B <strong>in</strong> establish<strong>in</strong>g gonadal vasculature<strong>in</strong> mice. Biol Reprod 74, 978–983.Author’s address (for correspondence): AS Cupp, A224i AnimalScience Department, 38th and Fair Street, University of Nebraska-L<strong>in</strong>coln, L<strong>in</strong>coln, NE 68583-0908, USA. E-mail: acupp2@unl.eduConflict of <strong>in</strong>terest: All authors declare no conflict of <strong>in</strong>terests.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 317–323 (2008); doi: 10.1111/j.1439-0531.2008.01180.xISSN 0936-6768Comparative Aspects of the Endotox<strong>in</strong>- and Cytok<strong>in</strong>e-Induced Endocr<strong>in</strong>e CascadeInfluenc<strong>in</strong>g Neuroendocr<strong>in</strong>e Control of Growth and <strong>Reproduction</strong> <strong>in</strong> Farm <strong>Animals</strong>BK Whitlock 1 , JA Daniel 2 , RR Wilborn 3 , TH Elsasser 4 , JA Carroll 5 and JL Sart<strong>in</strong> 11 Department of Anatomy, Physiology and Pharmacology, College of Veter<strong>in</strong>ary Medic<strong>in</strong>e, Auburn University, Auburn, AL, USA; 2 Department ofAnimal Science, Berry College, Mt. Berry, GA, USA; 3 Department of Cl<strong>in</strong>ical Sciences, College of Veter<strong>in</strong>ary Medic<strong>in</strong>e, Auburn University, Auburn,AL, USA; 4 Growth Biology Laboratory, USDA ⁄ ARS, Beltsville, MD, USA; 5 Livestock Issues Research Unit, USDA ⁄ ARS, Lubbock, TX, USAContentsDisease <strong>in</strong> animals is a well-known <strong>in</strong>hibitor of growth andreproduction. Earlier studies were <strong>in</strong>itiated to determ<strong>in</strong>e theeffects of endotox<strong>in</strong> on pituitary hormone secretion. Thesestudies found that <strong>in</strong> sheep, growth hormone (GH) concentrationwas elevated, whereas <strong>in</strong>sul<strong>in</strong>-like growth factor-I(IGF-I) was <strong>in</strong>hibited, as was lute<strong>in</strong>iz<strong>in</strong>g hormone (LH).Exam<strong>in</strong>ation of the site of action of endotox<strong>in</strong> <strong>in</strong> sheepdeterm<strong>in</strong>ed that somatotropes expressed the endotox<strong>in</strong> receptor(CD14) and that both endotox<strong>in</strong> and <strong>in</strong>terleuk<strong>in</strong>-Ibactivated GH secretion directly from the pituitary. In the faceof elevated GH, there is a reduction of IGF-I <strong>in</strong> all speciesexam<strong>in</strong>ed. As GH cannot activate IGF-I release dur<strong>in</strong>gdisease, there appears to be a downregulation of GH signall<strong>in</strong>gat the liver, perhaps related to altered nitration of Janus k<strong>in</strong>ase(JAK). In contrast to GH downregulation, LH release is<strong>in</strong>hibited at the level of the hypothalamus. New <strong>in</strong>sights havebeen ga<strong>in</strong>ed <strong>in</strong> determ<strong>in</strong><strong>in</strong>g the mechanisms by which diseaseperturbs growth and reproduction, particularly with regard tonitration of critical control pathways, with this perhaps serv<strong>in</strong>gas a novel mechanism central to lipopolysaccharide suppressionof all signall<strong>in</strong>g pathways. This pathway-based analysis iscritical to the develop<strong>in</strong>g novel strategies to reverse thedetrimental effect of disease on animal production.IntroductionGram-negative bacterial <strong>in</strong>fections such as Escherichiacoli (E. coli), Salmonella, Pseudomonas and Proteus tendto occur frequently <strong>in</strong> farm animals. Common portals ofentry for these Gram-negative (endotox<strong>in</strong>-conta<strong>in</strong><strong>in</strong>g)bacteria <strong>in</strong>clude, but are not limited to, the gastro<strong>in</strong>test<strong>in</strong>alsystem, the reproductive tract (especially uteri ofpostpartum animals), and the mammary gland. Infectionsof these systems or organs result <strong>in</strong> some of themost common and costly diseases of production ⁄ farmanimals such as enteritis, endometritis ⁄ metritis andmastitis. Regardless of the chronicity, <strong>in</strong>fections <strong>in</strong> farmanimals with bacteria conta<strong>in</strong><strong>in</strong>g endotox<strong>in</strong>, and thecytok<strong>in</strong>e and endocr<strong>in</strong>e changes that result, <strong>in</strong>evitablyimpact growth, metabolism and reproduction <strong>in</strong> anegative manner.Acute diarrhoea, often caused by Gram-negativebacteria such as enterotoxigenic E. coli and Salmonella,is a common disease <strong>in</strong> newborn calves and accounts formore than 50% of pre-wean<strong>in</strong>g deaths <strong>in</strong> <strong>in</strong>tensivelyraised calves (USDA 1996). However, pre-wean<strong>in</strong>gmortality of farm animals may be just the beg<strong>in</strong>n<strong>in</strong>g ofeconomic losses secondary to enteritis caused by Gramnegativebacteria. Other consequences of neonataldiarrhoea are as follows: (1) greater morbidity secondaryto <strong>in</strong>creased susceptibility to other pathogens, (2)impaired growth rates due to reduced food <strong>in</strong>take andmetabolic disturbances and (3) delayed puberty anddecreased reproductive productivity (production ofoffspr<strong>in</strong>g and milk) as a result of poor growth ratesand <strong>in</strong>adequate energy stores.The outcomes of bacterial <strong>in</strong>fection associated withthe postpartum uterus <strong>in</strong>clude puerperal metritis, cl<strong>in</strong>icalendometritis, pyometra and subcl<strong>in</strong>ical endometritis(Sheldon et al. 2006). Arcanobacterium pyogenes(A. pyogenes), E. coli and other Gram-negative bacteria,namely Fusobacterium necrophorum (F. necrophorum)and Bacteroides spp., are predom<strong>in</strong>ant <strong>in</strong> the uterus ofcl<strong>in</strong>ically diseased animals (Hirvonen et al. 1999). Thesecommon forms of reproductive tract diseases <strong>in</strong> farmanimals (especially dairy cows) may delay the completeregeneration of endometrium, disrupt the resumption ofcyclic ovarian function result<strong>in</strong>g <strong>in</strong> postponement of thefirst <strong>in</strong>sem<strong>in</strong>ation, <strong>in</strong>crease the numbers of <strong>in</strong>sem<strong>in</strong>ationsper conception and thus prolong the calv<strong>in</strong>g<strong>in</strong>terval and decrease the calv<strong>in</strong>g rate (Hussa<strong>in</strong> andDaniel 1991). It is clear that uter<strong>in</strong>e <strong>in</strong>fections andconsequential diseases have detrimental effects onreproductive performance of dairy cows. As mostcl<strong>in</strong>ical and reproductive consequences are attributedto the presence of A. pyogenes <strong>in</strong> comb<strong>in</strong>ation withorganisms like E. coli and other Gram-negative bacteria,a better understand<strong>in</strong>g <strong>in</strong> pathogenesis and the mechanisms<strong>in</strong>volved is of great practical and economicimportance.Mastitis is one of the major bacterial diseases <strong>in</strong>postpartum farm animals, especially dairy cows. In theearly weeks of lactation, Gram-negative bacteria may bethe predom<strong>in</strong>ant mastitis pathogen. Cl<strong>in</strong>ical cases ofGram-negative mastitis load the hosts with endotox<strong>in</strong>.In lactat<strong>in</strong>g cows, marked changes <strong>in</strong> plasma levels ofcerta<strong>in</strong> energy-related metabolites were reported simultaneouswith the endotox<strong>in</strong>-<strong>in</strong>duced endocr<strong>in</strong>e alterations.Concentrations of glucose tended to <strong>in</strong>crease<strong>in</strong>itially then subsequently decl<strong>in</strong>ed and there was atendency for <strong>in</strong>creased non-esterified fatty acid values,whereas plasma b-hydroxybutyrate (BHB) decreasedl<strong>in</strong>early <strong>in</strong> a dose-dependent manner after lipopolysaccharide(LPS) <strong>in</strong>fusion (Waldron et al. 2003).In addition to endotox<strong>in</strong> from Gram-negative bacteriacaus<strong>in</strong>g metabolic perturbations, abnormal metabolicstatus of farm animals can <strong>in</strong>fluence their responseto <strong>in</strong>fection. Epidemiological studies have demonstrated<strong>in</strong>terrelations among negative energy balance-relatedmetabolic disorders (hepatic lipidosis and ketosis), the<strong>in</strong>creased <strong>in</strong>cidence of cl<strong>in</strong>ical mastitis and theÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


318 BK Whitlock, JA Daniel, RR Wilborn, TH Elsasser, JA Carroll and JL Sart<strong>in</strong>subsequent decrease <strong>in</strong> reproductive performance <strong>in</strong>high-produc<strong>in</strong>g postpartum dairy cows (Valde et al.1997; Washburn et al. 2002). Some trials confirmed the<strong>in</strong>direct negative impact of cl<strong>in</strong>ical and subcl<strong>in</strong>icalmastitis on reproductive performance (Barker et al.1998), whereas others revealed direct mastitis-<strong>in</strong>ducedabnormalities <strong>in</strong> ovarian function (Moore et al. 1991;Hockett et al. 2000; Huszenicza et al. 2005). Understand<strong>in</strong>gthe mechanisms <strong>in</strong>volved <strong>in</strong> how endotox<strong>in</strong><strong>in</strong>teracts with reproduction, metabolism and endocr<strong>in</strong>ologymay lead to unique strategies to reverse thenegative effects of <strong>in</strong>fectious disease on farm animalsand humans.Understand<strong>in</strong>g Endotox<strong>in</strong> and Neuroendocr<strong>in</strong>eRegulationWhile the term ‘endotox<strong>in</strong>’ is often mistakenly appliedto any tox<strong>in</strong> derived from the microbes, the technicaldef<strong>in</strong>ition is specific for the group of LPS complexesextractable from or released from the outer membraneof Gram-negative pathogenic and non-pathogenic speciesof bacteria such as E. coli, Salmonella, Neisseria,Mannheimia, Pseudomonas and others. For the mostpart, LPS complexes are rather stabile and conf<strong>in</strong>edwith<strong>in</strong> the membrane of these bacteria under states ofwhat might be called ‘bacterial good health’. Forexample, upon <strong>in</strong>fection with a Gram-negative pathogen,several <strong>in</strong>teractions between the <strong>in</strong>vad<strong>in</strong>g bacteriaand cellular and biochemical factors <strong>in</strong> the immunesystem lead to the degeneration of the outer bacterialmembrane with the consequential release of LPS <strong>in</strong>tothe host’s <strong>in</strong>ternal environment. The most commoncauses for this release of LPS are bacterial autolysis,phagocytosis and digestion of bacteria by prowl<strong>in</strong>gactivated immune cells such as macrophages and neutrophils,and exogenous lysis facilitated by LPS activationof the complement cascade and lysosome activity.Of concern <strong>in</strong> human cl<strong>in</strong>ical medic<strong>in</strong>e is the recentconfirmation that massive septic crisis can be furthercomplicated acutely with the adm<strong>in</strong>istration of certa<strong>in</strong>antibiotics because of the mode of action of these drugsto facilitate cell wall breakdown with the result<strong>in</strong>grelease of LPS. In this situation, untimely adm<strong>in</strong>istrationof drugs with modes of action like that of thepenicill<strong>in</strong>s might precipitate an acute crisis through the<strong>in</strong>itiation of multiple organ failure. Though not acommon occurrence, the scenario should be recognizedas a potential confound<strong>in</strong>g factor <strong>in</strong> the treatment ofsepsis.Endotox<strong>in</strong> challenges metabolism through direct and<strong>in</strong>direct mechanisms. The direct effects of LPS, <strong>in</strong> theabsence of cytok<strong>in</strong>e production, on the release ofpituitary hormones critical to the ma<strong>in</strong>tenance ofmetabolism as well as reproduction were demonstratedby Coleman et al. (1993). In this regard, some nonimmunecells such as hepatocytes (Liu et al. 1998),adipocytes (Daniel et al. 2003) and pituitary cells(Daniel et al. 2005) differentially express the LPSb<strong>in</strong>d<strong>in</strong>g receptor CD14 on the plasma membrane,provid<strong>in</strong>g a mechanistic explanation as to how cellsmight respond directly to LPS. Moreover, we haveprelim<strong>in</strong>ary immunohistochemical evidence that theToll-like Receptor 4 is also expressed on somatotropes(Elsasser and Sart<strong>in</strong>, unpublished). While hepatocyteCD14 was shown to upregulate follow<strong>in</strong>g LPS challenge(Liu et al. 1998), somatotrope expression decreased andconstitutive presence on gonadotropes, lactotropes andcorticotropes was unchanged (Daniel et al. 2005). Thisrecent discovery of the downregulation of CD14 onsomatotropes after LPS release is consistent with anactivation of the endotox<strong>in</strong> receptor on the pituitary. Inregard to the so-called <strong>in</strong>direct mechanisms of actions,the classical pattern of pro<strong>in</strong>flammatory cytok<strong>in</strong>esreleased from a challenged immune system functions <strong>in</strong>both an endocr<strong>in</strong>e and paracr<strong>in</strong>e manner to <strong>in</strong>teractwith specific receptors for these cytok<strong>in</strong>es. This affectsnot only the metabolic character of target cells (liver,muscle and adipose), but also metabolic regulatoryorgans, the pituitary and pancreas <strong>in</strong> particular.As we have reviewed earlier (Elsasser et al. 2000;Daniel et al. 2002; Elsasser and Kahl 2002; Carroll2008), LPS elicits a well-timed elaboration of pro<strong>in</strong>flammatorycytok<strong>in</strong>es, prostagland<strong>in</strong> derivatives, catecholam<strong>in</strong>esand free radicals from neutrophils,monocytes and macrophages which, depend<strong>in</strong>g on theseverity of the response, largely halt anabolic processesand <strong>in</strong>itiate catabolic breakdown of tissue reserves.Most <strong>in</strong>fluential on metabolic processes and prototypicalof the anti-anabolic character of the endotox<strong>in</strong>pro<strong>in</strong>flammatory response is the release of tumournecrosis factor-a (TNF-a). This relatively short-termeffector has a unique <strong>in</strong>fluence on metabolism, but canalso be considered as an <strong>in</strong>itiator of further <strong>in</strong>flammatoryresponse. Data <strong>in</strong>dicate that the TNF-a response to<strong>in</strong>fused or bolus-adm<strong>in</strong>istered LPS is largely turned offafter approximately 4–6 h. Regardless of the adm<strong>in</strong>istrationmodel, the TNF-a event <strong>in</strong>itiates progression ofadditional pro<strong>in</strong>flammatory cytok<strong>in</strong>es such as IL-6, andalso elaborates anti-<strong>in</strong>flammatory cytok<strong>in</strong>es such as IL-4, c-<strong>in</strong>terferon and IL-10 (Fig. 1). Significant <strong>in</strong> thisprocess are the homeostatic survival mechanisms associatedwith the development of what is characterized asearly and late endotox<strong>in</strong> tolerance (West and Heagy2002; Elsasser et al. 2004). The response is dependent onPlasma cytok<strong>in</strong>e (pg/ml)1e+51e+41e+31e+21e+11e+01e–1IL-6IL-10 1 2 3 4 6Time (h)Fig. 1. Effect of LPS on plasma TNF-a, IL-6, IL-1 and INF-c <strong>in</strong>calves. Calves were treated with endotox<strong>in</strong> (arrow; 0.6 lg ⁄ kg BW),n = 5. Data are redrawn from unpublished data by the authors forillustrative purposesÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Endotox<strong>in</strong> and Neuroendocr<strong>in</strong>ology 319the timely generation of nitric oxide (NO) as driven byTNF-a and accompanied by translocation of NFjb tothe nucleus which <strong>in</strong> turn attenuates transcription of theTNF-a gene. The result is a necessary turndown orsuppression of pro<strong>in</strong>flammatory signall<strong>in</strong>g elementsthat, if left unchecked, leads to free radical tissuedamage (Zeisberger and Roth 1998).Sites of Endotox<strong>in</strong> Action on Growth Hormone(GH) and Lute<strong>in</strong>iz<strong>in</strong>g Hormone (LH)Both GH and LH are produced and secreted by thepituitary under the <strong>in</strong>fluence of releas<strong>in</strong>g hormones fromthe hypothalamus. Thus, LPS may <strong>in</strong>fluence circulat<strong>in</strong>gconcentrations of GH and LH directly by alter<strong>in</strong>gproduction and secretion at the pituitary, or <strong>in</strong>directlyby alter<strong>in</strong>g production and secretion of releas<strong>in</strong>ghormones at the level of the hypothalamus.Endotox<strong>in</strong> was reported to impair adenohypophysialLH release <strong>in</strong> rats (Rettori et al. 1994) and sheep(Coleman et al. 1993; Fig. 2). In cycl<strong>in</strong>g heifers receiv<strong>in</strong>gan experimental challenge 42 h after the PGF 2a (d<strong>in</strong>oprost)-<strong>in</strong>ducedluteolysis (Suzuki et al. 2001), LPSreduced the pulse frequency of LH for 6 h, and<strong>in</strong>creased the mean concentration and pulse amplitudeof LH. Plasma concentrations of cortisol andLH (ng/ml)GH (ng/ml)87654321–1 0 1 2 3 4 5 6 7 8 9 10Hours after treatment18161412108642ControlEndotox<strong>in</strong>Endotox<strong>in</strong> (0.4 µg/kg BW)Control0–1 0 1 2 3 4 5 6 7 8 9Hours after treatmentFig. 2. Effects of LPS on pulsatile patterns of LH and GH <strong>in</strong> sheeptreated with LPS (Coleman et al. 1993)10progesterone were simultaneously and transiently<strong>in</strong>creased due to the adrenocortical over-production ofthese hormones. Plasma oestradiol concentrations weredecreased and the preovulatory LH pulse was delayed orcompletely blocked. A similar disruption was demonstrated<strong>in</strong> the preovulatory rise of oestradiol and <strong>in</strong> thesecretory pattern of LH <strong>in</strong> ewes follow<strong>in</strong>g endotox<strong>in</strong>challenge (Battaglia et al. 2000). Endotox<strong>in</strong> absorbedfrom the uter<strong>in</strong>e lumen was reported to suppress theformation of the preovulatory LH peak and to <strong>in</strong>ducethe cystic degeneration of dom<strong>in</strong>ant follicles <strong>in</strong> postpartumcows (Peter et al. 1989). Also of particularimportance <strong>in</strong> farm animals is the f<strong>in</strong>d<strong>in</strong>g that neonatalexposure to LPS actually programs long-term sensitivityof the GnRH regulatory system, such that post-natalresponses to LPS produce a greater <strong>in</strong>hibition of GnRHand LH (Li et al. 2007).Endotox<strong>in</strong> suppresses circulat<strong>in</strong>g concentrations ofLH at the level of the hypothalamus (Coleman et al.1993). This is supported as follows by three primarypieces of evidence. Firstly, portal ve<strong>in</strong> cannulation<strong>in</strong>dicates that LPS results <strong>in</strong> reduced secretion of GnRHfrom the hypothalamus, thus suggest<strong>in</strong>g endotox<strong>in</strong><strong>in</strong>hibits circulat<strong>in</strong>g concentrations of LH by <strong>in</strong>hibit<strong>in</strong>ghypothalamic stimulation of LH secretion (Battagliaet al. 1997). Secondly, the <strong>in</strong>creased secretion of LH <strong>in</strong>response to LPS by dispersed pituitary cells (Colemanet al. 1993) further suggests that LPS acts at the level ofthe hypothalamus to <strong>in</strong>hibit circulat<strong>in</strong>g concentrationsof LH. F<strong>in</strong>ally, LPS challenge reduces electrical activity<strong>in</strong> areas of the hypothalamus associated with thegeneration of GnRH and thus LH pulses (Takeuchiet al. 1997; Yoo et al. 1997).In contrast to the effects on LH, endotox<strong>in</strong>’s effect oncirculat<strong>in</strong>g concentration of GH is more complicated. Inspecies where LPS decreases, the circulat<strong>in</strong>g concentrationof GH (cattle and rat), the effect is primarily at thelevel of the hypothalamus. However, <strong>in</strong> species whereLPS <strong>in</strong>creases circulat<strong>in</strong>g concentrations of GH (sheepand human), the effect appears to occur primarily at thepituitary. In pigs, an acute <strong>in</strong>crease <strong>in</strong> circulat<strong>in</strong>gconcentrations of GH follow<strong>in</strong>g an LPS challenge hasbeen reported (Parrott et al. 1995; Hevener et al. 1997).However, this effect was only short-lived, and subsequentLPS-<strong>in</strong>duced uncoupl<strong>in</strong>g of the GH ⁄ IGF-I axispersisted. Pituitary production and secretion of GH isprimarily under the regulation of GH releas<strong>in</strong>g hormone(GHRH) and somatostat<strong>in</strong> from the hypothalamus. Inspecies where GH concentration is decreased by LPS,the effect is likely mediated through cytok<strong>in</strong>e-<strong>in</strong>ducedstimulation of somatostat<strong>in</strong> production (Scarborough1990). Sheep <strong>in</strong>jected with LPS secrete GH dur<strong>in</strong>g thesame period when LH secretion is reduced (Fig. 2;Coleman et al. 1993). Challenge with LPS also results <strong>in</strong>an <strong>in</strong>crease <strong>in</strong> somatostat<strong>in</strong> concentrations <strong>in</strong> hypophysialportal blood with no change <strong>in</strong> the concentrations ofGHRH <strong>in</strong> sheep (Briard et al. 1998). Under normalphysiological conditions, an <strong>in</strong>crease <strong>in</strong> somatostat<strong>in</strong>accompanied with no change <strong>in</strong> GHRH would beexpected to result <strong>in</strong> decreased circulat<strong>in</strong>g concentrationsof GH. However, Briard et al. (1998) observed<strong>in</strong>creased circulat<strong>in</strong>g concentrations of GH associatedwith <strong>in</strong>creased hypophysial portal blood concentrationsÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


320 BK Whitlock, JA Daniel, RR Wilborn, TH Elsasser, JA Carroll and JL Sart<strong>in</strong>of somatostat<strong>in</strong> <strong>in</strong> sheep challenged with LPS (Briardet al. 1998). Thus, the effect of LPS to <strong>in</strong>crease circulat<strong>in</strong>gconcentrations of GH was not expla<strong>in</strong>ed byaltered hypothalamic function, and the effects of LPS to<strong>in</strong>crease circulat<strong>in</strong>g concentrations of GH appear to bemediated at the pituitary. Further support<strong>in</strong>g a pituitarysite of action, LPS challenge also results <strong>in</strong> <strong>in</strong>creasedsecretion of GH from dispersed pituitary cells (Colemanet al. 1993).Possible Mechanisms by Which Endotox<strong>in</strong>Alters LH and GHMultiple changes occur <strong>in</strong> response to endotox<strong>in</strong> challengewhich could potentially affect the decrease <strong>in</strong>circulat<strong>in</strong>g concentrations of LH. One key response to adisease challenge and stress <strong>in</strong> general is activation ofthe hypothalamic–pituitary–adrenal axis with an associated<strong>in</strong>crease <strong>in</strong> glucocorticoids, particularly cortisol.Indeed, adm<strong>in</strong>istration of cortisol will reduce circulat<strong>in</strong>gconcentrations of LH (Debus et al. 2002) and GH(Thompson et al. 1995). However, <strong>in</strong>hibition of cortisolsynthesis dur<strong>in</strong>g the LPS challenge does not preventLPS suppression of pulsatile GnRH and LH secretion(Debus et al. 2002). Thus, while cortisol may play a role<strong>in</strong> LPS-<strong>in</strong>duced suppression of circulat<strong>in</strong>g concentrationsof LH, other factors are also likely <strong>in</strong>volved.The endogenous opioid system may also be <strong>in</strong>volved<strong>in</strong> LPS suppression of circulat<strong>in</strong>g concentrations of LH.Central adm<strong>in</strong>istration of the opiate antagonist naloxoneblocked LPS suppression of circulat<strong>in</strong>g concentrationsof LH <strong>in</strong> monkeys (Xiao et al. 2000). In addition,adm<strong>in</strong>istration of naloxone to heifers follow<strong>in</strong>g LPStreatment resulted <strong>in</strong> <strong>in</strong>creased circulat<strong>in</strong>g concentrationsof LH (Kujjo et al. 1995). However, naloxone didnot prevent the decrease <strong>in</strong> circulat<strong>in</strong>g concentrations ofLH observed follow<strong>in</strong>g challenge with E. coli <strong>in</strong> sheep(Lesh<strong>in</strong> and Malven 1984). The <strong>in</strong>ability of an opiateantagonist to block E. coli-<strong>in</strong>duced LH suppression maybe associated with the differential pro<strong>in</strong>flammatorycytok<strong>in</strong>e profiles observed <strong>in</strong> LPS vs E. coli challengestudies. For <strong>in</strong>stance, <strong>in</strong> pigs, LPS is commonly knownto <strong>in</strong>duce the primary pro<strong>in</strong>flammatory cytok<strong>in</strong>es TNFa,IL-Ib and IL-6 (Carroll et al. 2003). However, <strong>in</strong> pigschallenged with live E. coli, TNF-a concentrations arenot elevated (Strauch et al. 2004) <strong>in</strong>dicat<strong>in</strong>g that theactivation of the acute phase immune response isdepend<strong>in</strong>g upon the immunological challenge. Thus,while the endogenous opioid system may play a role <strong>in</strong>LPS suppression of circulat<strong>in</strong>g concentrations of LH,other systems are <strong>in</strong>volved <strong>in</strong> the effect of Gramnegativebacterial challenge to suppress LH.As discussed earlier, challenge with LPS and Gramnegativebacteria results <strong>in</strong> activation of the immunesystem. Components of the <strong>in</strong>nate immune system maybe responsible for the suppression of circulat<strong>in</strong>g concentrationsof LH. Indeed, <strong>in</strong>vestigators have exam<strong>in</strong>edthe role of the cytok<strong>in</strong>es TNF-a and IL-1b as well asprostagland<strong>in</strong>s <strong>in</strong> LPS suppression of LH. Centraladm<strong>in</strong>istration of both TNF-a and IL-1b suppressedcirculat<strong>in</strong>g concentrations of LH (Daniel et al. 2005).Central adm<strong>in</strong>istration of IL-1b to monkeys also suppressedcirculat<strong>in</strong>g concentrations of LH (Xiao et al.2000). However, neither central nor peripheral adm<strong>in</strong>istrationof the cytok<strong>in</strong>e antagonists TNF-R1 norIL-1RA prevented the LPS-<strong>in</strong>duced suppression ofcirculat<strong>in</strong>g concentrations of LH (Xiao et al. 2000;Daniel et al. 2005). Thus, while <strong>in</strong>creased concentrationsof <strong>in</strong>flammatory cytok<strong>in</strong>es may play a role <strong>in</strong> LPSsuppression of circulat<strong>in</strong>g concentrations of LH, themechanism for LH <strong>in</strong>hibition is clearly multifactorial.In response to LPS challenge, prostagland<strong>in</strong> productionis also enhanced. Treatment with a prostagland<strong>in</strong>synthesis <strong>in</strong>hibitor (flurbiprofen) prevented endotox<strong>in</strong>suppression of LH and GnRH (Harris et al. 2000).Thus, prostagland<strong>in</strong> formation is a crucial step <strong>in</strong> LPSsuppression of pulsatile secretion of GnRH and LH.In contrast, the oestradiol-<strong>in</strong>duced surge of LH wasblocked by LPS, but the <strong>in</strong>hibition was found to functionvia prostagland<strong>in</strong> <strong>in</strong>dependent pathways (Breen et al.2004).The mechanisms by which LPS alters circulat<strong>in</strong>gconcentrations of GH are clearer than the means bywhich LPS suppresses LH. Early data suggested that <strong>in</strong>some species (rat), LPS adm<strong>in</strong>istration results <strong>in</strong> suppressedcirculat<strong>in</strong>g concentrations of GH. The reducedGH is due to <strong>in</strong>creased somatostat<strong>in</strong> release <strong>in</strong> responseto <strong>in</strong>flammatory cytok<strong>in</strong>es, specifically IL-1, TNF andIL-6 (Scarborough 1990). In contrast, other species<strong>in</strong>crease circulat<strong>in</strong>g concentrations of GH <strong>in</strong> response to<strong>in</strong>flammatory cytok<strong>in</strong>es and to LPS adm<strong>in</strong>istration. Inthis case, the site of action is the pituitary as opposed tothe hypothalamus. More recently, this disparate effect ofLPS between different species was evaluated by Priegoet al. (2003). Rats adm<strong>in</strong>istered low doses of LPS had<strong>in</strong>creased plasma GH, whereas high doses were <strong>in</strong>hibitoryto GH, <strong>in</strong>dicat<strong>in</strong>g that differences observed weredue to dose of LPS and not species differences. Thus, thetypical model <strong>in</strong> rats is a model of endotoxic shock,whereas the model <strong>in</strong> sheep and humans tends to modela less severe disease.Studies with dispersed ov<strong>in</strong>e pituitary cells have foundthat treatment with IL-1 stimulated GH synthesis andsecretion while TNF-a reduced GRH-stimulated GHrelease (Fry et al. 1998). Moreover, TNF-a will <strong>in</strong>hibitGH release from cultured bov<strong>in</strong>e pituitary cells (Elsasseret al. 1991). In addition, peripheral adm<strong>in</strong>istration ofTNF-a and IL-1b resulted <strong>in</strong> <strong>in</strong>creased circulat<strong>in</strong>gconcentrations of GH (Daniel et al. 2005). Intravenous(IV) but not <strong>in</strong>tracerebroventricular adm<strong>in</strong>istration ofthe cytok<strong>in</strong>e antagonists, sTNF-R1 or IL-1RA, preventedthe LPS-<strong>in</strong>duced <strong>in</strong>crease <strong>in</strong> circulat<strong>in</strong>g concentrationsof GH (Daniel et al. 2005). The differ<strong>in</strong>g effectsof TNF-a between <strong>in</strong> vivo and <strong>in</strong> vitro models suggestthat <strong>in</strong> vivo, TNF-a activates IL-1b release which <strong>in</strong> turnis a stimulus to GH. This <strong>in</strong>crease <strong>in</strong> <strong>in</strong>flammatorycytok<strong>in</strong>es <strong>in</strong> response to LPS plays a critical role <strong>in</strong>endotox<strong>in</strong>-<strong>in</strong>duced alterations <strong>in</strong> circulat<strong>in</strong>g concentrationsof GH. F<strong>in</strong>ally, Daniel et al. (2005) discoveredthat CD14 is expressed on somatotropes and is downregulated<strong>in</strong> response to LPS <strong>in</strong>jection, whereas thetypical dogma suggests that the major effects on therodent pituitary are via folliculostellate cells. In agreementwith previous research (Coleman et al. 1993), thesedata also provide evidence that GH can be released fromthe somatotrope <strong>in</strong> response to direct exposure to LPS.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Endotox<strong>in</strong> and Neuroendocr<strong>in</strong>ology 321Another mediator which may be <strong>in</strong>volved <strong>in</strong> the GHand LH response to LPS is ghrel<strong>in</strong>. Ghrel<strong>in</strong> is anendogenous ligand for the growth hormone secretagoguereceptor which stimulates GH secretion (Kojimaet al. 1999). In pigs, ghrel<strong>in</strong> has been reported to<strong>in</strong>crease body weight ga<strong>in</strong> and circulat<strong>in</strong>g concentrationsof GH and <strong>in</strong>sul<strong>in</strong> (Salfen et al. 2004). Recent data<strong>in</strong>dicate that LPS challenge results <strong>in</strong> <strong>in</strong>creased circulat<strong>in</strong>gconcentrations of ghrel<strong>in</strong> <strong>in</strong> humans, a specieswith <strong>in</strong>creased GH <strong>in</strong> response to LPS (Vila et al. 2007).Perhaps an <strong>in</strong>crease <strong>in</strong> ghrel<strong>in</strong> stimulates the <strong>in</strong>crease <strong>in</strong>GH observed follow<strong>in</strong>g an LPS challenge. In addition,ghrel<strong>in</strong> has been demonstrated to reduce circulat<strong>in</strong>gconcentrations of LH <strong>in</strong> rats (Furuta et al. 2001),monkeys (Vulliemoz et al. 2004), sheep (Iqbal et al.2006) and humans (Kluge et al. 2007). The possiblerelationship of ghrel<strong>in</strong> to the LPS <strong>in</strong>hibition of LH andstimulation of GH should be a focus of future research<strong>in</strong>volv<strong>in</strong>g the mechanisms of LPS action <strong>in</strong> rum<strong>in</strong>ants.Endotox<strong>in</strong> Disrupts GH Signall<strong>in</strong>gInitial studies demonstrated that the pro<strong>in</strong>flammatorymediators of LPS action with<strong>in</strong> the immune systemmediated a reduction <strong>in</strong> circulat<strong>in</strong>g plasma IGF-1concentrations and this reduction was <strong>in</strong>dependent ofthe decrease <strong>in</strong> voluntary food <strong>in</strong>take observed <strong>in</strong> theLPS-treated calves (Elsasser et al. 1995). The chronicdecl<strong>in</strong>e of this key anabolic hormone dur<strong>in</strong>g catabolicdisease suggested that calves suffer<strong>in</strong>g weight loss from<strong>in</strong>fectious diseases might benefit from the use ofanabolic hormones to decrease the catabolic milieu. Assuch, studies were conducted to evaluate the actions ofanabolic agents such as oestradiol and GH (Elsasseret al. 1998; Sart<strong>in</strong> et al. 1998). Elsasser et al. (1998)exam<strong>in</strong>ed the possibility that treatment with exogenousGH could be used to overcome the effects of sarcocystis<strong>in</strong>fection <strong>in</strong> cattle by <strong>in</strong>creas<strong>in</strong>g plasma IGF-I concentrationsand normaliz<strong>in</strong>g metabolism. However, GHhad no ability to normalize plasma concentrations ofIGF-I or other <strong>in</strong>dices of metabolism. S<strong>in</strong>ce no problemswere found with liver GH receptor functions, thislack of effect of GH was hypothesized to relate toaltered GH signall<strong>in</strong>g. In a series of follow-up studiesus<strong>in</strong>g the LPS model, Elsasser et al. (2004, 2007a,b)determ<strong>in</strong>ed that the activation of the major signaltransduction regulator of GH action, prote<strong>in</strong> tyros<strong>in</strong>ek<strong>in</strong>ase JAK-2, by GH was reduced by <strong>in</strong>fection, thusexpla<strong>in</strong><strong>in</strong>g the GH resistance observed dur<strong>in</strong>g disease.Moreover, the specific locus of the resistance was relatedto a reduced capacity for JAK-2 to be activated at itsk<strong>in</strong>ase epitope by phosphorylation (Fig. 3). Moredetailed studies have determ<strong>in</strong>ed that a major targetwhere JAK-2 is nitrated is the position normallyphosphorylated (… 1007 tyros<strong>in</strong>e- 1008 tyros<strong>in</strong>e…) <strong>in</strong> theGH activation of the signal transduction cascade, thusprovid<strong>in</strong>g a novel mechanism to expla<strong>in</strong> some verylocalized aspects of GH resistance <strong>in</strong> disease (Elsasseret al. 2007a,b). In this <strong>in</strong>stance, <strong>in</strong>creased nitration ofJAK-2 is associated with decreased phosphorylation,dimerization and translocation to the nucleus of the keynuclear gene transcription factor for GH activation ofIGF-1 production, STAT5b. The <strong>in</strong>terest<strong>in</strong>g feature ofFig. 3. Representative Western blot of liver homogenate prote<strong>in</strong>s fromcontrol (before LPS, lane 1) and after LPS (24 h, 2.5 lg ⁄ kg E. coli055:B5 LPS, lanes 2 and 3) biopsy samples. (Elsasser et al. 2007a)this nitration response is that while it appears to be atransient phenomenon <strong>in</strong> states of normal health, theformation of nitrated JAK-2 is an overt and prolongedoccurrence <strong>in</strong> chronic disease states. The demonstratedability to specifically modulate this nitration response byalter<strong>in</strong>g the generation of nitric oxide and superoxidemay prove useful to further pharmacological strategiesthat might be used to re-establish stabile metabolismand mitigate the impacts of disease stress on animalhealth.ConclusionMany diseases common to farm and production animalsare caused by <strong>in</strong>fections with Gram-negative bacteria.The consequences of these <strong>in</strong>fections are a result of theliberation of endotox<strong>in</strong> ⁄ LPS from the bacteria and thereaction of the immune system ⁄ <strong>in</strong>flammatory cells.Cytok<strong>in</strong>e (e.g. IL-I and TNF-a) production by <strong>in</strong>flammatorycells <strong>in</strong> response to LPS is a normal andnecessary function of the immune system <strong>in</strong> animals toprevent and alleviate <strong>in</strong>fections. However, the <strong>in</strong>flammatorycytok<strong>in</strong>es can also <strong>in</strong>itiate a cascade of eventsthat impair hormonal and metabolic homeostatic processesregulat<strong>in</strong>g growth, metabolism and reproduction.The purpose for impair<strong>in</strong>g these functions is most likelya consequence of the lofty nutrient ⁄ metabolic demandsfor the immune system’s response to <strong>in</strong>fections. In anattempt to redirect or conserve nutrients for immunefunctions, growth is impaired by direct or <strong>in</strong>directactions of cytok<strong>in</strong>es on the somatotropic axis. In adultanimals, nutrients may be conserved by <strong>in</strong>hibit<strong>in</strong>genergetically risky behaviour [i.e. reproduction (oestrus,pregnancy and lactation)] through manipulation of thehypothalamic–pituitary–gonadal axis at any po<strong>in</strong>t.More research is needed to completely understand themechanisms beh<strong>in</strong>d the effects of disease stress ongrowth, metabolism and reproduction. While previousdata on severe pro<strong>in</strong>flammatory-mediated dysfunctionwere associated with stark pathology, the nitrationconcept follows closely with perturbations associatedwith low level responses to immune challenge that donot culm<strong>in</strong>ate <strong>in</strong> death. For example, the implications ofthe recent data by Elsasser et al. (2007a,b) suggest anovel mechanism for <strong>in</strong>hibition of endocr<strong>in</strong>e signall<strong>in</strong>gwhich should be exam<strong>in</strong>ed as a possible unify<strong>in</strong>gmechanism by which LPS actions suppress key functions<strong>in</strong> cells. By understand<strong>in</strong>g the <strong>in</strong>tricacies of LPS-<strong>in</strong>ducedcytok<strong>in</strong>e release and how cytok<strong>in</strong>es affect farm animalproduction, we can target more precisely the strategiesneeded to combat <strong>in</strong>fectious agents as well as stabilizeÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


322 BK Whitlock, JA Daniel, RR Wilborn, TH Elsasser, JA Carroll and JL Sart<strong>in</strong>host responses to <strong>in</strong>fection <strong>in</strong> order to speed recoveryand improve animal welfare.ReferencesBarker AR, Schrick FN, Lewis MJ, Dowlen HH, Oliver SP,1998: Influence of cl<strong>in</strong>ical mastitis dur<strong>in</strong>g early lactation onreproductive performance of Jersey cows. J Dairy Sci 81,1285–1290.Battaglia DF, Bowen JM, Krasa HB, Thrun LA, Viguie C,Karsch FJ, 1997: Endotox<strong>in</strong> <strong>in</strong>hibits the reproductiveneuroendocr<strong>in</strong>e axis while stimulat<strong>in</strong>g adrenal steroids: asimultaneous view from hypophyseal portal and peripheralblood. Endocr<strong>in</strong>ology 138, 4273–4281.Battaglia DF, Krasa HB, Padmanabhan V, Viguie C, KarschFJ, 2000: Endocr<strong>in</strong>e alterations that underlie endotox<strong>in</strong><strong>in</strong>duceddisruption of the follicular phase <strong>in</strong> ewes. BiolReprod 62, 45–53.Breen KM, Bill<strong>in</strong>gs HJ, Debus N, Karsch FJ, 2004: Endotox<strong>in</strong><strong>in</strong>hibits the surge secretion of gonadotrop<strong>in</strong>-releas<strong>in</strong>g hormonevia a prostagland<strong>in</strong>-<strong>in</strong>dependent pathway. Endocr<strong>in</strong>ology145, 221–227.Briard N, Guillaume V, Frachebois C, Rico-Gomez M, SauzeN, Oliver C, Dutour A, 1998: Endotox<strong>in</strong> <strong>in</strong>jection <strong>in</strong>creasesgrowth hormone and somatostat<strong>in</strong> secretion <strong>in</strong> sheep.Endocr<strong>in</strong>ology 139, 2662–2669.Carroll JA 2008: Bi-directional communication: growth andimmunity <strong>in</strong> domestic livestock. J Anim Sci 86, (suppl. 14)E126–E137.Carroll JA, Ga<strong>in</strong>es AM, Spencer JD, Allee GL, Kattesh HG,Roberts MP, Zannelli ME, 2003: Effect of menhaden fish oilsupplementation and lipopolysaccharide exposure on nurserypigs. I. Effects on the immune axis when fed dietsconta<strong>in</strong><strong>in</strong>g spray-dried plasma. Domest Anim Endocr<strong>in</strong>ol24, 341–351.Coleman ES, Elsasser TH, Kemppa<strong>in</strong>en RJ, Coleman DA,Sart<strong>in</strong> JL, 1993: Effect of endotox<strong>in</strong> on pituitary hormonesecretion <strong>in</strong> sheep. Neuroendocr<strong>in</strong>ology 58, 111–122.Daniel JA, Whitlock BK, Wagner CG, Sart<strong>in</strong> JL, 2002:Regulation of the growth hormone and lute<strong>in</strong>iz<strong>in</strong>g hormoneresponse to endotox<strong>in</strong> <strong>in</strong> sheep. Domest Anim Endocr<strong>in</strong>ol23, 361–370.Daniel JA, Elsasser TH, Morrison CD, Keisler DH, WhitlockBK, Steele B, Pugh D, Sart<strong>in</strong> JL, 2003: Lept<strong>in</strong>, tumornecrosis factor-alpha (TNF), and CD14 <strong>in</strong> ov<strong>in</strong>e adiposetissue and changes <strong>in</strong> circulat<strong>in</strong>g TNF <strong>in</strong> lean and fat sheep.J Anim Sci 81, 2590–2599.Daniel JA, Elsasser TH, Mart<strong>in</strong>ez A, Steele B, Whitlock BK,Sart<strong>in</strong> JL, 2005: Interleuk<strong>in</strong>-1beta and tumor necrosisfactor-alpha mediation of endotox<strong>in</strong> action on growthhormone. Am J Physiol Endocr<strong>in</strong>ol Metab 289, E650–E657.Debus N, Breen KM, Barrell GK, Bill<strong>in</strong>gs HJ, Brown M,Young EA, Karsch FJ, 2002: Does cortisol mediate endotox<strong>in</strong>-<strong>in</strong>duced<strong>in</strong>hibition of pulsatile lute<strong>in</strong>iz<strong>in</strong>g hormoneand gonadotrop<strong>in</strong>-releas<strong>in</strong>g hormone secretion? Endocr<strong>in</strong>ology143, 3748–3758.Elsasser TH, Kahl S, 2002: Adrenomedull<strong>in</strong> has multiple roles<strong>in</strong> disease stress: development and remission of the <strong>in</strong>flammatoryresponse. Microsc Res Tech 57, 120–129.Elsasser TH, Caperna TJ, Fayer R, 1991: Tumor necrosisfactor-alpha affects growth hormone secretion by a directpituitary <strong>in</strong>teraction. Proc Soc Exp Biol Med 198, 547–554.Elsasser TH, Caperna TJ, Rumsey TS, 1995: Endotox<strong>in</strong>adm<strong>in</strong>istration decreases plasma <strong>in</strong>sul<strong>in</strong>-like growth factor(IGF)-I and IGF-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>-2 <strong>in</strong> Angus x Herefordsteers <strong>in</strong>dependent of changes <strong>in</strong> nutritional <strong>in</strong>take. JEndocr<strong>in</strong>ol 144, 109–117.Elsasser TH, Sart<strong>in</strong> JL, McMahon C, Romo G, Fayer R, KahlS, Blagburn B, 1998: Changes <strong>in</strong> somatotropic axis responseand body composition dur<strong>in</strong>g growth hormone adm<strong>in</strong>istration<strong>in</strong> progressive cachectic parasitism. Domest AnimEndocr<strong>in</strong>ol 15, 239–255.Elsasser TH, Kahl S, Rumsey TS, Blum JW, 2000: Modulationof growth performance <strong>in</strong> disease: reactive nitrogen compoundsand their impact on cell prote<strong>in</strong>s. Domest AnimEndocr<strong>in</strong>ol 19, 75–84.Elsasser TH, Kahl S, MacLeod C, Nicholson B, Sart<strong>in</strong> JL, LiC, 2004: Mechanisms underly<strong>in</strong>g growth hormone effects <strong>in</strong>augment<strong>in</strong>g nitric oxide production and prote<strong>in</strong> tyros<strong>in</strong>enitration dur<strong>in</strong>g endotox<strong>in</strong> challenge. Endocr<strong>in</strong>ology 145,3413–3423.Elsasser TH, Kahl S, Li C-J, Sart<strong>in</strong> JL, Garrett WM, RodrigoJ, 2007a: Caveolae nitration of JAK-2 at the 1007Y-1008Ysite: coord<strong>in</strong>at<strong>in</strong>g <strong>in</strong>flammatory response and metabolichormone readjustment with<strong>in</strong> the somatotropic axis. Endocr<strong>in</strong>ology148, 3792–3802.Elsasser TH, Li CJ, Caperna TJ, Kahl S, Schmidt WF, 2007b:Growth hormone (GH)-associated nitration of Janusk<strong>in</strong>ase-2 at the 1007Y-1008Y epitope impedes phosphorylationat this site: mechanism for and impact of a GH, AKT,and nitric oxide synthase axis on GH signal transduction.Endocr<strong>in</strong>ology 148, 3792–3802.Fry C, Gunter DR, McMahon CD, Steele B, Sart<strong>in</strong> JL, 1998:Cytok<strong>in</strong>e-mediated growth hormone release from culturedov<strong>in</strong>e pituitary cells. Neuroendocr<strong>in</strong>ology 68, 192–200.Furuta M, Funabashi T, Kimura F, 2001: Intracerebroventricularadm<strong>in</strong>istration of ghrel<strong>in</strong> rapidly suppresses pulsatilelute<strong>in</strong>iz<strong>in</strong>g hormone secretion <strong>in</strong> ovariectomized rats.Biochem Biophys Res Commun 288, 780–785.Harris TG, Battaglia DF, Brown ME, Brown MB, CarlsonNE, Viguie C, Williams CY, Karsch FJ, 2000: Prostagland<strong>in</strong>smediate the endotox<strong>in</strong>-<strong>in</strong>duced suppression ofpulsatile gonadotrop<strong>in</strong>-releas<strong>in</strong>g hormone and lute<strong>in</strong>iz<strong>in</strong>ghormone secretion <strong>in</strong> the ewe. Endocr<strong>in</strong>ology 141, 1050–1058.Hevener W, Almond GW, Armstrong JD, Richards RG, 1997:Effects of acute endotoxemia on serum somatotrop<strong>in</strong> and<strong>in</strong>sul<strong>in</strong>-like growth factor I concentrations <strong>in</strong> prepubertalgilts. Am J Vet Res 58, 1010–1013.Hirvonen J, Huszenicza G, Kulcsar M, Pyorala S, 1999:Acute-phase response <strong>in</strong> dairy cows with acute postpartummetritis. Theriogenology 51, 1071–1083.Hockett ME, Hopk<strong>in</strong>s FM, Lewis MJ, Saxton AM, DowlenHH, Oliver SP, Schrick FN, 2000: Endocr<strong>in</strong>e profiles ofdairy cows follow<strong>in</strong>g experimentally <strong>in</strong>duced cl<strong>in</strong>ical mastitisdur<strong>in</strong>g early lactation. Anim Reprod Sci 58, 241–251.Hussa<strong>in</strong> AM, Daniel RC, 1991: Bov<strong>in</strong>e endometritis: currentand future alternative therapy. Zentralbl Veter<strong>in</strong>armed A38, 641–651.Huszenicza Gy, Ja´nosi Sz, Kulcsa´r M,Ko´ro´di P, Reiczigel J,Kátai L, Peters AR, De Rensis F, 2005: Effects of cl<strong>in</strong>icalmastitis on ovarian function <strong>in</strong> postpartum dairy cows.Reprod Domest Anim 40, 199–204.Iqbal J, Kurose Y, Canny B, Clarke IJ, 2006: Effects of central<strong>in</strong>fusion of ghrel<strong>in</strong> on food <strong>in</strong>take and plasma levels ofgrowth hormone, lute<strong>in</strong>iz<strong>in</strong>g hormone, prolact<strong>in</strong>, and cortisolsecretion <strong>in</strong> sheep. Endocr<strong>in</strong>ology 147, 510–519.Kluge M, Schussler P, Uhr M, Yassouridis A, Steiger A, 2007:Ghrel<strong>in</strong> suppresses secretion of lute<strong>in</strong>iz<strong>in</strong>g hormone <strong>in</strong>humans. J Cl<strong>in</strong> Endocr<strong>in</strong>ol Metab 92, 3202–3205.Kojima M, Hosoda H, Date Y, Nakazato M, Matsuo H,Kangawa K, 1999: Ghrel<strong>in</strong> is a growth-hormone-releas<strong>in</strong>gacylated peptide from stomach. Nature 402, 656–660.Kujjo LL, Bosu WT, Perez GI, 1995: Opioid peptides<strong>in</strong>volvement <strong>in</strong> endotox<strong>in</strong>-<strong>in</strong>duced suppression of LH secre-Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Endotox<strong>in</strong> and Neuroendocr<strong>in</strong>ology 323tion <strong>in</strong> ovariectomized Holste<strong>in</strong> heifers. Reprod Toxicol 9,169–174.Lesh<strong>in</strong> LS, Malven PV 1984: Bacteremia-<strong>in</strong>duced changes <strong>in</strong>pituitary hormone release and effect of naloxone. Am JPhysiol 247 (5 Pt 1) E585–E591.Li XF, K<strong>in</strong>sey-Jones JS, Knox AM, Wu XQ, Tahs<strong>in</strong>soy D,Bra<strong>in</strong> SD, Lightman SL, O’Byrne KT, 2007: Neonatallipopolysaccharide exposure exacerbates stress-<strong>in</strong>duced suppressionof lute<strong>in</strong>iz<strong>in</strong>g hormone pulse frequency <strong>in</strong> adulthood.Endocr<strong>in</strong>ology 148, 5984–5990.Liu S, Khemlani LS, Shapiro RA, Johnson ML, Liu K, GellerDA, Watk<strong>in</strong>s SC, Goyert SM, Billiar TR, 1998: Expressionof CD14 by hepatocytes: upregulation by cytok<strong>in</strong>es dur<strong>in</strong>gendotoxemia. Infect Immun 66, 5089–5098.Moore DA, Cullor JS, Bondurant RH, Sischo WM, 1991:Prelim<strong>in</strong>ary field evidence for the association of cl<strong>in</strong>icalmastitis with altered <strong>in</strong>terestrus <strong>in</strong>tervals <strong>in</strong> dairy cattle.Theriogenology 36, 257–265.Parrott RF, Vellucci SV, Goode JA, Lloyd DM, Forsl<strong>in</strong>g ML,1995: Cyclo-oxygenase mediation of endotox<strong>in</strong>-<strong>in</strong>ducedfever, anterior and posterior pituitary hormone release,and hypothalamic c-Fos expression <strong>in</strong> the prepubertal pig.Exp Physiol 80, 663–674.Peter AT, Bosu WT, DeDecker RJ, 1989: Suppression ofpreovulatory lute<strong>in</strong>iz<strong>in</strong>g hormone surges <strong>in</strong> heifers after<strong>in</strong>trauter<strong>in</strong>e <strong>in</strong>fusions of Escherichia coli endotox<strong>in</strong>. Am JVet Res 50, 368–373.Priego T, Granado M, Iba´n˜ ez de Cáceres I, Martı´n AI, Villanu´aMA, Lo´pez-Caldero´n A., 2003: Endotox<strong>in</strong> at low dosesstimulates pituitary GH whereas it decreases IGF-I and IGFb<strong>in</strong>d<strong>in</strong>gprote<strong>in</strong>-3 <strong>in</strong> rats. J Endocr<strong>in</strong>ol 179, 107–117.Rettori V, Belova N, Kamat A, Lyson K, Gimeno M, McCannSM, 1994: Blockade by <strong>in</strong>terleuk<strong>in</strong>-1-alpha of nitricoxidergiccontrol of lute<strong>in</strong>iz<strong>in</strong>g hormone-releas<strong>in</strong>g hormone release<strong>in</strong> vivo and <strong>in</strong> vitro. Neuroimmunomodulation 1, 86–91.Salfen BE, Carroll JA, Keisler DH, Strauch TA, 2004: Effectsof exogenous ghrel<strong>in</strong> on feed <strong>in</strong>take, weight ga<strong>in</strong>, behavior,and endocr<strong>in</strong>e responses <strong>in</strong> weanl<strong>in</strong>g pigs. J Anim Sci 82,1957–1966.Sart<strong>in</strong> JL, Elsasser TH, Gunter DR, McMahon CD, 1998:Endocr<strong>in</strong>e modulation of physiological responses to catabolicdisease. Domest Anim Endocr<strong>in</strong>ol 15, 423–429.Scarborough DE 1990: Somatostat<strong>in</strong> regulation by cytok<strong>in</strong>es.Metabolism 39 (9 Suppl. 2) 108–111.Sheldon IM, Lewis GS, LeBlanc S, Gilbert RO, 2006: Def<strong>in</strong><strong>in</strong>gpostpartum uter<strong>in</strong>e disease <strong>in</strong> cattle. Theriogenology 65,1516–1530.Strauch TA, Carroll JA, Fangman TJ, Wiedmeyer CE,Hamback AK, 2004: The acute phase response of Escherichiacoli challenged pigs exhibit<strong>in</strong>g a febrile response <strong>in</strong> theabsence of elevated TNF-alpha. J Anim Vet Adv 3, 227–238.Suzuki C, Yoshioka K, Iwamura S, Hirose H, 2001: Endotox<strong>in</strong><strong>in</strong>duces delayed ovulation follow<strong>in</strong>g endocr<strong>in</strong>e aberrationdur<strong>in</strong>g the proestrous phase <strong>in</strong> Holste<strong>in</strong> heifers.Domest Anim Endocr<strong>in</strong>ol 20, 267–278.Takeuchi Y, Nagabukuro H, Kizumi O, Mori Y, 1997:Lipopolysaccharide-<strong>in</strong>duced suppression of the hypothalamicgonadotrop<strong>in</strong>-releas<strong>in</strong>g hormone pulse generator <strong>in</strong>ovariectomized goats. J Vet Med Sci 59, 93–96.Thompson K, Coleman ES, Hudmon A, Kemppa<strong>in</strong>en RJ,Soyoola EO, Sart<strong>in</strong> JL, 1995: Effects of short-term cortisol<strong>in</strong>fusion on growth hormone-releas<strong>in</strong>g hormone stimulationof growth hormone release <strong>in</strong> sheep. Am J Vet Res 56, 1228–1231.USDA 1996: Dairy ‘96. Animal Plant Health InspectionService. National Animal Health Monitor<strong>in</strong>g Service. Ft.Coll<strong>in</strong>s, CO, USA.Valde JP, Hird DW, Thurmond MC, Osteras O, 1997:Comparison of ketosis, cl<strong>in</strong>ical mastitis, somatic cell count,and reproductive performance between free stall and tie stallbarns <strong>in</strong> Norwegian dairy herds with automatic feed<strong>in</strong>g.Acta Vet Scand 38, 181–192.Vila G, Maier C, Riedl M, Nowotny P, Ludvik B, Luger A,Clodi M, 2007: Bacterial endotox<strong>in</strong> <strong>in</strong>duces biphasicchanges <strong>in</strong> plasma ghrel<strong>in</strong> <strong>in</strong> healthy humans. J Cl<strong>in</strong>Endocr<strong>in</strong>ol Metab 92, 3930–3934.Vulliemoz NR, Xiao E, Xia-Zhang L, Germond M, Rivier J,Fer<strong>in</strong> M, 2004: Decrease <strong>in</strong> lute<strong>in</strong>iz<strong>in</strong>g hormone pulsefrequency dur<strong>in</strong>g a five-hour peripheral ghrel<strong>in</strong> <strong>in</strong>fusion <strong>in</strong>the ovariectomized rhesus monkey. J Cl<strong>in</strong> Endocr<strong>in</strong>olMetab 89, 5718–5723.Waldron MR, Nishida T, Nonnecke BJ, Overton TR, 2003:Effect of lipopolysaccharide on <strong>in</strong>dices of peripheral andhepatic metabolism <strong>in</strong> lactat<strong>in</strong>g cows. J Dairy Sci 86, 3447–3459.Washburn SP, White SL, Green JT Jr, Benson GA, 2002:<strong>Reproduction</strong>, mastitis, and body condition of seasonallycalved Holste<strong>in</strong> and Jersey cows <strong>in</strong> conf<strong>in</strong>ement or pasturesystems. J Dairy Sci 85, 105–111.West MA, Heagy W, 2002: Endotox<strong>in</strong> tolerance: a review. CritCare Med 30(1 Suppl.), S64–S73.Xiao E, Xia-Zhang L, Fer<strong>in</strong> M, 2000: Inhibitory effects ofendotox<strong>in</strong> on LH secretion <strong>in</strong> the ovariectomized monkeyare prevented by naloxone but not by an <strong>in</strong>terleuk<strong>in</strong>-1receptor antagonist. Neuroimmunomodulation 7, 6–15.Yoo MJ, Nishihara M, Takahashi M, 1997: Tumor necrosisfactor-alpha mediates endotox<strong>in</strong> <strong>in</strong>duced suppression ofgonadotrop<strong>in</strong>-releas<strong>in</strong>g hormone pulse generator activity <strong>in</strong>the rat. Endocr J 44, 141–148.Zeisberger E, Roth J, 1998: Tolerance to pyrogens. Ann N YAcad Sci 856, 116–131.Author’s address (for correspondence): JL Sart<strong>in</strong>, Department ofAnatomy, Physiology and Pharmacology, College of Veter<strong>in</strong>aryMedic<strong>in</strong>e, Auburn University, Auburn, AL 36849, USA. E-mail:sartijl@vetmed.auburn.eduConflict of <strong>in</strong>terest: All authors declare no conflict of <strong>in</strong>terests.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 324–330 (2008); doi: 10.1111/j.1439-0531.2008.01173.xISSN 0936-6768Energy Metabolism and Lept<strong>in</strong>: Effects on Neuroendocr<strong>in</strong>e Regulation of<strong>Reproduction</strong> <strong>in</strong> the Gilt and SowCR Barb 1 , GJ Hausman 1 and CA Lents 21 USDA ⁄ ARS, Richard B. Russell Agriculture Research Center; 2 Animal and Dairy Science Department, University of Georgia, Athens, GA, USAContentsIt is well established that reproductive function is metabolicallygated. However, the mechanisms whereby energy storesand metabolic cues <strong>in</strong>fluence appetite, energy homeostasis andfertility are yet to be completely understood. Adipose tissue isno longer considered as only a depot to store excess energy.Recent f<strong>in</strong>d<strong>in</strong>gs have identified numerous genes, severalneurotrophic factors, <strong>in</strong>terleuk<strong>in</strong>s, <strong>in</strong>sul<strong>in</strong>-like growth factorb<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>-5, ciliary neurotrophic factor and neuropeptideY (NPY) as be<strong>in</strong>g expressed by adipose tissue dur<strong>in</strong>gpubertal development. These studies demonstrated for the firsttime the expression of several major adipok<strong>in</strong>es or cytok<strong>in</strong>es <strong>in</strong>pig adipose tissue which may <strong>in</strong>fluence local and centralmetabolism and growth. Lept<strong>in</strong> appears to be the primarymetabolic signal and is part of the adipose tissue-hypothalamicregulatory loop <strong>in</strong> the control of appetite, energy homeostasisand lute<strong>in</strong>iz<strong>in</strong>g hormone (LH) secretion. Lept<strong>in</strong>’s actions onappetite regulation are mediated by <strong>in</strong>hibition of hypothalamicNPY and stimulation of proopiomelanocort<strong>in</strong>. Its effects ongonadotrop<strong>in</strong>-releas<strong>in</strong>g hormone (GnRH) ⁄ LH secretion aremediated by NPY and kisspept<strong>in</strong>. Thus, lept<strong>in</strong> appears to bean important l<strong>in</strong>k between metabolic status, the neuroendocr<strong>in</strong>eaxis and subsequent fertility <strong>in</strong> the gilt and sow.IntroductionIt is generally accepted that there are two modes oflute<strong>in</strong>iz<strong>in</strong>g hormone (LH) secretion <strong>in</strong> the pig (Krael<strong>in</strong>gand Barb 1990), pulsatile secretion and surge secretion.These patterns of LH secretion reflect the pattern ofgonadotrop<strong>in</strong>-releas<strong>in</strong>g hormone (GnRH) released fromneurosecretory neurones with<strong>in</strong> the hypothalamus <strong>in</strong>tothe hypothalamic-hypophysial portal system (Williams1989). The importance of pulsatile GnRH ⁄ LH secretionwas demonstrated <strong>in</strong> studies that <strong>in</strong>duced precociousoestrus and ovulation <strong>in</strong> <strong>in</strong>tact pre-pubertal gilts withhourly <strong>in</strong>travenous (i.v.) <strong>in</strong>jections of GnRH (Lutz et al.1984; Press<strong>in</strong>g et al. 1992). In addition, hourly adm<strong>in</strong>istrationof GnRH to anestrous post-partum sows<strong>in</strong>duced oestrus and ovulation (Cox and Britt 1988).Thus, GnRH neurones secrete their product <strong>in</strong> anepisodic manner, but <strong>in</strong>teroceptive and exteroceptivefactors detected by the central nervous system aretranslated by neuroendocr<strong>in</strong>e mechanisms <strong>in</strong>to signalswhich alter the pattern of GnRH and subsequent LHsecretion. For example, <strong>in</strong>teroceptive signals, such asgonadal and adrenal steroids, metabolites, and otherneuronal signals act to modulate frequency and amplitudeof GnRH pulses.The importance of nutrition and metabolic state <strong>in</strong><strong>in</strong>itiat<strong>in</strong>g and ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g reproductive function andgrowth is well established (reviewed by Prunier et al.1993; Prunier and Quesnel 2000; Barb et al. 2002).Dietary nutrients <strong>in</strong>fluence expression of metabolicpathways that allow animals to achieve their full geneticpotential for reproduction and growth. These pathwaysare complex and <strong>in</strong>volve appetite regulation along withregulation of the reproductive and growth axis, as wellas gonadal function (Wade et al. 1996; Barb et al. 1999;Muller et al. 1999). Identification of blood-borne metabolicsignals that activate the GnRH ⁄ LH pulse generator,alter the pattern of growth hormone (GH)secretion, and regulate appetite rema<strong>in</strong>s elusive. Recentreports have identified adipose tissue as a source ofputative metabolic signals that regulate the neuroendocr<strong>in</strong>eaxis (Schwartz et al. 1996; Barb and Krael<strong>in</strong>g2004; Barb et al. 2006a).Nutrition and <strong>Reproduction</strong>Research has exam<strong>in</strong>ed backfat levels <strong>in</strong> gilts relative tofuture reproductive performances. Several reports demonstratedthat gilts with higher backfat subsequentlyhad a shorter wean<strong>in</strong>g to first oestrus <strong>in</strong>terval, largerlitter size, and higher farrow<strong>in</strong>g rate as second paritysows compared to gilts with lower amounts of backfat(Tummaruk et al. 2001). Whittemore (1996) suggestedthat primiparous sows should not be allowed to havebackfat depth fall below 14 mm or rise above 25 mm(lipid ⁄ prote<strong>in</strong> >1:1 and


Energy and <strong>Reproduction</strong> <strong>in</strong> the Pig 325lactation (Weldon et al. 1994; Whittemore 1996).Almeida et al. (2001) showed that moderate feedrestriction dur<strong>in</strong>g the luteal phase of the oestrous cycle<strong>in</strong> the gilt affects ovulation rate and the progesteronerise after the LH surge.The role of <strong>in</strong>sul<strong>in</strong> on LH secretion <strong>in</strong> the postpartumsow is somewhat controversial. Tokach et al.(1992) reported that <strong>in</strong>sul<strong>in</strong> levels dur<strong>in</strong>g early lactationwere correlated with LH peak amplitude and thusappear to be associated with reproductive function.Koketsu et al. (1998) reported greater lactation feed<strong>in</strong>take was associated with greater concentrations of<strong>in</strong>sul<strong>in</strong> and glucose, greater LH pulse frequency prior towean<strong>in</strong>g and shorter farrow<strong>in</strong>g-to-oestrus <strong>in</strong>terval. Inaddition, exogenous <strong>in</strong>sul<strong>in</strong> adm<strong>in</strong>istered on the day ofwean<strong>in</strong>g for 4 consecutive days <strong>in</strong> primiparous sowsdecreased the average <strong>in</strong>terval from wean<strong>in</strong>g to oestrusand <strong>in</strong>creased the percentage of sows <strong>in</strong> oestrus (Whitleyet al. 1998). One model used to study the effect of <strong>in</strong>sul<strong>in</strong>on the hypothalamic-pituitary axis is the diabetes<strong>in</strong>ducedanimal. In the diabetic ovariectomized gilt,withdrawal of <strong>in</strong>sul<strong>in</strong> therapy for 4 days prevented theoestradiol-<strong>in</strong>duced preovulatory-like LH surge, but didnot affect pulsatile LH secretion (Angell et al. 1996).This f<strong>in</strong>d<strong>in</strong>g <strong>in</strong>dicates that diabetes mellitus alters thesensitivity of the hypothalamic-pituitary axis to oestradioland pituitary responsiveness to GnRH. Pituitarycell culture experiments confirmed that the sensitivity ofthe pituitary gland to GnRH decreased after removal of<strong>in</strong>sul<strong>in</strong> therapy for 7 days <strong>in</strong> diabetic pigs (Angell et al.1996), suggest<strong>in</strong>g that <strong>in</strong>sul<strong>in</strong> may play a role <strong>in</strong>ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g pituitary responsiveness to GnRH. Incontrast, <strong>in</strong>sul<strong>in</strong> adm<strong>in</strong>istration on 5 consecutive daysprior to wean<strong>in</strong>g <strong>in</strong> feed-restricted sows did not affectwean<strong>in</strong>g to oestrus <strong>in</strong>terval or ovulation rate (Quesneland Prunier 1998). In primiparous sows, neither glucosenor <strong>in</strong>sul<strong>in</strong> concentrations were correlated with LHsecretion dur<strong>in</strong>g lactation, after wean<strong>in</strong>g or with thewean<strong>in</strong>g to oestrus <strong>in</strong>terval (van den Brand et al. 2000).Moreover, van den Brand et al. (2001) found <strong>in</strong>primiparous sows that plasma IGF-I concentrationson day 22 (wean<strong>in</strong>g) were positively correlated with LHpulse frequency while Quesnel et al. (1998) reported norelationship between IGF-I concentrations and LHpulse frequency before or after wean<strong>in</strong>g <strong>in</strong> primiparoussows. Thus, the def<strong>in</strong>itive role of IGF-I and <strong>in</strong>sul<strong>in</strong> onLH secretion and subsequent reproductive performance<strong>in</strong> the post-partum sow has yet to be determ<strong>in</strong>ed.Serum and milk lept<strong>in</strong> concentrations <strong>in</strong> the primaparousand multiparous lactat<strong>in</strong>g sow were positivelycorrelated with backfat thickness and level of dietaryenergy fed dur<strong>in</strong>g gestation, as well as feed consumption(Estienne et al. 2000, 2003). A positive correlation wasobserved among plasma <strong>in</strong>sul<strong>in</strong>, lept<strong>in</strong> and LH concentrations<strong>in</strong> lactat<strong>in</strong>g sows fed ad libitum but not <strong>in</strong> feedrestrictedsows. Moreover, the wean<strong>in</strong>g to oestrus<strong>in</strong>terval was greater <strong>in</strong> the feed-restricted sows comparedto controls (Mao et al. 1999). De Rensis et al. (2005)reported that <strong>in</strong> sows classified as fat, medium or th<strong>in</strong>based on backfat thickness at farrow<strong>in</strong>g, serum lept<strong>in</strong>concentrations were greater <strong>in</strong> fat sows compared tomedium and th<strong>in</strong> animals at wean<strong>in</strong>g. In addition, therewas no relationship between lept<strong>in</strong> concentrations andreproductive performance after wean<strong>in</strong>g. However,plasma lept<strong>in</strong> concentrations were associated withbackfat depth, and loss of backfat was associated withreproductive performance. These f<strong>in</strong>d<strong>in</strong>gs provide evidencethat circulat<strong>in</strong>g lept<strong>in</strong>, LH concentrations andfeed consumption dur<strong>in</strong>g lactation are <strong>in</strong>fluenced bydietary energy <strong>in</strong>take dur<strong>in</strong>g pregnancy or lactation <strong>in</strong>the sow, suggest<strong>in</strong>g that lept<strong>in</strong> may serve as a permissivemetabolic signal that may be necessary for activation ofthe reproductive axis <strong>in</strong> the post-partum sow.Growth and Metabolic SignalsIn addition to developmentally related maturation of theneuroendocr<strong>in</strong>e axis, permissive peripheral signals areassociated with atta<strong>in</strong>ment of a m<strong>in</strong>imum percentage ofbody fat (Frisch 1984); one example is lept<strong>in</strong> which mayplay a role <strong>in</strong> the tim<strong>in</strong>g of puberty (Barb et al. 2001a;Barb and Krael<strong>in</strong>g 2004). Dur<strong>in</strong>g the pre-pubertalperiod, expression of a number of hypothalamic genesassociated with appetite and growth regulation appearto be developmentally regulated. For example, hypothalamicexpression of the biological form of the lept<strong>in</strong>receptor (OB-rb), adipose tissue lept<strong>in</strong> expression andserum lept<strong>in</strong> concentrations <strong>in</strong>creased by 3.5 months ofage (Qian et al. 1999; L<strong>in</strong> et al. 2001). In addition,Kojima et al. (2007) demonstrated a positive relationshipbetween pre-wean<strong>in</strong>g weight and post-weanimghypothalamic agouti-related prote<strong>in</strong>, orex<strong>in</strong>, type-2orex<strong>in</strong> receptor and NPY gene expression. These genesmay play an important role <strong>in</strong> post-wean<strong>in</strong>g growth anddevelopment <strong>in</strong> the gilt. Furthermore, their encodedprote<strong>in</strong>s are well positioned anatomically to <strong>in</strong>teractwith GnRH (Krael<strong>in</strong>g and Barb 1990) and GHRH(Lesh<strong>in</strong> et al. 1994) neurones and appetite regulat<strong>in</strong>gneurones (Lawrence et al. 1999; Matteri 2001). Consistentwith this idea, central adm<strong>in</strong>istration of lept<strong>in</strong><strong>in</strong>creased GH secretion and suppressed feed <strong>in</strong>take <strong>in</strong>the pre-pubertal gilt (Barb et al. 1998) and <strong>in</strong> vitro lept<strong>in</strong>stimulated GnRH release from porc<strong>in</strong>e hypothalamicexplants (Barb et al. 2004). These changes <strong>in</strong> thegrowth ⁄ reproductive axis appear to be <strong>in</strong> concert withthe tim<strong>in</strong>g of puberty.We previously reported that metabolic response toacute feed deprivation occurred more rapidly <strong>in</strong> prepubertalgilts compared to mature gilts, likely becausepre-pubertal gilts have a higher metabolic rate, smallerenergy reserves and thus a greater nutrient <strong>in</strong>takerequirement for growth (Barb et al. 1997). An acute24 h fast <strong>in</strong>creased serum free fatty acid concentrations,and decreased lept<strong>in</strong> pulse frequency but not meanserum lept<strong>in</strong> concentrations <strong>in</strong> the ovariectomized prepubertalgilt (Barb et al. 2001a). Furthermore, shortterm feed restriction decreased lept<strong>in</strong> secretion and LHpulse frequency <strong>in</strong> the mature ovariectomized gilt(Whisnant and Harrell 2002) and decreased LH secretion<strong>in</strong> the <strong>in</strong>tact pre-pubertal gilt (Booth et al. 1996),while a 3-day fast reduced adipose tissue lept<strong>in</strong> mRNA<strong>in</strong> castrate male pigs (Spurlock et al. 1998). These resultssupport the idea that lept<strong>in</strong> may serve as a metabolicsignal <strong>in</strong> the activation of the reproductive axis.In pre-pubertal gilts short term feed restriction to33% of control diet for 8 days failed to affect LH orÓ 2008 No claim to orig<strong>in</strong>al government works


326 CR Barb, GJ Hausman and CA LentsTable 1. Microarray analysis of several fat depots demonstrated that thyroid hormone receptors were collectively upregulated while several otherreceptors (MCIR, FGF4) and lipogenic (LPL, SCD) enzymes were downregulated with fast<strong>in</strong>g aFat depot Gene Estimate SE T-value p-valuePerirenal Thyroid hormone receptor alpha 2 (Sus scrofa) )0.20083 0.051199 )3.92248 0.000254Mesenteric Thyroid hormone receptor, alpha )0.16131 0.047101 )3.42473 0.001195Leaf Thyroid hormone receptor, alpha )0.15323 0.030834 )4.96954 0.000007Leaf Thyroid hormone receptor alpha 2 (Sus scrofa) )0.31586 0.089853 )3.51524 0.000909Leaf Melanocort<strong>in</strong> 1 receptor 0.357846 0.175065 2.044075 0.045929Perirenal Melanocort<strong>in</strong> 1 receptor 0.396369 0.117805 3.364614 0.001431Leaf Fibroblast growth factor receptor 4 0.40214 0.121846 3.300404 0.001731Perirenal Fibroblast growth factor receptor 4 0.44749 0.139023 3.218823 0.002199Perirenal Stearoyl-CoA desaturase 0.914822 0.323623 2.826815 0.006618Lipoprote<strong>in</strong> lipase 0.43321 0.125486 3.452266 0.0011Mesenteric Stearoyl-CoA desaturase 1.40823 0.304109 4.630675 0.000024Lipoprote<strong>in</strong> lipase 0.14577 0.069703 2.091302 0.04131Leaf Stearoyl-CoA desaturase 0.170878 0.071794 2.380111 0.020937Lipoprote<strong>in</strong> lipase 0.380183 0.079236 4.798133 0.000013a Analysis of the microarray data was conducted with either LOWESS normalization based or an ANOVA normalization based method to determ<strong>in</strong>e the <strong>in</strong>fluence offast<strong>in</strong>g on gene expression.lept<strong>in</strong> secretion or to affect backfat lept<strong>in</strong>, Ob-rb,adipocyte fatty acid b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>, or the transcriptionfactors peroxisome proliferator-activated receptor-c2and CCAAT-enhancer-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>-a expression(Hart et al. 2007). However, based on ma<strong>in</strong>tenancerequirements for the pre-pubertal gilt (NRC 1998) used<strong>in</strong> that study, the feed restricted gilts were actually fed124% of the ma<strong>in</strong>tenance requirement. This may expla<strong>in</strong>the failure of feed restriction to affect LH or lept<strong>in</strong>concentrations. Although feed restriction did preventbody weight and backfat ga<strong>in</strong> adipocyte function wasaltered as evidenced by upregulation of thyroid hormonereceptor-a <strong>in</strong> perirenal, leaf and mesenteric fatdepots <strong>in</strong> feed restricted animals; this co<strong>in</strong>cided withelevated thyrox<strong>in</strong> concentration (GJ Hausman, CRBarb, HA Hart, unpublished data; Table 1). Thesechanges may <strong>in</strong> part represent an attempt to ma<strong>in</strong>ta<strong>in</strong>thermogenesis, as well as immune and neruoendocr<strong>in</strong>ehomeostasis.Adipose Tissue as an Endocr<strong>in</strong>e OrganAdipose tissue plays a more dynamic role thanpreviously thought <strong>in</strong> physiological mechanisms andwhole-body homeostasis. Studies now show that therole of adipose tissue <strong>in</strong>cludes respond<strong>in</strong>g to nutrient,neural and hormonal signals, and secret<strong>in</strong>g factors or‘adipok<strong>in</strong>es’ that control feed<strong>in</strong>g, thermogenesis, immunity,and neuroendocr<strong>in</strong>e function (review by Ahimaet al. 2006). The current evidence <strong>in</strong>dicates that of allthe adipose tissue secreted factors, or ‘adipok<strong>in</strong>es’,<strong>in</strong>terleuk<strong>in</strong>s (IL)-6, IL-8, plasm<strong>in</strong>ogen activator <strong>in</strong>hibitor-1,lept<strong>in</strong> and adiponect<strong>in</strong> can be considered trueendocr<strong>in</strong>e factors (review by Hauner 2005). Furthermore,the secretory function of adipose tissue isadversely <strong>in</strong>fluenced by both obesity (Hauner 2005)and impaired adipose tissue accretion (review byGuerre-Millo 2004). Therefore, body condition oradiposity by virtue of secreted adipok<strong>in</strong>es could impactor regulate, <strong>in</strong> part, physiological states such asreproductive condition or status. Adipose tissue constitutesthe largest amount of stored energy <strong>in</strong> the body(Loftus 1999), and regulation of energy expenditure<strong>in</strong>volves a balance of several factors, such as feed<strong>in</strong>gbehaviour, adipose tissue mass and activation ofcatabolic processes (e.g. lactation). Changes <strong>in</strong> bodyweight or nutritional status are characterized byalterations <strong>in</strong> serum concentrations of many hormonesand growth factors that regulate adipocyte functionand lept<strong>in</strong> secretion (Barb et al. 2001b). Recent microarrayanalyses by our laboratory revealed that 21 genesencod<strong>in</strong>g secreted prote<strong>in</strong>s were expressed 40-fold overbackground <strong>in</strong> neonatal porc<strong>in</strong>e adipose tissue andporc<strong>in</strong>e pre-adipocyte cultures (Hausman et al. 2006).Additionally, agouti gene expression was detected byRT-PCR <strong>in</strong> pig adipose tissue. Proteomic analysis ofadipocyte culture conditioned media identified severalsecreted prote<strong>in</strong>s, <strong>in</strong>clud<strong>in</strong>g several neurotrophic factors,IL-1A, IL-1B, IL-8, IL-6, IL-15, and IGF b<strong>in</strong>d<strong>in</strong>gprote<strong>in</strong>-5. Through microarray and RT-PCR analyses,we recently demonstrated that <strong>in</strong> addition to these,several other cytok<strong>in</strong>es, e.g., ciliary neurotrophic factorand NPY, are expressed by adipose tissue dur<strong>in</strong>gpubertal development (Hausman et al. 2007). Thesestudies demonstrate for the first time the expressionof several major adipok<strong>in</strong>es or cytok<strong>in</strong>es <strong>in</strong> pigadipose tissue which may <strong>in</strong>fluence local and centralmetabolism and growth. Thus, these reports supportthe idea that adipose tissue functions as an endocr<strong>in</strong>eorgan.Morphological studies have revealed that adiposetissue is <strong>in</strong>nervated by adrenergic nerve fibres (Hausmanand Richardson 1987). Further, immunocytochemicaldata revealed that most of the subpopulations of theadrenergic lept<strong>in</strong> receptor immuno-reactive (OBR-IR)neurones supply<strong>in</strong>g fat tissue <strong>in</strong> the pig were positive forNPY and tyros<strong>in</strong>e hydroxylase immunoactivity (Czajaet al. 2002). Moreover, immuno-positive neurones forOBR were located <strong>in</strong> the paraventricular nucleus,ventromedial nucleus, anterior hypothalamic area, preopticarea, arcuate nucleus and supraoptic nucleus(Czaja et al. 2003). Collectively, these studies providemorphological data demonstrat<strong>in</strong>g that hypothalamicOBR conta<strong>in</strong><strong>in</strong>g neurones are transsynaptically connectedto the perirenal fat depot. Therefore, the aboveevidence supports a direct l<strong>in</strong>k between hypothalamicÓ 2008 No claim to orig<strong>in</strong>al government works


Energy and <strong>Reproduction</strong> <strong>in</strong> the Pig 327Fig. 1. Schematic illustration of lept<strong>in</strong> coord<strong>in</strong>ation of energy homeostasisand neuroendocr<strong>in</strong>e function: Lept<strong>in</strong> is secreted <strong>in</strong> response tochanges <strong>in</strong> energy balance and acts on the hypothalamus to controlfood <strong>in</strong>take, reproduction and adipocyte function. Positive energybalance <strong>in</strong>creases blood lept<strong>in</strong> concentrations, suppresses expression ofneuropeptide Y (NPY) and agouti related peptide (AgRP), andupregulates proopiomelanocort<strong>in</strong> (POMC) and a-melanocyte stimulat<strong>in</strong>ghormone (a-MSH). Negative energy balance decreases bloodlept<strong>in</strong> concentrations, stimulates expression of NPY and AgRP anddownregulates POMC and a-MSH. Thick black l<strong>in</strong>es represent strongregulator tone and th<strong>in</strong> black l<strong>in</strong>es weak regulator tone. Modified fromLoftus (1999)neurones <strong>in</strong> the regulation of fat metabolism andreproduction.Under chang<strong>in</strong>g metabolic states, which may <strong>in</strong>creaseadipose tissue mass and circulat<strong>in</strong>g lept<strong>in</strong> concentrationsand stimulate catabolic pathways with<strong>in</strong> the hypothalamus,conversely decreased adipose mass and reducedblood lept<strong>in</strong> concentrations stimulate anabolic pathwaysand <strong>in</strong>hibit catabolic pathways (Fig. 1). These twopathways comprise a number of genes that not onlyregulate appetite and energy balance but also impact thereproductive axis either directly <strong>in</strong> the case of NPY, or<strong>in</strong>directly by alter<strong>in</strong>g adipocyte function and hence theexpression of genes and their prote<strong>in</strong> products. Forexample, hypothalamic NPY and proopiomelanocort<strong>in</strong>(POMC) associated products a-melanocyte stimulat<strong>in</strong>ghormone (a-MSH) and beta endorph<strong>in</strong> can affecthypothalamic GnRH release and subsequent feed<strong>in</strong>gbehaviour (Barb et al. 1994, 1998, 2006b; Crown et al.2007). Peripherally, adipocyte lept<strong>in</strong> expression andsecretion can suppress appetite (Barb et al. 1998),stimulate hypothalamic GnRH release (Barb et al.2004) and reverse the <strong>in</strong>hibitory effect of energy deprivationon LH secretion (Whisnant and Harrell 2002).Thus, as previously stated, reproductive function ismetabolically gated, but mechanisms <strong>in</strong>terfac<strong>in</strong>g energystores or metabolic cues and fertility are not completelyunderstood.Identification of targets of lept<strong>in</strong> <strong>in</strong> the hypothalamusIt is well established that reproductive function ismetabolically gated. However, the mechanisms wherebyenergy stores and metabolic cues <strong>in</strong>fluence fertility areyet to be completely understood. The effects of lept<strong>in</strong>appear to be mediated through modulation of hypothalamicNPY expression (Campfield et al. 1996). In thepig, the presence of biologically-active OBR <strong>in</strong> thehypothalamus and pituitary (L<strong>in</strong> et al. 2000) and thefact that lept<strong>in</strong> <strong>in</strong>creased LH secretion from pig pituitarycells and GnRH release from hypothalamic tissue <strong>in</strong>vitro (Barb et al. 2004) suggest that lept<strong>in</strong> acts throughthe hypothalamus. There is strong evidence from colocalizationof lept<strong>in</strong> receptor mRNA with NPY geneexpression that hypothalamic NPY is the primarypotential target for lept<strong>in</strong> <strong>in</strong> the pig (Czaja et al. 2002).Moreover, central adm<strong>in</strong>istration of NPY suppressedLH secretion and stimulated feed <strong>in</strong>take and reversedthe <strong>in</strong>hibitory action of lept<strong>in</strong> on feed <strong>in</strong>take (Barb et al.2006b). However, NPY alone may not mediate theaction of lept<strong>in</strong>, s<strong>in</strong>ce lept<strong>in</strong> failed to effect NPY releasefrom pig hypothalamic-preoptic area tissue fragments(Barb et al. 2004). Furthermore, metabolic signals may<strong>in</strong> part be communicated to GnRH neurones via otherneuropeptides such as galan<strong>in</strong>-like peptide (Rich et al.2007), a-MSH (Crown et al. 2007), b-endorph<strong>in</strong> (Barbet al. 1994), or kisspept<strong>in</strong> (Arregu<strong>in</strong>-Arevalo et al. 2007;Luque et al. 2007).The kisspept<strong>in</strong>s are potent stimulators of theGnRH ⁄ LH axis (Castellano et al. 2006; Arregu<strong>in</strong>-Arevalo et al. 2007; Luque et al. 2007). The kisspept<strong>in</strong>sare a group of structurally related peptides that areproducts of the kisspept<strong>in</strong>-1 (KiSS-1) gene (Ohtaki et al.2001; Kotani et al. 2001). Synthesized as a pre-prohormone,it is cleaved to liberate a 54 am<strong>in</strong>o acid peptidewhich can be proteolytically processed (Tak<strong>in</strong>o et al.2003) to shorter variants, all of which share the sameamidated C-term<strong>in</strong>us and reta<strong>in</strong> full biological activity.Kisspept<strong>in</strong>s act<strong>in</strong>g through their cognate receptor,GPR54, are thought to be an important determ<strong>in</strong>ate <strong>in</strong>the onset of puberty (Shahab et al. 2005; Smith andClarke 2007). Thus, hypothalamic kisspept<strong>in</strong> and itsreceptor, GPR54, may serve as an essential gatekeeperof GnRH neurones and hence of reproductive function.Kisspept<strong>in</strong>s play a role <strong>in</strong> the tim<strong>in</strong>g of puberty onset(Tena-Sempere 2006c) and act on the gonadotropic axisvia the release of hypothalamic GnRH (Tena-Sempere2006a; Dungan et al. 2006). The hypothalamic expressionof KiSS-1 gene is under the control of sex steroids,and KiSS-1 neurones are <strong>in</strong>volved <strong>in</strong> mediat<strong>in</strong>g thenegative and positive feedback effects of oestradiol ongonadotrop<strong>in</strong> secretion (Smith et al. 2005, 2006). Moreover,the hypothalamic KiSS-1 system may also conveythe modulatory action of metabolic signals to theGnRH neurones (Tena-Sempere 2006b; Luque et al.2007). A recent report demonstrated that short-termfast<strong>in</strong>g resulted <strong>in</strong> a decl<strong>in</strong>e <strong>in</strong> hypothalamic KiSS-1 andGPR54 mRNA levels at 12 and 24 h which preceded thereduction <strong>in</strong> GnRH gene expression at 48 h (Luqueet al. 2007). Moreover, lept<strong>in</strong> and not IGF-I or <strong>in</strong>sul<strong>in</strong>stimulated KiSS-1 expression <strong>in</strong> mouse hypothalamiccell l<strong>in</strong>e N6. Furthermore, hypothalamic KiSS-1 expressionwas decreased <strong>in</strong> NPY null mice and this wasreversed by NPY adm<strong>in</strong>istration (Luque et al. 2007).These reports support the idea that lept<strong>in</strong> and NPY arekey mediators of metabolic regulation of the hypothalamicKiSS-1 system and subsequent GnRH release.Lents et al. (2008) recently demonstrated that adm<strong>in</strong>istrationof kisspept<strong>in</strong> <strong>in</strong>to the lateral ventricle of thebra<strong>in</strong> stimulated LH and follicle stimulat<strong>in</strong>g hormone(FSH) secretion but not GH <strong>in</strong> the pre-pubertal gilt,Ó 2008 No claim to orig<strong>in</strong>al government works


328 CR Barb, GJ Hausman and CA LentsFig. 2. Schematic illustration of lept<strong>in</strong> coord<strong>in</strong>ation of energy homeostasisand neuroendocr<strong>in</strong>e function: Lept<strong>in</strong> is secreted <strong>in</strong> response tochanges <strong>in</strong> energy balance. Lept<strong>in</strong> acts on the hypothalamus to controlfood <strong>in</strong>take, reproduction and, by suppress<strong>in</strong>g expression of neuropeptideY (NPY) and agouti related peptide (AGRP) and upregulationof proopiomelanocort<strong>in</strong> (POMC), a-melanocyte stimulat<strong>in</strong>g hormone(a-MSH) and kisspept<strong>in</strong> (KiSS)while peripheral adm<strong>in</strong>istration of kisspept<strong>in</strong> <strong>in</strong>creasedserum concentrations of LH but not FSH or GH. Thesedata illustrate that kisspept<strong>in</strong> can activate the bra<strong>in</strong>pituitaryaxis and may be part of an importantmechanism regulat<strong>in</strong>g activation of the GnRH ⁄ LHrelease and <strong>in</strong>itiat<strong>in</strong>g onset of puberty <strong>in</strong> sw<strong>in</strong>e. Moreoverit is tempt<strong>in</strong>g to speculate that the kisspept<strong>in</strong>neuronal pathway may play a role <strong>in</strong> <strong>in</strong>terfac<strong>in</strong>gmetabolic status of the post-partum sow with theGnRH ⁄ gonadotropic axis and hence may affect thepost-wean<strong>in</strong>g fertility of the sow.In conclusion evidence presented supports the conceptthat metabolic signals, which reflect changes <strong>in</strong> energybalance, affect both the hypothalamic GnRH ⁄ LH pulsegenerator and appetite. Changes <strong>in</strong> fat metabolism <strong>in</strong>response to changes <strong>in</strong> feed <strong>in</strong>take and energy balancealter adipocyte function and circulat<strong>in</strong>g concentrationsof IGF-I and lept<strong>in</strong>. A preponderance of evidence citedabove suggests that lept<strong>in</strong> may serve as the primarymetabolic signal <strong>in</strong>teract<strong>in</strong>g with neuropeptides such askisspept<strong>in</strong> and NPY that l<strong>in</strong>k energy status with theneuroendocr<strong>in</strong>e axis and subsequent reproduction(Fig. 2).ReferencesAherne FX, Kirkwood RN, 1985: Nutrition and sow prolificacy.J Reprod Fertil 33, 169–183.Ahima RS, Qi Y, S<strong>in</strong>ghal NS, Jackson MB, Scherer PE, 2006:Bra<strong>in</strong> adipocytok<strong>in</strong>e action and metabolic regulation. Diabetes55, S145–S154.Almeida FR, Mao J, Novak S, Cosgrove JR, Foxcroft GR,2001: Effects of different patterns of feed restriction and<strong>in</strong>sul<strong>in</strong> treatment dur<strong>in</strong>g the luteal phase on reproductive,metabolic, and endocr<strong>in</strong>e parameters <strong>in</strong> cyclic gilts. J AnimSci 79, 200–212.Angell CA, Tubbs RC, Moore AB, Barb CR, Cox NM, 1996:Depressed lute<strong>in</strong>iz<strong>in</strong>g hormone response to estradiol <strong>in</strong> vivoand gonadotrop<strong>in</strong>-releas<strong>in</strong>g hormone <strong>in</strong> vitro <strong>in</strong> experimentallydiabetic sw<strong>in</strong>e. Domest Anim Endocr<strong>in</strong>ol 13, 453–463.Arregu<strong>in</strong>-Arevalo JA, Lents CA, Farmerie TA, Nett TM, ClayCM, 2007: KiSS-1 peptide <strong>in</strong>duces release of LH by a directeffect on the hypothalamus of ovariectomized ewes. AnimReprod Sci 101, 265–275.Barb CR, Krael<strong>in</strong>g RR, 2004: Role of lept<strong>in</strong> <strong>in</strong> the regulationof gonadotrop<strong>in</strong> secretion <strong>in</strong> farm animals. Anim ReprodSci 82–83, 155–167.Barb CR, Chang W-C, Lesh<strong>in</strong> LS, Rampacek GB, Krael<strong>in</strong>gRR, 1994: Opioid modulation of gonadotrop<strong>in</strong> releas<strong>in</strong>ghormone release from the hypothalamic preoptic area <strong>in</strong> thepig. Domest Anim Endocr<strong>in</strong>ol 11, 375–382.Barb CR, Krael<strong>in</strong>g RR, Rampacek GB, Dove CR, 1997:Metabolic changes dur<strong>in</strong>g the transition from the fed to theacute feed-deprived state <strong>in</strong> prepuberal and mature gilts. JAnim Sci 75, 781–789.Barb CR, Yan X, Aza<strong>in</strong> MJ, Krael<strong>in</strong>g RR, Rampacek GB,Ramsay TG, 1998: Recomb<strong>in</strong>ant porc<strong>in</strong>e lept<strong>in</strong> reduces feed<strong>in</strong>take and stimulates growth hormone secretion <strong>in</strong> sw<strong>in</strong>e.Domest Anim Endocr<strong>in</strong>ol 15, 77–86.Barb CR, Barrett JB, Krael<strong>in</strong>g RR, Rampacek GB, 1999: Roleof lept<strong>in</strong> <strong>in</strong> modulat<strong>in</strong>g neuroendocr<strong>in</strong>e function: a metabolicl<strong>in</strong>k between the bra<strong>in</strong>-pituitary and adipose tissue.Reprod Domest Anim 34, 111–125.Barb CR, Barrett JB, Krael<strong>in</strong>g RR, Rampacek GB, 2001a:Serum lept<strong>in</strong> concentrations, lute<strong>in</strong>iz<strong>in</strong>g hormone andgrowth hormone secretion dur<strong>in</strong>g feed and metabolic fuelrestriction <strong>in</strong> the prepuberal gilt. Domest Anim Endocr<strong>in</strong>ol20, 47–63.Barb CR, Hausman GJ, Houseknecht KL, 2001b: Biology oflept<strong>in</strong> <strong>in</strong> the pig. Domest Anim Endocr<strong>in</strong>ol 21, 297–317.Barb CR, Krael<strong>in</strong>g RR, Rampacek GB, 2002: Metabolicregulation of the neuroendocr<strong>in</strong>e axis <strong>in</strong> pigs. <strong>Reproduction</strong>Suppl 59, 203–217.Barb CR, Barrett JB, Krael<strong>in</strong>g RR, 2004: Role of lept<strong>in</strong> <strong>in</strong>modulat<strong>in</strong>g the hypothalamic-pituitary axis <strong>in</strong> the pig.Domest Anim Endocr<strong>in</strong>ol 26, 201–214.Barb CR, Hausman GJ, Ramsay TG, 2006a: Lept<strong>in</strong> <strong>in</strong> farmanimals. <strong>in</strong>: Castracane VD, Henson MC (eds), Lept<strong>in</strong>.Endocr<strong>in</strong>ology Update Series. Spr<strong>in</strong>ger Publish<strong>in</strong>g Group,New York, NY, pp. 263–308.Barb CR, Krael<strong>in</strong>g RR, Rampacek GB, Hausman GJ, 2006b:The role of neuropeptide Y and <strong>in</strong>teraction with lept<strong>in</strong> <strong>in</strong>regulat<strong>in</strong>g feed <strong>in</strong>take and lute<strong>in</strong>iz<strong>in</strong>g hormone and growthhormone secretion <strong>in</strong> the pig. <strong>Reproduction</strong> 131, 1127–1135.Booth PJ, Cosgrove JR, Foxcroft GR, 1996: Endocr<strong>in</strong>e andmetabolic responses to realimentation <strong>in</strong> feed-restrictedprepuberal gilts: associations among gonadotrop<strong>in</strong>s, metabolichormones, glucose, and uteroovarian development. JAnim Sci 74, 840–848.van den Brand H, Dieleman SJ, Soede NM, Kemp B, 2000:Dietary energy source at two feed<strong>in</strong>g levels dur<strong>in</strong>g lactationof primiparous sows: I. Effects on glucose, <strong>in</strong>sul<strong>in</strong>, andlute<strong>in</strong>iz<strong>in</strong>g hormone and on follicle development, wean<strong>in</strong>gto-estrus<strong>in</strong>terval, and ovulation rate. J Anim Sci 78, 396–404.van den Brand H, Prunier A, Soede NM, Kemp B, 2001: Inprimiparous sows, plasma <strong>in</strong>sul<strong>in</strong>-like growth factor-I canbe affected by lactational feed <strong>in</strong>take and dietary energysource and is associated with lute<strong>in</strong>iz<strong>in</strong>g hormone. ReprodNutr Dev 41, 27–39.Brendemuhl JH, Lewis AJ, Poe ER Jr, 1989: Influence ofenergy and prote<strong>in</strong> <strong>in</strong>take dur<strong>in</strong>g lactation on body compositionof primiparous sows. J Anim Sci 67, 1478–1488.Campfield LA, Smith FJ, Burn P, 1996: The OB prote<strong>in</strong>(lept<strong>in</strong>) pathway – a l<strong>in</strong>k between adipose tissue mass andcentral neural networks. Horm Metab Res 28, 619–632.Castellano JM, Navarro VM, Fernandez-Fernandez R,Castano JP, Malagon MM, Aguilar E, Dieguez C, MagniP, P<strong>in</strong>illa L, Tena-Sempere M, 2006: Ontogeny andÓ 2008 No claim to orig<strong>in</strong>al government works


Energy and <strong>Reproduction</strong> <strong>in</strong> the Pig 329mechanisms of action for the stimulatory effect of kisspept<strong>in</strong>on gonadotrop<strong>in</strong>-releas<strong>in</strong>g hormone system of the rat. MolCell Endocr<strong>in</strong>ol 257–258, 75–83.Clowes EJ, Aherne FX, Foxcroft GR, Baracos VE, 2003:Selective prote<strong>in</strong> loss <strong>in</strong> lactat<strong>in</strong>g sows is associated withreduced litter growth and ovarian function. J Anim Sci 81,753–764.Cox NM, Britt JH, 1988: Pulsatile adm<strong>in</strong>istration of gonadotrop<strong>in</strong>releas<strong>in</strong>g hormone to lactat<strong>in</strong>g sows: endocr<strong>in</strong>echanges associated with <strong>in</strong>duction of fertile estrus. BiolReprod 27, 1126–1137.Crown A, Clifton DK, Ste<strong>in</strong>er RA, 2007: Neuropeptidesignal<strong>in</strong>g <strong>in</strong> the <strong>in</strong>tegration of metabolism and reproduction.Neuroendocr<strong>in</strong>ology 86, 175–182.Czaja K, Lakomy M, Sienkiewicz W, Kaleczyc J, Pidsudko Z,Barb CR, Rampacek GB, Krael<strong>in</strong>g RR, 2002: Distributionof neurons conta<strong>in</strong><strong>in</strong>g lept<strong>in</strong> receptors <strong>in</strong> the hypothalamusof the pig. Biochem Biophys Res Commun 298, 333–337.Czaja K, Krael<strong>in</strong>g RR, Barb CR, 2003: Are hypothalamicneurons transsynaptically connected to porc<strong>in</strong>e adiposetissue? Biochem Biophys Res Commun 311, 482–485.De Rensis F, Gherpelli M, Superchi P, Kirkwood RN, 2005:Relationship between backfat depth and plasma lept<strong>in</strong>dur<strong>in</strong>g lactation and sow reproductive performance afterwean<strong>in</strong>g. Anim Reprod Sci 90, 95–100.Dungan HM, Clifton DK, Ste<strong>in</strong>er RA, 2006: M<strong>in</strong>ireview:kisspept<strong>in</strong> neurons as central processors <strong>in</strong> the regulation ofgonadotrop<strong>in</strong>-releas<strong>in</strong>g hormone secretion. Endocr<strong>in</strong>ology147, 1154–1158.Estienne MJ, Harper AF, Barb CR, Aza<strong>in</strong> MJ, 2000:Concentrations of lept<strong>in</strong> <strong>in</strong> serum and milk collected fromlactat<strong>in</strong>g sows differ<strong>in</strong>g <strong>in</strong> body condition [In ProcessCitation]. Domest Anim Endocr<strong>in</strong>ol 19, 275–280.Estienne MJ, Harper AF, Koz<strong>in</strong>k DM, Knight JW, 2003:Serum and milk concentrations of lept<strong>in</strong> <strong>in</strong> gilts fed a highorlow-energy diet dur<strong>in</strong>g gestation. Anim Reprod Sci 75,95–105.Frisch RE, 1984: Body fat, puberty and fertility. Biol Rev 59,161–188.Guerre-Millo M, 2004: Adipose tissue and adipok<strong>in</strong>es: forbetter or worse. Diabetes Metab 30, 13–19.Hart HA, Aza<strong>in</strong> MJ, Hausman GJ, Reeves DE, Barb CR,2007: Failure of short term feed restriction to affectlute<strong>in</strong>iz<strong>in</strong>g hormone and lept<strong>in</strong> secretion or subcutaneousadipose tissue expression of lept<strong>in</strong> <strong>in</strong> the prepuberal gilt. CanJ Anim Sci 87, 191–197.Hauner H, 2005: Secretory factors from human adipose tissueand their functional role. Proc Nutr Soc 64, 163–169.Hausman GJ, Richardson RL, 1987: Adrenergic <strong>in</strong>nervationof fetal pig adipose tissue. Histochemical and ultrastructuralstudies. Acta Anat (Basel) 130, 291–297.Hausman GJ, Poulos SP, Richardson RL, Barb CR, Andacht T,Mynatt R, Kirk H, 2006: Secreted prote<strong>in</strong>s and genes <strong>in</strong> fetaland neonatal pig adipose tissue and stromal-vascular cells.J Anim Sci 84, 1666–1681.Hausman GJ, Barb CR, Dean RG, 2007: Patterns of geneexpression <strong>in</strong> pig adipose tissue: transform<strong>in</strong>g growthfactors, <strong>in</strong>terferons, <strong>in</strong>terleuk<strong>in</strong>s and other cytok<strong>in</strong>es. JAnim Sci 85, 2445–2456.K<strong>in</strong>g RH, Mart<strong>in</strong> GB, 1989: Relationships between prote<strong>in</strong><strong>in</strong>take dur<strong>in</strong>g lactation, LH levels and oestrous activity <strong>in</strong>first-litter sows. Anim Reprod Sci 19, 283–292.K<strong>in</strong>g RH, Williams IH, 1984: The effect of nutrition on thereproductive performance of first litter sows 2. Prote<strong>in</strong> andenergy <strong>in</strong>takes dur<strong>in</strong>g lactation. Anim Prod 38, 249–256.Kojima CJ, Carroll JA, Matteri RL, Touchette KJ, Allee GL,2007: Effects of wean<strong>in</strong>g and wean<strong>in</strong>g weight on neuroendocr<strong>in</strong>eregulators of feed <strong>in</strong>take <strong>in</strong> pigs. J Anim Sci 85,2133–2139.Koketsu Y, Dial GD, Pettigrew JE, Xue J, Yang H, Lucia T,1998: Influence of lactation length and feed <strong>in</strong>take onreproductive performance and blood concentrations ofglucose, <strong>in</strong>sul<strong>in</strong> and lute<strong>in</strong>iz<strong>in</strong>g hormone <strong>in</strong> primiparoussows. Anim Reprod Sci 52, 153–163.Kotani M, Detheux M, Vandenbogaerde A, Communi D,Vanderw<strong>in</strong>den JM, Le Poul E, Brezillon S, Tyldesley R,Suarez-Huerta N, Vandeput F, Blanpa<strong>in</strong> C, Schiffmann SN,Vassart G, Parmentier M, 2001: The metastasis suppressorgene KiSS-1 encodes kisspept<strong>in</strong>s, the natural ligands of theorphan G prote<strong>in</strong>-coupled receptor GPR54. J Biol Chem276, 34631–34636.Krael<strong>in</strong>g RR, Barb CR, 1990: Hypothalamic control ofgonadotroph<strong>in</strong> and prolact<strong>in</strong> secretion <strong>in</strong> pigs. J ReprodFertil Suppl 40, 3–17.Lawrence CB, Turnbull AV, Rothwell NJ, 1999: Hypothalamiccontrol of feed<strong>in</strong>g. Curr Op<strong>in</strong> Neurobiol 9, 778–783.Lents CA, Heidorn NL, Barb CR, Ford JJ, 2008: Central andperipheral adm<strong>in</strong>istration of kisspept<strong>in</strong> activates gonadotrop<strong>in</strong>but not somatotrop<strong>in</strong> secretion <strong>in</strong> prepubertal gilts.<strong>Reproduction</strong> 135, 879–887.Lesh<strong>in</strong> LS, Barb CR, Kiser TE, Rampacek GB, Krael<strong>in</strong>g RR,1994: Growth hormone-releas<strong>in</strong>g hormone and somatostat<strong>in</strong>neurons with<strong>in</strong> the porc<strong>in</strong>e and bov<strong>in</strong>e hypothalamus.Neuroendocr<strong>in</strong>ology 59, 251–264.L<strong>in</strong> J, Barb CR, Matteri RL, Krael<strong>in</strong>g RR, Chen X,Me<strong>in</strong>ersmann RJ, Rampacek GB, 2000: Long form lept<strong>in</strong>receptor mRNA expression <strong>in</strong> the bra<strong>in</strong>, pituitary, andother tissues <strong>in</strong> the pig. Domest Anim Endocr<strong>in</strong>ol 19,53–61.L<strong>in</strong> J, Barb CR, Krael<strong>in</strong>g RR, Rampacek GB, 2001: Developmentalchanges <strong>in</strong> the long form lept<strong>in</strong> receptor andrelated neuropeptide gene expression <strong>in</strong> the pig bra<strong>in</strong>. BiolReprod 64, 1614–1618.Loftus TM, 1999: An adipocyte-central nervous systemregulatory loop <strong>in</strong> the control of adipose homeostasis. CellDev Biol 10, 11–18.Luque RM, K<strong>in</strong>emam RD, Tena-Sempere M, 2007: Regulationof hypothalamic expression of KiSS-1 and GPR54genes by metabolic factors: analyses us<strong>in</strong>g mouse modelsand a cell l<strong>in</strong>e. Endocr<strong>in</strong>ology 148, 4601–4611.Lutz JB, Rampacek GB, Krael<strong>in</strong>g RR, P<strong>in</strong>kert CA, 1984:Serum lute<strong>in</strong>iz<strong>in</strong>g hormone and estrogen profiles beforepuberty <strong>in</strong> the gilt. J Anim Sci 58, 686–691.Mao J, Zak LJ, Cosgrove JR, Shostak S, Foxcroft GR, 1999:Reproductive, metabolic, and endocr<strong>in</strong>e responses to feedrestriction and GnRH treatment <strong>in</strong> primiparous, lactat<strong>in</strong>gsows. J Anim Sci 77, 724–735.Matteri RL, 2001: Overview of central targets for appetiteregulation. J Anim Sci 79, E148–E158.Muller EE, Locatelli V, Cocchi D, 1999: Neuroendocr<strong>in</strong>econtrol of growth hormone secretion. Physiol Rev 79, 511–607.NRC, 1998: Nutrient Requirements <strong>in</strong> Sw<strong>in</strong>e. National AcademyPress, Wash<strong>in</strong>gton, DC.Ohtaki T, Sh<strong>in</strong>tani Y, Honda S, Matsumoto H, Hori A,Kanehashi K, Terao Y, Kumano S, Takatsu Y, Masuda Y,Ishibashi Y, Watanabe T, Asada M, Yamada T, Suenaga M,Kitada C, Usuki S, Kurokawa T, Onda H, Nishimura O,Fuj<strong>in</strong>o M, 2001: Metastasis suppressor gene KiSS-1 encodespeptide ligand of a G-prote<strong>in</strong>-coupled receptor. Nature 411,613–617.Press<strong>in</strong>g A, Dial GD, Esbenshade KL, Stroud CM, 1992:Hourly adm<strong>in</strong>istration of GnRH to prepubertal gilts:endocr<strong>in</strong>e and ovulatory responses from 70 to 190 days ofage. J Anim Sci 70, 232–242.Prunier A, Quesnel H, 2000: Nutritional <strong>in</strong>fluences on thehormonal control of reproduction <strong>in</strong> female pigs. LivestProd Sci 63, 1–16.Ó 2008 No claim to orig<strong>in</strong>al government works


330 CR Barb, GJ Hausman and CA LentsPrunier A, Dourmad JY, Etienne M, 1993: Feed<strong>in</strong>g level,metabolic parameters and reproductive performance ofprimiparous sows. Livest Prod Sci 37, 185–196.Qian H, Barb CR, Compton MM, Hausman GJ, Aza<strong>in</strong> MJ,Krael<strong>in</strong>g RR, Baile CA, 1999: Lept<strong>in</strong> mRNA expression andserum lept<strong>in</strong> concentrations as <strong>in</strong>fluenced by age, weight andestradiol <strong>in</strong> pigs. Domest Anim Endocr<strong>in</strong>ol 16, 135–143.Quesnel H, Prunier A, 1998: Effect of <strong>in</strong>sul<strong>in</strong> adm<strong>in</strong>istrationbefore wean<strong>in</strong>g on reproductive performance <strong>in</strong> feedrestricted primiparous sows. Anim Reprod Sci 51, 119–129.Quesnel H, Pasquier A, Mounier AM, Louveau I, Prunier A,1998: Influence of feed restriction <strong>in</strong> primiparous lactat<strong>in</strong>gsows on body condition and metabolic parameters. ReprodNutr Dev 38, 261–274.Quesnel H, Mejia-Guadarrama CA, Dourmad JY, Farmer C,Prunier A, 2005: Dietary prote<strong>in</strong> restriction dur<strong>in</strong>g lactation<strong>in</strong> primiparous sows with different live weights at farrow<strong>in</strong>g:I. Consequences on sow metabolic status and litter growth.Reprod Nutr Dev 45, 39–56.Rich N, Reyes P, Reap L, Goswami R, Fraley GS, 2007: Sexdifferences <strong>in</strong> the effect of prepubertal GALP <strong>in</strong>fusion ongrowth, metabolism and LH secretion. Physiol Behav 92,814–823.Schwartz MW, Seeley RJ, Campfield LA, Burn P, Bask<strong>in</strong> DG,1996: Identification of targets of lept<strong>in</strong> action <strong>in</strong> rathypothalamus. J Cl<strong>in</strong> Invest 98, 1101–1106.Shahab M, Mastronardi C, Sem<strong>in</strong>ara SB, Crowley WF, OjedaSR, Plant TM, 2005: Increased hypothalamic GPR54signal<strong>in</strong>g: a potential mechanism for <strong>in</strong>itiation of puberty<strong>in</strong> primates. Proc Natl Acad Sci U S A 102, 2129–2134.Shields RG, Mahan DC, Maxson PF, 1985: Effect of dietarygestation and lactation prote<strong>in</strong> levels on reproductionperformance and body composition of first litter femalesw<strong>in</strong>e. J Anim Sci 60, 179–189.Smith JT, Clarke IJ, 2007: Kisspept<strong>in</strong> expression <strong>in</strong> the bra<strong>in</strong>:catalyst for the <strong>in</strong>itiation of puberty. Rev Endocr MetabDisord 8, 1–9.Smith JT, Dungan HM, Stoll EA, Gottsch ML, Braun RE,Eacker SM, Clifton DK, Ste<strong>in</strong>er RA, 2005: Differentialregulation of KiSS-1 mRNA expression by sex steroids<strong>in</strong> the bra<strong>in</strong> of the male mouse. Endocr<strong>in</strong>ology 146, 2976–2984.Smith JT, Clifton DK, Ste<strong>in</strong>er RA, 2006: Regulation of theneuroendocr<strong>in</strong>e reproductive axis by kisspept<strong>in</strong>-GPR54signal<strong>in</strong>g. <strong>Reproduction</strong> 131, 623–630.Spurlock ME, Frank GR, Cornelius SG, Ji S, Willis GM,Bidwell CA, 1998: Obese gene expression <strong>in</strong> porc<strong>in</strong>e adiposetissue is reduced by food deprivation but not by ma<strong>in</strong>tenanceor subma<strong>in</strong>tenance <strong>in</strong>take. J Nutr 128, 677–682.Tak<strong>in</strong>o T, Koshikawa N, Miyamori H, Tanaka M, Sasaki T,Okada Y, Seiki M, Sato H, 2003: Cleavage of metastasissuppressor gene product KiSS-1 prote<strong>in</strong> ⁄ metast<strong>in</strong> by matrixmetalloprote<strong>in</strong>ases. Oncogene 22, 4617–4626.Tena-Sempere M, 2006a: GPR54 and kisspept<strong>in</strong> <strong>in</strong> reproduction.Hum Reprod Update 12, 631–639.Tena-Sempere M, 2006b: KiSS-1 and reproduction: focus onits role <strong>in</strong> the metabolic regulation of fertility. Neuroendocr<strong>in</strong>ology83, 275–281.Tena-Sempere M, 2006c: The roles of kisspept<strong>in</strong>s andG prote<strong>in</strong>-coupled receptor-54 <strong>in</strong> pubertal development.Curr Op<strong>in</strong> Pediatr 18, 442–447.Tokach MD, Pettigrew JE, Dial GD, Wheaton JE, CrookerBA, Johnston LJ, 1992: Characterization of lute<strong>in</strong>iz<strong>in</strong>ghormone secretion <strong>in</strong> the primiparous, lactat<strong>in</strong>g sow: relationshipto blood metabolites and return-to-estrus <strong>in</strong>terval.J Anim Sci 70, 2195–2201.Tummaruk P, Lundeheim N, E<strong>in</strong>arsson S, Dal<strong>in</strong> AM, 2001:Effect of birth litter size, birth parity number, growth rate,backfat thickness and age at first mat<strong>in</strong>g of gilts on theirreproductive performance as sows. Anim Reprod Sci 66,225–237.Wade GN, Schneider JE, Li HY, 1996: Control of fertility bymetabolic cues. Am J Physiol 270, E1–E19.Weldon WC, Lewis AJ, Louis GF, Kovar JL, Giesemann MA,Miller PS, 1994: Postpartum hypophagia <strong>in</strong> primiparoussows: I. Effects of gestation feed<strong>in</strong>g level on feed <strong>in</strong>take,feed<strong>in</strong>g behavior, and plasma metabolite concentrationsdur<strong>in</strong>g lactation. J Anim Sci 72, 387–394.Whisnant CS, Harrell RJ, 2002: Effect of short-term feedrestriction and refeed<strong>in</strong>g on serum concentrations of lept<strong>in</strong>,lute<strong>in</strong>iz<strong>in</strong>g hormone and <strong>in</strong>sul<strong>in</strong> <strong>in</strong> ovariectomized gilts.Domest Anim Endocr<strong>in</strong>ol 22, 73–80.Whitley NC, Payne DB, Zhang H, Cox NM, 1998: The<strong>in</strong>fluence of <strong>in</strong>sul<strong>in</strong> adm<strong>in</strong>istration after wean<strong>in</strong>g the firstlitter on ovulation rate and embryo survival <strong>in</strong> sows.Theriogenology 50, 479–485.Whittemore CT, 1996: Nutrition reproduction <strong>in</strong>teractions <strong>in</strong>primiparous sows. Livest Prod Sci 46, 65–83.Williams LT, 1989: Signal transduction by the platelet-derivedgrowth factor receptor. Science 243, 1564–1570.Author’s address (for correspondence): C Richard Barb, USDA ⁄ ARS,Richard B, Russell Agriculture Research Center, PO Box 5677,Athens, GA 30604, USA. E-mail: richard.barb@ars.usda.govConflict of <strong>in</strong>terest: This research was supported by USDA funds(C.R.B.; G.J.H.) and State funds allocated to the Georgia AgriculturalExperiment Station (C.A.L.) and <strong>in</strong> part by a National ResearchInitiative Competitive Grant (no. 2005-35023-15949 and 2005-35203-16852) from the USDA Cooperative State Research, Education, andExtension Service awarded to C.A.L. Mention of trade name,proprietary product, or specific equipment does not constitute aguarantee or warranty by the US Department of Agriculture or TheUniversity of Georgia and does not imply its approval to the exclusionof other products which may be suitable. The authors declare thatthere is no conflict of <strong>in</strong>terest that would prejudice their impartiality ofthis scientific work.Ó 2008 No claim to orig<strong>in</strong>al government works


Reprod Dom Anim 43 (Suppl. 2), 331–337 (2008); doi: 10.1111/j.1439-0531.2008.01181.xISSN 0936-6768Somatic Cell Nuclear Transfer <strong>in</strong> HorsesCesare Galli 1,2 , Ir<strong>in</strong>a Lagut<strong>in</strong>a 1 , Roberto Duchi 1 , Silvia Colleoni 1 and Giovanna Lazzari 11 Laboratorio di Tecnologie della Riproduzione, Istituto Sperimentale Italiano Lazzaro Spallanzani, CIZ srl, Cremona, Italy; 2 Dip. Cl<strong>in</strong>icoVeter<strong>in</strong>ario, Universita` di Bologna, Cremona, ItalyContentsThe clon<strong>in</strong>g of equids was achieved <strong>in</strong> 2003, several years afterthe birth of Dolly the sheep and also after the clon<strong>in</strong>g ofnumerous other laboratory and farm animal species. The delaywas because of the limited development <strong>in</strong> the horse of moreclassical-assisted reproductive techniques required for successfulclon<strong>in</strong>g, such as oocyte maturation and <strong>in</strong> vitro embryoproduction. When these technologies were developed, theapplication of clon<strong>in</strong>g also became possible and cloned horseoffspr<strong>in</strong>g were obta<strong>in</strong>ed. This review summarizes the ma<strong>in</strong>technical procedures that are required for clon<strong>in</strong>g equids andthe present status of this technique. The first step is competentoocyte maturation, this is followed by oocyte enucleation andreconstruction, us<strong>in</strong>g either zona-enclosed or zona-freeoocytes, by efficient activation to allow high cleavage ratesand f<strong>in</strong>ally by a suitable <strong>in</strong> vitro embryo culture technique.Clon<strong>in</strong>g of the first equid, a mule, was achieved us<strong>in</strong>g an<strong>in</strong> vivo-matured oocytes and immediate transfer of the reconstructedembryo, i.e. at the one cell stage, to the recipientoviduct. In contrast, the first horse offspr<strong>in</strong>g was obta<strong>in</strong>edus<strong>in</strong>g a complete <strong>in</strong> vitro procedure from oocyte maturation toembryo culture to the blastocyst stage, followed by nonsurgicaltransfer. Later studies on equ<strong>in</strong>e clon<strong>in</strong>g report highefficiency relative to that for other species. Cloned equidoffspr<strong>in</strong>g reported to date appear to be normal and those thathave reached puberty have been confirmed to be fertile. Insummary, horse clon<strong>in</strong>g is now a reproducible technique thatoffers the opportunity to preserve valuable genetics andnotably to generate copies of castrated champions andtherefore, offspr<strong>in</strong>g from those champions that would beimpossible to obta<strong>in</strong> otherwise.IntroductionClon<strong>in</strong>g mammals by somatic cell nuclear transfer(SCNT) has become a common technology <strong>in</strong> recentyears, follow<strong>in</strong>g the work of Wilmut and co-workers(Wilmut et al. 1997). Today, a decade later, mostdomestic and laboratory species have been cloned,<strong>in</strong>clud<strong>in</strong>g the horse (Campbell et al. 2007). Initially, <strong>in</strong>farm animals, the application of SCNT was focussed onthe multiplication of superior genotypes of high geneticvalue. Very soon, yet, it became evident that thetechnique could offer, ma<strong>in</strong>ly because of its peculiarfailures, an important biological model for basicresearch as well as a wealth of opportunities forbiomedical research (Hochedl<strong>in</strong>ger and Jaenisch 2006).It is <strong>in</strong> this latter field that most of the potentialapplications lie today. They range from the creation ofgenetically modified large animals to the derivation ofgenetically matched embryonic stem cells derived fromcloned embryos (Rideout et al. 2002).With regard to Equids, the birth of three mule foalscloned from foetal cells (Woods et al. 2003) and onehorse foal cloned from adult somatic cells (Galli et al.2003a) were reported <strong>in</strong> 2003 and additional cloned foalshave also been documented (Lagut<strong>in</strong>a et al. 2005;H<strong>in</strong>richs et al. 2006, 2007). The relatively late applicationof SCNT to horse reproduction is a consequence ofthe limited <strong>in</strong>formation available <strong>in</strong> the publishedstudies on assisted reproductive techniques <strong>in</strong> Equids,<strong>in</strong> particular oocyte maturation, activation and <strong>in</strong> vitroculture of early pre-implantation embryos. The clon<strong>in</strong>gof the mule by Woods and co-workers (Woods et al.2003) relied on the use of <strong>in</strong> vivo matured oocytes thatwere transferred to the oviducts of recipient maresimmediately after nuclear transfer and activation. Incontrast, Galli (Galli et al. 2003a), Lagut<strong>in</strong>a (Lagut<strong>in</strong>aet al. 2005) and H<strong>in</strong>richs (H<strong>in</strong>richs et al. 2006, 2007)have all reported clon<strong>in</strong>g of horse embryos up to theblastocyst stage carried out completely <strong>in</strong> vitro.In general, few studies are available <strong>in</strong> the publishedstudies on equ<strong>in</strong>e <strong>in</strong> vitro embryo production and it isonly recently that reports have been published oncomplete <strong>in</strong> vitro production of equ<strong>in</strong>e pre-implantationembryos by means of <strong>in</strong> vitro oocyte maturation,fertilization by <strong>in</strong>tracytoplasmic sperm <strong>in</strong>jection (ICSI)and <strong>in</strong> vitro culture (Galli et al. 2002a; Lazzari et al.2002a; Choi et al. 2004; H<strong>in</strong>richs et al. 2005). Theselatter reports demonstrate the feasibility of apply<strong>in</strong>g acompletely <strong>in</strong> vitro procedure to obta<strong>in</strong> horse ICSIblastocysts able to establish a pregnancy (Galli et al.2002a,b; Lazzari et al. 2002a) and develop <strong>in</strong>to live termoffspr<strong>in</strong>g (Galli et al. 2007).One additional reason for the limited development ofassisted reproduction techniques <strong>in</strong> Equids is the lack of<strong>in</strong>terest demonstrated by the horse <strong>in</strong>dustry. SCNT, <strong>in</strong>particular, has received a sceptical welcome <strong>in</strong> mostequestrian discipl<strong>in</strong>es. Some of the mayor players <strong>in</strong> thefield (such as the American Quarter Horse Associationand the Jockey Club) are opposed to the use of clon<strong>in</strong>gand will not register offspr<strong>in</strong>g produced by this technique(Church 2006).The ref<strong>in</strong>ement of the <strong>in</strong> vitro culture conditionssuitable for equ<strong>in</strong>e oocyte maturation and embryogrowth (H<strong>in</strong>richs et al. 2005; Galli et al. 2007), thedevelopment of adequate horse oocyte activation protocols(Lazzari et al. 2002b) and the application of apiezoelectric device (Westhus<strong>in</strong> et al. 2003) or zona-freemanipulation for embryo reconstruction (Booth et al.2001; Oback et al. 2003; Lagut<strong>in</strong>a et al. 2007), have allbeen the fundamental steps for def<strong>in</strong><strong>in</strong>g a successful<strong>in</strong> vitro procedure for SCNT <strong>in</strong> the horse.Although nuclear transfer, <strong>in</strong> general, and SCNT, <strong>in</strong>particular, are rout<strong>in</strong>e techniques <strong>in</strong> several laboratoriesworldwide, both <strong>in</strong> farm animals and <strong>in</strong> laboratoryspecies, they are very demand<strong>in</strong>g. The major hurdle <strong>in</strong>Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


332 C Galli, I Lagut<strong>in</strong>a, R Duchi, S Colleoni and G Lazzariclon<strong>in</strong>g, and not only for beg<strong>in</strong>ners is to master all thelengthy and complicated procedures <strong>in</strong>volved <strong>in</strong> SCNTto a high level of efficiency and reproducibility <strong>in</strong> orderto obta<strong>in</strong> scientifically mean<strong>in</strong>gful results (Galli et al.2003b; Ritchie 2006).In pr<strong>in</strong>ciple, all techniques used today <strong>in</strong> mammalsare based on those described by Willadsen (Willadsen1986). Basically, there is a requirement for maturedoocytes, perhaps <strong>in</strong> large numbers and of good quality,the oocytes then need to be enucleated before the donorcell is fused or <strong>in</strong>jected <strong>in</strong> the enucleated oocyte andf<strong>in</strong>ally the reconstructed embryo must be activated. Atthis po<strong>in</strong>t, embryos can be transferred to the oviduct ofrecipients for further development. Yet, <strong>in</strong> large animals,it is preferable to culture <strong>in</strong> vitro the clonedembryos up to the blastocyst stage, so that a nonsurgicaluter<strong>in</strong>e transfer can be performed (Galli et al.2003b).Source of OocytesOocytes can be harvested from the ovaries of live donorsby ovum pick up (OPU) or from the ovaries ofslaughtered mares (Zhang et al. 1989), the latter be<strong>in</strong>gthe only economically susta<strong>in</strong>able source of oocytes forSCNT. On average, it is possible to recover three to fouroocytes per abattoir ovary when compared with oocyteretrieval from live donors by OPU, <strong>in</strong> which, therecovery is slightly more variable and can be from threeto six oocytes per session of OPU (Galli et al. 2007). Thematuration rate of horse oocytes is also quite variable,averag<strong>in</strong>g between 25% and 70% <strong>in</strong> published studies(Carneiro et al. 2001; Dell’Aquila et al. 2001, 2003;Bogh et al. 2002; Choi et al. 2002; Lorenzo et al. 2002).The recovery of oocytes from abattoir horse ovariesrequires <strong>in</strong>cision and scrap<strong>in</strong>g of the follicle wall with acurette and extensive wash<strong>in</strong>g to detach the cumulusoocytecomplexes (COCs). Another <strong>in</strong>terest<strong>in</strong>g aspect,peculiar to the horse, is the frequent collection ofoocytes with expanded cumulus; <strong>in</strong> our laboratory thisaccounts for approximately a third of the recoveredCOCs. In other species such as rum<strong>in</strong>ants and pigs, anexpanded cumulus is l<strong>in</strong>ked to collection from atreticfollicles and these oocytes are generally discardedimmediately because of their extremely low developmentalcapacity (de Loos et al. 1989). In the horse, yet,oocytes with an expanded cumulus mature normally andhave normal developmental competence. Table 1 presentsthe data from a large study conducted <strong>in</strong> ourlaboratory to measure the efficiency of oocyte collectionfrom abattoir ovaries and the expected maturation ratefrom compact and expanded COCs. The data <strong>in</strong>dicatethat each ovary conta<strong>in</strong>s 5.3 follicles on an average witha diameter above 5 mm that are suitable for oocytecollection. If these follicles are scraped and washed, therecovery is 3.8 oocytes correspond<strong>in</strong>g to approximately70% efficiency. The need to scrape the follicles derivesfrom the tight connections between the cumulus and themembrana granulosa and between the latter and thefollicle wall. Once the oocytes are collected and selectedon the basis of cumulus morphology, they are transferredto maturation medium and allowed to mature for24 h. In our culture conditions, as shown <strong>in</strong> Table 1(Galli et al. 2007), the maturation rate ranges from51.1% to 60% for compact and expanded COCs,respectively. This difference is statistically significantand <strong>in</strong> agreement with other studies (H<strong>in</strong>richs andWilliams 1997; H<strong>in</strong>richs and Schmidt 2000), <strong>in</strong> which,oocytes with expanded cumulus were found to be morecapable to complete maturation than were oocytes witha compact cumulus. Yet, the ability of oocytes withexpanded cumuli to develop to the blastocyst stage wasnot statistically different from oocytes with a compactcumulus (see Table 2). In calculat<strong>in</strong>g the maturationrate, the authors <strong>in</strong>cluded the large number of degenerat<strong>in</strong>goocytes that are found after IVM. In fact, at thetime of collection, the oocytes that will degeneratedur<strong>in</strong>g maturation cannot be identified; this is anotherpeculiarity of the equ<strong>in</strong>e species when oocytes arecollected from abattoir ovaries. At the end of maturation,after the removal of the surround<strong>in</strong>g somatic cells,degenerated oocytes are easily detected. If these oocytesthat degenerate <strong>in</strong> culture are not <strong>in</strong>cluded <strong>in</strong> thecalculation, the percentage of mature oocytes <strong>in</strong>creasesconsiderably <strong>in</strong>to the range of 70–85% and is comparableto that obta<strong>in</strong>ed from OPU oocytes from livemares. As no lysis is observed with OPU oocytes, thisf<strong>in</strong>d<strong>in</strong>g implies that the postmortem modificationsoccurr<strong>in</strong>g <strong>in</strong> the large equ<strong>in</strong>e ovaries are responsiblefor the degeneration of abattoir oocytes. Indeed, whenequ<strong>in</strong>e oocytes were placed <strong>in</strong>to maturation immediatelyafter slaughter, the rate of maturation was higher andthe rate of degeneration lower than those for oocytesrecovered after transport of ovaries to the laboratoryTable 2. Effect of cumulus morphology on the developmental competenceof equ<strong>in</strong>e oocytes matured <strong>in</strong> vitro, fertilized by ICSI andcultured <strong>in</strong> vivo <strong>in</strong> the sheep oviduct (Galli et al. 2007)CumulusmorphologyNo. <strong>in</strong>jected(MII)No.cleavedCleavagerate (%)No. ofblastocysts% Blastocyst ⁄MII<strong>in</strong>jectedCompact 150 110 73.3 a 43 28.6 aExpanded 73 43 58.9 b 13 17.8 aChi square test: values with<strong>in</strong> columns with different letters differ (p < 0.05).Table 1. Maturation competence of horse oocytes derived from expanded or compact COCs (Galli et al. 2007)No. ofovariesNo. of follicles(per ovary)No. of COCsexpanded(per ovary)No. of COCscompact(per ovary)No. of COCs matured after IVMNo. of COCs degenerated after IVMExpanded (%) Compact (%) Expanded (%) Compact (%)603 3204 (5.3) 590 (1) 1672 (2.8) 354 (60.0) a 855 (51.1) b 177 (30.0) 558 (33.4)Chi square test: values with different letters differ (p < 0.05).Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Clon<strong>in</strong>g <strong>in</strong> Horses 333Table 3. Effect of maturation media on maturation, cleavage rate andICSI embryo development (Galli et al. 2007)MaturationmediumNo. of No. ofoocytes degeneratedNo. ofmetaphaseII (%)No. of<strong>in</strong>jectedNo. ofcleaved(% of<strong>in</strong>jected)No. ofblastocysts(% of<strong>in</strong>jected)TCM 199 434 105 205 (47.2 a) 191 111 (58.1 a) 23 (12.0 a)DMEM ⁄ F12 338 71 159 (45.6 a) 140 108 (77.1 b) 37 (26.4 b)Replicates = 6.Chi square test. Numbers with<strong>in</strong> columns with different letters differ (p < 0.05).Table 4. Effect of maturation media on maturation, cleavage rate andNT embryo development (Galli et al. 2007)MaturationmediumNo. ofoocytesNo. oflysedNo. ofmetaphaseII (%)No. ofNTembryosNo. ofcleaved(% of NT)No. ofblastocysts(% of NT)TCM 199 164 35 64 (39.0 a) 41 38 (92.7 a) 4 (9.77 a)DMEM ⁄ F12 166 35 67 (40.4 a) 47 46 (97.9 a) 13 (27.7 b)Chi square test. Numbers with<strong>in</strong> columns with different letters differ (p < 0.05).(H<strong>in</strong>richs et al. 2005). In that study, high rates ofdegeneration <strong>in</strong> culture were seen only <strong>in</strong> oocytes with acompact cumulus; evaluation of oocyte chromat<strong>in</strong>configurations led to the conclusion that these oocyteswere juvenile and that their chromat<strong>in</strong> was damageddur<strong>in</strong>g storage with<strong>in</strong> the ovary.It is known that <strong>in</strong> vitro maturation conditionsdramatically <strong>in</strong>fluence the development of fertilizedoocytes to the blastocyst stage. Different media are usedfor maturation of equ<strong>in</strong>e oocytes, although the mostcommon rema<strong>in</strong>s medium TCM199, probably becauseof its widespread use for bov<strong>in</strong>e oocyte maturation. Inthe last 2 years, the authors have tested other media<strong>in</strong>clud<strong>in</strong>g DMEM-F12 supplemented with 15% serumreplacement (Invitrogen sri, San Giuliano Milanese,Italy) <strong>in</strong>stead of TCM 199 supplemented with 10%foetal calf serum (FCS). Hormones and growth factorswere added to both media as reported earlier (Lagut<strong>in</strong>aet al. 2005). We found a clear advantage <strong>in</strong> the use of aDMEM-F12 based medium not only on cleavage ratebut also on embryo development to the blastocyst stage(Table 3). Prelim<strong>in</strong>ary experiments have confirmed similarf<strong>in</strong>d<strong>in</strong>gs <strong>in</strong> horse SCNT (Table 4). This positiveeffect is evident also on blastocyst development, which issignificantly higher <strong>in</strong> the DMEM-F12 group than <strong>in</strong> theTCM199 group. This remarkable improvement <strong>in</strong>creasesthe efficiency of equ<strong>in</strong>e embryo production andmakes it possible to use a totally <strong>in</strong> vitro systemfollow<strong>in</strong>g OPU <strong>in</strong> the horse. The time required forcomplet<strong>in</strong>g IVM ranges from 22 to 24 h for expandedoocytes and 26–36 h for compacted onesSource of Donor CellsA variety of donor cells from different orig<strong>in</strong> (foetal andadult) and various tissues have been used for SCNTwithout observ<strong>in</strong>g significant differences <strong>in</strong> the overallefficiency (Oback and Wells 2002). In the horse, foetalfibroblasts (Li et al. 2003; Lagut<strong>in</strong>a et al. 2005; WoodsTable 5. Cleavage and blastocyst development after nuclear transferwith confluent or roscovit<strong>in</strong>e-treated fibroblasts (H<strong>in</strong>richs et al. 2006)TreatmentNo. of<strong>in</strong>jected(MII)No. of Cleavage No. of D +cleaved rate 7 blastocystsNo. of D +8 blastocystsTotalblastocyst(%)Confluent 55 40 73 1 1 2 (3.6)Roscovit<strong>in</strong>e 56 44 79 0 2 2 (3.6)et al. 2003) as well as adult fibroblasts and granulosacells have been used (Vanderwall et al. 2004; Lagut<strong>in</strong>aet al. 2005). For recover<strong>in</strong>g adult fibroblasts, sk<strong>in</strong>biopsies are taken under local anaesthesia on the neckor the chest of the donor horse. The recovered tissue isthen m<strong>in</strong>ced <strong>in</strong>to small pieces that are plated <strong>in</strong>TCM199 ⁄ DMEM (1:1) culture medium supplementedwith 10% FCS. In 2–3 weeks, the fibroblasts outgrowthe tissue and form a confluent monolayer. Uponsubsequent sub-culture and expansion, the cells arefrozen for later use or prepared for the SCNT procedureat early passages (Li et al. 2003). There is a considerablevariation <strong>in</strong> the success rate of horse SCNT because ofthe specific cell l<strong>in</strong>e source of donor nuclei (Galli et al.2003a; Lagut<strong>in</strong>a et al. 2005) as reported for other species(Oback and Wells 2002). The cell cycle of the donor cellis crucial for success and G0 or G1 stages have beenused <strong>in</strong> horses. This stages can be achieved throughserum starvation (reduction of FCS to 0.5%), growth toconfluency (contact <strong>in</strong>hibition) or the use of k<strong>in</strong>ase<strong>in</strong>hibitors like Roscovit<strong>in</strong>e (Table 5) (Oback and Wells2002; H<strong>in</strong>richs et al. 2006, 2007). Given the relativelylow number of experiments performed, it cannot beascerta<strong>in</strong>ed if any of these approaches is better than theother.Embryo Reconstruction and ActivationIn our laboratory, the oocytes are stripped of theircumulus after 22–24 h of maturation. First, by pipett<strong>in</strong>gdur<strong>in</strong>g exposure to hyaluronidase; second, <strong>in</strong> 0.25%tryps<strong>in</strong> for 1.5 m<strong>in</strong> and f<strong>in</strong>ally, <strong>in</strong> SOF-Hepes conta<strong>in</strong><strong>in</strong>g10% FCS. For reconstruction of NT-embryos, theauthors use both zona-enclosed and zona-free methods(Oback et al. 2003). For the zona-enclosed method,oocytes with an extruded polar body are sta<strong>in</strong>ed withHoechst 33342 <strong>in</strong> the presence of cytochalas<strong>in</strong> B(5 lg ⁄ ml). Enucleation is performed by the aspirationof the polar body and metaphase II plate <strong>in</strong> a m<strong>in</strong>imalvolume of ooplasm under UV light. All manipulationsare carried out <strong>in</strong> SOF-Hepes with 6 mg ⁄ ml BSA(Gardner et al. 1994), except fusion. Individual nucleardonor cells are then transferred <strong>in</strong>to the perivitell<strong>in</strong>espace of zona-enclosed cytoplasts us<strong>in</strong>g the enucleationpipette. The cytoplast–karyoplast couplets are transferred<strong>in</strong>to 0.3 M mannitol (50 lM Ca and 100 lM Mg)solution and fused one or two times with<strong>in</strong> 15–30 m<strong>in</strong><strong>in</strong>terval by a double DC-pulse of 1.8–2.4 kV ⁄ cm appliedfor 30 ls at 26–27 h of maturation. Because of theheterogeneous characteristics of the horse zona pellucida,the fusion rate is often <strong>in</strong>consistent or low. Li et al.(2002) have found that even high voltage DC pulses of2.2–2.5 kV ⁄ cm were able to fuse less than 60% ofÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


334 C Galli, I Lagut<strong>in</strong>a, R Duchi, S Colleoni and G Lazzaricouplets, the cleavage rate was 35–50% and onlyapproximately 2.5% of NT-embryos reached the blastocyststage. H<strong>in</strong>richs (H<strong>in</strong>richs et al. 2006, 2007) usedpiezoelectric micromanipulation both to enucleate themetaphase oocyte and to <strong>in</strong>ject the somatic cell directly<strong>in</strong>to the enucleated oocyte, thus overcom<strong>in</strong>g the limitationof cell fusion. The horse oocyte tolerates themicro<strong>in</strong>jection procedure very well as demonstrated alsoby the success of the ICSI procedure (Galli et al. 2007).For the zona-free method, the zona pellucida ofoocytes with extruded polar bodies is digested with0.5% pronase <strong>in</strong> PBS. Zona-free oocytes are enucleatedunder UV light with a blunt micropipette (with perpendicularbreak). All manipulations are performed <strong>in</strong>SOF-Hepes conta<strong>in</strong><strong>in</strong>g 10% FCS. Subsequently, zonafreecytoplasts are <strong>in</strong>dividually washed for few seconds<strong>in</strong> 300 lg ⁄ ml phytohemagglut<strong>in</strong><strong>in</strong> P <strong>in</strong> TCM 199-Hepesand then quickly dropped over a s<strong>in</strong>gle donor cell (Vajtaet al. 2003) settled to the bottom of a drop of TCM 199with 0.5% of FCS. Formed cell couplets are washed <strong>in</strong>0.3 M mannitol (50 lM Ca and 100 lM Mg) solutionand fused one or two times at 15–30 m<strong>in</strong> <strong>in</strong>tervals by as<strong>in</strong>gle DC-pulse of 1.2 kV ⁄ cm applied for 30 ls at 26–27 h of maturation. With such an approach, the fusionrate approximates 100% and because of the lowerelectric field required, both cleavage rate and postcleavagedevelopment to the blastocyst stage are higher(Lagut<strong>in</strong>a et al. 2005, 2007). Activation is usuallyperformed 2–4 h after fusion. In our experience, horseoocytes are difficult to activate and the standardprotocols used <strong>in</strong> rum<strong>in</strong>ants do not yield a high rate ofcleavage. The authors found that the use of ionomyc<strong>in</strong>(a calcium ionophore) followed by the synergistic effectof 6-DMAP (1 mM) and Cycloheximide (5 lg ⁄ ml)resulted <strong>in</strong> over 90% of parthenogenetic activation(Lazzari et al. 2002b; Galli et al. 2007) and cleavage ofcloned embryos (Lagut<strong>in</strong>a et al. 2005). Woods (Woodset al. 2003) <strong>in</strong>creased the calcium level dur<strong>in</strong>g activationof <strong>in</strong> vivo matured oocytes. H<strong>in</strong>richs (H<strong>in</strong>richs et al.2006, 2007) extensively compared different activationprotocols <strong>in</strong>clud<strong>in</strong>g the <strong>in</strong>jection of sperm extract.Sperm extract <strong>in</strong> comb<strong>in</strong>ation with ionomyc<strong>in</strong>, whilenot statistically different from other treatments, providedthe highest blastocyst development rate althoughthis was not superior to that reported by Lagut<strong>in</strong>a(Lagut<strong>in</strong>a et al. 2005) us<strong>in</strong>g the association of ionomyc<strong>in</strong>with both 6-DMAP and cycloheximide.Embryo CultureThe progress <strong>in</strong> <strong>in</strong> vitro maturation and ICSI technologyhas <strong>in</strong>creased efforts to design suitable culture systemsfor early cleavage-stage embryos. Very little is knownabout the specific <strong>in</strong> vitro culture requirements of equ<strong>in</strong>eembryos. The assessment of culture conditions has beencarried out with embryos produced <strong>in</strong> vitro by ICSI andus<strong>in</strong>g various media. Many different culture conditionshave been reported for pre-implantation development ofICSI fertilized horse oocytes, <strong>in</strong>clud<strong>in</strong>g def<strong>in</strong>ed mediasuch as G1.2 (Choi et al. 2002), DMEM-F12 and CZB(Choi et al. 2004) and modified SOF (Galli et al. 2002a).Earlier work evaluated co-culture with somatic cells<strong>in</strong>clud<strong>in</strong>g Vero cells (Dell’Aquila et al. 1997), oviductepithelial cells (Battut et al. 1991), cumulus cells (Liet al. 2001), granulosa cells (Rosati et al. 2002) orculture <strong>in</strong> conditioned media (Choi et al. 2001). In mostof these systems, yet, the blastocyst rates rema<strong>in</strong>ed low,rang<strong>in</strong>g from 4% to 16% of <strong>in</strong>jected oocytes. Incontrast, the culture of presumptive zygotes follow<strong>in</strong>gICSI, <strong>in</strong> vivo either <strong>in</strong> the mare oviduct or <strong>in</strong> thesurrogate sheep oviduct, allowed much higher development,be<strong>in</strong>g approximately 30% (Galli et al. 2002b,2007; Lazzari et al. 2002b; Tremoleda et al. 2003; Choiet al. 2004) (Table 6). When cell number counts werecompared among <strong>in</strong> vivo produced embryos and thoseproduced by <strong>in</strong> vitro culture <strong>in</strong> a modified SOF medium,both on day 7 of development, <strong>in</strong> vitro producedembryos had significantly lower cell numbers, resembl<strong>in</strong>ga day 5 embryo rather than a day 7 (Tremoledaet al. 2003). This difference is now taken <strong>in</strong>to accountwhen embryos are transferred to synchronized recipients(see Section ‘Embryo Transfer and Offspr<strong>in</strong>g’).The results reported above where obta<strong>in</strong>ed us<strong>in</strong>g aTCM 199-based medium for oocyte maturation. Interest<strong>in</strong>gly,the authors found that matur<strong>in</strong>g the oocytes <strong>in</strong>a DMEM-F12-based medium allows to <strong>in</strong>crease blastocystdevelopment to a much closer rate to <strong>in</strong> vivo culture,be<strong>in</strong>g <strong>in</strong> the range of 26% when compared with 27–50%for the sheep oviduct system, both for ICSI and forSCNT embryos (Galli et al. 2007). Yet, other laboratorieshave achieved equivalent blastocyst development(25–35%) with oocytes matured <strong>in</strong> M199, us<strong>in</strong>gDMEM-F12 medium for embryo culture under a mixedgas atmosphere (H<strong>in</strong>richs et al. 2005; Choi et al.2006a,b).In our laboratory, equ<strong>in</strong>e embryos are cultured <strong>in</strong>20 ll microdrops under oil with up to 20 embryos perdrop of mSOF. The culture of zona-free embryos isperformed <strong>in</strong>dividually <strong>in</strong> 5 ll drops or <strong>in</strong> the WOWsystem (Vajta et al. 2000). Fifty per cent of the media isreplaced on D 4 with mSOF and on D6 withDMEM ⁄ F12 supplemented with 5%FCS and 5% SR(serum replacement).The residual difference <strong>in</strong> embryo development ratesbetween the <strong>in</strong> vitro and <strong>in</strong> vivo culture environments<strong>in</strong>dicates the need for further improvement of theTable 6. Oocyte recovery rate by OPU and effect of <strong>in</strong> vitro or <strong>in</strong> vivo sheep oviduct culture on embryos development (Galli et al 2007)No. ofOPUsNo. of oocytes(no. per OPU)No. metaphase II(% of oocytes)(no. per OPU)No. of cleaved(% of <strong>in</strong>jected)(no. per OPU)No. of comp.morulae ⁄ blastocysts(% of <strong>in</strong>jected) (no. per OPU)Sheep oviduct culture 20 60 (3.0) 46 (76.7) (2.3) 41 (89.1% a) (2.1) 23 (50.0 a) (1.2)In vitro culture 12 46 (4.1) 36 (73.5) (3.0) 25 (69.4% a) (2.1) 5 (13.9 b) (0.4)Student’s t-test. Numbers with<strong>in</strong> columns with different letters are significantly different (p < 0.05).Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Clon<strong>in</strong>g <strong>in</strong> Horses 335embryo culture system. In our experience, the viabilityof cloned embryos is somewhat lower than their IVFcounterpart and for this reason more than one SCNTembryo is transferred <strong>in</strong>to each recipient <strong>in</strong> our laboratory.Embryo Transfer and Offspr<strong>in</strong>gTo date, a few groups have reported the birth of foalsfollow<strong>in</strong>g ICSI of <strong>in</strong> vitro matured oocytes, after eithersurgical transfer of early embryos to the oviduct(Cochran et al. 1998) or <strong>in</strong> vitro embryo culture to theblastocyst stage and transcervical transfer to the uterus(Li et al. 2001; H<strong>in</strong>richs et al. 2005; Galli et al. 2007).This has been true also for SCNT embryos where to datethree labs have reported <strong>in</strong> the scientific literature, thebirth of a dozen equids follow<strong>in</strong>g oviduct transfer ofearly cleav<strong>in</strong>g embryos (Woods et al. 2003) or uter<strong>in</strong>etransfer of blastocyst stage embryos (Galli et al. 2003a;Lagut<strong>in</strong>a et al. 2005; H<strong>in</strong>richs et al. 2006, 2007). Ashorse clon<strong>in</strong>g is enter<strong>in</strong>g the commercial arena, additionalanimals are currently be<strong>in</strong>g cloned or have beencloned by commercial companies. While one suchcompany has announced the birth of cloned foals tothe popular press, <strong>in</strong>formation on the methods used andtheir efficiency is not <strong>in</strong> the public doma<strong>in</strong>.In our laboratory, recipient mares are exam<strong>in</strong>ed twoto three times a week by ultrasound to determ<strong>in</strong>e the dayof ovulation. A total of 3–7 (mostly 5) days afterovulation, each mare receives up to four nuclear transferembryos at the blastocyst stage on day 7 or day 8 ofdevelopment by non-surgical transfer. On day 17, afterovulation, the animals are exam<strong>in</strong>ed for pregnancydiagnosis and thereafter, are scanned weekly throughoutthe first trimester of pregnancy and later at monthly<strong>in</strong>tervals until foal<strong>in</strong>g.Table 7 summarizes the results reported <strong>in</strong> thepublished studies for the transfer of SCNT equidembryos. The work of Woods and Vanderwall for muleand horse SCNT, respectively, refer to the surgicaltransfer to the recipient mare oviduct of one-cellnuclear transfer embryos. Results <strong>in</strong>dicate a total ofthree mule foals born from 305 zygotes transferred, outof 21 pregnancies diagnosed (Woods et al. 2003) andno live foals from 63 zygotes transferred and 7pregnancies diagnosed (Vanderwall et al. 2004). Inour laboratory, the authors have transferred 118embryos (morulae or blastocysts), fresh or frozen, <strong>in</strong>to46 recipients, obta<strong>in</strong><strong>in</strong>g 13 pregnancies with oneembryonic vesicle each and 3 live foals, i.e. an embryosurvival rate of 11% and a foal<strong>in</strong>g rate of 23% (3 ⁄ 13)(Galli et al. 2003a; Lagut<strong>in</strong>a et al. 2005). H<strong>in</strong>richs et al.(2006, 2007) reported the transfer of a total of 42blastocyst-stage embryos for 20 pregnancies and 11 livefoals. In our experience, the viability of clonedembryos is lower than for ICSI embryos. In fact,dur<strong>in</strong>g the same period, <strong>in</strong> which the SCNT embryoswere transferred a parallel study was conducted transferr<strong>in</strong>gICSI embryos both from abattoir and OPUoocytes cultured under the same conditions (Galli et al.2007). Overall, from 34 embryos produced by ICSItransferred <strong>in</strong>to 21 recipients, the authors obta<strong>in</strong>ed 13pregnancies at day 17, <strong>in</strong>dicat<strong>in</strong>g a survival rate of38% with<strong>in</strong> this study. The best result <strong>in</strong>volved oocytescollected by OPU, with embryos cultured <strong>in</strong> vitro andcryopreserved, <strong>in</strong> which 9 vesicles were observed onday 17 out of 13 embryos transferred <strong>in</strong>to 11 recipients.In this case, the survival rate after transfer was 69%and the subsequent foal<strong>in</strong>g rate was 88% (7 ⁄ 8) ofestablished pregnancies. Therefore, results confirm thelower viability of SCNT embryos when compared withfertilized embryos as already described <strong>in</strong> other species.Pregnancy losses with equ<strong>in</strong>e SCNT embryos occurthroughout gestation (Table 7) as described for rum<strong>in</strong>antsbut at a much lower rate. No large offspr<strong>in</strong>gsyndrome has been described <strong>in</strong> equids. The femalehorse produced by Galli (Galli et al. 2003a) (Fig. 1) iscurrently 4 years old and its foal was born on the 17 thMarch 2008 and it is normal and <strong>in</strong> good health. Themale produced by Lagut<strong>in</strong>a (Lagut<strong>in</strong>a et al. 2005) hasproduced semen for artificial <strong>in</strong>sem<strong>in</strong>ation and it hasconfirmed to be fertile. Altogether, these results suggestthat cloned horses are reproductively normal, as hasbeen shown <strong>in</strong> other species.Table 7. Pre- and post-implantation development of SCNT embryosreported <strong>in</strong> the published studiesAuthorNo. ofoocytesfusedNo. ofoocytescleavedNo. ofblastocystsNo. oftransferredNo. of No. ofpregnancies foalsWoods et al.(2003)Galli et al.(2003)Vanderwall et al.(2004)Lagut<strong>in</strong>a et al.(2005)H<strong>in</strong>richs et al.(2006)H<strong>in</strong>richs et al.(2007)307 NA NA 305 a 21 3841 753 21 17 4 172 NA NA 62 a 7 01508 1339 101 101 9 2567 457 14 10 4 2589 480 35 26 16 7a Transferred immediately after fusion to the oviduct.Fig. 1. Prometea, the world’s first cloned horse, born <strong>in</strong> 2003, <strong>in</strong> frontof her nuclear donor who was also her surrogate motherÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


336 C Galli, I Lagut<strong>in</strong>a, R Duchi, S Colleoni and G LazzariConclud<strong>in</strong>g RemarksAssisted-reproductive techniques have been developed<strong>in</strong> recent years and efficient techniques for oocytematuration, ICSI, embryo culture and SCNT are nowavailable. Their application for breed<strong>in</strong>g purposes hasonly started to be explored because of the limitednumber of laboratories currently able to master them.Interest<strong>in</strong>gly, cloned equ<strong>in</strong>e offspr<strong>in</strong>g reported so farappear not to exhibit the placental, foetal and neonatalpathologies described <strong>in</strong> other species. Furthermore, thefirst two cloned horses that have reached puberty haveshown normal reproductive performance and offspr<strong>in</strong>gfrom both are expected soon. For these reasons, thehorse breed<strong>in</strong>g <strong>in</strong>dustry is watch<strong>in</strong>g with some <strong>in</strong>terest,although often mixed with suspicion, the performance ofthe first cloned horses because clon<strong>in</strong>g offers theunprecedented opportunity of generat<strong>in</strong>g copies ofcastrated champions and, therefore, offspr<strong>in</strong>g thatwould be impossible to obta<strong>in</strong> otherwise (Church2006). This possibility promises to dramatically <strong>in</strong>creasethe impact of superior male genotypes <strong>in</strong> those horsebreeds that will accept clon<strong>in</strong>g <strong>in</strong> their breed<strong>in</strong>g schemes.AcknowledgementsThis work has been supported <strong>in</strong> part by grants from MUR (TECLA),Cariplo Foundation (NOBEL), the European Commission (Xenome,LSHB-CT-2006-037377) and the European Science Foundation (EuroStellsERAS-CT-2003-980409).ReferencesBattut I, Bezard J, Palmer E, 1991: Establishment of equ<strong>in</strong>eoviduct cell monolayers for co-culture with early equ<strong>in</strong>eembryos. J Reprod Fertil Suppl 44, 393–403.Bogh IB, Bezard J, Duchamp G, Baltsen M, Gerard N, DaelsP, Greve T, 2002: Pure preovulatory follicular fluid promotes<strong>in</strong> vitro maturation of <strong>in</strong> vivo aspirated equ<strong>in</strong>eoocytes. Theriogenology 57, 1765–1779.Booth PJ, Tan SJ, Reipurth R, Holm P, Callesen H, 2001:Simplification of bov<strong>in</strong>e somatic cell nuclear transfer byapplication of a zona-free manipulation technique. Clon<strong>in</strong>gStem Cells 3, 139–150.Campbell KH, Fisher P, Chen WC, Choi I, Kelly RD, Lee JH,Xhu J, 2007: Somatic cell nuclear transfer: past, present andfuture perspectives. Theriogenology 68(Suppl. 1), S214–S231.Carneiro G, Lorenzo P, Pimentel C, Pegoraro L, Bertol<strong>in</strong>i M,Ball B, Anderson G, Liu I, 2001: Influence of <strong>in</strong>sul<strong>in</strong>-likegrowth factor-I and its <strong>in</strong>teraction with gonadotrop<strong>in</strong>s,estradiol, and fetal calf serum on <strong>in</strong> vitro maturation andparthenogenic development <strong>in</strong> equ<strong>in</strong>e oocytes. Biol Reprod65, 899–905.Choi YH, Chung YG, Seidel GE Jr, Squires EL, 2001:Developmental capacity of equ<strong>in</strong>e oocytes matured andcultured <strong>in</strong> equ<strong>in</strong>e trophoblast-conditioned media. Theriogenology56, 329–339.Choi YH, Love CC, Chung YG, Varner DD, Westhus<strong>in</strong> ME,Burghardt RC, H<strong>in</strong>richs K, 2002: Production of nucleartransfer horse embryos by Piezo-driven <strong>in</strong>jection of somaticcell nuclei and activation with stallion sperm cytosolicextract. Biol Reprod 67, 561–567.Choi YH, Love LB, Westhus<strong>in</strong> ME, H<strong>in</strong>richs K, 2004:Activation of equ<strong>in</strong>e nuclear transfer oocytes: methodsand tim<strong>in</strong>g of treatment <strong>in</strong> relation to nuclear remodel<strong>in</strong>g.Biol Reprod 70, 46–53.Choi YH, Love LB, Varner DD, H<strong>in</strong>richs K, 2006a: Blastocystdevelopment <strong>in</strong> equ<strong>in</strong>e oocytes with low meiotic competenceafter suppression of meiosis with roscovit<strong>in</strong>e prior to <strong>in</strong> vitromaturation. Zygote 14, 1–8.Choi YH, Love LB, Varner DD, H<strong>in</strong>richs K, 2006b: Hold<strong>in</strong>gimmature equ<strong>in</strong>e oocytes <strong>in</strong> the absence of meiotic <strong>in</strong>hibitors:effect on germ<strong>in</strong>al vesicle chromat<strong>in</strong> and blastocystdevelopment after <strong>in</strong>tracytoplasmic sperm <strong>in</strong>jection. Theriogenology66, 955–963.Church SL, 2006: Nuclear transfer saddles up. Nat Biotechnol24, 605–607.Cochran R, Me<strong>in</strong>tjes M, Reggio B, Hylan D, Carter J, P<strong>in</strong>toC, Paccamonti D, Godke RA, 1998: Live foals producedfrom sperm-<strong>in</strong>jected oocytes derived from pregnant mares.J Equ<strong>in</strong>e Vet Sci 18, 736–740.Dell’Aquila ME, Cho YS, M<strong>in</strong>oia P, Tra<strong>in</strong>a V, LacalandraGM, Maritato F, 1997: Effects of follicular fluid supplementationof <strong>in</strong>-vitro maturation medium on the fertilizationand development of equ<strong>in</strong>e oocytes after <strong>in</strong>-vitrofertilization or <strong>in</strong>tracytoplasmic sperm <strong>in</strong>jection. HumReprod 12, 2766–2772.Dell’Aquila ME, Masterson M, Maritato F, H<strong>in</strong>richs K, 2001:Influence of oocyte collection technique on <strong>in</strong>itial chromat<strong>in</strong>configuration, meiotic competence, and male pronucleusformation after <strong>in</strong>tracytoplasmic sperm <strong>in</strong>jection (ICSI) ofequ<strong>in</strong>e oocytes. Mol Reprod Dev 60, 79–88.Dell’Aquila ME, Albrizio M, Maritato F, M<strong>in</strong>oia P, H<strong>in</strong>richsK, 2003: Meiotic competence of equ<strong>in</strong>e oocytes and pronucleusformation after <strong>in</strong>tracytoplasmic sperm <strong>in</strong>jection(ICSI) as related to granulosa cell apoptosis. Biol Reprod68, 2065–2072.Galli C, Crotti G, Tur<strong>in</strong>i P, Duchi R, Mari G, Zavaglia G,Duchamp G, Daels P, Lazzari G, 2002a: Frozen-thawedembryos produced by ovum pick up of immature oocytesand ICSI are capable to establish pregnancies <strong>in</strong> the horse.Theriogenology 58, 705–708.Galli C, Maclellan LJ, Crotti G, Tur<strong>in</strong>i P, Ponderato N, DuchiR, Merlo B, Mari G, Barbac<strong>in</strong>i S, Lazzari G, 2002b:Development of equ<strong>in</strong>e oocytes matured <strong>in</strong> vitro <strong>in</strong> differentmedia and fertilised by ICSI. Theriogenology 57, 718.Galli C, Lagut<strong>in</strong>a I, Crotti G, Colleoni S, Tur<strong>in</strong>i P, PonderatoN, Duchi R, Lazzari G, 2003a: Pregnancy: a cloned horseborn to its dam tw<strong>in</strong>. Nature 424, 635.Galli C, Lagut<strong>in</strong>a I, Lazzari G, 2003b: Introduction to clon<strong>in</strong>gby nuclear transplantation. Clon<strong>in</strong>g Stem Cells 5, 223–232.Galli C, Colleoni S, Duchi R, Lagut<strong>in</strong>a I, Lazzari G, 2007:Developmental competence of equ<strong>in</strong>e oocytes and embryosobta<strong>in</strong>ed by <strong>in</strong> vitro procedures rang<strong>in</strong>g from <strong>in</strong> vitromaturation and ICSI to embryo culture, cryopreservationand somatic cell nuclear transfer. Anim Reprod Sci 98, 39–55.Gardner DK, Lane M, Spitzer A, Batt PA, 1994: Enhancedrates of cleavage and development for sheep zygotescultured to the blastocyst stage <strong>in</strong> vitro <strong>in</strong> the absence ofserum and somatic cells: am<strong>in</strong>o acids, vitam<strong>in</strong>s, and cultur<strong>in</strong>gembryos <strong>in</strong> groups stimulate development. Biol Reprod50, 390–400.H<strong>in</strong>richs K, Schmidt AL, 2000: Meiotic competence <strong>in</strong> horseoocytes: <strong>in</strong>teractions among chromat<strong>in</strong> configuration, folliclesize, cumulus morphology, and season. Biol Reprod 62,1402–1408.H<strong>in</strong>richs K, Williams KA, 1997: Relationships among oocytecumulusmorphology, follicular atresia, <strong>in</strong>itial chromat<strong>in</strong>configuration, and oocyte meiotic competence <strong>in</strong> the horse.Biol Reprod 57, 377–384.H<strong>in</strong>richs K, Choi YH, Love LB, Varner DD, Love CC,Walckenaer BE, 2005: Chromat<strong>in</strong> configuration with<strong>in</strong> thegerm<strong>in</strong>al vesicle of horse oocytes: changes post mortem andÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Clon<strong>in</strong>g <strong>in</strong> Horses 337relationship to meiotic and developmental competence. BiolReprod 72, 1142–1150.H<strong>in</strong>richs K, Choi YH, Love CC, Chung YG, Varner DD,2006: Production of horse foals via direct <strong>in</strong>jection ofroscovit<strong>in</strong>e-treated donor cells and activation by <strong>in</strong>jection ofsperm extract. <strong>Reproduction</strong> 131, 1063–1072.H<strong>in</strong>richs K, Choi YH, Varner DD, Hartman DL, 2007:Production of cloned horse foals us<strong>in</strong>g roscovit<strong>in</strong>e-treateddonor cells and activation with sperm extract and ⁄ orionomyc<strong>in</strong>. <strong>Reproduction</strong> 134, 319–325.Hochedl<strong>in</strong>ger K, Jaenisch R, 2006: Nuclear reprogramm<strong>in</strong>gand pluripotency. Nature 441, 1061–1067.Lagut<strong>in</strong>a I, Lazzari G, Duchi R, Colleoni S, Ponderato N,Tur<strong>in</strong>i P, Crotti G, Galli C, 2005: Somatic cell nucleartransfer <strong>in</strong> horses: effect of oocyte morphology, embryoreconstruction method and donor cell type. <strong>Reproduction</strong>130, 559–567.Lagut<strong>in</strong>a I, Lazzari G, Duchi R, Tur<strong>in</strong>i P, Tessaro I, BrunettiD, Colleoni S, Crotti G, Galli C, 2007: Comparative aspectsof somatic cell nuclear transfer with conventional and zonafreemethod <strong>in</strong> cattle, horse, pig and sheep. Theriogenology67, 90–98.Lazzari G, Crotti G, Tur<strong>in</strong>i P, Duchi R, Mari G, Zavaglia G,Barbac<strong>in</strong>i S, Galli C, 2002a: Equ<strong>in</strong>e embryos at thecompacted morula and blastocyst stage can be obta<strong>in</strong>edby <strong>in</strong>tracytoplasmic sperm <strong>in</strong>jection (ICSI) of <strong>in</strong> vitromatured oocytes with frozen-thawed spermatozoafrom semen of different fertilities. Theriogenology 58, 709–712.Lazzari G, Mari G, Galli C, 2002b: Synergistic effect ofcycloheximide and 6-DMAP on activation of equ<strong>in</strong>e andbov<strong>in</strong>e oocytes. J Reprod Fertil Abstr Ser 28, 73.Li X, Morris LH, Allen WR, 2001: Influence of co-culturedur<strong>in</strong>g maturation on the developmental potential of equ<strong>in</strong>eoocytes fertilized by <strong>in</strong>tracytoplasmic sperm <strong>in</strong>jection(ICSI). <strong>Reproduction</strong> 121, 925–932.Li X, Morris LH, Allen WR, 2002: In vitro development ofhorse oocytes reconstructed with the nuclei of fetal andadult cells. Biol Reprod 66, 1288–1292.Li X, Tremoleda JL, Allen WR, 2003: Effect of the numberof passages of fetal and adult fibroblasts on nuclearremodell<strong>in</strong>g and first embryonic division <strong>in</strong> reconstructedhorse oocytes after nuclear transfer. <strong>Reproduction</strong> 125, 535–542.de Loos F, van Vliet C, van Maurik P, Kruip TA, 1989:Morphology of immature bov<strong>in</strong>e oocytes. Gamete Res 24,197–204.Lorenzo PL, Liu IK, Carneiro GF, Conley AJ, Enders AC,2002: Equ<strong>in</strong>e oocyte maturation with epidermal growthfactor. Equ<strong>in</strong>e Vet J 34, 378–382.Oback B, Wells D, 2002: Donor cells for nuclear clon<strong>in</strong>g: manyare called, but few are chosen. Clon<strong>in</strong>g Stem Cells 4, 147–168.Oback B, Wiersema AT, Gaynor P, Laible G, Tucker FC,Oliver JE, Miller AL, Troskie HE, Wilson KL, Forsyth JT,Berg MC, Cockrem K, McMillan V, Tervit HR, Wells DN,2003: Cloned cattle derived from a novel zona-free embryoreconstruction system. Clon<strong>in</strong>g Stem Cells 5, 3–12.Rideout WM III, Hochedl<strong>in</strong>ger K, Kyba M, Daley GQ,Jaenisch R, 2002: Correction of a genetic defect by nucleartransplantation and comb<strong>in</strong>ed cell and gene therapy. Cell109, 17–27.Ritchie WA, 2006: Nuclear transfer <strong>in</strong> sheep. Methods MolBiol 325, 11–23.Rosati I, Berl<strong>in</strong>guer F, Bogliolo L, Leoni G, Ledda S, NaitanaS, 2002: The effect of co-culture on the development of <strong>in</strong>vitro matured equ<strong>in</strong>e oocytes after <strong>in</strong>tracytoplastic sperm<strong>in</strong>jection. Equ<strong>in</strong>e Vet J 34, 673–678.Tremoleda JL, Stout TA, Lagut<strong>in</strong>a I, Lazzari G, Bevers MM,Colenbrander B, Galli C, 2003: Effects of <strong>in</strong> vitro productionon horse embryo morphology, cytoskeletal characteristics,and blastocyst capsule formation. Biol Reprod 69,1895–1906.Vajta G, Peura TT, Holm P, Paldi A, Greve T, Trounson AO,Callesen H, 2000: New method for culture of zona-<strong>in</strong>cludedor zona-free embryos: the Well of the Well (WOW) system.Mol Reprod Dev 55, 256–264.Vajta G, Lewis IM, Trounson AO, Purup S, Maddox-HyttelP, Schmidt M, Pedersen HG, Greve T, Callesen H, 2003:Handmade somatic cell clon<strong>in</strong>g <strong>in</strong> cattle: analysis of factorscontribut<strong>in</strong>g to high efficiency <strong>in</strong> vitro. Biol Reprod 68, 571–578.Vanderwall DK, Woods GL, Aston KI, Bunch TD, Li G,Meerdo LN, White KL, 2004: Cloned horse pregnanciesproduced us<strong>in</strong>g adult cumulus cells. Reprod Fertil Dev 16,675–679.Westhus<strong>in</strong> M, H<strong>in</strong>richs K, Choi YH, Sh<strong>in</strong> T, Liu L, KraemerD, 2003: Clon<strong>in</strong>g companion animals (horses, cats, anddogs). Clon<strong>in</strong>g Stem Cells 5, 301–317.Willadsen SM, 1986: Nuclear transplantation <strong>in</strong> sheepembryos. Nature 320, 63–65.Wilmut I, Schnieke AE, McWhir J, K<strong>in</strong>d AJ, CampbellKH1997: Viable offspr<strong>in</strong>g derived from fetal and adultmammalian cells. Nature 385, 810–813.Woods GL, White KL, Vanderwall DK, Li GP, Aston KI,Bunch TD, Meerdo LN, Pate BJ, 2003: A mule cloned fromfetal cells by nuclear transfer. Science 301, 1063.Zhang JJ, Boyle MS, Allen WR, Galli C, 1989: Recent studieson <strong>in</strong> vivo fertilization of <strong>in</strong> vitro matured horse oocytes.Equ<strong>in</strong>e Vet J 8, 101–104.Author’s address (for correspondence): Cesare Galli, Laboratorio diTecnologie della Riproduzione, CIZ srl, Via Porcellasco 7 ⁄ f, 26100Cremona, Italy. E-mail: cesaregalli@ltrciz.itConflict of <strong>in</strong>terest: All authors declare no conflict of <strong>in</strong>terests.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 338–346 (2008); doi: 10.1111/j.1439-0531.2008.01182.xISSN 0936-6768Application and Commercialization of Flow Cytometrically Sex-Sorted SemenD Rath 1 and LA Johnson 21 Institute of Farm Animal Genetics, Mariensee (FLI), Neustadt, Germany; 2 Mount Airy, MD, USAContentsThe current technology to sort X and Y chromosomebear<strong>in</strong>g sperm population requires <strong>in</strong>dividual identificationand selection of spermatozoa <strong>in</strong> a modified high-speed flowcytometer. For farm animal species, the technology iscapable of produc<strong>in</strong>g sexed sperm at greater than 90%purity. However, only <strong>in</strong> the bov<strong>in</strong>e, the technology hasreached a developmental level that allows its commercialapplication. Meanwhile, the demand for female calves hasgrown rapidly, which encourages the demand for sex-sortedsemen from high genetic value bulls. The success of thetechnology will depend ma<strong>in</strong>ly on the fertiliz<strong>in</strong>g capacity ofthe sorted spermatozoa, as this is the most affect<strong>in</strong>g andeconomically relevant factor. To date, fertility is still variableand is quite dependant on post-sort process<strong>in</strong>g. Newprocess<strong>in</strong>g techniques are under <strong>in</strong>vestigation and will likelybe able to improve the fertility rates after AI with sex-sortedsemen. It is of great importance to select the right bulls andto test the sorted samples on a rout<strong>in</strong>e basis. In addition tothe demand for sex-sorted semen by the cattle <strong>in</strong>dustry, thereis also a significant demand expressed by pig farmers.However, it is still unknown if the use of sex-sorted sementhrough commercial pig AI will be economically feasible. Forthe pig, the comb<strong>in</strong>ation of <strong>in</strong> vitro fertilization with sexedsemen and non-surgical embryo transfer is an alternative thatmerits further scientific attention. Recent developments <strong>in</strong>ov<strong>in</strong>e AI and ET will make it very likely that commercialsheep <strong>in</strong>dustry will adopt the sex<strong>in</strong>g technology <strong>in</strong> theirbreed<strong>in</strong>g concepts.IntroductionControll<strong>in</strong>g the sex of offspr<strong>in</strong>g prior to conceptionpermits the livestock <strong>in</strong>dustry to produce the optimalproportion of males and females to take advantage ofsex-limited and sex-<strong>in</strong>fluenced traits, thus provid<strong>in</strong>geconomically flexible management practices for theproducer. Controll<strong>in</strong>g sex ratio permits faster geneticprogress, higher productivity, improves animal welfareby decreas<strong>in</strong>g obstetric difficulties <strong>in</strong> cattle, avoid<strong>in</strong>gcastration <strong>in</strong> pigs, and produc<strong>in</strong>g less environmentalimpact due to the elim<strong>in</strong>ation of the unwanted sexbefore they grow to adulthood. Sex pre-selection <strong>in</strong>animals must be effective and efficient, must result <strong>in</strong>fertility equal to or better than with unsexed semen, andmust be reasonably <strong>in</strong>expensive and convenient to bewidely applied (Foote and Miller 1971). Currently, theonly known means of effectively produc<strong>in</strong>g separatepopulations of X and Y sperm <strong>in</strong> mammals is throughthe use of DNA differentiation by high-speed flowcytometry (Beltsville Sperm Sex<strong>in</strong>g Technology; Johnsonet al. 1989). Applicable to virtually all mammals,this method can produce populations of X or Y spermwith greater than 90% purity and subsequent offspr<strong>in</strong>gwhose phenotypic sex is consistent with <strong>in</strong>itial purityof the sorted sperm population. Limitations of thetechnology are entw<strong>in</strong>ed with the physiology andanatomy of the specific species <strong>in</strong> terms of the numberof sperm required for fertilization and subsequentproduction of offspr<strong>in</strong>g. The current technology requiresthat each sperm be separately <strong>in</strong>terrogated for DNAcontent, thus limit<strong>in</strong>g the number of sorted X or Ysperm <strong>in</strong> cattle, sheep, sw<strong>in</strong>e and horses to approximately12–20 million sperm per hour (Johnson andWelch 1999; de Graaf et al. 2007c; Heer 2007).As flow cytometers and computers were developed andimproved, it soon became apparent that flow cytometryheld the key to separation of viable X and Y chromosomebear<strong>in</strong>g sperm based on their DNA content, which couldthen be used for fertilization. Successful development of aviable sperm sex<strong>in</strong>g procedure encompassed four steps toachieve a verifiable method of controll<strong>in</strong>g sex ratio <strong>in</strong>mammals. These steps were:(1) modification of a commercially available flowcytometer ⁄ cell sorter <strong>in</strong>to a sperm cell sorter byadd<strong>in</strong>g a forward fluorescence detector and a bevelledsample <strong>in</strong>jection needle to accommodate sperm orientationand m<strong>in</strong>imize DNA variability (Johnson andP<strong>in</strong>kel 1986),(2) development of a method to sta<strong>in</strong> liv<strong>in</strong>g spermwith a vital fluorescent dye (Hoechst 33342, Sigma-Aldrich, Mu¨ nchen, Germany) <strong>in</strong> order to ma<strong>in</strong>ta<strong>in</strong>viability through the sort<strong>in</strong>g process and to the timeof fertilization (Johnson et al. 1987a),(3) merg<strong>in</strong>g the analytical and sort<strong>in</strong>g capability of thesperm sorter to produce separate populations of liv<strong>in</strong>gX and Y sperm based on differential DNA content(Johnson et al. 1989) and(4) development of a method to re-analyse separatesorted populations of X and Y liv<strong>in</strong>g sperm for theirDNA content to verify the purity of X or Y sperm <strong>in</strong>the laboratory (Johnson et al. 1987b; Welch andJohnson 1999) to be a predictor of ultimate phenotypicsex of the live offspr<strong>in</strong>g (Johnson et al. 1989;Johnson 1991) and prove the efficacy of us<strong>in</strong>g DNA todifferentiate X and Y sperm that would be used toachieve fertilization.The limitations of s<strong>in</strong>gle-cell measurement technologyhave been followed throughout the development of thetechnology and cont<strong>in</strong>ued even to this day <strong>in</strong> commercialapplication. Consistent with these limitations, themethod is applied most easily <strong>in</strong> the bov<strong>in</strong>e (2 millionsperm per AI; Seidel et al. 1997), ov<strong>in</strong>e (1–5 millionsperm per AI; de Graaf et al. 2007c) but hardly at all <strong>in</strong>sw<strong>in</strong>e (50–100 million sperm per AI; Rath et al. 2003;Vazquez et al. 2003; Grossfeld et al. 2005). Even withthe obvious limitations, the technology has begun toflourish <strong>in</strong> commercial bov<strong>in</strong>e practice. It is estimatedÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Commercialization of Sex-Sorted Semen 339that more than 2 million sexed calves have beenproduced s<strong>in</strong>ce the technology was first applied tocommercial practice <strong>in</strong> 2000. Although the license tobeg<strong>in</strong> commercialization us<strong>in</strong>g AI was granted by theUS Department of Agriculture <strong>in</strong> 1996 (Patent#5,135,759) to XY-Inc. Fort Coll<strong>in</strong>s, CO, USA, commercialproduction of bov<strong>in</strong>e sexed sperm did not beg<strong>in</strong>until 2000 (Cogent Breed<strong>in</strong>g Ltd., Chester, UK). However,<strong>in</strong>sufficient development of the technology by theparent company hampered faster and wider commercialapplication. With<strong>in</strong> the past year, XY-Inc. was sold to asub-licensee (Sex<strong>in</strong>g Technologies, Navasota, TX,USA). Prior to this sale, Sex<strong>in</strong>g Technologies had some20 sorters operat<strong>in</strong>g <strong>in</strong> North America, which has nowbeen expanded to more than 50 sorters <strong>in</strong> numerouslocations <strong>in</strong> North and South America, Europe andother parts of the world. These recent developmentssuggest that sexed bov<strong>in</strong>e sperm will soon be available tomost cattle producers around the world. Intensiveresearch is still required to further improve the exist<strong>in</strong>gtechnology or f<strong>in</strong>d alternatives and ⁄ or comb<strong>in</strong>ationswith other biotechniques. Increased technology developmentis critical <strong>in</strong> order to develop the largecommercial opportunity that exists for sexed semencattle and <strong>in</strong> other livestock species, especially sheep,sw<strong>in</strong>e and horses. In this review, the current status of thesex-sort<strong>in</strong>g technology associated with commercialapplications and the consequences for farm animalagriculture are summarized.The Limitations of S<strong>in</strong>gle-Cell MeasurementTechnologyThe current technology requires s<strong>in</strong>gle sperm identification,<strong>in</strong>dividual recognition of the orient<strong>in</strong>g position <strong>in</strong>front of the laser beam and s<strong>in</strong>gle droplet charg<strong>in</strong>g.Although there has been significant progress <strong>in</strong> thenumber of sperms sorted per unit time <strong>in</strong> the last12 years (1–2 million sperm per hour to the current levelof approximately 20 million sperm per hour), theprocess rema<strong>in</strong>s <strong>in</strong>efficient <strong>in</strong> the production of spermfor AI. Standard AI dose numbers of fresh or frozenunsexed sperm are out of reach for this technology.Efficient utilization of sex-sorted semen requires asignificant reduction <strong>in</strong> spermatozoa per AI dose orfor use <strong>in</strong> <strong>in</strong> vitro fertilization (IVF) and other biotechniques.For example, 2 million live spermatozoa frommany bulls or for some bulls even below seem to besufficient for AI. The usability of these bulls for sexsort<strong>in</strong>gdepends only partly on the strength of theirspermatozoa to survive the sort<strong>in</strong>g process. It is wellknown that the fertiliz<strong>in</strong>g ability with unsortedspermatozoa varies among <strong>in</strong>dividual bulls when lowsperm concentrations are used for AI (Den Daas et al.1998).In addition to the effects of reduced sperm number,high dilution effects dim<strong>in</strong>ish the fertiliz<strong>in</strong>g potentialof spermatozoa as the protect<strong>in</strong>g and regulat<strong>in</strong>gsubstances of sem<strong>in</strong>al plasma are also diluted orelim<strong>in</strong>ated (Maxwell and Johnson 1999; Centurionet al. 2003).The differential sperm DNA content is identified withthe Bis-benzimide Hoechst 33342 that emits a bluefluorescence when excited with UV light. Hoechst 33342was selected based on the fact that it was a vital sta<strong>in</strong>and one that effectively was able to penetrate the liv<strong>in</strong>gsperm membrane and b<strong>in</strong>d to the DNA of the highlycondensed chromat<strong>in</strong> of the sperm nucleus (Johnsonet al. 1987a). In several experiments, the <strong>in</strong>fluence of theHoechst 33342 on DNA <strong>in</strong>tegrity was tested. Johnsonet al. (1989) postulated that fluorochrome dyes reduceembryonic viability by mid-gestation. This co<strong>in</strong>cideswith the f<strong>in</strong>d<strong>in</strong>gs of Sp<strong>in</strong>aci et al. (2005) with boarspermatozoa. Co-<strong>in</strong>cubation with Hoechst dye as well asthe sort<strong>in</strong>g process itself dim<strong>in</strong>ished the percentage oflive spermatozoa. The damaged ability to fertilize andcarry the embryo to term may be the result of thecomb<strong>in</strong>ed effects of the dye and UV laser or of either<strong>in</strong>dividually. Higher laser <strong>in</strong>tensity is more damag<strong>in</strong>gthan the lower laser <strong>in</strong>tensity as shown for rabbitspermatozoa by Johnson et al. (1996). Recently, Schenkand Seidel (2007) reported a similar f<strong>in</strong>d<strong>in</strong>g for bov<strong>in</strong>esemen. However, less laser <strong>in</strong>tensity dim<strong>in</strong>ishes theresolution and <strong>in</strong>directly the sort rates. Guthrie et al.(2002) saw no differences on embryo development whenpig spermatozoa were illum<strong>in</strong>ated with 125 or 25 mWlaser power, but optimum resolution dur<strong>in</strong>g sort<strong>in</strong>gbetween X and Y <strong>in</strong>tact sperm required at least 125 mWlaser power.Catt et al. (1997) labelled human spermatozoa onmicroscope slides with the Hoechst dye and exposedthem to UV laser light. No changes were found <strong>in</strong> thefrequency of endogenous DNA nicks. This is <strong>in</strong>agreement with a recent study from Parrilla et al.(2004), <strong>in</strong>dicat<strong>in</strong>g no genotoxic effects of Hoechst33342 <strong>in</strong> porc<strong>in</strong>e spermatozoa. Boe-Hansen et al.(2005) used the neutral Comet assay and the sperm condensationstructure assay (SCSA) to evaluatesperm chromosome <strong>in</strong>tegrity. Both tests showed thatsperm <strong>in</strong>tegrity is improved <strong>in</strong> the sorted populationwhen compared with unsorted semen. This f<strong>in</strong>d<strong>in</strong>g isno doubt due to the presence of FD#40 red food dye,which is added to the pre-sort sample to elim<strong>in</strong>atemembrane damaged sperm from the sort<strong>in</strong>g process(Johnson et al. 1999). Similar results were obta<strong>in</strong>ed byDe Ambrogi et al. (2006), who found no effect on thedefragmentation <strong>in</strong>dex after sort<strong>in</strong>g, and de Graafet al. (2007a,b) gett<strong>in</strong>g even better fertilization and ETresults with sorted semen when compared with highdiluted controls.A significant factor dur<strong>in</strong>g sort<strong>in</strong>g is also therepeated electrical charg<strong>in</strong>g and electrostatic deviation.Membranes of the mid-piece of the sperm tail aresensitive to the electric field and may undergo depolarization.Furthermore, we believe that mitochondrialactivity is reduced due to the presence of reactiveoxygen species (ROS) produced by electric forces(Kl<strong>in</strong>c and Rath 2007; Kl<strong>in</strong>c et al. 2007). Similarpositive results were reported for sex-sorted boarspermatozoa when media were supplemented withPSP I ⁄ II heterodimers (Garcı´ a et al. 2007), which<strong>in</strong>creased motility and mitochondrial activity(Centurion et al. 2003). However, <strong>in</strong> a study reportedby de Graaf et al. (2007b), they were unable to seesimilar benefits to ram semen antioxidants or sem<strong>in</strong>alplasma.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


340 D Rath and LA JohnsonCommercial Application of Sex-SortedSpermatozoa <strong>in</strong> Livestock ProductionExcellent reviews on the commercialization, especially ofbov<strong>in</strong>e sexed semen, have been published by Amann(1999), Seidel (2003a,b), Garner (2006) and Garner andSeidel (2008). Although their calculation models arema<strong>in</strong>ly directed to the American farm situation, they arehelpful guidel<strong>in</strong>es for other countries too. The situation<strong>in</strong> other species had been summarized by Maxwell et al.(2004) and for pigs by Johnson et al. (2005). Spermsort<strong>in</strong>g based on the Beltsville Sperm Sex<strong>in</strong>g Technologyhas reached a certa<strong>in</strong> technical standard that makes itrobust enough for use under practical conditions, even ifit has clear limitations as described above. Its utilizationdiffers significantly among species ma<strong>in</strong>ly because of thebiological differences related to site of semen deposition,length of cycle, demand on sperm numbers, sortabilityand freezability of spermatozoa as well as commercialdemands and management conditions.The Commercial Use of Bov<strong>in</strong>e Sex-SortedSemenStage of applicabilityDue to the high sort<strong>in</strong>g <strong>in</strong>dex (131), an approximation ofthe ability to flow cytometrically sort sperm consist<strong>in</strong>g ofthe head profile area (lm 2 ) · X-Y sperm DNA difference(%; Garner 2006), bov<strong>in</strong>e spermatozoa are more suitablefor high-speed sort<strong>in</strong>g than other species. The methodhas reached a standard that is likely to make it profitable<strong>in</strong> dairy herds soon. However, impairments to thefertiliz<strong>in</strong>g capacity of sorted bull sperm are obviousand impairment of fertiliz<strong>in</strong>g capacity because of theimpact of sort<strong>in</strong>g and high dilution on capacitation andsem<strong>in</strong>al plasma components cont<strong>in</strong>ue to be limitations tobe dealt with. Further improvement of the pre- and postsorthandl<strong>in</strong>g of the semen is critical and requires<strong>in</strong>tensive research to m<strong>in</strong>imize negative effects on thepost-thaw lifespan of sexed spermatozoa as the fertilephase has direct implications on AI regimens. In a recentstudy employ<strong>in</strong>g a modified sperm handl<strong>in</strong>g protocoldur<strong>in</strong>g sort<strong>in</strong>g and freez<strong>in</strong>g and <strong>in</strong> comb<strong>in</strong>ation with anew extender for both sort<strong>in</strong>g and freez<strong>in</strong>g (Sexcess Ò ,Masterr<strong>in</strong>d GmbH, Verden, Germany), we were able toextend the post-thaw viability and progressive motilityfor several hours as shown by thermo tolerance test andcalv<strong>in</strong>g rates (Kl<strong>in</strong>c 2005; Kl<strong>in</strong>c et al. 2007). In this AIstudy, sorted semen (2 million live sperm ⁄ straw) wasthawed and <strong>in</strong>sem<strong>in</strong>ated <strong>in</strong> accordance with the exist<strong>in</strong>gprotocols for conventional semen (thaw<strong>in</strong>g at 37°C for20 s; AI 12–24 h after onset of heat; semen deposition <strong>in</strong>the uter<strong>in</strong>e body or if possible without force <strong>in</strong>to theipsilateral horn). Pregnancy and calv<strong>in</strong>g rates were equalto controls. Similar data were also obta<strong>in</strong>ed for sortedliquid semen stored for up to 72 h (Kl<strong>in</strong>c and Rath 2007).Significant technical improvements <strong>in</strong> sperm quality aftersort<strong>in</strong>g were developed us<strong>in</strong>g the latest laser technologyand replac<strong>in</strong>g the water-cooled Argon gas laser with apulsed solid-state laser. Shorter exposure to the laserlight seems to further reduce sperm stress, and observationswith ram spermatozoa <strong>in</strong>dicate <strong>in</strong>creased fertilityrates (de Graaf et al. 2007c). Another stress factor is thehydrodynamic pressure. The percentage of live bull andstallion spermatozoa <strong>in</strong>creased significantly when thefluid pressure was lowered from 2.07 to 2.59 mm Hg and<strong>in</strong>creased developmental rates of bov<strong>in</strong>e IVF embryos(Campos-Chillon and de la Torre 2003; Suh et al. 2005).For many reasons, it may be helpful to sort conventionallyfrozen semen after thaw<strong>in</strong>g and refreeze thesorted samples. Such methods are under <strong>in</strong>vestigation.Holl<strong>in</strong>shead et al. (2004) showed for the first time thatbull semen survives an adapted protocol from ram(Holl<strong>in</strong>shead et al. 2003) employ<strong>in</strong>g a gradient centrifugationto separate egg yolk from the thawed samples andto enrich <strong>in</strong>tact sperm prior to sort<strong>in</strong>g. Maxwell et al.(2007) tested PureSperm Ò (Labotect, Go¨ tt<strong>in</strong>gen, Germany)as effective gradient for high diluted, unsorted bullsperm, and <strong>in</strong> a recent study, Parrilla et al. (unpublished)showed that PureSperm and Bovipure Ò (Labotect,Go¨ tt<strong>in</strong>gen, Germany) are suitable for refreez<strong>in</strong>g sexedbull sperm and to produce embryos to the same extent aswith unsorted sperm from the same ejaculates.Altogether, the technical improvements made <strong>in</strong> therecent years allow for the ma<strong>in</strong>tenance of an acceptablestandard of bov<strong>in</strong>e semen quality. Nevertheless, it is selfevidentto control the sorted sperm quality on a dailybasis. Some suggested prerequisites for sex-sorted bullsemen of optimal quality are as follows:(1) Bulls must be pre-tested and those fail<strong>in</strong>g withunsorted low-dose <strong>in</strong>sem<strong>in</strong>ation should be excludedfrom their use to produce sex-sorted sperm.(2) A complete spermiogram should be made fromeach ejaculate before sort<strong>in</strong>g. The test should <strong>in</strong>clude:macroscopical evaluation, mass movement categorization,s<strong>in</strong>gle sperm motility estimation, evaluation ofsperm concentration and a complete morphologicalevaluation test, especially as a faster acrosome reactionis commonplace <strong>in</strong> sex-sorted sperm (Mocé et al.2006).(3) After sort<strong>in</strong>g, aliquots of each batch should betested for s<strong>in</strong>gle sperm motility, motility pattern <strong>in</strong> athermal tolerance test preferably for 6 h at 38°C,precise morphological evaluation for acrosome <strong>in</strong>tegrityand total morphology, FACS Analysis (FITC-PNA ⁄ Syto17 ⁄ PI; SCSA), and a general test ofmicrobiological contam<strong>in</strong>ation. These tests may beperformed with sorted sperm of the unwanted sex.Additionally, one straw from each batch of thewanted sex needs to be reanalysed for sort<strong>in</strong>g purityand the total number of spermatozoa per straw.Ballester et al. (2007) confirmed that it is not sufficientto test semen quality directly after thaw<strong>in</strong>g. Incubationof the samples at 38°C revealed a reduction ofl<strong>in</strong>earity, sperm viability (Sem<strong>in</strong>aphtharhodafluor)and acrosome <strong>in</strong>tegrity.It is recommended to put at least 2 million livespermatozoa <strong>in</strong> a straw as the absolute m<strong>in</strong>imum.Andersson et al. (2004, 2006) clearly showed thatmore than 2 million spermatozoa are necessary toobta<strong>in</strong> satisfactory pregnancy and calv<strong>in</strong>g rates.Caution must also be exercised to account for high<strong>in</strong>dividual bull effects. In order to avoid losses of thehighly diluted semen, the liquid column <strong>in</strong> the strawshould be divided <strong>in</strong>to a first segment conta<strong>in</strong><strong>in</strong>gÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Commercialization of Sex-Sorted Semen 341extender only, which will close the cotton plug, and asecond segment conta<strong>in</strong><strong>in</strong>g the sexed spermatozoa.Both segments need to be divided by an air bubble.Automatic systems to fill the straws <strong>in</strong> this manner arecommercially available.(4) It is recommended that only heifers are <strong>in</strong>sem<strong>in</strong>atedwith sex-sorted semen at the present time. Fromseveral studies on our research farm, we have no<strong>in</strong>dication that neither tim<strong>in</strong>g nor variability ofovulation nor site of <strong>in</strong>sem<strong>in</strong>ation is responsible forthe large differences <strong>in</strong> non-return rates betweenheifers and cows.On the female side, it is a prerequisite to provideoptimal herd management to atta<strong>in</strong> high female fertility(Seidel 2003a,b). Virtually, all fertility results to datefrom the AI of sex-sorted semen <strong>in</strong> cattle have been fromheifers. Us<strong>in</strong>g AI with sorted semen <strong>in</strong> cows frequentlyproduces unacceptable non-return rates, possibly becauseof the <strong>in</strong>ability to consistently p<strong>in</strong>po<strong>in</strong>t ovulationfor optimum AI.Several groups (Cran et al. 1994; Merton et al. 1997;Lu et al. 1999) reported similar cleavage rates butreduced blastocyst development follow<strong>in</strong>g fertilization<strong>in</strong> vitro with sorted bov<strong>in</strong>e sperm cells. In a recentstudy, we demonstrated for the first time that bov<strong>in</strong>eIVF-embryos derived from sex-sorted spermatozoadisplay a reduction <strong>in</strong> the relative abundance ofdevelopmentally important genes like Gluc-3 andG6PD, compared with their counterparts derived fromunsorted semen (Morton et al. 2007). It requiresfurther <strong>in</strong>-depth study as epigenetic changes alreadyoccurr<strong>in</strong>g <strong>in</strong> the early embryo are thought to be<strong>in</strong>volved <strong>in</strong> postnatal abnormalities. These are deleteriouseffects of sperm sex<strong>in</strong>g on spermatozoa beyondthose previously recognized and reflected <strong>in</strong> the developmentalcompetence of embryos. Similarly, cleavagerates after IVF with sex-sorted spermatozoa were 30%below those of unsorted spermatozoa of the sameejaculate, blastocyst formation on day 8 was 30–40%lower than for the controls (Bermejo-Alvarez et al.2007) and cell cycles were reduced (Beyhan et al. 1999)or disturbed <strong>in</strong> tim<strong>in</strong>g (Cran et al. 1993; Lu et al. 1999;Morton et al. 2005). On the contrary, Seidel et al.(1999) found no excess embryonic loss between 1 and2 months of gestation <strong>in</strong> heifers <strong>in</strong>sem<strong>in</strong>ated withsorted sperm.Commercial demandsThe demand for sex-sorted semen may vary betweencountries. Based on the American market situation,advantages are foreseeable for the decoupl<strong>in</strong>g ofproduction of replacement heifers and cows from thenumber of culled cows. More heifers are available forherd replacement and by this a stronger selection willbe possible. A higher degree of female selection willhave an impact on the genetic development of thepopulation as females will contribute with up to 15%on genetic selection, which was based so far on sires(Weigel and Barlass 2003). As the number of replacementanimals is satisfied, herd size may grow andmore females are sold. In parallel, the milk productioncan rise and the market price may decrease as benefitfor consumers. In addition, the costs for progenytest<strong>in</strong>g and embryo transfer will decrease and eventuallythe economic benefit for genetic markers will beenhanced as more sibl<strong>in</strong>gs of the same sex can beproduced of the best bulls and cows. The more thetechnology develops and more reasonable pric<strong>in</strong>g isable to be applied the more the prices for replacementand export heifers will decrease. Therefore, only theearly users will benefit most by sell<strong>in</strong>g surplus heifersat substantially higher prices than the cost to raise orreplace (De Vries et al. 2008). It is essential that ascommercialization <strong>in</strong>creases, more data on fertility,embryonic losses, foetal deformations, abortions andheifer health can be collected. Such <strong>in</strong>formation willhelp to evaluate the technology correctly for itspractical use.Status of commercial implementationThe technology is the most highly developed for bov<strong>in</strong>esemen and was <strong>in</strong>troduced <strong>in</strong>to commercial application<strong>in</strong> the United K<strong>in</strong>gdom <strong>in</strong> 2000. Recently, severalAmerican AI centres have contracted for the technologyand offer sexed semen from their bulls worldwide.As this manuscript is written, there are about 50sorters <strong>in</strong> the USA and three new centres <strong>in</strong> Europestart<strong>in</strong>g production of sexed bull semen. At the currentsituation, the high price for sexed semen may beadequate as long as the demand is high for heifers butits value will decrease with decreas<strong>in</strong>g milk, heifer andcull cow prices, with <strong>in</strong>creas<strong>in</strong>g feed costs and lowerpric<strong>in</strong>g of conventional semen. If the sex-sorted semenis highly fertile and female prices are $200 (approximately130€) above males, Seidel (2003a) considered anadditional price of $21.56 per AI dose as affordable formarket<strong>in</strong>g.In a recent study, Ettema et al. (2007) calculated thesituation <strong>in</strong> Denmark as an example for the Europeanmarket. They developed a spreadsheet model <strong>in</strong>clud<strong>in</strong>gprice for spr<strong>in</strong>g<strong>in</strong>g heifers, replacement costs, price perbeef calf, price of sexed semen, conception rates withsexed semen, replacement rates, sex ratio of sexedsemen, <strong>in</strong>cidence of dystocia and stillbirth. The ma<strong>in</strong>factors affect<strong>in</strong>g price are lowered fertility, the high costof equipment, necessity for skilled personnel and thecost for <strong>in</strong>tellectual property. From this data, it isobvious that the net return to assets (NRA) will benegative for the first year and it will take 3–4 yearsbefore herds have reached a steady state with respect torais<strong>in</strong>g and sell<strong>in</strong>g replacements.In summary, sex-sorted bull semen is <strong>in</strong> high demand.The success of a widespread application depends on itsprice. It has to be moderate enough to allow areasonable profit for farmers, and it has to be highenough to make its production lucrative (Seidel et al.2003). A prerequisite for <strong>in</strong>tegration <strong>in</strong> genetic programmesis the quality of the sorted semen. Underoptimal conditions, fertility is beg<strong>in</strong>n<strong>in</strong>g to reach satisfactorylevels though semen variability and managementfactors cont<strong>in</strong>ue to be critically important to success <strong>in</strong>commercial practice.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


342 D Rath and LA JohnsonThe Commercial Use of Ov<strong>in</strong>e Sex-SortedSemenStage of applicabilityIn sheep, <strong>in</strong>sem<strong>in</strong>ation with less than 1 million motileunsexed sperm has been demonstrated to result <strong>in</strong>acceptable fertility (Walker et al. 1984; 5 million motile,Eppleston et al. 1986) but the results vary considerably(Salamon and Maxwell 2000). Lambs from sex-sortedsemen were produced via laparoscopy by Cran et al.(1997). Subsequent experiments with 1–16 million sexsortedfrozen-thawed spermatozoa produced disappo<strong>in</strong>t<strong>in</strong>glevels of fertility (Holl<strong>in</strong>shead et al. 2002,2003). Even <strong>in</strong>sem<strong>in</strong>ations with up to 40 million totalsorted, cryopreserved spermatozoa were <strong>in</strong>conclusive(Holl<strong>in</strong>shead et al. 2003). However, <strong>in</strong> a very recentstudy us<strong>in</strong>g an adapted protocol, de Graaf et al. (2007c)demonstrated that dosages of 1 or 5 million sortedmotile ram sperm gave superior fertility to non-sortedsperm when laparoscopically <strong>in</strong>sem<strong>in</strong>ated. These resultshave significance for the future commercialization of sexpre-selection technology <strong>in</strong> sheep as a reduction <strong>in</strong> them<strong>in</strong>imum effective sperm number will allow a correspond<strong>in</strong>gdecrease <strong>in</strong> the associated cost per dose and setaside a larger percentage of the sheep <strong>in</strong>dustry to utilizesex pre-selection. In another trial, the same groupshowed for the first time for any species that frozenthawedspermatozoa, after sex-sort<strong>in</strong>g and a secondcryopreservation step are capable of produc<strong>in</strong>g offspr<strong>in</strong>gof the predicted sex follow<strong>in</strong>g AI (de Graaf et al. 2006).Sort<strong>in</strong>g of frozen semen and subsequent refreez<strong>in</strong>g withthe birth of lambs had only been described so far afterIVF and ET (Holl<strong>in</strong>shead et al. 2004). Sex-sorted ramsperm can be used effectively with <strong>in</strong>tra-cytoplasmics<strong>in</strong>gle sperm <strong>in</strong>jection (ICSI; Catt et al. 1996; Holl<strong>in</strong>sheadet al. 2002) and IVF (Rhodes et al. 1994; Maxwellet al. 2004; Morton et al. 2004). de Graaf et al. (2007a)also showed that the potential of sex-sorted frozenthawedram semen is equal to unsorted semen whenused for laparoscopic <strong>in</strong>sem<strong>in</strong>ation, or <strong>in</strong>trauter<strong>in</strong>e<strong>in</strong>sem<strong>in</strong>ation <strong>in</strong> superovulated ewes and subsequentembryo transfer of morula and blastocysts.Status of commercial implementationCommercialization of sex-sorted ram spermatozoa forlaparoscopical <strong>in</strong>sem<strong>in</strong>ation may play a role <strong>in</strong> the nearfuture, though it is questionable, whether its use will besufficiently profitable to make it rout<strong>in</strong>ely available. Thelatest results of the University of Sydney group (deGraaf et al. 2006, 2007c) are very promis<strong>in</strong>g and theimplementation of sex-sorted ram sperm <strong>in</strong> the commercialpractice may have a significant impact on theAustralian and New Zealand markets.The Commercial Use of Porc<strong>in</strong>e SexSortedSemenStage of applicabilityIn pr<strong>in</strong>ciple, porc<strong>in</strong>e sperm are as easy to sort as otherlivestock species. Its sort<strong>in</strong>g <strong>in</strong>dex (115) ranges betweenbull and ram sperm (Garner 2006). The primarylimitations are the large amount of sperm necessaryfor AI and the sensitivity of sorted sperm aga<strong>in</strong>st highdilution and cryopreservation. Factors associated withprocess<strong>in</strong>g boar sperm for sex<strong>in</strong>g lead to capacitationlike membrane changes (Ashworth et al. 1994; Maxwellet al. 1997, 1998; Barrios et al. 2000). Sem<strong>in</strong>al plasma asa constituent of the sample medium and of the catchmedium has been shown to be beneficial to decapacitatesorted spermatozoa (Caballero et al. 2004).Embryos from IVF with sexed boar semen were firstproduced 15 years ago (Rath et al. 1993). Subsequently,offspr<strong>in</strong>g were produced after sex-sort<strong>in</strong>g and IVF ofnon-frozen spermatozoa (Rath and Niemann 1996,1997; Abeydeera et al. 1998). In addition, ICSI hasbeen used successfully to produce male offspr<strong>in</strong>g (Probstand Rath 2003).A major step forward was made with the idea of lowdose<strong>in</strong>sem<strong>in</strong>ation (for review see Rath 2002) and withthe development of a flexible catheter (Firflex Ò , M<strong>in</strong>itu¨b, Tiefenbach, Germany) for deep <strong>in</strong>trauter<strong>in</strong>e <strong>in</strong>sem<strong>in</strong>ation(DIU) <strong>in</strong> Spa<strong>in</strong> (Mart<strong>in</strong>ez et al. 2001, 2002;Cuello et al. 2005). Progeny have been produced afterDIU of non-frozen bulk-sorted (Vazquez et al. 2003)and sex-sorted non-frozen sperm (Rath et al. 2003;Grossfeld et al. 2005), us<strong>in</strong>g as few as 50 · 10 6 spermatozoa<strong>in</strong> a volume of 2 ml (Rath et al. 2003). However,the method is only available for sows and not forprimiparous gilts. Plac<strong>in</strong>g the semen directly <strong>in</strong> front ofthe uterotubal junction may avoid major sperm lossesdur<strong>in</strong>g uter<strong>in</strong>e transportation. Unilateral <strong>in</strong>sem<strong>in</strong>ation issufficient to f<strong>in</strong>d adequate number of embryos <strong>in</strong> bothhorns (Tummaruk et al. 2007).Boar semen has been sex-sorted, frozen, thawed andsurgically <strong>in</strong>sem<strong>in</strong>ated <strong>in</strong>to the oviduct to produce liveoffspr<strong>in</strong>g (Johnson et al. 2000). However, a repeatablemethod for freez<strong>in</strong>g sex-sorted boar sperm rema<strong>in</strong>s to bedeveloped. Although attempts to freeze sex-sorted boarsemen resulted <strong>in</strong> sufficient post-thaw sperm quality, nopregnancies were able to go to term after non-surgicalDUI (Bathgate et al. 2008). Similarly, embryos derivedfrom IVF with sex-sorted frozen sperm and transferrednon-surgically did not develop to term (Bathgate et al.2007).Commercial demandsIn sw<strong>in</strong>e, the greatest general demand for sex-sortedsemen is to produce female piglets for meat production.Several European countries will soon beg<strong>in</strong> to forbidcastration of male piglets; therefore a shift <strong>in</strong> sex ratioswill be a necessity for pig producers under suchregulations.Status of commercial implementationThe Beltsville Sperm Sex<strong>in</strong>g Technology, as it iscurrently used <strong>in</strong> research and commercial practice, isunable to produce sufficient numbers of sperm to makesw<strong>in</strong>e AI feasible at the present time. In its current stageof development, sex-sorted pig sperm could be used <strong>in</strong>specialized commercial practice, e.g. nuclear herds, ifcomb<strong>in</strong>ed with IVF ⁄ ET and ⁄ or laparoscopic <strong>in</strong>sem<strong>in</strong>ation(Rath et al. 1999; Vazquez et al. 2006; Garcı´ a et al.2007). Optimum utilization of sex-sorted boar spermÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Commercialization of Sex-Sorted Semen 343would suggest that it would be advantageous andessential to freeze sex-sorted boar spermatozoa so thatthe greatest flexibility for <strong>in</strong>sem<strong>in</strong>ation and ovulationcan be exercised.The Commercial Use of Equ<strong>in</strong>e Sex-SortedSemenSemen from horses can be sorted even though theefficiency is lower than <strong>in</strong> other livestock species.However, the value of a s<strong>in</strong>gle dose may cover allproduction costs. Buchanan et al. (2000) reported a40% pregnancy rate <strong>in</strong> mares after AI of 25 · 10 6 sexsortednon-frozen spermatozoa and pregnancies haveeven been obta<strong>in</strong>ed with as few as 5 · 10 6 liquid orfrozen-thawed spermatozoa us<strong>in</strong>g hysteroscopic <strong>in</strong>sem<strong>in</strong>ation(L<strong>in</strong>dsey et al. 2002a, b). Recently, a filly wasborn after hysteroscopic <strong>in</strong>sem<strong>in</strong>ation with low numbersof frozen-thawed sexed spermatozoa <strong>in</strong>to the uterotubaljunction. Highly diluted unsorted semen from the sameejaculates did no better. Undiluted semen <strong>in</strong>sem<strong>in</strong>ated ata dose of 5 · 10 8 spermatozoa yielded satisfactoryconception rates (Clulow et al. 2007). Therefore, thepoor fertility after hysteroscopic <strong>in</strong>sem<strong>in</strong>ation with lowdoses of sex-sorted or non-sorted spermatozoa may bedirectly attributable to the low-dose <strong>in</strong>sem<strong>in</strong>ation conditionswith frozen-thawed rather than sex-sorted spermatozoa.In order to improve the post-thaw quality ofsex-sorted stallion semen, different approaches werefocused on cushion centrifugation before and aftersort<strong>in</strong>g as well as pre-selection of a stable spermpopulation us<strong>in</strong>g a PureSperm gradient (Knop et al.2005; Heer 2007) and different automated freez<strong>in</strong>gprotocols (Buss 2006; Clulow et al. 2007). The qualityof the sorted frozen semen was improved stepwise and<strong>in</strong>sem<strong>in</strong>ation trials have yet to be performed. The majordifficulty with equ<strong>in</strong>e semen is its variability amongstallions and ejaculates. The commercialization of sexedstallion semen will depend on the consistency of thefertility after sort<strong>in</strong>g and freez<strong>in</strong>g, a broader number ofstallions with sortable ejaculates and improvements ofthe current hysteroscopic <strong>in</strong>sem<strong>in</strong>ation technique.ReferencesAbeydeera LR, Johnson LA, Welch GR, Wang WH, BoquestAC, Cantley TC, Rieke A, Day BN, 1998: Birth of pigletspreselected for gender follow<strong>in</strong>g <strong>in</strong> vitro fertilization of <strong>in</strong>vitro matured pig oocytes by X and Y chromosome bear<strong>in</strong>gspermatozoa sorted by high speed flow cytometry. Theriogenology50, 981–988.Amann RP, 1999: Issues affect<strong>in</strong>g commercialization of sexedsperm. Theriogenology 52, 1441–1457.Andersson M, Taponen J, Kosk<strong>in</strong>en E, Dahlbom M, 2004:Effect of <strong>in</strong>sem<strong>in</strong>ation with doses of 2 or 15 million frozenthawedspermatozoa and semen deposition site on pregnancyrate <strong>in</strong> dairy cows. Theriogenology 61, 1583–1588.Andersson M, Taponen J, Kommeri M, Dahlbom M, 2006:Pregnancy rates <strong>in</strong> lactat<strong>in</strong>g Holste<strong>in</strong>-Friesian cows afterartificial <strong>in</strong>sem<strong>in</strong>ation with sexed sperm. Reprod DomestAnim 41, 95–97.Ashworth PJC, Harrison RAP, Miller NGA, Plummer JM,Watson PF, 1994: Survival of ram spermatozoa at highdilution: protective effect of simple constituents of culturemedia as compared with sem<strong>in</strong>al plasma. Reprod Fertil Dev6, 173–180.Ballester J, Johannisson A, Saravia F, Håård M, GustafssonH, Bajramovic D, Rodriguez-Mart<strong>in</strong>ez H, 2007: Post-thawviability of bull AI-doses with low-sperm numbers. Theriogenology68, 934–943.Barrios B, Pérez-Pe´ R, Gallego M, Tato A, Osada J, Mu<strong>in</strong>˜ o-Blanco T, Cebrián-Pérez JA, 2000: Sem<strong>in</strong>al plasma prote<strong>in</strong>srevert the cold-shock damage on ram sperm membrane. BiolReprod 63, 1531–1537.Bathgate R, Morton KM, Eriksson BM, Rath D, Sieg B,Maxwell WM, Evans G, 2007: Non-surgical deep <strong>in</strong>trauter<strong>in</strong>etransfer of <strong>in</strong> vitro produced porc<strong>in</strong>e embryosderived from sex-sorted frozen-thawed boar sperm. AnimReprod Sci 99, 82–92.Bathgate R, Grossfeld R, Susetio D, Ruckholdt M, HeasmanK, Rath D, Evans G, Maxwell WM, 2008: Early pregnancyloss <strong>in</strong> sows after low dose, deep uter<strong>in</strong>e artificial <strong>in</strong>sem<strong>in</strong>ationwith sex-sorted, frozen-thawed sperm. Anim ReprodSci 104, 440–444.Bermejo-Alvarez P, Rizos D, Rath D, Lonergan P, Gutierrez-Adan A, 2007: Epigenetic differences between male andfemale bov<strong>in</strong>e blastocysts produced <strong>in</strong> vitro. Physiol Genomics17, 264–272.Beyhan Z, Johnson LA, First NL, 1999: Sexual dimorphism <strong>in</strong>IVM-IVF bov<strong>in</strong>e embryos produced from X and Y chromosome-bear<strong>in</strong>gspermatozoa sorted by high speed flowcytometry. Theriogenology 52, 35–48.Boe-Hansen GB, Morris ID, Annette B, Ersbølla K, Greve T,Christensen P, 2005: DNA <strong>in</strong>tegrity <strong>in</strong> sexed bull spermassessed by neutral Comet assay and sperm chromat<strong>in</strong>structure assay. Theriogenology 63, 1789–1802.Buchanan BR, Seidel GE, McCue PM, Schenk JL, HerickhoffLA, Squires EL, 2000: Insem<strong>in</strong>ation of mares with lownumbers of either unsexed or sexed spermatozoa. Theriogenology53, 1309–1321.Buss H, 2006: Improvement of the freezability of sex-sortedstallion spermatozoa. Thesis, University of Veter<strong>in</strong>aryMedic<strong>in</strong>e Hannover, Hannover, Germany. (<strong>in</strong> German).Caballero I, Vazquez JM, Centurio´ n F, Rodrı´ guez-Mart<strong>in</strong>ezH, Parrilla I, Roca J, Cuello C, Mart<strong>in</strong>ez EA, 2004:Comparative effects of autologous and homologous sem<strong>in</strong>alplasma on the viability of largely extended boar spermatozoa.Reprod Domest Anim 39, 370–375.Campos-Chillon LF, de la Torre JF, 2003: Effect on concentrationof sexed bov<strong>in</strong>e sperm sorted at 40 and 50 psi ondevelopmental capacity of <strong>in</strong> vitro produced embryos.Theriogenology 59, 506. (Abstract).Catt SL, Catt JW, Gomez MC, Maxwell WMC, Evans G,1996: Birth of a male lamb derived from <strong>in</strong> vitro maturedoocyte fertilized by <strong>in</strong>tracytoplasmic <strong>in</strong>jection of a s<strong>in</strong>gle‘male’ sperm. Vet Rec 139, 494–495.Catt SL, Sakkas D, Bizzaro D, Bianchi PG, Maxwell WMC,Evans G, 1997: Hoechst sta<strong>in</strong><strong>in</strong>g and exposure to UV laserdur<strong>in</strong>g flow cytometric sort<strong>in</strong>g does not affect the frequencyof detected endogenous DNA nicks <strong>in</strong> abnormal and normalhuman sperm. Mol Hum Reprod 3, 821–825.Centurion F, Vazquez JM, Calvete JJ, Roca J, Sanz L, ParrillaI, Garcia EM, Mart<strong>in</strong>ez EA, 2003: Influence of porc<strong>in</strong>espermadhes<strong>in</strong>s on the susceptibility of boar spermatozoa tohigh dilution. Biol Reprod 69, 640–646.Clulow JR, Buss H, Sieme H, Rodger JA, Cawdell-Smith AJ,Evans G, Rath D, Morris LH, Maxwell WM2007: Fieldfertility of sex-sorted and non-sorted frozen-thawed stallionspermatozoa. Anim Reprod Sci. doi:10.1016/j.anireprosci.2007.08.015.(Epub ahead of pr<strong>in</strong>t)Cran DG, Johnson LA, Miller NGA, Cochrane D, Polge CE,1993: Production of bov<strong>in</strong>e calves follow<strong>in</strong>g separation ofÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


344 D Rath and LA JohnsonX- and Y-bear<strong>in</strong>g sperm and <strong>in</strong> vitro fertilization. Vet Rec132, 40–41.Cran DG, Cochrane DJ, Johnson LA, Wei H, Lu KH, PolgeC, 1994: Separation of X- and Y-chromosome bear<strong>in</strong>gbov<strong>in</strong>e sperm by flow cytometry for use <strong>in</strong> IVF. Theriogenology41, 183. (Abstract).Cran DG, McKelvey WAC, K<strong>in</strong>g ME, Dolman DF, McEvoyTG, Broadbent PJ, Rob<strong>in</strong>son JJ, 1997: Production of lambsby low dose <strong>in</strong>trauter<strong>in</strong>e <strong>in</strong>sem<strong>in</strong>ation with flow cytometricallysorted and unsorted sperm. Theriogenology 47, 267.(Abstract).Cuello C, Berthelot F, Mart<strong>in</strong>at-Botte´ F, Venturi E, GuillouetP, Vázquez JM, Roca J, Martı´ nez EA, 2005: Piglets bornafter non-surgical deep <strong>in</strong>trauter<strong>in</strong>e transfer of vitrifiedblastocysts <strong>in</strong> gilts. Anim Reprod Sci 85, 275–286.De Ambrogi M, Sp<strong>in</strong>aci M, Galeati G, Taman<strong>in</strong>i C, 2006:Viability and DNA fragmentation <strong>in</strong> differently sorted boarspermatozoa. Theriogenology 66, 1994–2000.De Vries A, Overton M, Fetrow J, Leslie K, Eicker S, RogersG, 2008: Explor<strong>in</strong>g the impact of sexed semen on thestructure of the dairy <strong>in</strong>dustry. J Dairy Sci 91, 847–856.Den Daas JH, De Jong G, Lansbergen LM, Van Wagtendonk-De Leeuw AM, 1998: The relationship between thenumber of spermatozoa <strong>in</strong>sem<strong>in</strong>ated and the reproductiveefficiency of <strong>in</strong>dividual dairy bulls. J Dairy Sci 81, 1714–1723.Eppleston J, Maxwell WMC, Battye KM, Roberts EM,1986: Effect of thawed motility and <strong>in</strong>trauter<strong>in</strong>e dose ofmotile sperm on fertility <strong>in</strong> ewes. Proc Aust Soc ReprodBiol 18, 19.Ettema JF, Hoag DL, Seidel DE Jr, 2007: Economic opportunitiesfor sexed semen on commercial dairies. West DairyNews 7, 67–68.Foote RH, Miller P, 1971: What might sex ratio control mean<strong>in</strong> the animal world? In: Kiddy CA, Hafs HD(eds), SexRatio at Birth Prospects for Control. American Society ofAnimal Science, Champaign, pp. 1–9.Garcı´ a EM, Vázquez JM, Parrilla I, Calvete JJ, Sanz L,Caballero I, Roca J, Vazquez JL, Martı´ nez EA, 2007:Improv<strong>in</strong>g the fertiliz<strong>in</strong>g ability of sex sorted boar spermatozoa.Theriogenology 68, 771–778.Garner DL, 2006: Flow cytometric sex<strong>in</strong>g of mammaliansperm. Theriogenology 65, 943–957.Garner DL, Seidel GE Jr, 2008: History of commercializ<strong>in</strong>gsexed semen for cattle. Theriogenology 69, 886–895.de Graaf SP, Evans G, Maxwell WM, O’Brien JK, 2006: Invitro characteristics of fresh and frozen-thawed ram spermatozoaafter sex-sort<strong>in</strong>g and re-freez<strong>in</strong>g. Reprod FertilDev 18, 867–874.de Graaf SP, Beilby K, O’Brien JK, Osborn D, Down<strong>in</strong>g JA,Maxwell WM, Evans G, 2007a: Embryo production fromsuperovulated sheep <strong>in</strong>sem<strong>in</strong>ated with sex-sorted ram spermatozoa.Theriogenology 67, 550–555.de Graaf SP, Evans G, Gillan L, Guerra MMP, Maxwell WM,O’Brien JK, 2007b: The <strong>in</strong>fluence of antioxidant, cholesteroland sem<strong>in</strong>al plasma on the <strong>in</strong> vitro quality of sortedand non-sorted ram spermatozoa. Theriogenology 67, 217–227.de Graaf SP, Evans G, Maxwell WM, Down<strong>in</strong>g JA, O’BrienJK, 2007c: Successful low dose <strong>in</strong>sem<strong>in</strong>ation of flowcytometrically sorted ram spermatozoa <strong>in</strong> sheep. ReprodDomest Anim 42, 648–653.Grossfeld R, Kl<strong>in</strong>c P, Sieg B, Rath D, 2005: Production ofpiglets with sexed semen employ<strong>in</strong>g a non-surgical <strong>in</strong>sem<strong>in</strong>ationtechnique. Theriogenology 63, 2269–2277.Guthrie HD, Johnson LA, Garrett WM, Welch GR, Dobr<strong>in</strong>skyJR, 2002: Flow cytometric sperm sort<strong>in</strong>g: effects ofvary<strong>in</strong>g laser power on embryo development <strong>in</strong> sw<strong>in</strong>e. MolReprod Dev 61, 87–92.Heer P, 2007: Adjustment of cryopreservation of stallionspermatozoa to the Beltsville Sperm Sex<strong>in</strong>g Technology.Doctorial Thesis, University of Veter<strong>in</strong>ary Medic<strong>in</strong>e Hannover,Hannover, Germany (<strong>in</strong> German).Holl<strong>in</strong>shead FK, O’Brien JK, Maxwell WMC, Evans G, 2002:Production of lambs of predeterm<strong>in</strong>ed sex after the <strong>in</strong>sem<strong>in</strong>ationof ewes with low numbers of frozen-thawed sortedX- or Y-chromosome-bear<strong>in</strong>g spermatozoa. Reprod FertilDev 14, 503–508.Holl<strong>in</strong>shead FK, Gillian L, O’Brien JK, Evans G, MaxwellWMC, 2003: In vitro and <strong>in</strong> vivo assessment of functionalcapacity of flow cytometrically sorted ram spermatozoaafter freez<strong>in</strong>g and thaw<strong>in</strong>g. Reprod Fertil Dev 15, 351–359.Holl<strong>in</strong>shead FK, Evans G, Evans KM, Catt SL, MaxwellWMC, O’Brien JK, 2004: Birth of lambs of a predeterm<strong>in</strong>edsex after <strong>in</strong> vitro production of embryos us<strong>in</strong>gfrozen-thawed sex-sorted and re-frozen-thawed ram spermatozoa.<strong>Reproduction</strong> 127, 557–568.Johnson LA, 1991: Sex preselection <strong>in</strong> sw<strong>in</strong>e: altered sex ratios<strong>in</strong> offspr<strong>in</strong>g follow<strong>in</strong>g surgical <strong>in</strong>sem<strong>in</strong>ation of flow sortedX- and Y-bear<strong>in</strong>g sperm. Reprod Domest Anim 26, 309–314.Johnson LA, P<strong>in</strong>kel D, 1986: Modification of a laser-basedflow cytometer for high-resolution DNA analysis of mammalianspermatozoa. Cytometry 7, 268–273.Johnson LA, Welch GR, 1999: Sex preselection: high-speedflow cytometric sort<strong>in</strong>g of X- and Y-sperm for maximumefficiency. Theriogenology 52, 1134–1323.Johnson LA, Flook JP, Look MV, 1987a: Flow cytometry ofX and Y chromosome-bear<strong>in</strong>g sperm for DNA us<strong>in</strong>g animproved preparation method and sta<strong>in</strong><strong>in</strong>g with Hoechst33342. Gamete Res 17, 203–212.Johnson LA, Flook JP, Look MV, P<strong>in</strong>kel D, 1987b: Flowsort<strong>in</strong>g of X and Y chromosome-bear<strong>in</strong>g spermatozoa <strong>in</strong>totwo populations. Gamete Res 16, 1–9.Johnson LA, Flook JP, Hawk HW, 1989: Sex preselection <strong>in</strong>rabbits: live births from X and Y sperm separated by DNAand cell sort<strong>in</strong>g. Biol Reprod 41, 199–203.Johnson LA, Cran DG, Welch GR, Polge C, 1996: Genderpreselection <strong>in</strong> mammals. In: Miller RH, Pursel VG,Norman HD(eds), Beltsville Symposium XX: Biotechnology’sRole <strong>in</strong> the Genetic Improvement of Farm <strong>Animals</strong>.American Society of Animal Science, Savoy, pp. 151–164.Johnson LA, Welch GR, Rens W, 1999: The Beltsville spermsex<strong>in</strong>g technology: high-speed sperm sort<strong>in</strong>g gives improvedsperm output for <strong>in</strong> vitro fertilization and AI. J Anim Sci77(Suppl. 2), 213–220.Johnson LA, Guthrie HD, Fiser P, Maxwell WMC, WelchGR, Garrett WM2000: Cryopreservation of flow cytometricallysorted boar sperm: effects on <strong>in</strong> vivo embryodevelopment. J Anim Sci 78 (Suppl. 1), 198. (Abstract).Johnson LA, Rath D, Vazquez JM, Maxwell WMC, Dobr<strong>in</strong>skyJR, 2005: Preselection of sex of offspr<strong>in</strong>g <strong>in</strong> sw<strong>in</strong>e forproduction: current status of the process and its application.Theriogenology 63, 615–624.Kl<strong>in</strong>c P, 2005: Improved fertility of flow cytometrically sexselected bull spermatozoa. Doctoral thesis, University ofVeter<strong>in</strong>ary Medic<strong>in</strong>e Hannover, Hannover, Germany.Kl<strong>in</strong>c P, Rath D, 2007: Reduction of oxidative stress <strong>in</strong> bov<strong>in</strong>espermatozoa dur<strong>in</strong>g flow cytometric sort<strong>in</strong>g. Reprod DomestAnim 42, 63–67.Kl<strong>in</strong>c P, Frese D, Osmers H, Rath D, 2007: Insem<strong>in</strong>ation withsex sorted fresh bov<strong>in</strong>e spermatozoa processed <strong>in</strong> thepresence of antioxidative substances. Reprod Domest Anim42, 58–62.Knop K, Hoffmann N, Rath D, Sieme H, 2005: Effects ofcushioned centrifugation technique on sperm recovery andsperm quality <strong>in</strong> stallions with good and poor semenfreezability. Anim Reprod Sci 89, 294–297.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Commercialization of Sex-Sorted Semen 345L<strong>in</strong>dsey AC, Morris LHA, Allen WR, Schenk JL, Squires EL,Bruemmer JE, 2002a: Hysteroscopic <strong>in</strong>sem<strong>in</strong>ation of mareswith low numbers of nonsorted or flow sorted spermatozoa.Equ<strong>in</strong>e Vet J 34, 128–132.L<strong>in</strong>dsey AC, Schenk JL, Graham JK, Bruemmer JE, SquiresEL, 2002b: Hysteroscopic <strong>in</strong>sem<strong>in</strong>ation of low numbers offlow sorted fresh and frozen ⁄ thawed stallion spermatozoa.Equ<strong>in</strong>e Vet J 34, 121–127.Lu KH, Cran DG, Seidel GE, 1999: In vitro fertilization withflow-cytometrically sorted bov<strong>in</strong>e sperm. Theriogenology52, 1393–1405.Mart<strong>in</strong>ez EA, Vazquez JM, Roca J, Lucas X, Gil MA, ParrillaI, 2001: Successful non-surgical deep <strong>in</strong>trauter<strong>in</strong>e <strong>in</strong>sem<strong>in</strong>ationwith small numbers of spermatozoa <strong>in</strong> sows. <strong>Reproduction</strong>122, 289–296.Mart<strong>in</strong>ez EA, Vazquez JM, Roca J, Lucas X, Gil MA, ParrillaI, Vazquez JL, Day BN, 2002: M<strong>in</strong>imum number ofspermatozoa required for normal fertility after deep <strong>in</strong>trauter<strong>in</strong>e<strong>in</strong>sem<strong>in</strong>ation <strong>in</strong> non-sedated sows. <strong>Reproduction</strong>123, 163–170.Maxwell WMC, Johnson LA, 1999: Physiology of spermatozoaat high dilution rates: the <strong>in</strong>fluence of sem<strong>in</strong>al plasma.Theriogenology 52, 1353–1362.Maxwell WMC, Welch GR, Johnson LA, 1997: Viability andmembrane <strong>in</strong>tegrity of spermatozoa after dilution and flowcytometric sort<strong>in</strong>g <strong>in</strong> the presence or absence of sem<strong>in</strong>alplasma. Reprod Fertil Dev 8, 1165–1178.Maxwell WMC, Long CR, Johnson LA, Dobr<strong>in</strong>sky JR, WelchGR, 1998: The relationship between membrane status andfertility of boar spermatozoa after flow cytometric sort<strong>in</strong>g <strong>in</strong>the presence or absence of sem<strong>in</strong>al plasma. Reprod FertilDev 10, 433–440.Maxwell WMC, Evans G, Holl<strong>in</strong>shead FK, Bathgate R, deGraaf SP, Eriksson BM, Gillan L, Morton KM, O’BrienJK, 2004: Integration of sperm sex<strong>in</strong>g technology <strong>in</strong>to theART toolbox. Anim Reprod Sci 82, 79–95.Maxwell WM, Parrilla I, Caballero I, Garcia E, Roca J,Mart<strong>in</strong>ez EA, Vazquez JM, Rath D, 2007: Reta<strong>in</strong>edfunctional <strong>in</strong>tegrity of bull spermatozoa after double freez<strong>in</strong>gand thaw<strong>in</strong>g us<strong>in</strong>g PureSperm density gradient centrifugation.Reprod Domest Anim 42, 489–494.Merton JS, Har<strong>in</strong>g RM, Stap J, Hoebe RA, Aten JA, 1997:Effect of flow cytometrically sorted frozen ⁄ thawed semen onsuccess rate of <strong>in</strong> vitro bov<strong>in</strong>e embryo production. Theriogenology47, 295. (Abstract).Moce´ E, Graham JK, Schenk JL, 2006: Effect of sex-sort<strong>in</strong>gon the ability of fresh and cryopreserved bull sperm toundergo an acrosome reaction. Theriogenology 66, 929–936.Morton KM, Catt SL, Holl<strong>in</strong>shead FK, Maxwell WMC,Evans G2004: Lambs born after <strong>in</strong> vitro embryo productionfrom prepubertal lamb oocytes and frozen-thawed unsortedand sex-sorted spermatozoa. Reprod Fertil Dev 16, 260.(Abstract).Morton KM, Catt SL, Maxwell WM, Evans G, 2005: Anefficient method of ovarian stimulation and <strong>in</strong> vitro embryoproduction from prepubertal lambs. Reprod Fertil Dev 17,701–706.Morton KM, Herrmann D, Sieg B, Struckmann C, MaxwellWM, Rath D, Evans G, Lucas-Hahn A, Niemann H,Wrenzycki C, 2007: Altered mRNA expression patterns <strong>in</strong>bov<strong>in</strong>e blastocysts after fertilisation <strong>in</strong> vitro us<strong>in</strong>g flowcytometricallysex-sorted sperm. Mol Reprod Dev 74, 931–940.Parrilla I, Vazquez JM, Cuello C, Gil MA, Roca J, DiBerard<strong>in</strong>o D, Martınez EM, 2004: Hoechst 33342 sta<strong>in</strong>and u.v. laser exposure do not <strong>in</strong>duce genotoxic effects<strong>in</strong> flow-sorted boar spermatozoa. <strong>Reproduction</strong> 128, 615–621.Probst S, Rath D, 2003: Production of piglets us<strong>in</strong>g <strong>in</strong>tracytoplasmicsperm <strong>in</strong>jection (ICSI) with flowcytometricallysorted boar semen and artificially activated oocytes. Theriogenology59, 961–973.Rath D, 2002: Low dose <strong>in</strong>sem<strong>in</strong>ation <strong>in</strong> the sow. A review.Reprod Domest Anim 37, 1291–1304.Rath D, Niemann H, 1996: In vitro production of sw<strong>in</strong>eembryos. Dtsch Tierarztl Wochenschr 103, 331–336.Rath D, Niemann H, 1997: In vitro fertilization of porc<strong>in</strong>eoocytes with fresh and frozen-thawed ejaculated or frozenthawedepididymal semen obta<strong>in</strong>ed from identical boars.Theriogenology 47, 785–793.Rath D, Johnson LA, Welch GR, 1993: In vitro culture ofporc<strong>in</strong>e embryos: development to blastocysts after <strong>in</strong> vitrofertilization (IVF) with flow cytometrically sorted andunsorted semen. Theriogenology 39, 293. (Abstract).Rath D, Long CR, Dobr<strong>in</strong>sky JR, Welch GR, Schreier LL,Johnson LA, 1999: In vitro production of sexed embryos forgender preselection: high-speed sort<strong>in</strong>g of X-chromosomebear<strong>in</strong>gsperm to produce pigs after embryo transfer. J AnimSci 77, 3346–3352.Rath D, Ruiz S, Sieg B, 2003: Birth of female piglets follow<strong>in</strong>g<strong>in</strong>trauter<strong>in</strong>e <strong>in</strong>sem<strong>in</strong>ation of a sow us<strong>in</strong>g flow cytometricallysexed boar semen. Vet Rec 152, 400–401.Rhodes SL, Maxwell WMC, Evans G1994: DNA analysis andsort<strong>in</strong>g of ram spermatozoa by flow cytometry andsubsequent fertilization of <strong>in</strong> vitro matured sheep oocytes.In: Proceed<strong>in</strong>gs of the 7th International Symposium ofSpermatology, Cairns, Australia, pp. 9.26. (Abstract).Salamon S, Maxwell WMC, 2000: Storage of ram semen.Anim Reprod Sci 62, 77–111.Schenk JL, Seidel GE Jr, 2007: Pregnancy rates <strong>in</strong> cattle withcryopreserved sexed spermatozoa: effects of laser <strong>in</strong>tensity,sta<strong>in</strong><strong>in</strong>g conditions and catalase. Soc Reprod Fertil Suppl64, 165–177.Seidel GE Jr, 2003a: Economics of select<strong>in</strong>g for sex: the mostimportant genetic trait. Theriogenology 59, 585–598.Seidel GE Jr, 2003b: Sex<strong>in</strong>g mammalian sperm-<strong>in</strong>tertw<strong>in</strong><strong>in</strong>g ofcommerce, technology, and biology. Anim Reprod Sci 79,145–156.Seidel GE Jr, Allen CH, Johnson LA, Holland MD, Br<strong>in</strong>k Z,Welch GR, Graham JK, Cattell MB, 1997: Uter<strong>in</strong>e horn<strong>in</strong>sem<strong>in</strong>ation of heifers with very low numbers of non-frozenand sexed spermatozoa. Theriogenology 48, 1255–1264.Seidel GE Jr, Schenk JL, Herickhoff LA, Doyle SP, Br<strong>in</strong>k Z,Green RD, Cran DG, 1999: Insem<strong>in</strong>ation of heifers withsexed sperm. Theriogenology 52, 1407–1420.Seidel GE Jr, Br<strong>in</strong>k Z, Schenk JL, 2003: Use of heterospermic<strong>in</strong>sem<strong>in</strong>ation with fetal sex as the genetic marker to studyfertility of sexed sperm. Theriogenology 59, 515.Sp<strong>in</strong>aci M, De Ambrogi M, Volpe S, Galeati G, Taman<strong>in</strong>i C,Seren E, 2005: Effect of sta<strong>in</strong><strong>in</strong>g and sort<strong>in</strong>g on boar spermmembrane <strong>in</strong>tegrity, mitochondrial activity and <strong>in</strong> vitroblastocyst development. Theriogenology 64, 191–201.Suh TK, Schenk JL, Seidel GE Jr, 2005: High pressure flowcytometric sort<strong>in</strong>g damages sperm. Theriogenology 64,1035–1048.Tummaruk P, Sumransap P, Techakumphu M, KunavongkritA, 2007: Distribution of spermatozoa and embryos <strong>in</strong> thefemale reproductive tract after unilateral deep <strong>in</strong>tra uter<strong>in</strong>e<strong>in</strong>sem<strong>in</strong>ation <strong>in</strong> the pig. Reprod Domest Anim 42, 603–609.Vazquez JM, Mart<strong>in</strong>ez EA, Parrilla I, Roca J, Gil MA,Vazquez JL, 2003: Birth of piglets after deep <strong>in</strong>trauter<strong>in</strong>e<strong>in</strong>sem<strong>in</strong>ation with flow cytometrically sorted boar spermatozoa.Theriogenology 59, 1605–1614.Vazquez JM, Mart<strong>in</strong>ez EA, Parrilla I, Cuello C, Gil MA,Garcia E, 2006: Improv<strong>in</strong>g the efficiency of laparoscopic<strong>in</strong>traoviductal <strong>in</strong>sem<strong>in</strong>ation with sex sorted boar spermatozoa.Reprod Fertil Dev 18, 283. (Abstract).Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


346 D Rath and LA JohnsonWalker SK, Smith DH, Little DL, Warnes GM, Qu<strong>in</strong>n P,Seamark RF, 1984: Artificial <strong>in</strong>sem<strong>in</strong>ation and transfer ofembryos by laparoscopy. In: L<strong>in</strong>dsay DR, Pearce DT(eds),<strong>Reproduction</strong> <strong>in</strong> Sheep. Australian Academy of Science andAustralian Wool Corporation, Canberra, pp. 306–309.Weigel KA, Barlass KA, 2003: Results of a producer surveyregard<strong>in</strong>g crossbreed<strong>in</strong>g on US dairy farms. J Dairy Sci 86,4148–4154.Welch GR, Johnson LA, 1999: Sex preselection: laboratoryvalidation of the sperm sex ratio of flow sorted X- and Y-sperm by sort reanalysis for DNA. Theriogenology 52,1343–1352.Author’s address (for correspondence): D. Rath, Institute of FarmAnimal Genetics, Mariensee (FLI), 31535 Neustadt, Germany.E-mail: detlef.rath@fli.bund.deConflict of <strong>in</strong>terest: All authors declare no conflict of <strong>in</strong>terests.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 347–354 (2008); doi: 10.1111/j.1439-0531.2008.01183.xISSN 0936-6768Low-Dose Insem<strong>in</strong>ation <strong>in</strong> Pigs: Problems and PossibilitiesJM Vazquez, J Roca, MA Gil, C Cuello, I Parrilla, I Caballero, JL Vazquez and EA Martı´nezDepartment of Animal Medic<strong>in</strong>e and Surgery, Faculty of Veter<strong>in</strong>ary Medic<strong>in</strong>e, Campus de Esp<strong>in</strong>ardo, University of Murcia, Murcia, Spa<strong>in</strong>ContentsLow-dose AI procedures are required by the pig <strong>in</strong>dustry toefficiently utilize emerg<strong>in</strong>g sperm technologies, such ascryopreservation and sex-sort<strong>in</strong>g. Currently, several differentprocedures for <strong>in</strong>sem<strong>in</strong>at<strong>in</strong>g with a low or very low numberof spermatozoa have been described. Deep <strong>in</strong>trauter<strong>in</strong>e<strong>in</strong>sem<strong>in</strong>ation allows the deposition of the spermatozoa <strong>in</strong>the depth of the uter<strong>in</strong>e horn, allow<strong>in</strong>g a significantreduction <strong>in</strong> the number of spermatozoa <strong>in</strong>sem<strong>in</strong>ated withma<strong>in</strong>tenance of optimal reproductive performance. Intraoviductallaparoscopic <strong>in</strong>sem<strong>in</strong>ation has been recentlyapplied <strong>in</strong> pigs. This technique has proved to be applicablewith diluted and sex-sorted spermatozoa. This reviewdiscusses several problems encountered dur<strong>in</strong>g the developmentof deep <strong>in</strong>trauter<strong>in</strong>e <strong>in</strong>sem<strong>in</strong>ation and <strong>in</strong>tra-oviductallaparoscopic <strong>in</strong>sem<strong>in</strong>ation of pigs and provides potentialsolutions for the practical application of both the technologies.IntroductionLow-dose <strong>in</strong>sem<strong>in</strong>ation techniques are recommended <strong>in</strong>pigs when the available number of spermatozoa islimited, s<strong>in</strong>ce standard pig artificial <strong>in</strong>sem<strong>in</strong>ation (AI)protocols employ 3 · 10 9 spermatozoa per dose deposited<strong>in</strong>tracervically two or three times dur<strong>in</strong>g oestrus.Under these standard conditions, one ejaculate can beused to <strong>in</strong>sem<strong>in</strong>ate only a limited number of sows, thusconstra<strong>in</strong><strong>in</strong>g the efficient use of the boars. The standardAI technique is either unsuitable or <strong>in</strong>efficient whenapplied to the emerg<strong>in</strong>g sperm technologies, such asfrozen-thawed spermatozoa and sperm sex<strong>in</strong>g. For thesereasons, a new procedure has been developed fordeposit<strong>in</strong>g spermatozoa deep <strong>in</strong>to the uter<strong>in</strong>e horn(DUI), which allows a reduction of the number ofspermatozoa per dose (Mart<strong>in</strong>ez et al. 2001a,b, 2002).When fresh semen is used under field conditions, thesperm dose can be reduced to 150 · 10 6 spermatozoawith acceptable fertility results (Mart<strong>in</strong>ez et al. 2002,2006). This practical procedure should offer a greatbenefit for the optimization of the use of fresh semenfrom superior boars or <strong>in</strong> sanitary cont<strong>in</strong>gencies whenthe number of doses to be used is decreased. Currently,the DUI technique has the potential to achieve highfertilityresults us<strong>in</strong>g as few as 1–2 · 10 9 total frozenthawedspermatozoa (Roca et al. 2003, 2006). The DUItechnology has the potential to counteract factorslimit<strong>in</strong>g rout<strong>in</strong>e application of frozen-thawed spermatozoa,such as the normally high number of spermatozoarequired per dose and the low fertility achieved.The most sought after reproductive technology is preconceptionsex pre-selection. The only accurate andpotentially cost-effective approach for achiev<strong>in</strong>g sex preselection<strong>in</strong>volves separat<strong>in</strong>g the X- from the Y-chromosomebear<strong>in</strong>g spermatozoa us<strong>in</strong>g flow cytometry andsperm sort<strong>in</strong>g (Johnson et al. 2005). However, thenumber of available flow-sorted spermatozoa is toolow for an extended use of the technology <strong>in</strong> pigproduction, even us<strong>in</strong>g DUI methodology (Vazquezet al. 2003, 2005). Laparoscopic <strong>in</strong>sem<strong>in</strong>ation <strong>in</strong>to theoviduct (ILI) might be an alternative method, at leastwhen applied under specialized production situations(Vazquez et al. 2006).The follow<strong>in</strong>g review identifies objections and problems,which appeared dur<strong>in</strong>g the development of theDUI and ILI procedures, and describes potentialsolutions for practical application of both technologies.Low Dose: Deep Intrauter<strong>in</strong>e Insem<strong>in</strong>ationTwo ma<strong>in</strong> objections have been raised to the procedureof DUI <strong>in</strong> sows: (1) putative damage to the cervix andthe uter<strong>in</strong>e wall by the <strong>in</strong>sem<strong>in</strong>ation device dur<strong>in</strong>g itsadvancement along the lumen of the cervical canal andthe uterus and (2) the <strong>in</strong>cidence of the unilateralfertilization when 150 · 10 6 fresh spermatozoa are used.Putative damage of the cervix and uter<strong>in</strong>e wall by the DUIcatheterIt has been suggested that the DUI catheter may causedamage to the cervix and the uterus mucosa dur<strong>in</strong>g its<strong>in</strong>sertion <strong>in</strong> the sow reproductive tract, potentiallycompromis<strong>in</strong>g subsequent fertility. Us<strong>in</strong>g a fibre opticendoscope procedure developed <strong>in</strong> our laboratories fornon-surgical DUI <strong>in</strong> non-sedated sows (Mart<strong>in</strong>ez et al.2001a), slight bleed<strong>in</strong>g of the cervical canal wasobserved <strong>in</strong> 3 out of 33 sows (9.1%) dur<strong>in</strong>g the <strong>in</strong>sertionof the endoscope. In addition, dur<strong>in</strong>g removal of theendoscope from the genital tract, a visible mark on theendometrium of the first uter<strong>in</strong>e curvature was found <strong>in</strong>seven animals (21.2%). The endometrial damage wasnot accompanied by <strong>in</strong>ternal or external bleed<strong>in</strong>g <strong>in</strong> anysow and did not have a detrimental effect on fertility ofhormonally treated oestrous sows used <strong>in</strong> that study.Recently, results from Australia <strong>in</strong>dicated that a highproportion of sows (27%) bled dur<strong>in</strong>g or after DUI andthat 22% of the <strong>in</strong>sem<strong>in</strong>ations resulted <strong>in</strong> some form ofbleed<strong>in</strong>g from the reproductive tract (Bathgate et al.2007). In that paper, the type of bleed<strong>in</strong>g was dividedsubjectively <strong>in</strong>to three categories: cervical (blood wasseen on the catheter or at anytime dur<strong>in</strong>g <strong>in</strong>sem<strong>in</strong>ation),uter<strong>in</strong>e (blood not noticed dur<strong>in</strong>g <strong>in</strong>sem<strong>in</strong>ation, butbecame evident 12 or more hours after <strong>in</strong>sem<strong>in</strong>ation)and both (both types of blood were observed). Theauthors did not f<strong>in</strong>d significant differences <strong>in</strong> non-returnrates, farrow<strong>in</strong>g rates and litter sizes between sows withor without bleed<strong>in</strong>g. The high proportion of sows withÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


348 JM Vazquez, J Roca, MA Gil, C Cuello, I Parrilla, I Caballero, JL Vazquez and EA Martı´nezbleed<strong>in</strong>g as a result of the <strong>in</strong>sertion of the DUI catheterobta<strong>in</strong>ed by the Australian group is difficult to expla<strong>in</strong> ifwe take <strong>in</strong>to account other researches where the<strong>in</strong>cidence of bleed<strong>in</strong>g dur<strong>in</strong>g or after DUI was low ornon-existent (Mart<strong>in</strong>ez et al. 2001a, 2002, 2006; Dayet al. 2003; Rath et al. 2003; Roca et al. 2003; Vazquezet al. 2003; Bolar<strong>in</strong> et al. 2005a; Grossfeld et al. 2005;Wongtawan et al. 2006). In a recent trial performed on acommercial farm <strong>in</strong> Spa<strong>in</strong>, we could detect blood on theDUI catheter after <strong>in</strong>sem<strong>in</strong>ation <strong>in</strong> only 2 out of 95 sows<strong>in</strong>sem<strong>in</strong>ated which was similar to that observed <strong>in</strong> sows<strong>in</strong>sem<strong>in</strong>ated with the traditional AI method (1 out of95). Each of those three sows farrowed a high number ofpiglets (Mart<strong>in</strong>ez et al. 2006). More recently, similardata on bleed<strong>in</strong>g have been obta<strong>in</strong>ed by our group us<strong>in</strong>gDUI methodology (6 sows with blood out of 108) andstandard AI (4 sows with blood out of 114) with no<strong>in</strong>fluence on performance reproductive of the sows(unpublished data).While it seems clear that the presence of blood dur<strong>in</strong>gor after <strong>in</strong>sem<strong>in</strong>ation does not exert a detrimental effecton fertility results after DUI, the contradictory results <strong>in</strong>relation to the <strong>in</strong>cidence of bleed<strong>in</strong>g as a consequence ofthe <strong>in</strong>sertion of a DUI catheter rema<strong>in</strong> to be expla<strong>in</strong>ed.Several factors could be implicated <strong>in</strong> the differencesobserved <strong>in</strong> relation to the bleed<strong>in</strong>g phenomenon. First,it is evident that dur<strong>in</strong>g the development of any newtechnology, problems dur<strong>in</strong>g the period of tra<strong>in</strong><strong>in</strong>g areexpected. Deep <strong>in</strong>to the uter<strong>in</strong>e horn technologyrequires more patience and skill when compared tostandard AI procedures. In addition, improper manipulationof the device when plac<strong>in</strong>g the catheter <strong>in</strong>to theuterus could result <strong>in</strong> damage of the cervix and ⁄ orthe uterus. The device should always be <strong>in</strong>serted carefullyand not forced when some resistance is encountered.Although the DUI procedure can be performed safelyand quickly, a m<strong>in</strong>imum period of tra<strong>in</strong><strong>in</strong>g is necessary<strong>in</strong> order to achieve optimal results. Second, the differencesmay be due to disparity between the commercialcatheters and the experimental prototype used <strong>in</strong> thepublished reports. While the experimental prototypeswere carefully hand-made, some abnormalities wereobserved <strong>in</strong> several parts of the DUI catheter <strong>in</strong> somebatches of the first commercial catheters constructed.This limitation has been solved by a later ref<strong>in</strong>ement ofthe DUI catheters carried out by a German company(M<strong>in</strong>itube, Tiefenbach, Germany). Currently, the <strong>in</strong>cidenceof bleed<strong>in</strong>g after the DUI procedure us<strong>in</strong>g thecurrent commercial DUI catheter (M<strong>in</strong>itube) is low andsimilar to that observed after the standard AI technique.In our first studies, us<strong>in</strong>g a fibre optic endoscope, avisible mark on the endometrium of the first uter<strong>in</strong>ecurvature was visible <strong>in</strong> a high proportion of sows(Mart<strong>in</strong>ez et al. 2001a). Recently, Bathgate and coworkersperformed an experiment <strong>in</strong> order to exam<strong>in</strong>eendometrial trauma result<strong>in</strong>g from the DUI technique(Bathgate et al. 2007). In this experiment, five sows duefor slaughter were weaned at a time to match the onsetof oestrus with their arrival at the abattoir. Less than1 h prior to slaughter, the DUI catheter was <strong>in</strong>serted asif the sow was to be <strong>in</strong>sem<strong>in</strong>ated, without deposition ofthe semen. Immediately after slaughter, the entirereproductive tract was removed from each sow andtransported to the laboratory where the tracts weredissected and <strong>in</strong>spected for damage to the uter<strong>in</strong>eendometrium. The authors observed slight damage tothe endometrial l<strong>in</strong><strong>in</strong>g <strong>in</strong> all sows and that none of thesesows bled dur<strong>in</strong>g or after the <strong>in</strong>sem<strong>in</strong>ation. These resultsare <strong>in</strong> agreement with our previous observations us<strong>in</strong>gthe endoscope (Mart<strong>in</strong>ez et al. 2001a). While <strong>in</strong> ourstudy the endometrial damage did not affect the fertilityof hormonally treated oestrous sows, the implications ofany damage to the reproductive tract of the sows<strong>in</strong>duced by <strong>in</strong>sertion of the DUI catheter needs to beconsidered, as suggested by Bathgate et al. (2007).Consequently, two experiments have been conductedby our group. In the first experiment (Bolar<strong>in</strong> et al.2005b), the effect on future fertility of DUI catheter<strong>in</strong>sertion <strong>in</strong>to the uter<strong>in</strong>e horn dur<strong>in</strong>g deep <strong>in</strong>trauter<strong>in</strong>e<strong>in</strong>sem<strong>in</strong>ation was evaluated. A total of 159 weaned sows(parity 2–8) were twice DUI-<strong>in</strong>sem<strong>in</strong>ated <strong>in</strong> a commercialfarm dur<strong>in</strong>g 10 months with 1–2 · 10 9 frozenthawedspermatozoa. N<strong>in</strong>ety-n<strong>in</strong>e sows (62.7%) werepregnant and 97 (61.0%) farrowed with a mean littersize of 9.6 ± 0.28. Reproductive data on pigs <strong>in</strong>sem<strong>in</strong>atedby standard AI before and after DUI wererecorded and compared (Table 1). No differences(p > 0.05) were found <strong>in</strong> pregnancy and farrow<strong>in</strong>grates nor litter size for AI before and after DUI.Moreover, ‘not <strong>in</strong> pig’ rates were also similar. Fromthese results, we concluded that the <strong>in</strong>sertion of the DUIcatheter <strong>in</strong>to the uter<strong>in</strong>e horn did not affect subsequentfertility of the sows.In the second experiment (Rodriguez-Vilar et al.2006), the effect of <strong>in</strong>sertion of the DUI device <strong>in</strong>to oneuter<strong>in</strong>e horn on the reproductive performance ofmultiparous sows was evaluated. A total of 51 sowswere conventionally <strong>in</strong>sem<strong>in</strong>ated twice with cooledsemen (3 · 10 9 sperm ⁄ 100 ml doses) at 0 and 24 hafter onset of oestrus. At the time of the first<strong>in</strong>sem<strong>in</strong>ation, the sows were randomly divided <strong>in</strong>totwo groups. In one group, a DUI device was <strong>in</strong>sertedat the same time that conventional <strong>in</strong>sem<strong>in</strong>ation wascarried out (DUI group). The rema<strong>in</strong><strong>in</strong>g 23 sows wereonly conventionally <strong>in</strong>sem<strong>in</strong>ated (control group). Therewere no differences (p > 0.05) between the DUI andcontrol groups for pregnancy and farrow<strong>in</strong>g rates orlitter size (Table 2). A supplementary study with ahigher number of sows (n = 186) offered similarresults (unpublished data). These results show thatthe <strong>in</strong>sertion of the DUI catheter <strong>in</strong>to the uterus doesnot affect the reproductive performance of multiparoussows.Table 1. Reproductive performance of sows traditionally <strong>in</strong>sem<strong>in</strong>atedwith 3 · 10 9 fresh spermatozoa ⁄ 100 ml of extender before and afterthe use of the DUI procedure (from Bolar<strong>in</strong> et al. 2005b)ParameterStandard AIbefore DUI <strong>in</strong>sertion(n = 159)Standard AIafter DUI <strong>in</strong>sertion(n = 159)Pregnancy rate (%) 87.4 86.2Farrow<strong>in</strong>g rate (%) 84.9 84.3Total piglets born11.1 ± 0.17 11.5 ± 0.15(mean ± SEM)Not <strong>in</strong> pig (%) 2.9 2.2Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Low-Dose Insem<strong>in</strong>ation <strong>in</strong> Pigs 349Table 2. Effect of the <strong>in</strong>sertion of the DUI catheter <strong>in</strong> sowstraditionally <strong>in</strong>sem<strong>in</strong>ated with 3 · 10 9 fresh spermatozoa ⁄ 100 ml ofextender on fertility parameters (from Rodriguez-Vilar et al. 2006)ParameterStandard AI plusDUI catheter <strong>in</strong>sertion(n = 28)Standard AIwithout DUI catheter<strong>in</strong>sertion (n = 23)Pregnancy rate (%) 89.3 91.3Farrow<strong>in</strong>g rate (%) 85.7 89.3Total piglets born(mean ± SEM)11.0 ± 0.54 11.1 ± 0.53It may be concluded that: (1) the <strong>in</strong>cidence of bleed<strong>in</strong>gafter the DUI procedure is low and similar to thatobserved after standard AI when practiced by suitablytra<strong>in</strong>ed personnel and with appropriate DUI cathetersand (2) the <strong>in</strong>sertion of the DUI catheter through thecervix <strong>in</strong>to the uterus does not produce significantdamage to the cervix and ⁄ or uter<strong>in</strong>e wall and does notadversely affect the present and ⁄ or future reproductiveperformance of the sows.Incidence of the unilateral fertilization follow<strong>in</strong>g DUIwith 150 · 106 spermatozoaThe earliest experiments carried out <strong>in</strong> our laboratory,compar<strong>in</strong>g DUI with standard AI (3 · 10 9 spermatozoa<strong>in</strong> 80–100 ml), <strong>in</strong>dicated that DUI allows a 20-foldreduction <strong>in</strong> the number of fresh spermatozoa <strong>in</strong>sem<strong>in</strong>ated,and at least a 16–20-fold reduction <strong>in</strong> the dosevolume, without affect<strong>in</strong>g farrow<strong>in</strong>g rate or litter size <strong>in</strong>weaned sows, hormonally <strong>in</strong>duced to ovulate (Mart<strong>in</strong>ezet al. 2002). However, prelim<strong>in</strong>ary studies us<strong>in</strong>g theDUI procedure on weaned, untreated sows under farmconditions <strong>in</strong>dicated that although pregnancy and farrow<strong>in</strong>grates were not affected, litter sizes after DUI(150 · 10 6 fresh spermatozoa ⁄ 5 ml) could be up to 1.0piglet less (p < 0.01) than after standard AI (3 · 10 9spermatozoa ⁄ 95 ml) (Vazquez et al. 2001; Day et al.2003). More recent data us<strong>in</strong>g spontaneously ovulat<strong>in</strong>gweaned sows confirmed these results (Mart<strong>in</strong>ez et al.2006).Various experiments have been conducted to determ<strong>in</strong>ethe possible factors implicated <strong>in</strong> such a reductionof prolificacy. In one of them (Mart<strong>in</strong>ez et al. 2006), 42weaned sows were <strong>in</strong>sem<strong>in</strong>ated at 12, 24 and 36 h afterthe onset of spontaneous oestrus us<strong>in</strong>g one of thefollow<strong>in</strong>g two regimes: (1) DUI (treatment) with150 · 10 6 fresh spermatozoa <strong>in</strong> 5 ml of BTS extenderor (2) standard cervical AI (control) with 2.85 · 10 9fresh spermatozoa <strong>in</strong> 95 ml of BTS extender. On day 6,after onset of oestrus, the proximal segment of the<strong>in</strong>dividual uter<strong>in</strong>e horns of the sows were surgicallyflushed under anaesthesia to retrieve ova ⁄ embryos andevaluate the success of fertilization per horn (e.g.occurrence of effective uni- vs bilateral sperm transportrender<strong>in</strong>g uni- or bilateral, complete or partial fertilization).Retrieved embryos were assessed for cleavage andnumber of accessory spermatozoa. Although identicaloverall fertilization rates were achieved <strong>in</strong> both <strong>in</strong>sem<strong>in</strong>ationgroups, the percentage of sows with partialbilateral fertilization and unilateral fertilization wasmarkedly higher (p < 0.05) <strong>in</strong> the DUI group (35%)compared with the control (standard AI) group (5%),with a consequent lower (p < 0.001) percentage ofviable early embryos after DUI. The number of spermbound to the zona pellucida (ZP) was higher <strong>in</strong> embryosfrom control sows than <strong>in</strong> DUI sows, <strong>in</strong>dicat<strong>in</strong>g that thenumber of spermatozoa reach<strong>in</strong>g the sperm reservoirslargely depends on the number of spermatozoa <strong>in</strong>sem<strong>in</strong>ated,as reported <strong>in</strong> other species (Morton and Glover1974; Nadir et al. 1993).Our results also <strong>in</strong>dicate a high <strong>in</strong>dividual variability<strong>in</strong> the number of accessory spermatozoa <strong>in</strong> the ZP ofembryos from sows <strong>in</strong>sem<strong>in</strong>ated on the same day andwith sem<strong>in</strong>al doses orig<strong>in</strong>at<strong>in</strong>g from the same ejaculates.This variability was also observed with<strong>in</strong> sow betweenuter<strong>in</strong>e horns, regardless of the AI method used (Fig. 1).In addition, no accessory spermatozoa were found <strong>in</strong>oocytes retrieved from sows with unilateral fertilization(Fig. 2). This <strong>in</strong>dicates that spermatozoa were not ableto reach the contralateral oviduct <strong>in</strong> these sows andsuggests that <strong>in</strong>tr<strong>in</strong>sic characteristics of the sow (i.e.myometrial contractility and ⁄ or local phagocytosis by<strong>in</strong>vad<strong>in</strong>g leucocytes) could be implicated <strong>in</strong> the <strong>in</strong>cidenceof unilateral fertilizations. It is known that once thesperm dose is deposited deeply <strong>in</strong>to a uter<strong>in</strong>e horn,spermatozoa are able to reach the contralateral oviductand to fertilize a high proportion of oocytes (Mart<strong>in</strong>ezet al. 2002; Tummaruk et al. 2007) although spermatozoacan be recovered from only one oviduct via theflush<strong>in</strong>g technique (Tummaruk et al. 2007). It could bespeculated that <strong>in</strong> some sows, the number of spermatozoareach<strong>in</strong>g the contralateral oviduct might be too lowto establish an efficient sperm population <strong>in</strong> the reservoirto fertilize the oocytes, and as a consequence, theFig. 1. Individual variations <strong>in</strong> the number of spermatozoa bound tothe ZP of normal embryos <strong>in</strong> sows <strong>in</strong>sem<strong>in</strong>ated three times dur<strong>in</strong>goestrus either by deep <strong>in</strong>trauter<strong>in</strong>e (DUI; 150 · 10 6 spermatozoa⁄ 5 ml) or <strong>in</strong> contemporaries <strong>in</strong>sem<strong>in</strong>ated cervically (AI;2.85 · 10 9 spermatozoa ⁄ 95 ml). The sows were <strong>in</strong>sem<strong>in</strong>ated on thesame day us<strong>in</strong>g doses from the same batch of semen. Each dose ofsemen used for DUI was taken directly from the same bottle used to<strong>in</strong>sem<strong>in</strong>ate the contemporary control sow. *Differences betweenuter<strong>in</strong>e horns of the same sow (p £ 0.002). (a) Differences comparedto the same uter<strong>in</strong>e horn of the contemporary sow (at least p < 0.01).Unilateral fertilization was observed <strong>in</strong> sow DUI 3. In that sow, allstructures recovered from uter<strong>in</strong>e horn 2 were oocytes with nospermatozoa bound to the ZP and the differences <strong>in</strong> sperm countbetween her uter<strong>in</strong>e horns or <strong>in</strong> relation to uter<strong>in</strong>e horn 2 of hercontemporary control AI sow were not analysed (from Mart<strong>in</strong>ez et al.2006)Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


350 JM Vazquez, J Roca, MA Gil, C Cuello, I Parrilla, I Caballero, JL Vazquez and EA Martı´nezFig. 2. Number of spermatozoa bound to the ZP of normal embryos<strong>in</strong> deep <strong>in</strong>trauter<strong>in</strong>e <strong>in</strong>sem<strong>in</strong>ated sows (DUI; 0.15 · 10 9 spermatozoa⁄ 5 ml) with unilateral fertilization and <strong>in</strong> contemporary controlsows (AI; 2.85 · 10 9 spermatozoa ⁄ 95 ml). Each dose of semen usedfor DUI was taken directly from the same bottle used to <strong>in</strong>sem<strong>in</strong>ate thecontemporary control sow. Sows from both groups were <strong>in</strong>sem<strong>in</strong>atedthree times dur<strong>in</strong>g oestrus. *Differences between uter<strong>in</strong>e horns of thesame sow (at least p < 0.002). (a) Differences compared to the sameuter<strong>in</strong>e horn of the contemporary sow (at least p < 0.01). Allstructures recovered from uter<strong>in</strong>e horn 2 <strong>in</strong> DUI sows were oocyteswith no spermatozoa bound to the ZP and differences between uter<strong>in</strong>ehorns of the same sow or <strong>in</strong> relation to the uter<strong>in</strong>e horn 2 of AI sowswere not analysed (from Mart<strong>in</strong>ez et al. 2006)oocytes of that oviduct are not fertilized. Although twodifferent pathways (transperitoneal and <strong>in</strong>trauter<strong>in</strong>e)have been confirmed for the transport of spermatozoa<strong>in</strong>to the reproductive tract after the DUI procedure(Mart<strong>in</strong>ez et al. 2005), more <strong>in</strong>vestigations are needed tounderstand the mechanisms by which spermatozoacolonize the oviducts (reviewed by Rodriguez-Mart<strong>in</strong>ezet al. 2005) after DUI with low numbers of spermatozoa.From the above results, it can be concluded that thelower litter size after DUI with 150 · 10 6 fresh spermatozoa<strong>in</strong> 5 ml of BTS extender is related to a decreaseddistribution of spermatozoa, lead<strong>in</strong>g to a higher <strong>in</strong>cidenceof partial bilateral and unilateral fertilization thanwith standard AI.The discrepancies exist<strong>in</strong>g between our first experimentsus<strong>in</strong>g hormonally <strong>in</strong>duced oestrous sows (Mart<strong>in</strong>ezet al. 2001a, 2002) and other studies us<strong>in</strong>g sows <strong>in</strong>spontaneous oestrus (Vazquez et al. 2001; Day et al.2003; Mart<strong>in</strong>ez et al. 2006), <strong>in</strong> relation to the differencesobserved <strong>in</strong> the litter size between DUI and standard AIgroups, could be related to the ovulatory response to thehormonal treatment. It is possible that the hormonaltreatment of sows <strong>in</strong>sem<strong>in</strong>ated <strong>in</strong> the depth of oneuter<strong>in</strong>e horn with 150 million spermatozoa <strong>in</strong>creased thenumber of ovulations from each ovary and, as a result, agreater number of oocytes could have been fertilized <strong>in</strong>relation to standard AI sows (spontaneous oestroussows), even with unilateral or partial bilateral fertilization.Consequently, the litter sizes were similar betweenboth groups of sows.There are several possibilities for solv<strong>in</strong>g the problemof unilateral fertilization after DUI. It is evident that thenumbers of spermatozoa, volume of <strong>in</strong>sem<strong>in</strong>ates and ⁄ orthe place of deposition of semen plays a major role.Thus, we have observed that fertilization rates and thepercentage of bilateral fertilization after DUI with600 · 10 6 spermatozoa <strong>in</strong> 20 ml of BTS extender didnot differ from those of the standard AI group (97.8%and 100% vs 98.5% and 100%, respectively) (Mart<strong>in</strong>ezet al. 2005). Other factors, such as the <strong>in</strong>sem<strong>in</strong>ation–ovulation <strong>in</strong>terval, uter<strong>in</strong>e contractility and phagocytosis,could also be implicated <strong>in</strong> the <strong>in</strong>cidence ofunilateral fertilization when DUI is used under fieldconditions. Therefore, hormonal treatments to controlovulation, stimulants of myometrial contractility andthe addition of substances for decreas<strong>in</strong>g local phagocytosiscould be ways to <strong>in</strong>crease the litter size whensows <strong>in</strong> spontaneous oestrus are <strong>in</strong>sem<strong>in</strong>ated us<strong>in</strong>g DUIwith 150 · 10 6 fresh spermatozoa.In conclusion, the <strong>in</strong>cidence of unilateral fertilization<strong>in</strong> sows <strong>in</strong> spontaneous oestrus after DUI with 150 · 10 6fresh spermatozoa <strong>in</strong> 5 ml of extender can be overcomeby <strong>in</strong>creas<strong>in</strong>g the number of spermatozoa and volume ofthe dose to 600 · 10 6 and 20 ml, respectively. Otherpotential remedies that <strong>in</strong>fluence uter<strong>in</strong>e contractibilityand local phagocytosis rema<strong>in</strong> to be explored.Very Low Dose: Laparoscopic Insem<strong>in</strong>ationThe deposition of the spermatozoa higher <strong>in</strong> thereproductive tract than the uterus (uterotubal junctionor oviduct) allows a greater proportion of spermatozoato survive and colonize the oviduct; and therefore, fewerwould be necessary to achieve the same probability offertility than with standard AI (Watson 2000). Techniquesto deposit a very low number of spermatozoa <strong>in</strong> asmall volume of <strong>in</strong>sem<strong>in</strong>ate would <strong>in</strong>crease the efficiencyof <strong>in</strong>sem<strong>in</strong>ation when us<strong>in</strong>g spermatozoa with a highadded value (e.g. sex-sorted semen or sperm-mediatedgene transfer) (Rath 2002; Johnson et al. 2005). Insem<strong>in</strong>ation<strong>in</strong>to the uterotubal junction or the oviductwould reduce the number of spermatozoa required tothe m<strong>in</strong>imum possible, decreas<strong>in</strong>g the loss of spermatozoaby phagocytosis or backflow.Successful fertility results have been obta<strong>in</strong>ed whenfrozen-thawed (Maxwell and Salamon 1977) or sexsortedspermatozoa (Johnson 1991) were depositeddirectly <strong>in</strong>to the oviduct by laparotomy. Moreover, itis well known that optimal fertility can be achieved withonly 1 · 10 7 spermatozoa after surgical <strong>in</strong>sem<strong>in</strong>ationnear the utero-tubal junction, <strong>in</strong>stead of the 3 · 10 9spermatozoa normally <strong>in</strong>sem<strong>in</strong>ated <strong>in</strong> the cervix (Kruegeret al. 1999). The number of spermatozoa requiredfor <strong>in</strong>sem<strong>in</strong>ation is directly dependant on the place ofdeposition of these spermatozoa (Vazquez et al. 2005).The disadvantage of a protocol based on laparotomy isthat the <strong>in</strong>vasive procedure cannot be applied on farms.Surgical laparotomy results <strong>in</strong> distress for the sows and<strong>in</strong> post-operative adhesions that complicate the subsequent<strong>in</strong>sem<strong>in</strong>ations.Laparoscopy is a less-<strong>in</strong>vasive technique than laparotomyfor deposit<strong>in</strong>g semen directly <strong>in</strong>to the uterus oroviduct and could be performed on farm by technicallytra<strong>in</strong>ed personnel. This procedure provides sharp andmagnified images of the genital tract. Although laparoscopyhas been rout<strong>in</strong>ely applied to commercial<strong>in</strong>sem<strong>in</strong>ations of sheep with a low number of spermatozoa(reviewed by Maxwell and Watson 1996), theÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Low-Dose Insem<strong>in</strong>ation <strong>in</strong> Pigs 351application <strong>in</strong> pigs should be further evaluated before itcan be applied on farm. The effectiveness (safety andsuccess) of this <strong>in</strong>sem<strong>in</strong>ation procedure as a solution forthe application of spermatozoa with a high added value(very low dose) is discussed.Laparoscopic procedure <strong>in</strong> pigsLaparoscopy is an alternative procedure that allowsaccess <strong>in</strong>to the abdom<strong>in</strong>al cavity. This methodology isless <strong>in</strong>vasive than laparotomy s<strong>in</strong>ce a large <strong>in</strong>cision isreplaced by several smaller <strong>in</strong>cisions. The laparoscopicprocedure reduces the exposure of the peritoneal cavity,and the handl<strong>in</strong>g of the bowel is avoided (Garry andFrancog 2006).However, laparoscopic <strong>in</strong>sem<strong>in</strong>ation hasthe disadvantage that, although m<strong>in</strong>imally <strong>in</strong>vasive, it isalso a surgical procedure. Moreover, it is important totake <strong>in</strong>to consideration that laparoscopy deprives theoperator of three-dimensional vision when comparedwith full laparotomy procedures.While laparoscopic <strong>in</strong>sem<strong>in</strong>ation has been successfullyused <strong>in</strong> millions of humans and animals worldwide, theexperience with pigs is limited. We first developed amodel <strong>in</strong> sows to carry out laparoscopic <strong>in</strong>sem<strong>in</strong>ation.The sows, under general anaesthesia, were placed <strong>in</strong> aTrendelenburg position at an angle of approximately20–30° above horizontal. A 2-cm mid-l<strong>in</strong>e <strong>in</strong>cision wasperformed at the umbilical level, where a Veres needle(Veres Insufflation Needle, Aesculap Inc., Center Valley,USA) was <strong>in</strong>serted <strong>in</strong>to the peritoneal cavity to <strong>in</strong>sufflatecarbon dioxide to establish the pneumoperitoneum andto distend the abdom<strong>in</strong>al cavity to ensure the visualizationof the genital structures.Subsequently, a trocar-canula was <strong>in</strong>serted <strong>in</strong> the same<strong>in</strong>cision to allow the passage of the laparoscope, ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>gthe gas pressure by a device attached to the trocarcanula.The need to <strong>in</strong>sert the trocar-canula as well as thedistension of the cavity are weaknesses of laparoscopy.Initially, <strong>in</strong> our experimental trials, approximately 200laparoscopic <strong>in</strong>sem<strong>in</strong>ations were conducted us<strong>in</strong>g thisprocedure. The two ma<strong>in</strong> disadvantages observed were:(1) the time taken to <strong>in</strong>sufflate carbon dioxide <strong>in</strong>to theperitoneal cavity and (2) the risk of gastro<strong>in</strong>test<strong>in</strong>al(bowel or vessel) <strong>in</strong>jury dur<strong>in</strong>g the laparoscopy.Gas <strong>in</strong>sufflation provides a safe space to <strong>in</strong>sert theadditional trocars as well as allow<strong>in</strong>g the clear visualizationof the genital structures. Our experience <strong>in</strong>dicatesthat the gas pressure required for <strong>in</strong>sem<strong>in</strong>ation <strong>in</strong>pigs should be 13–15 mmHg. The time needed to reachthis pressure <strong>in</strong> pigs was 8–10 m<strong>in</strong> due to the diameter ofthe Veres needle and the pressure output of thelaparoscopic <strong>in</strong>sufflator. Consequently, <strong>in</strong>creas<strong>in</strong>g thecarbon dioxide flow rate should decrease the timerequired to reach the correct <strong>in</strong>tra-abdom<strong>in</strong>al pressure.The <strong>in</strong>cidence of the gastro<strong>in</strong>test<strong>in</strong>al <strong>in</strong>jury wasapproximately 0.5–1% based on the diagnosis madedur<strong>in</strong>g exploration of the abdom<strong>in</strong>al cavity. The bl<strong>in</strong>d<strong>in</strong>sertion of the Veres needle and the first trocar maycause complications, especially <strong>in</strong> large and fat sows, asobserved with obese human patients. It is also importantto take <strong>in</strong>to consideration that this technique requiresspecific <strong>in</strong>strumentation and, especially, a skilled operator.A number of modifications to the laparoscopicprocedure have been developed based on these limitations.As an alternative to the Veres needle, opticaltrocars can be safely positioned under direct visualization.These trocars have been shown <strong>in</strong> humans to besafe and easy to handle, offer<strong>in</strong>g several advantages overthe use of the Veres needle and the m<strong>in</strong>i-laparotomy(Str<strong>in</strong>g et al. 2001). In our laboratory, we carried out anexperiment us<strong>in</strong>g a 12-mm Optiview trocar (EthiconEndo-Surgery, C<strong>in</strong>c<strong>in</strong>nati, OH, USA) <strong>in</strong>stead of theVeres needle. This trocar and a 0° laparoscope are<strong>in</strong>serted and advanced <strong>in</strong>to the wound, allow<strong>in</strong>g the tipof the trocar to be viewed while it is enter<strong>in</strong>g theabdom<strong>in</strong>al cavity. After the cavity is entered, the handpiece of the Optiview (Ethicon Endo-Surgery) isremoved and replaced by the 0° laparoscope, and theabdom<strong>in</strong>al cavity directly <strong>in</strong>flated to 13–15 mmHg withcarbon dioxide. Us<strong>in</strong>g this procedure, less than 3 m<strong>in</strong>are required to <strong>in</strong>sufflate the abdom<strong>in</strong>al cavity, and nogastro<strong>in</strong>test<strong>in</strong>al <strong>in</strong>juries have been observed after the<strong>in</strong>sem<strong>in</strong>ation of nearly 500 sows.Once the reproductive tract has been located, thelaparoscope allows the <strong>in</strong> situ observation of ovarianstatus. Two accessory posts are placed <strong>in</strong> the right andleft part of the hemi-abdomen, which provides accessfor laparoscopic Duval forceps for manipulation ofthe uter<strong>in</strong>e horn and grasp<strong>in</strong>g of the uterus or theoviduct for the <strong>in</strong>sem<strong>in</strong>ation needle. To carry out the<strong>in</strong>sem<strong>in</strong>ation, the needle is <strong>in</strong>serted through the UTJor oviduct wall with a fast, precise and controlledmovement and spermatozoa are <strong>in</strong>jected <strong>in</strong>to thelumen. In some cases (low percentage,


352 JM Vazquez, J Roca, MA Gil, C Cuello, I Parrilla, I Caballero, JL Vazquez and EA Martı´nezAlthough the number of spermatozoa deposited bylaparotomy is very low when compared with the numberrequired for standard AI, 10 · 10 6 could still be a highnumber when valuable spermatozoa (e.g. sex-sorted) areused. Furthermore, failures of the <strong>in</strong>sem<strong>in</strong>ation havebeen observed <strong>in</strong> 29% of the animals and no fertilizationwas observed <strong>in</strong> one uter<strong>in</strong>e horn (Fant<strong>in</strong>ati et al. 2005).In our own experience, once the UTJ is identified, theneedle is advanced to touch the uter<strong>in</strong>e part and then<strong>in</strong>serted <strong>in</strong>to the uter<strong>in</strong>e lumen. However, it is easy toaccidently <strong>in</strong>troduce the tip of the needle <strong>in</strong>to the uter<strong>in</strong>ewall, rather than <strong>in</strong>to the lumen, result<strong>in</strong>g <strong>in</strong> <strong>in</strong>sem<strong>in</strong>ationfailure. The difficulty <strong>in</strong> feel<strong>in</strong>g the location of thetip of the needle makes it very hard to ensure its correctposition<strong>in</strong>g <strong>in</strong> the reproductive tract, even for welltra<strong>in</strong>edoperators. Only after expulsion of the semen,does the absence of progressive swell<strong>in</strong>g of uter<strong>in</strong>e tip, aswell as the resistance encountered while attempt<strong>in</strong>g toeject the semen, <strong>in</strong>dicate that semen has been deposited<strong>in</strong>correctly.A variation of this technique has been developed <strong>in</strong>our laboratory by deposition of semen directly <strong>in</strong>to theoviduct. This laparoscopic <strong>in</strong>sem<strong>in</strong>ation method is easyand fast <strong>in</strong> pigs when done by technically tra<strong>in</strong>edpersonnel. The ma<strong>in</strong> advantage is that the number ofspermatozoa deposited can be much lower than thatrequired <strong>in</strong> the UTJ as demonstrated by the goodfertility when frozen-thawed (Maxwell and Salamon1977) or sex-sorted spermatozoa (Johnson 1991) weredeposited directly <strong>in</strong>to the oviduct by laparotomy.Moreover, the correct position<strong>in</strong>g of the tip of theneedle <strong>in</strong> the lumen of the oviduct is much easier toachieve than for UTJ <strong>in</strong>sem<strong>in</strong>ations. Nevertheless, it isimportant to take <strong>in</strong>to consideration that entry ofspermatozoa <strong>in</strong>to the oviduct is normally a highlyselective process, be<strong>in</strong>g dependent ma<strong>in</strong>ly on the <strong>in</strong>tr<strong>in</strong>sicmotility of each <strong>in</strong>dividual spermatozoan. Thisselectivity is abolished when the spermatozoa aredeposited directly <strong>in</strong>to the oviduct.Efficiency of the <strong>in</strong>tra-oviductal <strong>in</strong>sem<strong>in</strong>ation techniqueIt is well known that the polyspermy block from theporc<strong>in</strong>e oocyte is low when compared with other species(Day 2000) and, <strong>in</strong> vitro, the sperm : oocyte ratio affectsthe percentage of polyspermic oocytes (Gil et al. 2004,2007). From more than 50 · 10 9 spermatozoa deposited<strong>in</strong>to the sows after natural mat<strong>in</strong>g, only tens to hundredsof sperm reached the oviduct (Hunter 2002). Consequently,a high percentage of polyspermic oocytes couldbe expected after <strong>in</strong>tra-oviductal laparoscopic <strong>in</strong>sem<strong>in</strong>ationwhen compared with conventional <strong>in</strong>sem<strong>in</strong>ation,s<strong>in</strong>ce a higher than physiological number of spermatozoaare deposited <strong>in</strong> the oviduct. As expected, a highpercentage of oocytes were penetrated after <strong>in</strong>traoviductallaparoscopic <strong>in</strong>sem<strong>in</strong>ation with 0.3, 0.5 or1 · 10 6 spermatozoa per oviduct <strong>in</strong> our laboratory.Surpris<strong>in</strong>gly, however, regardless of the high number ofspermatozoa deposited <strong>in</strong> the oviductal ampulla, the<strong>in</strong>cidence of polyspermic penetration was very low when0.3 or 0.5 · 10 6 spermatozoa were <strong>in</strong>sem<strong>in</strong>ated beforeovulation (Table 3). These results show the potentialeffectiveness of laparoscopic <strong>in</strong>sem<strong>in</strong>ation as a methodTable 3. Percentages of penetrated and monospermic oocytes <strong>in</strong> sowsat an <strong>in</strong>duced ovulation follow<strong>in</strong>g <strong>in</strong>tra-oviductal laparoscopic <strong>in</strong>sem<strong>in</strong>ationof 0.3, 0.5 or 1 million spermatozoa per oviductInsem<strong>in</strong>ationDose (·10 6 )% Oocytespenetrated% Monospermy0.3 91.8 ± 1.5 88.1 ± 2.1 a0.5 92.2 ± 1.8 86.1 ± 2.1 a1.0 95.0 ± 2.1 68.6 ± 29 bDifferent letters with<strong>in</strong> the same column represent a significant difference(p < 0.05).for obta<strong>in</strong><strong>in</strong>g pregnancies with very low numbers ofspermatozoa. There was evidence of polyspermy onlywhen 1 · 10 6 spermatozoa were <strong>in</strong>sem<strong>in</strong>ated.It is well known that the presence of the gametes <strong>in</strong>the oviduct modifies the amount of prote<strong>in</strong> <strong>in</strong> theoviductal fluid. N<strong>in</strong>eteen prote<strong>in</strong>s have been identified<strong>in</strong> vitro that are specifically regulated by spermatozoa,four prote<strong>in</strong>s regulated specifically by oocytes, and oneprote<strong>in</strong> that is commonly regulated by both spermatozoaand oocytes (Georgiou et al. 2005). The oviductalenvironment, and consequently the fertilization process,also depends on the hormones secreted by the follicles(Hunter 2002). We have recently identified a number ofprote<strong>in</strong>s that are regulated by the presence of spermatozoaor oocytes <strong>in</strong> the oviduct <strong>in</strong> vivo (Georgiou et al.2007). Therefore, the oviductal environment may modulatethe penetration of oocytes by spermatozoa as aconsequence of their deposition directly <strong>in</strong>to the oviduct.All of the prote<strong>in</strong>s regulated by the gametes arewell known to <strong>in</strong>fluence gamete maturation, viabilityand function. Consequently, the deposition of thespermatozoa before, dur<strong>in</strong>g or after the arrival of theoocyte <strong>in</strong> the oviductal ampulla should be an importantvariable to be considered <strong>in</strong> the analysis of the efficiencyof <strong>in</strong>tra-oviductal laparoscopic <strong>in</strong>sem<strong>in</strong>ation.Recently, we have evaluated the effect of the tim<strong>in</strong>g of<strong>in</strong>sem<strong>in</strong>ation <strong>in</strong> relation to ovulation (pre-ovulatory,ovulat<strong>in</strong>g or ovulated follicles) on the penetration andpolyspermic fertilization of oocytes (Table 4). The rateof polyspermy was very high when sows with ovulatedoocytes were <strong>in</strong>sem<strong>in</strong>ated directly <strong>in</strong>to the oviduct.While polyspermy has been associated with aged oocytes(Hunter 2002), <strong>in</strong> this experiment, it seemed to be morerelated to the oviductal environment than the age ofthe oocytes, s<strong>in</strong>ce all of the sows ovulated only 1–3 hbefore <strong>in</strong>sem<strong>in</strong>ation. When the spermatozoa were present<strong>in</strong> the oviduct before ovulation (pre-ovulatory<strong>in</strong>sem<strong>in</strong>ations), the rate of polyspermy was very low.Table 4. Percentages of penetrated and monospermic oocytes <strong>in</strong> sowsat an <strong>in</strong>duced ovulation follow<strong>in</strong>g <strong>in</strong>tra-oviductal laparoscopic <strong>in</strong>sem<strong>in</strong>ation(0.3 spermatozoa per oviduct) before, dur<strong>in</strong>g or after ovulationInsem<strong>in</strong>ation timerelative to ovulation% Oocytespenetrated% MonospermyBefore 98.1 ± 1.3 97.5 ± 1.1 aDur<strong>in</strong>g 89.3 ± 2.2 87.8 ± 2.1 bAfter 94.3 ± 2.7 49.8 ± 2.9 cDifferent letters with<strong>in</strong> the same column represent a significant difference(p < 0.05).Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Low-Dose Insem<strong>in</strong>ation <strong>in</strong> Pigs 353However, if the oocytes were present at the time of<strong>in</strong>sem<strong>in</strong>ation (peri-ovulatory <strong>in</strong>sem<strong>in</strong>ation), polyspermywas significantly higher (p < 0.05).Us<strong>in</strong>g the <strong>in</strong>tra-oviductal laparoscopic <strong>in</strong>sem<strong>in</strong>ationprocedure, pregnancy rates higher than 80% have beenobta<strong>in</strong>ed with only 0.3 · 10 6 diluted or sex-sortedspermatozoa per oviduct (Vazquez et al. 2006). Highfertility has also been reported after <strong>in</strong>tra-oviductallaparoscopic <strong>in</strong>sem<strong>in</strong>ation with sex-sorted spermatozoa<strong>in</strong>sem<strong>in</strong>ated <strong>in</strong> presence of the PSP-I ⁄ PSP-II heterodimerextracted from boar sem<strong>in</strong>al plasma (Garcia et al.2007).ConclusionsUs<strong>in</strong>g appropriate <strong>in</strong>sem<strong>in</strong>ation procedures, it is nowfeasible to achieve high fertility rates <strong>in</strong> pigs with low orvery low doses of spermatozoa. When compared withstandard AI, post-cervical <strong>in</strong>sem<strong>in</strong>ation allows a threefoldreduction <strong>in</strong> the number of cooled spermatozoa anddeep <strong>in</strong>trauter<strong>in</strong>e <strong>in</strong>sem<strong>in</strong>ation allows a substantialreduction <strong>in</strong> the number of cooled (5–20-fold) orfrozen-thawed (sixfold) spermatozoa. Intra-oviductal<strong>in</strong>sem<strong>in</strong>ation by laparoscopy successfully allows <strong>in</strong>sem<strong>in</strong>ationswith only 0.3 · 10 6 sex-sorted spermatozoa. Allthese <strong>in</strong>sem<strong>in</strong>ation procedures may be useful tools forthe pig <strong>in</strong>dustry.AcknowledgementsThe authors wish to thank Professor W.M.C. Maxwell for helpfuldiscussion and critical read<strong>in</strong>g of the manuscript. F<strong>in</strong>ancial supportfrom CICYT (AGF98-0533; AGL2001-0471; AGL2004-07546 andAGF2005-00760), CDTI (B288 ⁄ 98; 03.180; 03-402, 04-0231), INIA(RZ01-019) and Fundacion Séneca de Murcia (PB ⁄ 74 ⁄ FS ⁄ 02,03002 ⁄ PI ⁄ 05 and 04543 ⁄ GERM ⁄ 07).ReferencesBathgate R, Eriksson BM, Thomson PC, Maxwell WMC,Evans G, 2007: Field fertility of frozen-thawed boar spermat low doses us<strong>in</strong>g non-surgical, deep uter<strong>in</strong>e <strong>in</strong>sem<strong>in</strong>ation.Anim Reprod Sci. doi:10.1016/j.anireprosci.2007.01.008.Bolar<strong>in</strong> A, Roca J, Rodriguez-Mart<strong>in</strong>ez H, Hernandez M,Vazquez JM, Mart<strong>in</strong>ez EA, 2005a: Dissimilarities <strong>in</strong> sows’ovarian status at the <strong>in</strong>sem<strong>in</strong>ation time could expla<strong>in</strong>differences <strong>in</strong> fertility between farms when frozen thawedsemen is used. Theriogenology 65, 669–680.Bolar<strong>in</strong> A, Hernandez M, Vazquez JM, Mart<strong>in</strong>ez EA, Roca J,2005b: Catheter <strong>in</strong>sertion <strong>in</strong>to uterus horn dur<strong>in</strong>g deep<strong>in</strong>trauter<strong>in</strong>e <strong>in</strong>sem<strong>in</strong>ation does not compromise futurefertility potential of sows. Reprod Domest Anim 40, 340(abstract).Brussow KP, Torner H, Ratky J, Manabe N, Tuchscherer A,2006: Experimental evidence for the <strong>in</strong>fluence of cumulus–oocyte-complexes on sperm release from the porc<strong>in</strong>e oviductalsperm reservoir. J Reprod Dev 52, 249–257.Day BN, 2000: Reproductive biotechnologies: current status <strong>in</strong>porc<strong>in</strong>e reproduction. Anim Reprod Sci 60–61, 161–172.Day BN, Mathias K, Didion BA, Mart<strong>in</strong>ez EA, Caamano JN,2003: Deep <strong>in</strong>trauter<strong>in</strong>e <strong>in</strong>sem<strong>in</strong>ation <strong>in</strong> sows: first field trial<strong>in</strong> USA commercial farm with a newly developed device.Theriogenology 59, 213 (Abstract).Fant<strong>in</strong>ati P, Zannoni A, Bernard<strong>in</strong>i C, Webster N, LavitranoM, Forni M, Seren E, Bacci ML, 2005: Laparoscopic<strong>in</strong>sem<strong>in</strong>ation technique with low numbers of spermatozoa <strong>in</strong>superovulated prepuberal gilts for biotechnological application.Theriogenology 63, 806–817.Garcia EM, Vazquez JM, Parrilla I, Calvete JJ, Sanz L,Caballero I, Roca J, Vazquez JL, Mart<strong>in</strong>ez EA, 2007:Improv<strong>in</strong>g the fertiliz<strong>in</strong>g ability of sex sorted boar spermatozoa.Theriogenology 68, 771–778.Garry R, Francog MDF, 2006: Laparoscopic surgery. BestPract Cl<strong>in</strong> Obstet Gynaecol 20, 89–104.Georgiou AS, Sostaric E, Wong CH, Snijders AP, Wright PC,Moore HD, Fazeli A, 2005: Gametes alter the oviductalsecretory proteome. Mol Cell Proteomics 4, 1785–1796.Georgiou AS, Snijders APL, Sostaric E, Afaltoonian R,Vazquez JL, Vazquez JM, Roca J, Mart<strong>in</strong>ez EA, WrightPC, Fazeli A, 2007: Modulation of the oviductal environmentby gametes. J Proteome Res 6, 4656–4666.Gil MA, Ruiz M, Cuello C, Vazquez JM, Roca J, Mart<strong>in</strong>ezEA, 2004: Influence of sperm: oocyte ratio dur<strong>in</strong>g <strong>in</strong> vitrofertilization of <strong>in</strong> vitro matured cumulus-<strong>in</strong>tact pig oocyteson fertilization parameters and embryo development. Theriogenology61, 551–560.Gil MA, Alm<strong>in</strong>ana C, Cuello C, Parrilla I, Roca J, VazquezJM, Mart<strong>in</strong>ez EA, 2007: Brief co<strong>in</strong>cubation of gametes <strong>in</strong>porc<strong>in</strong>e <strong>in</strong> vitro fertilization: Role of sperm: oocyte ratio andpost-co<strong>in</strong>cubation medium. Theriogenology 67, 620–626.Grossfeld R, Kl<strong>in</strong>c P, Sieg B, Rath D, 2005: Production ofpiglets with sexed semen employ<strong>in</strong>g a non-surgical <strong>in</strong>sem<strong>in</strong>ationtechnique. Theriogenology 63, 2269–2277.Hunter RHF, 2002: Vital aspects of Fallopian tube physiology<strong>in</strong> pigs. Reprod Domest Anim 37, 186–190.Johnson LA, 1991: Sex preselection <strong>in</strong> sw<strong>in</strong>e: altered sex ratio<strong>in</strong> offspr<strong>in</strong>g follow<strong>in</strong>g surgical <strong>in</strong>sem<strong>in</strong>ation of flow sortedX- and Y-bear<strong>in</strong>g sperm. Reprod Domest Anim 26, 309–314.Johnson LA, Rath D, Vazquez JM, Maxwell WMC, Dobr<strong>in</strong>skyJR, 2005: Preselection of sex of offspr<strong>in</strong>g <strong>in</strong> sw<strong>in</strong>e forproduction: current status of the process and its application.Theriogenology 63, 615–624.Krueger C, Rath D, Johnson LA, 1999: Low dose <strong>in</strong>sem<strong>in</strong>ation<strong>in</strong> synchronized gilts. Theriogenology 52, 1363–1373.Mart<strong>in</strong>ez EA, Vazquez JM, Roca J, Lucas X, Gil MA, ParrillaI, Vazquez JL, Day BN, 2001a: Successful non-surgical deep<strong>in</strong>trauter<strong>in</strong>e <strong>in</strong>sem<strong>in</strong>ation with low number of spermatozoa<strong>in</strong> sows by a fiberoptic endoscope technique. <strong>Reproduction</strong>122, 289–296.Mart<strong>in</strong>ez EA, Vazquez JM, Roca J, Lucas X, Gil MA,Vazquez JL, 2001b: Deep <strong>in</strong>trauter<strong>in</strong>e <strong>in</strong>sem<strong>in</strong>ation andembryo transfer <strong>in</strong> pigs. <strong>Reproduction</strong> 58(Suppl.), 301–311.Mart<strong>in</strong>ez EA, Vazquez JM, Roca J, Lucas X, Gil MA, ParrillaI, Vazquez JL, Day BN, 2002: M<strong>in</strong>imal number of spermatozoarequired for normal fertility after deep <strong>in</strong>trauter<strong>in</strong>e<strong>in</strong>sem<strong>in</strong>ation <strong>in</strong> non-sedated sows. <strong>Reproduction</strong> 123, 163–170.Mart<strong>in</strong>ez EA, Vazquez JM, Roca J, Cuello C, Gil MA, ParrillaI, Vazquez JL, 2005: An update on reproductive technologieswith potential short-term application <strong>in</strong> pig production.Reprod Domest Anim 40, 300–309.Mart<strong>in</strong>ez EA, Vazquez JM, Parrilla I, Cuello C, Gil MA,Rodriguez-Mart<strong>in</strong>ez H, Roca J, Vazquez JL, 2006: Incidenceof unilateral fertilizations after low dose deep <strong>in</strong>trauter<strong>in</strong>e<strong>in</strong>sem<strong>in</strong>ation <strong>in</strong> spontaneously ovulat<strong>in</strong>g sows underfield conditions. Reprod Dom Anim 41, 41–47.Maxwell WMC, Salamon S, 1977: Fertility of boar semen afterlong term frozen storage. Theriogenology 8, 206 (abstract).Maxwell WMC, Watson PF, 1996: Recent progress <strong>in</strong> thepreservation of ram semen. Anim Reprod Sci 42, 55–65.Morton DB, Glover JD, 1974: Sperm transport <strong>in</strong> the femalerabbit: the effect of <strong>in</strong>sem<strong>in</strong>ate volume and sperm density.J Reprod Fertil 38, 139–142.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


354 JM Vazquez, J Roca, MA Gil, C Cuello, I Parrilla, I Caballero, JL Vazquez and EA Martı´nezNadir S, Saacke RG, Bame JH, Mull<strong>in</strong>s J, Degelos S, 1993:Effect of freez<strong>in</strong>g semen and dosage of sperm on number ofaccessory sperm, fertility and embryo quality <strong>in</strong> artificially<strong>in</strong>sem<strong>in</strong>ated cattle. J Anim Sci 71, 199–204.Rath D, 2002: Low Dose <strong>in</strong>sem<strong>in</strong>ation <strong>in</strong> the Sow-A review.Rep Dom Anim 37, 201–205.Rath D, Ruiz S, Sieg B, 2003: Birth of female piglets follow<strong>in</strong>g<strong>in</strong>trauter<strong>in</strong>e <strong>in</strong>sem<strong>in</strong>ation of a sow us<strong>in</strong>g flow cytometricallysexed boar semen. Vet Rec 152, 400–401.Roca J, Carvajal G, Lucas X, Vazquez JM, Mart<strong>in</strong>ez EA,2003: Fertility of weaned sows after deep <strong>in</strong>trauter<strong>in</strong>e<strong>in</strong>sem<strong>in</strong>ation with a reduced number of frozen-thawedspermatozoa. Theriogenology 60, 77–87.Roca J, Vazquez JM, Gil MA, Cuello C, Parrilla I, Mart<strong>in</strong>ezEA, 2006: Challenges <strong>in</strong> pig artificial <strong>in</strong>sem<strong>in</strong>ation. ReprodDom Anim 41(Suppl. 2), 43–53.Rodriguez-Mart<strong>in</strong>ez H, Saravia F, Wallgren M, Tienthai P,Johannisson A, Vazquez JM, Mart<strong>in</strong>ez EA, Roca J, Sanz L,Calvete JJ, 2005: Boar spermatozoa <strong>in</strong> the oviduct. Theriogenology63, 514–535.Rodriguez-Vilar L, Lo´pez-Sánchez C, Boları´n A, Herna´ndezM, Vazquez JM, Mart<strong>in</strong>ez EA, Roca J, 2006: Effects of<strong>in</strong>sertion of deep <strong>in</strong>trauter<strong>in</strong>e <strong>in</strong>sem<strong>in</strong>ation device <strong>in</strong>to oneuter<strong>in</strong>e horn on the reproductive performance of multiparoussows. Reprod Dom Anim 41 (Suppl. 2), 124 (abstract).Str<strong>in</strong>g A, Berber E, Foroutani A, Macho JR, Pearl JM,Siperste<strong>in</strong> AE, 2001: Use of optical access trocar for safe andrapid entry <strong>in</strong> various laparoscopic procedures. Surg Endosc15, 570–573.Tummaruk P, Sumransap P, Techakumphu M, KunavongkritA, 2007: Distribution of spermatozoa and embryos <strong>in</strong> thefemale reproductive tract after unilateral deep <strong>in</strong>tra uter<strong>in</strong>e<strong>in</strong>sem<strong>in</strong>ation <strong>in</strong> the pig. Reprod Domest Anim 42, 603–609.Vazquez JM, Martı´nez EA, Parrilla I, Cuello C, Gil MA,Lucas X, Roca J, Vazquez JL, Didion BA, Day BN2001:Deep <strong>in</strong>trauter<strong>in</strong>e <strong>in</strong>sem<strong>in</strong>ation <strong>in</strong> natural post-wean<strong>in</strong>goestrus sows. In: Sixth International Conference on Pig<strong>Reproduction</strong>, Columbia, MO, USA, pp. 132.Vazquez JM, Mart<strong>in</strong>ez EA, Parrilla I, Roca J, Gil MA,Vazquez JL, 2003: Birth of piglets after deep <strong>in</strong>trauter<strong>in</strong>e<strong>in</strong>sem<strong>in</strong>ation with flow cytometrically sorted spermatozoa.Theriogenology 59, 1605–1614.Vazquez JM, Mart<strong>in</strong>ez EA, Roca J, Gil MA, Parrilla I, CuelloC, Carvajal G, Lucas X, Vazquez JL, 2005: Improv<strong>in</strong>g theefficiency of sperm technologies <strong>in</strong> pigs: the value of deep<strong>in</strong>trauter<strong>in</strong>e <strong>in</strong>sem<strong>in</strong>ation. Theriogenology 63, 536–547.Vazquez JM, Mart<strong>in</strong>ez EA, Parrilla I, Cuello C, Gil MA,Garcia E, Caballero I, Alm<strong>in</strong>ana C, Roca J, Vazquez JL,2006: Laparoscopic <strong>in</strong>traoviductal <strong>in</strong>sem<strong>in</strong>ation <strong>in</strong> pigs: Anew tool for improv<strong>in</strong>g the efficiency of sex sorted spermatozoa.Reprod Domest Anim 41, 298 (abstract).Watson PF2000: The causes of reduced fertility with cryopreservedsemen. Anim Reprod Sci 60–61, 481–492.Wongtawan T, Saravia F, Wallgren M, Caballero I, Rodriguez-Mart<strong>in</strong>ezH, 2006: Fertility after deep <strong>in</strong>tra-uter<strong>in</strong>eartificial <strong>in</strong>sem<strong>in</strong>ation of concentrated low-volume boarsemen doses. Theriogenology 65, 773–787.Author’s address (for correspondence): JM Vazquez, Department ofAnimal Medic<strong>in</strong>e and Surgery, Faculty of Veter<strong>in</strong>ary Medic<strong>in</strong>e,Campus de Esp<strong>in</strong>ardo, University of Murcia, E-30071, Murcia, Spa<strong>in</strong>.E-mail: vazquez@um.esConflict of <strong>in</strong>terest: All authors declare that the f<strong>in</strong>ancial supportwith<strong>in</strong> the past two years has been obta<strong>in</strong>ed from: CICYT (AGF98-0533; AGL2001-0471; AGL2004-07546 and AGF2005-00760), CDTI(B288/98; 03.180; 03-402, 04-0231), INIA (RZ01-019), FundacionSéneca de Murcia (PB/74/FS/02, 03002/PI/05 and (04543/GERM/07).The authors have no further conflict of <strong>in</strong>terests.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 355–358 (2008); doi: 10.1111/j.1439-0531.2008.01184.xISSN 0936-6768Production of Transgenic Farm <strong>Animals</strong> by Viral Vector-Mediated Gene TransferCBA Whitelaw, SG Lillico and T K<strong>in</strong>gThe Rosl<strong>in</strong> Institute and Royal (Dick) School of Veter<strong>in</strong>ary Studies, University of Ed<strong>in</strong>burgh, Ed<strong>in</strong>burghUKContentsTransgenic technology holds considerable promise to advanceunderstand<strong>in</strong>g <strong>in</strong> biomedical and agricultural systems withsome believ<strong>in</strong>g that one day transgenic animals may directlycontribute to farm<strong>in</strong>g and breed<strong>in</strong>g practice. Nevertheless,applications <strong>in</strong> livestock have been restricted <strong>in</strong> part by the<strong>in</strong>efficiency of the technology. The recent development oflentivirus vectors for transgene delivery may overcome some ofthis limitation. This presentation describes these vectors, theiradvantages and limitations.History of Transgenesis <strong>in</strong> LivestockThe first transgenic livestock were generated more thantwo decades ago (Hammer et al. 1985). As then the useof the pronuclear micro<strong>in</strong>jection technique, although<strong>in</strong>efficient, has been the most commonly used methodfor generat<strong>in</strong>g non-rodent transgenic animals. Pronuclear<strong>in</strong>jection <strong>in</strong>volves the direct <strong>in</strong>jection of thetransgene DNA <strong>in</strong>to one of the two pronuclei <strong>in</strong> thezygote (Simons et al. 1988; Nottle et al. 2001). Although<strong>in</strong>efficient, with usually only 1% of <strong>in</strong>jected eggsresult<strong>in</strong>g <strong>in</strong> a transgenic founder animal, the reliabilityof pronuclear <strong>in</strong>jection has ensured that it rema<strong>in</strong>s themost used method.The <strong>in</strong>efficiency of pronuclear <strong>in</strong>jection has been thema<strong>in</strong> driver <strong>in</strong> the search for more efficient methods.These <strong>in</strong>vestigations have resulted <strong>in</strong> a range of techniqueseach display<strong>in</strong>g different benefits and limitations(Clark and Whitelaw 2003). The use of sperm as adelivery route to generate transgenic livestock is attractivebecause of its simplicity but suffers from apparentvariability, while nuclear transfer us<strong>in</strong>g transgenic cellshas the advantage that all founder animals had to betransgenic but is currently restricted because of lowfoetal survival and high neonatal mortality.Five years after, pronuclear <strong>in</strong>jection was first demonstrated<strong>in</strong> livestock, an excit<strong>in</strong>g method was developedthat <strong>in</strong>volved sperm-mediated gene transfer(Lavitrano et al. 1989). The concept of simply mix<strong>in</strong>gthe transgene with sperm was very appeal<strong>in</strong>g because ofits simplicity. Unfortunately, this method has not seenwide uptake because of apparent variability <strong>in</strong> success(Br<strong>in</strong>ster et al. 1989; Noseno 2003). Recently, modifications<strong>in</strong>clud<strong>in</strong>g the use of <strong>in</strong>tracytoplasmic sperm<strong>in</strong>jection (Moreira et al. 2007) or the direct <strong>in</strong>jection ofthe transgene <strong>in</strong>to testes (Coward et al. 2007) may yetrek<strong>in</strong>dle <strong>in</strong>terest <strong>in</strong> this approach.Then came a huge technological advance; nucleartransfer (Wilmut et al. 1997; Campbell et al. 2006) –commonly called clon<strong>in</strong>g – offers the precise modificationto the genome. In this method, the donor cell usedfor reconstitut<strong>in</strong>g the embryo is first made transgenic.Both random (Schnieke et al. 1997) and sequencetargeted (McCreath et al. 2000) transgene <strong>in</strong>tegrationis possible. This approach to transgenesis is therefore,somewhat analogous to the long established use ofembryonic stem cells <strong>in</strong> mice (Capecchi 1989). Incontrast, pronuclear micro<strong>in</strong>jection only offers random<strong>in</strong>tegration of the transgene, result<strong>in</strong>g <strong>in</strong> the transgenesuffer<strong>in</strong>g from position-effects, which make expressionof the transgene unpredictable (al-Shawi et al. 1990;Wilson et al. 1990). Therefore, the precise nature of thegenetic modification possible us<strong>in</strong>g nuclear transfermakes this approach to alter<strong>in</strong>g the activity of a specificgene very attractive. Yet, the method rema<strong>in</strong>s <strong>in</strong>efficientand technically demand<strong>in</strong>g (Campbell et al. 2007); andfor many hypothesis driven studies, this <strong>in</strong>efficiencyrenders this method too expensive both <strong>in</strong> cost andanimal numbers.Recently, livestock have been produced us<strong>in</strong>g anothermethod, one that has the benefit of be<strong>in</strong>g very efficient.This method utilizes a viral vector and builds on theprogress achieved <strong>in</strong> the development of gene therapystrategies. The use of viral vectors <strong>in</strong> transgenesis is notnew, with the first ever transgenic mice carry<strong>in</strong>g aretroviral vector transgene (Jaenisch 1976). Yet, thesefirst generation vectors were often transcriptionallysilenced (Pannell and Ellis 2001). The new vectors arebased on a specific type of retrovirus called a lentivirusand are both robustly expressed and offer spectaculartransgenesis efficiencies (Pfeifer 2004; Whitelaw 2004).This presentation describes these vectors, their advantagesand limitations. It does not address what transgenicanimals might be used for: this is left to theimag<strong>in</strong>ation of the reader.Lentivirus Vectors – Pros and ConsLentiviruses are <strong>in</strong>fectious retroviral pathogens thatcause a number of diseases <strong>in</strong> a range of species<strong>in</strong>clud<strong>in</strong>g man. Therefore, a lentivirus had to be disabledfor it to be used as a vector (Amado and Chen 1999;Nald<strong>in</strong><strong>in</strong> and Verma 2000; Loewen and Poeschla 2005).Lentivirus vectors are generated by deletion of key genesfrom the viral genome <strong>in</strong>volved <strong>in</strong> packag<strong>in</strong>g andreplication of the virus from the viral genome. In thismethod, the replication-defective vector is only able togo through the first phase of the <strong>in</strong>fectious cycle once,and not able to produce <strong>in</strong>fectious virus. Vector particlesare produced by <strong>in</strong>troduc<strong>in</strong>g the vector DNA <strong>in</strong>to apackag<strong>in</strong>g cell l<strong>in</strong>e <strong>in</strong> trans with the miss<strong>in</strong>g prote<strong>in</strong>s bytransfection strategies (Pfeifer 2004).An additional key feature is that exogenous transcriptionelements can be <strong>in</strong>serted <strong>in</strong>to the vector (Pfeifer2004). This allows control of the spatial and temporaltransgene expression profile. Apart from the <strong>in</strong>troducedÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


356 CBA Whitelaw, SG Lillico and T K<strong>in</strong>gpromoter element, the vector is otherwise transcriptionallysilent. This is achieved by <strong>in</strong>troduc<strong>in</strong>g mutations<strong>in</strong>to the viral genome transcription control sequences(the long-term<strong>in</strong>al repeats) to generate a self-<strong>in</strong>activat<strong>in</strong>gvector (Pfeifer 2004). This dist<strong>in</strong>guishes the lentivirusvector from other retrovirus vectors, which rely on viraltranscriptional elements for transgene activity (Pfeifer2004).With regard to transgenesis, lentivirus vectors haveseveral advantages (Pfeifer 2004; Whitelaw 2004).A dist<strong>in</strong>guish<strong>in</strong>g property of lentivirus vectors is theirability to <strong>in</strong>fect both divid<strong>in</strong>g and non-divid<strong>in</strong>g cells. Itis this property that has promoted their development asgene delivery vectors s<strong>in</strong>ce they can be delivered to eitherthe egg or zygote. Second, delivery of the vector,although still requir<strong>in</strong>g the use of micro<strong>in</strong>jection equipment,is less damag<strong>in</strong>g to the egg ⁄ zygote when comparedwith pronuclear micro<strong>in</strong>jection result<strong>in</strong>g <strong>in</strong> aconsiderable proportion of <strong>in</strong>jected embryos develop<strong>in</strong>gto term. Exploit<strong>in</strong>g the natural efficiency of <strong>in</strong>fectionafforded to virus by evolution, the vector fuses with thecell (egg or zygote) and is <strong>in</strong>ternalized. This means thatthe lentivirus vector can be <strong>in</strong>troduced (Fig. 1), by<strong>in</strong>jection <strong>in</strong>to the perivitell<strong>in</strong>e space of the zygote (Loiset al. 2002; Ritchie et al. 2007) or by co-culture with azona-free (or laser microdrilled) zygote (Ewerl<strong>in</strong>g et al.2006). This delivery method results <strong>in</strong> a very highproportion of <strong>in</strong>jected embryos surviv<strong>in</strong>g. Recent datasuggests that many embryo manipulation methods causeDNA damage and thus limited embryo development(Yamauchi et al. 2007). Lentivirus vector-mediatedtransgene delivery appears not to suffer from thisproblem, perhaps because of the ‘natural’ route of<strong>in</strong>fection that they use to deliver the transgene.A further and significant property of lentivirus vectorsis the spectacular efficiency of transgenesis that can beachieved. Transgenic mice (Lois et al. 2002; Pfeifer et al.LTRPromoter - transgeneFig. 1. Diagram of lentiviral vector delivery methods. A prototypicalvector is shown long-term<strong>in</strong>al repeats encompass<strong>in</strong>g the promotertransgeneof choice. Packaged viral vectors can then be either coculturedwith zona denuded (or microdrilled) zygotes, which requires<strong>in</strong>dividual cultur<strong>in</strong>g of each embryo or <strong>in</strong>jected <strong>in</strong>to the perivitell<strong>in</strong>espace of an oocyte or zygote. Both methods have been demonstrated toproduce live transgenic animalsLTR2002), rats (Lois et al. 2002; Dann 2007; Michalkiewiczet al. 2007), poultry (McGrew et al. 2004; Lillico et al.2007), pigs (Hofmann et al. 2003; Whitelaw et al. 2004),cattle (Hofmann et al. 2004), sheep (Whitelaw, unpublished)and recently rabbits (Zsuzsa Bosze, personalcommunication) have been made us<strong>in</strong>g this method. Formany of these species, transgenesis rates of up to 100%of <strong>in</strong>jected embryos carry<strong>in</strong>g an <strong>in</strong>tegrated transgenehave been achieved. This had to be compared with thestandard pronuclear <strong>in</strong>jection method, where 5% ofmouse zygotes and 1% of livestock zygotes result <strong>in</strong> atransgenic founder animal. Furthermore, unlike pronuclear<strong>in</strong>jection, most of these <strong>in</strong>tegration events carrytranscriptionally active transgenes. Taken together, theadvantageous properties of lentivirus vectors make thema very attractive transgene delivery route, especially forspecies, where the more standard methods are verycostly <strong>in</strong> terms of animal numbers and <strong>in</strong> the f<strong>in</strong>ancialsupport required.Unfortunately like all good th<strong>in</strong>gs, lentivirus vectorshave properties, which can constra<strong>in</strong> their application(Pfeifer 2004; Whitelaw et al. 2004). Perhaps, the mostsignificant concern is the reduced ‘cargo’ capacity. Thesevectors can only carry small transgenes (up to 8 kb),although <strong>in</strong> practice, transgenes bigger than 6 kb oftendisplay poor packag<strong>in</strong>g efficiency and low vector titres.Second, the vector <strong>in</strong>tegrates as a s<strong>in</strong>gle copy butmultiple <strong>in</strong>tegrations can occur. This leads to segregationissues <strong>in</strong> subsequent generations. Yet, s<strong>in</strong>ce founderanimals carry different copies of the transgene, this canbe an advantage for founder population studies, offer<strong>in</strong>ga dose–response to transgene activity.Integration is random, as <strong>in</strong> the pronuclear <strong>in</strong>jectionmethod. This can result <strong>in</strong> position-effect phenomenonthat is often associated with pronuclear <strong>in</strong>jection.Position-effects can result <strong>in</strong> aberrant or ectropic transgeneactivity, and poorly express<strong>in</strong>g or silent transgeneloci (Wilson et al. 1990). In our experience, the authorshave rarely seen these <strong>in</strong>appropriate transgene expressionprofiles, although others have (Hofmann et al.2006). The authors have seen expression <strong>in</strong> 4-year-oldpigs at the same level they displayed as a young pig; <strong>in</strong>sheep, pigs and chickens the vast majority of transgenicanimals that they produced us<strong>in</strong>g lentiviral vectorsexpress the transgene; although the authors haveobserved transgenerational reduction <strong>in</strong> expression(after the 6th generation) <strong>in</strong> a few l<strong>in</strong>es of transgenemice (the majority of l<strong>in</strong>es do not show silenc<strong>in</strong>g), theauthors have yet to see any transgenerational silenc<strong>in</strong>g<strong>in</strong> chickens and pigs. Random <strong>in</strong>tegration also raises thepossibility of functional impairment of the host genomeat the site of <strong>in</strong>tegration. Lentiviral transgene <strong>in</strong>tegrationsites have yet to be mapped <strong>in</strong> animals but do lieclose to genes <strong>in</strong> studies on human cells and, although atvery low frequency, can cause gene activation (Themiset al. 2005; Mont<strong>in</strong>i et al. 2006). To overcome thislimitation, there is a considerable effort be<strong>in</strong>g directed todesign lentivirus vectors that can target transgene<strong>in</strong>tegration to specific sequences with<strong>in</strong> the genome(Balciunas et al. 2006; Philippe et al. 2006; Clark et al.2007; Lombardo et al. 2007; Sh<strong>in</strong>ohara et al. 2007).F<strong>in</strong>ally, it must be remembered that VSV-G pseudotyped(Pfeifer 2004) lentivirus vectors have the potentialÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Lentiviral Transgenesis 357to <strong>in</strong>fect the handler. This function is lost upon<strong>in</strong>tegration s<strong>in</strong>ce these vectors are replication-defective.Nevertheless this method, at least the <strong>in</strong>itial laboratorybasedsteps of virus preparation and handl<strong>in</strong>g, requiresgreater focus on biosafety than other method oftransgenesis.Livestock Transgenesis – The Best has Yet toComeLentivirus vectors represent a useful new method fortransgene delivery, especially to non-rodent species,offer<strong>in</strong>g unprecedented numbers of transgenic founderanimals. In essence, regardless of what species, eachlitter will carry transgenic founder animals. This meansthat for many studies, evaluation of transgene functioncan be carried out <strong>in</strong> the founder population. Overall,the efficiency and founder study strategy results <strong>in</strong> theuse of significantly fewer animals per study anddramatically reduces the timel<strong>in</strong>e of a study. Therefore,there are substantial cost sav<strong>in</strong>gs mak<strong>in</strong>g it feasible todesign studies that were previously restricted to mousebasedexperiments. Perhaps, the possibility of comb<strong>in</strong><strong>in</strong>glentivirus vector efficiency with the excit<strong>in</strong>g potentialof RNA-<strong>in</strong>terference offers an excit<strong>in</strong>g future for transgeniclarge animals (Clark and Whitelaw 2003; :Tiscornia et al. 2003; Sh<strong>in</strong> et al. 2006).Already this method of transgene delivery hasyielded successful applications, exemplified by thetargeted production of two therapeutic prote<strong>in</strong>s <strong>in</strong>transgenic hens eggs (Lillico et al. 2007); and more areunderway. The authors are aware of studies look<strong>in</strong>gto deliver RNAi-based systems to combat viral <strong>in</strong>fection<strong>in</strong> chickens and sheep, and attempts to produceanimal models of human disease focuss<strong>in</strong>g on diseasesof the eye, lung and throat. In addition, studiesutiliz<strong>in</strong>g lentiviral vectors are now allow<strong>in</strong>g mechanisticstudies of basic biological traits, for examplephotoperiodism <strong>in</strong> sheep, to be <strong>in</strong>vestigated <strong>in</strong> largeanimals, experiments that were simply not feasibleus<strong>in</strong>g standard transgene delivery methods <strong>in</strong> thesespecies.Lentivirus vectors suffer from two drawbacks; limitedcargo capacity and random <strong>in</strong>tegration. It is not easy tosee how the carry<strong>in</strong>g capacity of viral vectors can besignificantly improved; this aspect is likely to be overcomethrough the generation of robust methods forartificial chromosome transfer (Kuroiwa et al. 2002).Yet, to prevent <strong>in</strong>sertional mutagenesis or positioneffects,the targeted <strong>in</strong>tegration of viral vectors is under<strong>in</strong>tense <strong>in</strong>vestigation and is an achievable goal. As theauthors learnt more about how to harness sequenceb<strong>in</strong>d<strong>in</strong>g activities of factors such z<strong>in</strong>c-f<strong>in</strong>ger b<strong>in</strong>d<strong>in</strong>gprote<strong>in</strong>s, they might able to overcome this limitation.Alternatively, manipulat<strong>in</strong>g the b<strong>in</strong>d<strong>in</strong>g specificity affordedby recomb<strong>in</strong>ase enzymatic activities is likely toresult <strong>in</strong> sequence-directed transgene <strong>in</strong>tegration strategies(Balciunas et al. 2006; Clark et al. 2007; Lombardoet al. 2007; Sh<strong>in</strong>ohara et al. 2007). These technologiesare just around the corner but perhaps the best is yet tocome. With the research activity <strong>in</strong> this area, <strong>in</strong> concertto the energy be<strong>in</strong>g applied to the development of genetherapy delivery methods, there is the real prospect thatefficient, sequence-specific transgene <strong>in</strong>tegration <strong>in</strong> theegg or zygote will be available soon.AcknowledgementsThe authors recognize the support and discussion with both presentand past members of this group. All animal work at The Rosl<strong>in</strong>Institute is performed under licence from the UK Home Office. Work<strong>in</strong> the authors’ group is supported by a number of sources,<strong>in</strong> particular, a SFHEFC Strategic Research Development Grant‘Development of Novel Technologies to Fight Viral Diseases <strong>in</strong> Farm<strong>Animals</strong>’, the European Commission NEST029025 project INTEGRAand the BBSRC.ReferencesAmado RG, Chen IS, 1999: Lentiviral vectors – the promise ofgene therapy <strong>in</strong> reach. Science 285, 674–676.Balciunas D, Wangensteen KJ, Wilber A, Bell J, Geurts A,Sivasubbu S, Wang X, Hackett PB, Largaespada DA,McIvor RS, Ekker SC, 2006: Harness<strong>in</strong>g a high cargocapacitytransposon for genetic applications <strong>in</strong> vertebrates.PLoS Genet 2, e169.Br<strong>in</strong>ster RL, Sandgren EP, Behr<strong>in</strong>ger RR, Palmiter RD, 1989:No simple solution for mak<strong>in</strong>g transgenic mice. Cell 59,239–241.Campbell KH, McWhir J, Ritchie WA, Wilmut I, 2006: Sheepcloned by nuclear transfer from a cultured cell l<strong>in</strong>e. Nature380, 64–68.Campbell KH, Fisher P, Chen WC, Choi I, Kelly RD, Lee JH,Xhu J, 2007: Somatic cell nuclear transfer: past, present andfuture perspectives. Theriogenology 68(Suppl. 1), S214–S231.Capecchi MR, 1989: Alter<strong>in</strong>g the genome by homologousrecomb<strong>in</strong>ation. Science 244, 1288–1292.Clark AJ, Whitelaw CBA, 2003: A future for transgeniclivestock. Nat Rev Genet 4, 825–833.Clark KJ, Carlson DF, Foster LK, Kong BW, Foster DN,Fahrenkrug SC, 2007: Enzymatic eng<strong>in</strong>eer<strong>in</strong>g of the porc<strong>in</strong>egenome with transposons and recomb<strong>in</strong>ases. BMC Biotechnol17, 42.Coward K, Kubota H, Parr<strong>in</strong>gton J, 2007: In vivo genetransfer <strong>in</strong>to testis and sperm: developments and futureapplication. Arch Androl 53, 187–197.Dann CT, 2007: New technology for an old favorite: lentiviraltransgenesis and RNAi <strong>in</strong> rats. Transgenic Res 16, 571–580.Ewerl<strong>in</strong>g S, Hofmann A, Klose R, Weppert M, Brem G, R<strong>in</strong>kK, Pfeifer A, Wolf E, 2006: Evaluation of laser-assistedlentiviral transgenesis <strong>in</strong> bov<strong>in</strong>e. Transgenic Res 15, 447–454.Hammer RE, Pursel VG, Rexroad CE, Wall RJ, Bolt DJ,Ebert KM, Palmiter RD, Br<strong>in</strong>ster RL, 1985: Production oftransgenic rabbits, sheep and pigs by micro<strong>in</strong>jection. Nature315, 680–683.Hofmann A, Kessler B, Ewerl<strong>in</strong>g S, Weppert W, Vogg B,Ludwig H, Stojkovic M, Boelhauve M, Brem G, Wolf E,Pfeifer A, 2003: Efficient transgenesis <strong>in</strong> farm animals bylentiviral vectors. EMBO Rep 4, 1054–1060.Hofmann A, Zakhartchenko V, Weppert M, Sebald H,Wenigerk<strong>in</strong>d H, Brem G, Wolf E, Pfeifer A, 2004: Generationof transgenic cattle by lentiviral gene transfer <strong>in</strong>tooocytes. Biol Reprod 71, 405–409.Hofmann A, Kessler B, Ewerl<strong>in</strong>g S, Kabermann A, Brem G,Wolf E, Pfeifer A, 2006: Epigenetic regulation of lentiviraltransgene vectors <strong>in</strong> a large animal model. Mol Ther 13, 59–66.Jaenisch R, 1976: Germ l<strong>in</strong>e <strong>in</strong>tegration and Mendeliantransmission of the exogenous Moloney leukemia virus.Proc Natl Acad Sci USA 73, 1260–1264.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


358 CBA Whitelaw, SG Lillico and T K<strong>in</strong>gKuroiwa Y, Kas<strong>in</strong>athan P, Choi YJ, Naeem R, Tomizuka K,Sullivan EJ, Knott JG, Duteau A, Goldsby RA, OsborneBA, Ishida I, Robl JM, 2002: Cloned transchromosomiccalves produc<strong>in</strong>g human immunoglobul<strong>in</strong>. Nat Biotechnol20, 889–894.Lavitrano M, Camaioni A, Fazio VM, Dolci S, Farace MG,Spadafora C, 1989: Sperm cells as vectors for <strong>in</strong>troduc<strong>in</strong>gforeign DNA <strong>in</strong>to eggs: genetic transformation of mice. Cell57, 717–723.Lillico SG, Sherman A, McGrew MJ, Roberston CD, Smith J,Haslam C, Barnard P, Radcliffe PA, Mitrophanous KA,Elliot EA, Sang H, 2007: Oviduct-specific expression of twotherapeutic prote<strong>in</strong>s <strong>in</strong> transgenic hens. Proc Natl Acad SciUSA 104, 1771–1776.Loewen N, Poeschla EM, 2005: Lentiviral vectors. AdvBiochem Eng Biotechnol 99, 169–191.Lois C, Hong EJ, Pease S, Brown EJ, Baltimore D, 2002:Germl<strong>in</strong>e transmission and tissue-specific expression oftransgenes delivered by lentiviral vectors. Science 295,868–872.Lombardo A, Genovese P, Peausejour CM, Colleoni S, leeYL, Kim KA, Ando D, Urnow FD, Galli C, Gregory PD,Holmes MC, Nald<strong>in</strong>i L, 2007: Gene edit<strong>in</strong>g <strong>in</strong> human stemcells us<strong>in</strong>g z<strong>in</strong>c f<strong>in</strong>ger nucleases and <strong>in</strong>tegrase-defectivelentiviral vector delivery. Nat Biotechnol 25, 1298–1306.McCreath KJ, Howcroft J, Campbell KH, Colman A,Schneike AE, K<strong>in</strong>d AJ, 2000: Production of gene-targetedsheep by nuclear transfer from cultured somatic cells.Nature 405, 1066–1069.McGrew MJ, Sherman A, Ellard FM, Lillico SG, GilhooleyHJ, K<strong>in</strong>gsman AJ, Mitrophanous KA, Sang H, 2004:Efficient production of germl<strong>in</strong>e transgenic chickens us<strong>in</strong>glentiviral vectors. EMBO Rep 5, 728–733.Michalkiewicz M, Michalkiewicz T, Geurts AM, Roman RJ,Slocum GR, S<strong>in</strong>ger O, Wihrauch D, Greene AS, KaldunskiM, Verma IM, Jacob HJ, Cowley AW Jr, 2007: Efficienttransgenic rat production by a lentiviral vector. Am JPhysiol Heart Circ Physiol 293, H881–H894.Mont<strong>in</strong>i E, Cesana D, Shcnidt M, Sanvito F, Ponzoni M,Bartholomae C, Sergi Sergi L, Benedicenti F, Ambrosi A, DiSerio C, Doglioni C, von Kalle C, Nald<strong>in</strong>i L, 2006:Hematopoietic stem cell gene transfer <strong>in</strong> a tumor-pronemouse model uncovers low genotoxicity of lentiviral vector<strong>in</strong>tegration. Nat Biotechnol 24, 687–696.Moreira PN, Pozueta J, Perez-Crespo M, Valdivieso F,Gutierrez-Adan A, Montoliu L, 2007: Improv<strong>in</strong>g the generationof genomic-type transgenic mice by ICSI. TransgenicRes 16, 163–168.Nald<strong>in</strong><strong>in</strong> L, Verma IM, 2000: Lentiviral vectors. Adv VirusRes 55, 599–609.Noseno N, 2003: Piglets add some colour to transgenic story.Nature 424, 604.Nottle MB, Haskard KA, Verma PJ, Du ZT, Grupen CG,McIlfatrick SM, Ashman RJ, Harrison SJ, Barlow H,Wigley PL, Lyons IG, Cowan PJ, Crawford RJ, TolstoshevPL, Pearse MJ, Rob<strong>in</strong>s AJ, d’Apice AJ, 2001: Effect ofDNA concentration on transgenesis rates <strong>in</strong> mice and pigs.Transgenic Res 10, 523–531.Pannell D, Ellis J, 2001: Silenc<strong>in</strong>g of gene expression: implicationsfor design of retrovirus vectors. Rev Med Virol 11, 205–217.Pfeifer A, 2004: Lentiviral transgenesis. Transgenic Res 13, 513–522.Pfeifer A, Ikawa M, Dyn Y, Verma IM, 2002: Transgenesis bylentiviral vectors: lack of gene silenc<strong>in</strong>g <strong>in</strong> mammalianembryonic stem cells and preimplantation embryos. ProcNatl Acad Sci USA 99, 2140–2145.Philippe S, Sarkis C, Barkats M, Mammeri H, Ladroue C,Petit C, Mallet J, Serguera C, 2006: Lentiviral vectors with adefective <strong>in</strong>tegrase allow efficient and susta<strong>in</strong>ed transgeneexpression <strong>in</strong> vitro and <strong>in</strong> vivo. Proc Natl Acad Sci USA103, 17684–17689.Ritchie WA, Neil C, K<strong>in</strong>g T, Whitelaw CBA, 2007: Transgenicembryos and mice produced from low titre lentiviral vectors.Transgenic Res 16, 661–664.Schnieke AE, K<strong>in</strong>d AJ, Ritchie WA, Mycock K, Scott AR,Ritchie M, Wilmut I, Colman A, Campbell KH, 1997:Human factor IX transgenic sheep produced by transfer ofnuclei from transfected fetal fibroblasts. Science 278, 2130–2133.al-Shawi R, K<strong>in</strong>naird J, Burke J, Bishop JO, 1990: Expressionof a foreign gene <strong>in</strong> a l<strong>in</strong>e of transgenic mice is modulated bya chromosomal position effect. Mol Cell Biol 10, 1192–1198.Sh<strong>in</strong> KJ, Wall EA, Zavzavadjian JR, Santat LA, Liu J, HwangJI, Rebres R, Roacj T, Seaman W, Simon MI, Fraser ID,2006: A s<strong>in</strong>gle lentiviral vector platform for microRNAbasedconditional RNA <strong>in</strong>terference and coord<strong>in</strong>ated transgeneexpression. Proc Natl Acad Sci USA 103, 13759–13764.Sh<strong>in</strong>ohara ET, Kam<strong>in</strong>ski JM, Segal DJ, Pelczar P, Kolhe R,Ryan T, Costes CJ, Fraser MJ, Handler AM, YamagimachiR, Moisyadi S, 2007: Active <strong>in</strong>tegration: new strategies fortransgenesis. Transgenic Res 16, 333–339.Simons JP, Wilmut I, Clark AJ, Archibald AL, Bishop JO,Lathe R, 1988: Gene transfer <strong>in</strong>to sheep. Biotechnology 6,179–183.Themis M, Wadd<strong>in</strong>gton SN, Schmidt M, von Kalle C, WangY, Al-Allaf F, Gregory LG, Novsarkar M, Holder MV,Buckley SM, Dighe N, Ruthe AT, Mistry A, Bigger B,Rahim A, Nguyen TH, Trono D, Thrasher AJ, Coutelle C,2005: Oncogenesis follow<strong>in</strong>g delivery of a nonprimatelentiviral gene therapy vector to fetal and neonatal mice.Mol Ther 12, 763–771.Tiscornia G, S<strong>in</strong>ger O, Ikawa M, Verma IM, 2003: A generalmethod for gene knockdown <strong>in</strong> mice by us<strong>in</strong>g lentiviralvectors express<strong>in</strong>g small <strong>in</strong>terfer<strong>in</strong>g RNA. Proc Natl AcadSci USA 100, 1844–1848.Whitelaw CBA, 2004: Transgenic livestock made easy. TrendsBiotechnol 22, 157–159.Whitelaw CBA, Radcliffe PA, Ritchie WA, Carlisle A, EllardFM, Pena RN, Rowe J, Clark AJ, K<strong>in</strong>g TJ, MitrophanousKA, 2004: Efficient generation of transgenic pigs us<strong>in</strong>gequ<strong>in</strong>e <strong>in</strong>fectious anaemia virus (EIAV) derived vector.FEBS Lett 571, 233–236.Wilmut I, Schnieke AE, McWhir J, K<strong>in</strong>d AJ, Campbell KH,1997: Viable offspr<strong>in</strong>g derived from fetal and adult mammaliancells. Nature, 385, 810–813.Wilson C, Bellen HJ, Gehr<strong>in</strong>g WJ, 1990: Position effects oneukaryotic gene expression. Annu Rev Cell Biol 6, 679–714.Yamauchi Y, Doe B, Ajduk A, Ward MA, 2007: GenomicDNA damage <strong>in</strong> mouse transgenesis. Biol Reprod 77, 803–812.Author’s address (for correspondence): C Bruce A Whitelaw, TheRosl<strong>in</strong> Institute and Royal (Dick) School of Veter<strong>in</strong>ary Studies,University of Ed<strong>in</strong>burgh, Rosl<strong>in</strong>, Midlothian, Ed<strong>in</strong>burgh EH25 9PS,UK. E-mail: bruce.whitelaw@rosl<strong>in</strong>.ed.ac.ukConflict of <strong>in</strong>terest: CBA Whitelaw has received both National andInternational research fund<strong>in</strong>g from various public bodies to pursuethis research; the rema<strong>in</strong><strong>in</strong>g authors declare no conflict of <strong>in</strong>terests.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 359–367 (2008); doi: 10.1111/j.1439-0531.2008.01185.xISSN 0936-6768Use of Microarray Technology to Profile Gene Expression Patterns Important for<strong>Reproduction</strong> <strong>in</strong> CattleACO Evans 1 , N Forde 1 , GM O’Gorman 1 , AE Zielak 2 , P Lonergan 1 and T Fair 11 School of Agriculture, Food Science and Veter<strong>in</strong>ary Medic<strong>in</strong>e, and the Conway Institute, University College Dubl<strong>in</strong>, Dubl<strong>in</strong>, Ireland; 2 Institute ofAnimal Breed<strong>in</strong>g, Faculty of Biology and Animal Science, Wroclaw University of Environmental and Life Sciences, Wroclaw, PolandContentsFertility <strong>in</strong> cattle is a major component of many agriculturalenterprises and there is pressure to devise methods to improvethis. A number of approaches are ongo<strong>in</strong>g, one of which is tobetter understand the cellular and molecular events of thedevelopment of reproductive tissues and to use these as targetsfor develop<strong>in</strong>g new strategies. Microarray technologies nowallow us the potential to determ<strong>in</strong>e the transcriptional profileof expressed genes <strong>in</strong> a given tissue. This review focuses on thetypes of microarrays available for studies <strong>in</strong> cattle andconcludes that genes associated with one or more of thecellular processes of cell survival ⁄ death, <strong>in</strong>tracellular signall<strong>in</strong>g,transcription and translation, cell division and proliferationand cellular metabolism are the ma<strong>in</strong> transcriptionalpathways that control the development of ovarian follicles,oocytes, early embryos and the uter<strong>in</strong>e endometrium about thetime of the establishment of pregnancy.IntroductionFertility rates <strong>in</strong> cattle (pregnancies per <strong>in</strong>sem<strong>in</strong>ation)are depend<strong>in</strong>g on the types of animals be<strong>in</strong>g bred andthe production system that they are <strong>in</strong>; however, ratesof more than 50% <strong>in</strong> multiparous animals (i.e. notheifers) are not often achieved (Disk<strong>in</strong> et al. 2006).Strategies that aim to improve fertility rates depend onimprov<strong>in</strong>g our understand<strong>in</strong>g of organ, cellular andmolecular events that control reproduction and it is forthis reason that there is a great <strong>in</strong>terest <strong>in</strong> determ<strong>in</strong><strong>in</strong>gthe relationship between the expression of genes <strong>in</strong>tissues and their consequences for fertility. A numberof recent publications have reviewed the potential formicroarrays and bov<strong>in</strong>e reproduction (Niemann et al.2007; Smith and Rosa 2007; Wrenzycki et al. 2007;Bonnet et al. 2008; Mitko et al. 2008). In this review,we focus on the different microarray approaches andtechnologies that are <strong>in</strong> use and then go on to reviewthe genomics of ovarian follicle development, oocytedevelopment, embryo development and uter<strong>in</strong>e functionabout the time of the establishment of pregnancy<strong>in</strong> cattle.Microarrays and Functional GenomicsThe term genomics was proposed <strong>in</strong> 1987 as thebr<strong>in</strong>g<strong>in</strong>g together of molecular biology, cell biology andgenetics (McKusick and Ruddle 1987) and refers to thestudy of whole sets of genes and their <strong>in</strong>teractions. Thestudy of the expression of particular genes at specifictimes <strong>in</strong> cells, that leads to a description of theirfunction, is termed functional genomics. Advances <strong>in</strong>the scale and sophistication of DNA microarrays haveheralded a new era away from the traditional gene-bygeneapproach and towards the study of complex<strong>in</strong>terrelated processes that frequently occur <strong>in</strong> biologicalsystems. Although many different microarraysystems have been developed, the most commonly usedsystems are complementary DNA (cDNA) microarrays,oligonucleotide microarrays and high-density(Affymetrix-style) fabricated short oligonucleotidemicroarrays.Complementary DNA (cDNA) microarraysThe earliest and still most widely used form of geneexpression arrays are DNA microarrays where a presynthesizedprobe (cDNA or oligonucleotide) is spottedonto a support (Hager 2006). The first generation ofmicroarrays used cDNA as probes on glass slides(Schena et al. 1995). These early microarrays weregenerally available for human, rodent and other majormodel organisms, while those for cattle were developedlater and cont<strong>in</strong>ue to be developed. Many of thesebov<strong>in</strong>e cDNA microarrays are customized or specificfor particular research areas of <strong>in</strong>terest, which has anadvantage of produc<strong>in</strong>g ‘bespoke’ <strong>in</strong>-house microarrays.Probes for cDNA microarrays are usuallyproducts of the polymerase cha<strong>in</strong> reaction (PCR)generated from cDNA libraries or clone collections,us<strong>in</strong>g either vector-specific or gene-specific primers, andare pr<strong>in</strong>ted onto glass slides or nylon membranes asspots at def<strong>in</strong>ed locations (Schulze and Downward2001). On most slides, genes are spotted <strong>in</strong> duplicate ortriplicate, where the cDNA <strong>in</strong> each spot is usually awhole gene or expressed sequence tag (EST) and isoptimally approximately 300–800 nucleotides long(Bryant et al. 2004). The availability of cDNA clonesets for array<strong>in</strong>g has depended on large-scale ESTsequenc<strong>in</strong>g projects that have resulted <strong>in</strong> millions ofEST sequences be<strong>in</strong>g deposited <strong>in</strong> GenBank; subsequentlythe complexity of these collections is reducedby the assembly of related sequences <strong>in</strong>to clusters(Lyons 2003). Overall, cDNA microarrays have beenvital for the <strong>in</strong>itial development and dissem<strong>in</strong>ation ofmicroarray technology. However, a range of technicalissues arise dur<strong>in</strong>g their production; these <strong>in</strong>clude phagecontam<strong>in</strong>ation, <strong>in</strong>correct annotation and errors <strong>in</strong> highthroughputproduction of probe sets generated fromtens of thousands of bacterial clones. These problemshave prompted a search for an alternative type of probeand spotted oligonucleotide microarrays have been onepopular alternative.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


360 ACO Evans, N Forde, GM O’Gorman, AE Zielak, P Lonergan and T FairOligonucleotide microarraysThe reliability of synthesis and fall<strong>in</strong>g production costsof pre-synthesized long (50–70 mer) oligonucleotideshave made them a viable alternative probe choice <strong>in</strong>microarray production (Lyons 2003). Improvements <strong>in</strong>slide surface chemistry and, <strong>in</strong> particular, the ability toattach oligonucleotide probes to slides at a sufficientconcentration has resulted <strong>in</strong> the successful developmentof the oligonucleotide microarray platform (Hollowayet al. 2002). In addition, genes that are expressed <strong>in</strong> lowabundance and are not represented <strong>in</strong> available cDNAlibraries can be <strong>in</strong>cluded provided that sequence <strong>in</strong>formationis available, as is the case for many organismsnow. The design of oligonucleotides is highly critical andalgorithms have been developed to <strong>in</strong>crease theirperformance (Kreil et al. 2006). A study by Hugheset al. (2001) exam<strong>in</strong>ed oligonucleotide sensitivity andspecificity, and found that 60-mer oligonucleotides werereliable when complex biological samples were used andsuggested that microarrays with one s<strong>in</strong>gle carefullyselected oligonucleotide per gene correlated closely withcDNA microarrays (Hughes et al. 2001). Overall, synthesizedoligonucleotide probes have become the mostpopular format <strong>in</strong> many DNA microarray facilities.When good oligonucleotide probe design practices areemployed they can potentially discrim<strong>in</strong>ate betweenhighly similar targets, such as splice variants or genefamilies (Kreil et al. 2006).The process<strong>in</strong>g steps for both types of spottedmicroarrays, cDNA and oligonucleotide, are the same<strong>in</strong> many respects. Two samples (mRNA to convertedcDNA), a ‘test’ and a ‘control’ are differentiallyfluorescently labelled with fluorochrome dyes. Depend<strong>in</strong>gon the experimental design selected, one sample maybe a reference that is used <strong>in</strong> all hybridizations and actsas an <strong>in</strong>ternal control. The labell<strong>in</strong>g of the samples canoccur dur<strong>in</strong>g the reverse transcription (direct method)or afterwards (<strong>in</strong>direct method). The two samples arethen comb<strong>in</strong>ed and allowed to hybridize to the immobilizedtargets on the microarray under sensitive andspecific conditions. Competitive hybridization yields aquantitative ratio of the two fluorescent dyes, determ<strong>in</strong>edafter the slide is scanned and gives <strong>in</strong>formationabout the comparative over- or under-expression ofgenes <strong>in</strong> that particular experimental sample (Bryantet al. 2004).Affymetrix GeneChips ÒThe high-density, synthesized <strong>in</strong> situ or fabricatedoligonucleotide platform employs the most sophisticatedmicroarray technology available to date. Inparticular, the Affymetrix GeneChip Ò has been widelyaccepted and is considered the gold standard <strong>in</strong> globalgene expression analysis (http://www.affymetrix.com).The GeneChip Ò technology platform consists of highdensitymicroarrays, which are used with standardizedassays, reagents, <strong>in</strong>strumentation, data managementand analysis tools (McGall and Christians 2002). TheGeneChip Ò bov<strong>in</strong>e genome microarray was designedbased on content from bov<strong>in</strong>e UniGene Build 57(March 24, 2004) and bov<strong>in</strong>e mRNAs deposited <strong>in</strong>the GenBank database. Specifically, the microarrayconta<strong>in</strong>s 24 027 (Bos taurus) probe sets that representmore than 23 000 (Bos taurus) transcripts, <strong>in</strong>clud<strong>in</strong>gassemblies from approximately 19 000 UniGene Clusters.The bov<strong>in</strong>e GeneChip Ò also has probe features formultiple hybridization controls, poly-A controls andhousekeep<strong>in</strong>g (control or reference) genes. GeneChip Òmicroarrays consist of 25-mer oligonucleotides (probes)that are synthesized at specific locations on a coatedquartz surface. The 25-mer probe length confers highspecificity, while a number of probes are used to spaneach gene conferr<strong>in</strong>g high sensitivity and reproducibility.For each probe on the microarray that perfectlymatches its target sequence (perfect match or PM),there is also a paired ‘mismatch’ probe (MM). The MMprobe conta<strong>in</strong>s a s<strong>in</strong>gle mismatch located directly <strong>in</strong> themiddle of the 25-base probe sequence. Informationfrom the PM and MM probe sets may be summarizedus<strong>in</strong>g either the MAS5 Statistical algorithm, or alternativelyRobust Multichip Analysis (RMA) can beperformed which ignores MM values with<strong>in</strong> theAffymetrix Ò Expression ConsoleÔ Software package.A recent review describes the GeneChip Ò technologyplatform <strong>in</strong>clud<strong>in</strong>g its <strong>in</strong>dividual components andapplications <strong>in</strong> more detail (Dalma-Weiszhausz et al.2006). The preparation of RNA samples for geneexpression profil<strong>in</strong>g on Affymetrix GeneChips Ò requiresa number of sequential steps that results <strong>in</strong> thesynthesis of a biot<strong>in</strong>-labelled cRNA sample. AftercRNA fragmentation, the sample is ready to behybridized to the Affymetrix GeneChip Ò . Follow<strong>in</strong>ghybridization, probe arrays are washed, sta<strong>in</strong>ed andscanned.Bio<strong>in</strong>formaticsCollated raw microarray expression data must besubjected to a computational pipel<strong>in</strong>e compris<strong>in</strong>g <strong>in</strong>itiallyof quality control validation and filter<strong>in</strong>g. Anappropriate normalization methodology, for the experimentand data set <strong>in</strong> question, is then applied to removesystemic bias. These biases can be <strong>in</strong>troduced byunequal quantities of start<strong>in</strong>g RNA, differences <strong>in</strong>labell<strong>in</strong>g or detection efficiencies between the fluorescentdyes used, and systematic biases <strong>in</strong> the measuredexpression levels (Quackenbush 2002). Downstreamstatistical analysis can subsequently be performed togenerate mean<strong>in</strong>gful lists of differentially expressedgenes. Free open source software packages are available,<strong>in</strong>clud<strong>in</strong>g, e.g. TM4 (http://www.tm4.org/); which <strong>in</strong>cludesa collection of software solutions (MicroarrayData Manager (MADAM), TIGR_Spotf<strong>in</strong>der, MicroarrayData Analysis System (MIDAS), and MultiexperimentViewer (MeV)), to capture, manage and analyzedata from DNA microarray experiments. Alternatively,the Bioconductor software project (http://www.bioconductor.org/;based on the R programm<strong>in</strong>g language) hasa range of powerful statistical and graphical methods forthe analysis of genomic data (see also De Bruyne et al.2007; Grant et al. 2007) for further <strong>in</strong>formation on theanalysis and management of microarray expressiondata).Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Use of Microarray Technology for <strong>Reproduction</strong> <strong>in</strong> Cattle 361Global Gene Expression Dur<strong>in</strong>g OvarianFollicle DevelopmentFolliculogenesis starts dur<strong>in</strong>g foetal life when millions ofprimordial germ cells surrounded by pregranulosa cellsdevelop and form a life-time supply of follicles. At as yetpoorly understood <strong>in</strong>tervals, these follicles develop fromthe rest<strong>in</strong>g pool to grow and mature. Dur<strong>in</strong>g thisdevelopment, most follicles undergo atresia and die, witha small proportion develop<strong>in</strong>g to the later stages beyond5 mm <strong>in</strong> diameter and few develop<strong>in</strong>g to ovulatory sizeand fewer still go<strong>in</strong>g on to ovulate. These later stages offollicle development are the most susceptible to manipulationand have been the focus of many studies over theyears (reviewed <strong>in</strong> Mihm and Evans 2008).It is clear that antral follicle growth occurs <strong>in</strong>successive waves of development <strong>in</strong> cattle that are eachcharacterized by the growth of a cohort of follicles thatdevelop <strong>in</strong> parallel until the growth of one folliclerapidly <strong>in</strong>creases and becomes dom<strong>in</strong>ant (this follicle isselected to become dom<strong>in</strong>ant) with the other folliclescease grow<strong>in</strong>g, beg<strong>in</strong> to regress and undergo atresia(subord<strong>in</strong>ate follicles). The hormonal changes andassociated regulation of growth, selection and atresiaof follicles has been well studied. After selection, theparameters that differentiate the dom<strong>in</strong>ant follicle fromsubord<strong>in</strong>ate follicles are larger diameter, enhancedoestradiol production, more LH receptors <strong>in</strong> granulosacells, and an <strong>in</strong>crease <strong>in</strong> IGF b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> (IGFBP)protease activity <strong>in</strong> follicular fluid, low concentrations ofIGFBPs and <strong>in</strong>creased concentrations of free IGF <strong>in</strong>follicular fluid (Fortune et al. 2004; Webb and Campbell2007; Mihm and Evans 2008). As with other areas ofbiomedical science, the importance of the expression ofgenes <strong>in</strong> controll<strong>in</strong>g follicle development <strong>in</strong> cattle hasbeen studied one gene at a time (the candidate geneapproach) and has been highly reveal<strong>in</strong>g [reviewed by(Mihm and Bleach 2003)]. Alternatively, study<strong>in</strong>g theexpression of many genes simultaneously has beenachieved us<strong>in</strong>g suppressive subtraction hybridization(Sisco et al. 2003), serial analysis of gene expression(SAGE) (Mihm et al. 2008) or microarrays (see next).Microarrays have been used to study the progressionof ovarian follicle development and have shown changes<strong>in</strong> gene expression associated with a decrease <strong>in</strong> FSHdependence and an <strong>in</strong>crease <strong>in</strong> LH dependence asdom<strong>in</strong>ant follicles cont<strong>in</strong>ue to develop after selection(Mihm et al. 2006). Cont<strong>in</strong>ued development of thedom<strong>in</strong>ant follicle was associated with decreased mRNAexpression for genes known to be <strong>in</strong>duced by FSH <strong>in</strong>granulosa cells (FSHR, ESR2, INHA, ACVR1 andCCND2), <strong>in</strong>creased mRNA expression for the LHreceptor (LHCGR) <strong>in</strong> granulosa cells and alterations<strong>in</strong> mRNA expression of numerous genes potentially<strong>in</strong>volved <strong>in</strong> survival and apoptosis of granulosa andtheca cells (SIVA, FADD, TIAF1, LASS4 andTNFSF8) (Mihm et al. 2006). In another study, microarrayswere used to characterize the global pattern ofgene expression <strong>in</strong> aberrant persistent dom<strong>in</strong>ant follicles(low fertility) and concluded that alteration of theexpression of genes <strong>in</strong>volved <strong>in</strong> energy and prote<strong>in</strong>metabolism (e.g. phosphofructok<strong>in</strong>ase and fructose-1,6-bisphosphatase), am<strong>in</strong>o acid transport (e.g. am<strong>in</strong>o acidtransporter A2) and apoptosis (e.g. caspase-3, Bcl-2 andBcl-x) <strong>in</strong> granulosa cells may negatively impact on theirability to support a healthy oocyte and thereby contributeto decreased fertility (L<strong>in</strong>genfelter et al. 2008).In a number of experiments, we have used bov<strong>in</strong>ecDNA microarrays to ask what genes <strong>in</strong> granulosa andtheca cells contribute to selection of the dom<strong>in</strong>ant fromsubord<strong>in</strong>ate follicles. We identified 261 genes ⁄ ESTs thatwere differentially expressed between dom<strong>in</strong>ant andsubord<strong>in</strong>ate follicles and focused our attentions offamilies (ontologies) of genes. First, we exam<strong>in</strong>ed genes<strong>in</strong>volved <strong>in</strong> apoptosis and showed that dom<strong>in</strong>ant folliclesurvival was associated with genes <strong>in</strong>volved <strong>in</strong> the<strong>in</strong>hibition of apoptosis (aromatase, LH receptor, oestradiolreceptor b, DICE-1 tumour suppressor prote<strong>in</strong> andMCL-1) and the regression of subord<strong>in</strong>ate follicles wasassociated with enhanced expression of genes <strong>in</strong>volved <strong>in</strong>the apoptotic pathways (Betaglycan, COX1, TNFalpha,caspase-activated DNase, DRAK-2, caspase 13,P58(IPK), Apaf-1, BTG-3 and TS-BCLL) (Evans et al.2004). We also identified 83 genes <strong>in</strong>volved <strong>in</strong> signaltransduction (largely hormone receptors and <strong>in</strong>tracellularsignall<strong>in</strong>g molecules) that were differentially expressedbetween dom<strong>in</strong>ant and subord<strong>in</strong>ate folliclesand after a number of additional experiments hypothesizedimportant roles for CAMk<strong>in</strong>ase1 and EphA4 <strong>in</strong>theca cells and BCAR1 <strong>in</strong> granulosa cells for thedevelopment of dom<strong>in</strong>ant follicles and for betaglycanand FIBP <strong>in</strong> granulosa cells of regress<strong>in</strong>g subord<strong>in</strong>atefollicles (Forde et al. 2008a). The fate of cells is regulated<strong>in</strong> part by the transcription of new genes and we alsoexam<strong>in</strong>ed the relative expression of genes cod<strong>in</strong>g fortranscription factors <strong>in</strong> follicles dur<strong>in</strong>g the development.We identified 34 genes that code for transcription factors<strong>in</strong> granulosa and theca cells with good evidence tosuggest that genes for CEBP-b, SRF, FKHRL1,NCOR1 and Midnol<strong>in</strong> may be important <strong>in</strong> determ<strong>in</strong><strong>in</strong>gthe fate of follicle cells (Zielak et al. 2007a). In summary,these studies used a microarray approach to identify alarge number of genes which <strong>in</strong>dicates that ovarianfollicle development <strong>in</strong>volves the concerted actions ofgroups of molecules with diverse functions. However, themicroarray approach also lead to the discovery of genesthat have not previously been thought to be <strong>in</strong>volved <strong>in</strong>follicle development, for example, the prion prote<strong>in</strong>(Forde et al. 2007) and completely novel (some as yetunidentified) genes (Zielak et al. 2007b).Global Gene Expression Dur<strong>in</strong>g OocyteMaturationPrior to embryonic genome activation, embryo development<strong>in</strong> cattle is driven by maternal mRNAs andprote<strong>in</strong>s produced dur<strong>in</strong>g the oocyte growth phase.Intr<strong>in</strong>sic oocyte developmental competence, as assessedby development to the blastocyst stage has beenpositively associated with a number of factors, <strong>in</strong>clud<strong>in</strong>gfollicular environment (Lonergan et al. 2004) and thesite of oocyte maturation (Rizos et al. 2002). Numerousstudies have been carried out to <strong>in</strong>vestigate the affect ofthese factors on the oocyte transcriptome us<strong>in</strong>g a widerange of gene expression analysis techniques such asÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


362 ACO Evans, N Forde, GM O’Gorman, AE Zielak, P Lonergan and T Faircandidate gene PCR, global gene expression microarrayanalysis and cDNA subtraction.The first global analysis of the bov<strong>in</strong>e oocyte transcriptomepre- and post-meiotic maturation wasreported <strong>in</strong> 2003 (Dalbies-Tran and Mermillod 2003).Us<strong>in</strong>g the Atlas Human 1.2 cDNA expression array(Clontech Laboratories, Mounta<strong>in</strong> View, CA, USA),the authors identified approximately 300 genes withknown identities that were expressed <strong>in</strong> the bov<strong>in</strong>eoocyte, of which 70 were differentially expressed dur<strong>in</strong>gmeiotic maturation, and these genes were associatedwith cell cycle regulation, DNA transcription andapoptosis regulation. More recently, us<strong>in</strong>g the samecross-species hybridization approach, Adjaye et al.(2007) compared mRNA transcript profiles of bov<strong>in</strong>eoocytes and blastocysts us<strong>in</strong>g a human cDNA arrayconsist<strong>in</strong>g of over 15 000 probe sets. A total of 164 geneswere exclusively expressed <strong>in</strong> the oocyte. Among thegenes that were either exclusive to, or enriched <strong>in</strong>oocytes, the authors identified 31 genes that wereoocyte-conserved across mouse and human oocyte datasets. The genes <strong>in</strong>cluded germ cell-specific transcriptssuch as DAZL and NASP and a panel of genesimplicated <strong>in</strong> cell proliferation such as FER, KIT andMAPRE1 (Adjaye et al. 2007). S<strong>in</strong>ce then with access tothe Affymetrix Bov<strong>in</strong>e GeneChip, we recently carriedout a global mRNA analysis of bov<strong>in</strong>e immature and<strong>in</strong> vitro matured oocytes (Carter et al. 2007; Fair et al.2007). We have identified at least 8000 genes that areexpressed <strong>in</strong> bov<strong>in</strong>e oocytes, over 800 of which aredifferentially expressed (‡2-fold) dur<strong>in</strong>g meiotic maturation(Fair et al. 2007). The result<strong>in</strong>g differentiallyexpressed and present ⁄ absent gene lists were classifiedaccord<strong>in</strong>g to their gene ontology (GO); cellular location,biological process and molecular function. In general,the same GO categories were represented <strong>in</strong> the twooocytestages. However, 25% of the genes were upregulatedand 75% were downregulated <strong>in</strong> the <strong>in</strong> vitromatured oocytes when compared with their immaturecounterparts. Similarly, Misirlioglu et al. (2006) alsoused the Affymetrix Bov<strong>in</strong>e GeneChip to compare geneexpression of bov<strong>in</strong>e matured oocytes (MII) and eightcell-stage embryos. Genes controll<strong>in</strong>g DNA methylationand metabolism were upregulated <strong>in</strong> MII oocytes(Misirlioglu et al. 2006). When the two MII oocyte genelists were analyzed, they not only appeared to confirmeach other but also the f<strong>in</strong>d<strong>in</strong>gs of Dalbies-Tran andMermillod (2003), as a number of common genes thatwere primarily <strong>in</strong>volved <strong>in</strong> metabolism, transport andcell death regulation were identified.To date many models of bov<strong>in</strong>e oocyte developmentalcompetence have been described (Lonergan et al. 2003;Fair et al. 2004; Sirard et al. 2006). Recently, a broadrang<strong>in</strong>g study employed a comb<strong>in</strong>ation of SSH, cDNAmicroarray analysis and quantitative PCR to <strong>in</strong>terrogatemRNA transcript profiles <strong>in</strong> oocytes from several suchmodels, <strong>in</strong>clud<strong>in</strong>g; oocytes cultured with or withoutrFSH, early- vs late-cleav<strong>in</strong>g embryos, and oocytes fromdifferent follicle sizes. The gene candidates CCNB2,PTTG1, H2A, CKS1, PSMB2, SKIIP, CDC5L, RGS16and PRDX1, represent<strong>in</strong>g cell cycle, cell metabolism,cell signall<strong>in</strong>g and gene expression functional genecategories were identified as significantly associated withdevelopmental competence (Mourot et al. 2006). Focus<strong>in</strong>gjust on follicle status, Ghanem et al. (2007) comparedtranscript abundance of bov<strong>in</strong>e oocytes retrievedfrom small grow<strong>in</strong>g follicles vs dom<strong>in</strong>ant folliclesrecovered dur<strong>in</strong>g the first follicular wave us<strong>in</strong>g theBlueChip cDNA microarray (Sirard et al. 2006; Ghanemet al. 2007). Fifty-one differentially regulated geneswere identified, of which genes regulat<strong>in</strong>g prote<strong>in</strong>biosynthesis (RPLP0, RPL8, RPL24, ARL6IP, RpS14,RpS15, RpS4x and RPS3A) were enriched <strong>in</strong> the morecompetent oocytes, i.e. oocytes recovered from grow<strong>in</strong>gfollicles, other over expressed genes fell <strong>in</strong>to a number offunctional classifications, <strong>in</strong>clud<strong>in</strong>g the follow<strong>in</strong>g: translationelongation, ATP b<strong>in</strong>d<strong>in</strong>g, NADH dehydrogenaseactivity, cytoskeleton, calcium ion b<strong>in</strong>d<strong>in</strong>g. In addition,us<strong>in</strong>g the BlueChip cDNA microarray, a recent reportdescribes a novel non-<strong>in</strong>vasive approach to identify<strong>in</strong>gdevelopmentally competent and <strong>in</strong>competent bov<strong>in</strong>eoocytes and zygotes (Dessie et al. 2007). In this study,bov<strong>in</strong>e oocytes and zygotes were dielectrophoreticallyseparated accord<strong>in</strong>g to their speed of migration to theopposite electrode. The authors reported that themajority of differentially regulated genes were moreabundant <strong>in</strong> the very fast mov<strong>in</strong>g oocytes and zygotes,and fell ma<strong>in</strong>ly under functional categories associatedwith regulation of prote<strong>in</strong> biosynthesis and metabolicregulation. Among these transcripts, RPL2, RPL8,RPL35 and RPLP0 were more abundant <strong>in</strong> the veryfast mov<strong>in</strong>g developmentally superior oocytes, confirm<strong>in</strong>gthe f<strong>in</strong>d<strong>in</strong>gs of Mourot et al. (2006) described earlier.F<strong>in</strong>ally, compar<strong>in</strong>g RNA isolated from germ<strong>in</strong>al vesiclestage oocytes collected from adult vs pre-pubertalanimals, as model of high and low oocyte competence,on a bov<strong>in</strong>e cDNA array represent<strong>in</strong>g over 15 000unique genes (Suchyta et al. 2003), Patel et al. (2007)described significant over-representation of transcriptsencod<strong>in</strong>g for genes <strong>in</strong> hormone secretion classificationwith<strong>in</strong> adult oocytes. Further analysis revealed follistat<strong>in</strong>as a maternal mRNA marker of high developmentalcompetence <strong>in</strong> two models (Patel et al. 2007).Global Gene Expression Dur<strong>in</strong>g EmbryoDevelopmentMammalian pre-implantation embryos are very sensitiveto the environment <strong>in</strong> which they develop. In vitro,the period of post-fertilization embryo culture is themost critical period affect<strong>in</strong>g blastocyst quality assessed<strong>in</strong> terms of cryotolerance, gene expression pattern andability to establish a pregnancy (Corcoran et al. 2006;Lonergan et al. 2006). Precise control of gene expressiondur<strong>in</strong>g this phase of development is particularly importantas several critical developmental events occurdur<strong>in</strong>g this period <strong>in</strong>clud<strong>in</strong>g: (i) the first cleavagedivision, the tim<strong>in</strong>g of which has been associated withthe developmental competence <strong>in</strong> a variety of mammalianspecies, (ii) embryonic genome activation, when theembryo transfers from a reliance on maternal RNAderived from the oocyte to expression of its owngenome, (iii) morula compaction, which <strong>in</strong>volves theestablishment of the first <strong>in</strong>timate cell-to-cell contacts <strong>in</strong>the embryo and (iv) blastocyst formation, <strong>in</strong>volv<strong>in</strong>g thedifferentiation of two cell types, the trophectoderm andÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Use of Microarray Technology for <strong>Reproduction</strong> <strong>in</strong> Cattle 363the <strong>in</strong>ner cell mass and subsequent elongation. Analysisof expression patterns of developmentally importantgenes provides a useful tool to assess the normality of<strong>in</strong> vitro produced embryos alongside their <strong>in</strong> vitroderivedcounterparts and may provide a bluepr<strong>in</strong>taga<strong>in</strong>st which to manipulate culture conditions <strong>in</strong> orderto improve embryo quality.Gene expression studies <strong>in</strong> cattle embryos have beenconducted employ<strong>in</strong>g qualitative, semi-quantitative andquantitative approaches. Most studies have focused on asmall set of ‘candidate’ genes putatively important forembryogenesis <strong>in</strong>clud<strong>in</strong>g growth factors, metabolicenzymes, transcription factors <strong>in</strong>volved <strong>in</strong> the regulationof early developmental events and impr<strong>in</strong>ted geneproducts (see Wrenzycki et al. 2005 for review). A muchmore limited, but <strong>in</strong>creas<strong>in</strong>g, number of studies haveused cDNA microarrays to exam<strong>in</strong>e global gene expressionchanges <strong>in</strong> cattle embryos (Pfister-Genskow et al.2005; Smith et al. 2005; Corcoran et al. 2006; Somerset al. 2006; Adjaye et al. 2007).Ushizawa et al. (2004) used a custom-designed uteroplacentalcDNA microarray to study gene expression <strong>in</strong>bov<strong>in</strong>e embryos on days 7, 14 and 21, extra-embryonicmembranes on day 28 and foetuses on day 28 ofpregnancy. Comparison of day 7 and day 14 revealedmost genes <strong>in</strong>creased dur<strong>in</strong>g this period, and a smallnumber of genes exhibit<strong>in</strong>g altered expression decreasedas gestation progressed (Ushizawa et al. 2004). Theexpression of trophoblast cell-specific molecules such asplacental lactogens, prolact<strong>in</strong>-related prote<strong>in</strong>s, <strong>in</strong>terferon-tauand adhesion molecules was enhanced dur<strong>in</strong>gthe pre-implantation period suggest<strong>in</strong>g a pivotal role <strong>in</strong>the preparation needed for implantation.We recently compared mRNA expression across awide range of biological processes <strong>in</strong> bov<strong>in</strong>e blastocystsderived from either culture <strong>in</strong> vivo or <strong>in</strong> vitro us<strong>in</strong>gbov<strong>in</strong>e cDNA microarrays (BOTL). The vast majority(85%) of the differentially expressed transcripts betweenthe two embryo populations were downregulated <strong>in</strong><strong>in</strong> vitro cultured blastocysts, suggest<strong>in</strong>g that the primaryreason why <strong>in</strong> vitro embryos are of <strong>in</strong>ferior developmentalcompetence compared to <strong>in</strong> vivo cultured may bebecause of a deficiency of the mach<strong>in</strong>ery associated withtranscription and translation (Corcoran et al. 2006).Based on previous data show<strong>in</strong>g the feasibility andhigh reproducibility of cross species hybridizationapproach us<strong>in</strong>g a human cDNA array to study geneexpression <strong>in</strong> bov<strong>in</strong>e tissues (Adjaye et al. 2004), Adjayeet al. (2007) used a human 15 529 chip (the ENSEMBLchip) to compare gene expression <strong>in</strong> bov<strong>in</strong>e oocytes andblastocysts; 164 and 1324 genes were expressed exclusivelyat the oocyte or blastocyst stage, respectively,while 419 genes expressed <strong>in</strong> both stages. Few studieshave attempted to correlate the differences <strong>in</strong> mRNAabundance observed at the blastocyst stage, such asthose outl<strong>in</strong>ed earlier, with the ability of the embryo toestablish a pregnancy. One such recent study (El-Sayedet al. 2006) addressed the relationship between transcriptionalprofile of embryos and the pregnancy successbased on gene expression analysis of biopsies fromblastocysts taken prior to transfer to recipients. Microarraydata (BlueChip) analysis revealed a total of 52differentially regulated genes between embryos result<strong>in</strong>g<strong>in</strong> a calf delivery vs those not result<strong>in</strong>g <strong>in</strong> a pregnancy(El-Sayed et al. 2006). Biopsies from embryos that weretransferred to recipients and resulted <strong>in</strong> calf deliverywere enriched with genes necessary for implantation(COX2 and CDX2), carbohydrate metabolism(ALOX15), signal transduction (PLAU) and placentaldevelopment (PLAC8), while those fail<strong>in</strong>g to establish apregnancy were enriched with transcripts for an <strong>in</strong>flammatorycytok<strong>in</strong>e (TNF), prote<strong>in</strong> am<strong>in</strong>o acid b<strong>in</strong>d<strong>in</strong>g(EEF1A1), transcription factors (MSX1 and PTTG1),glucose metabolism (PGK1 and AKR1B1) and CD9which is an <strong>in</strong>hibitor of implantation (El-Sayed et al.2006).Bov<strong>in</strong>e embryos produced by nuclear transfer (NT)are capable of develop<strong>in</strong>g to the blastocyst stage with anefficiency of 30–50% which is comparable to thatachieved with rout<strong>in</strong>e IVF (Yang et al. 2007). However,follow<strong>in</strong>g somatic cell clon<strong>in</strong>g and transfer to surrogaterecipients a very limited percentage (0.5–5%) of theembryos completes full-term development. This isma<strong>in</strong>ly due to a high frequency of post-implantationdevelopmental arrest. Such losses are predom<strong>in</strong>antlydur<strong>in</strong>g the first trimester of pregnancy but can occurmuch later (Heyman et al. 2002) and are often associatedwith aberrant placental development (Hill et al.2000). The high <strong>in</strong>cidence of pregnancy loss andneonatal death follow<strong>in</strong>g somatic cell NT (SCNT) arehypothesized to result from <strong>in</strong>complete nuclear reprogramm<strong>in</strong>g;several authors have conv<strong>in</strong>c<strong>in</strong>gly demonstratedthat the donor somatic cell is reprogrammedsuch that expression pattern at the blastocyst stage issubstantially different from that of the somatic cell priorto NT (Smith et al. 2005; Beyhan et al. 2007). Therehave been several recent cDNA microarray studiesexam<strong>in</strong><strong>in</strong>g differential gene expression <strong>in</strong> NT-derivedembryos (Pfister-Genskow et al. 2005; Smith et al. 2005;Somers et al. 2006) <strong>in</strong> an effort to expla<strong>in</strong> the poorgestational success.In one of the first reports compar<strong>in</strong>g <strong>in</strong>dividualembryos produced by NT and IVF us<strong>in</strong>g cDNAmicroarray technology for any species, Pfister-Genskowet al. (2005) found 18 genes differentially expressedbetween NT- and IVF-produced embryos <strong>in</strong>clud<strong>in</strong>gthree <strong>in</strong>termediate-filament prote<strong>in</strong> genes (cytokerat<strong>in</strong>8, cytokerat<strong>in</strong> 19 and viment<strong>in</strong>), three metabolic genes(phosphoribosyl pyrophosphate synthetase 1, mitochondrialacetoacetyl-coenzyme A thiolase and alpha-glucosidase),two lysosomal-related genes (prosapos<strong>in</strong> andlysosomal-associated membrane prote<strong>in</strong> 2), and a geneassociated with stress responses (heat shock prote<strong>in</strong> 27)along with major histocompatibility complex class I,nidogen 2, a putative transport prote<strong>in</strong>, heterogeneousnuclear ribonuclear prote<strong>in</strong> K, mitochondrial 16S rRNA,and ES1 (a zebrafish orthologue of unknown function).Smith et al. (2005) used microarray technology toanalyze global differences <strong>in</strong> gene expression betweenblastocysts derived from (i) natural fertilization <strong>in</strong> vivo,(ii) <strong>in</strong> vitro fertilization or (iii) NT. Perhaps surpris<strong>in</strong>gly,the NT embryos most closely resembled true <strong>in</strong> vivoembryos <strong>in</strong> terms of numbers of differentially expressedgenes. They reported a low variability <strong>in</strong> the pattern ofmRNA expression among <strong>in</strong>dividual <strong>in</strong> vivo-derivedblastocysts and among NT blastocysts when comparedÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


364 ACO Evans, N Forde, GM O’Gorman, AE Zielak, P Lonergan and T Fairwith <strong>in</strong> vitro produced blastocysts suggest<strong>in</strong>g a relativelyuniform reprogramm<strong>in</strong>g mechanism <strong>in</strong> the NT embryosby the blastocyst stage. Despite this apparent similarity<strong>in</strong> gene expression pattern between NT- and AI-derivedblastocysts, the low pregnancy rate and developmentalanomalies associated with NT suggest that reprogramm<strong>in</strong>gof the donor genome is <strong>in</strong>complete. To <strong>in</strong>vestigatethe possible causes of these losses, transcript profil<strong>in</strong>g oftrophoblast and embryonic disc tissue collected fromday 25 conceptuses derived by NT or AI was conductedus<strong>in</strong>g a microarray consist<strong>in</strong>g of 13 257 oligonucleotides-derivedfrom cattle gene sequences. Approximately9000 genes were differentially expressed between trophoblastand disc demonstrat<strong>in</strong>g major gene expressiondifferences <strong>in</strong> embryonic and extra-embryonic tissues.Compar<strong>in</strong>g NT with AI, 188 genes were differentiallyexpressed <strong>in</strong> trophoblast and 10 <strong>in</strong> the embryonic disc,provid<strong>in</strong>g evidence that nuclear reprogramm<strong>in</strong>g isdefective <strong>in</strong> a large proportion of NT-derived clonesand that aberrant gene expression <strong>in</strong> the trophoblastcontributes to pregnancy failure at day 25 and beyond(Everts et al. 2005).Us<strong>in</strong>g Affymetrix GeneChip bov<strong>in</strong>e arrays, Beyhanet al. (2007) compared global gene expression of twodifferent somatic cell l<strong>in</strong>es whose capacity to generatehealth NT-derived calves is significantly different, thecloned blastocysts derived from the cell l<strong>in</strong>es and IVFblastocysts <strong>in</strong> order to elucidate some of the key genesresponsible for successful reprogramm<strong>in</strong>g. The majorityof genes <strong>in</strong> SCNT blastocysts had comparable expressionpatterns to IVF blastocysts, with the exception of asmall number of genes whose relative expression valueswere similar to their correspond<strong>in</strong>g donor cells, <strong>in</strong>dicat<strong>in</strong>ga failure of reprogramm<strong>in</strong>g <strong>in</strong> SCNT blastocysts(Beyhan et al. 2007).Global Gene Expression <strong>in</strong> the Uter<strong>in</strong>eEndometriumThe study of endometrial function is necessary tounderstand the factors associated with the establishmentof pregnancy and to get an <strong>in</strong>sight <strong>in</strong>to the causes ofearly embryo loss. The endometrium is responsible forthe secretion of the numerous cytok<strong>in</strong>es, growth factorsand prote<strong>in</strong>s that together make are collectively termedthe histotroph. The histotroph is secreted from theglandular epithelium <strong>in</strong> the endometrium <strong>in</strong>to the lumenof the uterus and it is <strong>in</strong> this environment that theembryo develops. Studies <strong>in</strong>volv<strong>in</strong>g endometrial geneexpression <strong>in</strong> cattle have ma<strong>in</strong>ly focused on the changesthat occur at different stages of the oestrous cycle. Somestudies have exam<strong>in</strong>ed the mRNA expression profiles ofbov<strong>in</strong>e epithelial cells of the ipsilateral vs the contralateraloviduct (Bauersachs et al. 2003) and cells from theipsilateral oviduct (Bauersachs et al. 2004) or endometrium(Bauersachs et al. 2005) at oestrus (low progesterone)and dioestrus (high progesterone), to identifychanges <strong>in</strong> gene expression at different stages of theoestrous cycle <strong>in</strong> cattle.Microarray studies that give an <strong>in</strong>sight <strong>in</strong>to thechanges that occur <strong>in</strong> gene expression <strong>in</strong> the endometrium,which have a functional consequence for embryosurvival, have been performed <strong>in</strong> humans (Horcajadaset al. 2006), mice (Kashiwagi et al. 2007), sheep (Chenet al. 2007) and pigs (Ross et al. 2007). Although there isscant literature on the use of microarrays to studyendometrial gene expression <strong>in</strong> cattle, two papers(Bauersachs et al. 2006; Kle<strong>in</strong> et al. 2006) have takentwo different animal model approaches to address thedifferences <strong>in</strong> endometrial gene expression betweenpregnant and cycl<strong>in</strong>g animals on day 18. In one study,monozygotic tw<strong>in</strong> recipients were used along with<strong>in</strong> vitro-derived embryos and suppression subtractivehybridization to enrich the cDNA for transcripts thatwere upregulated <strong>in</strong> the endometrium of pregnantanimals before microarray hybridization (Kle<strong>in</strong> et al.2006). They identified 87 different genes ⁄ mRNAs, ofwhich 80 were known bov<strong>in</strong>e genes or were <strong>in</strong>ferredfrom human orthologues. Ontological classification ofthese genes showed that the largest class was type I<strong>in</strong>terferon-stimulated genes. Of these differentially expressedgenes, several have already been described asbe<strong>in</strong>g <strong>in</strong>volved <strong>in</strong> changes <strong>in</strong> endometrial gene expression<strong>in</strong> other species. For example, <strong>in</strong>terferon-stimulatedgene-15 has shown to be expressed <strong>in</strong> the endometriumof cows (Aust<strong>in</strong> et al. 2004), sheep (Johnson et al. 2000),pigs (Joyce et al. 2003), mice (Aust<strong>in</strong> et al. 2003) andhumans and baboons (Beb<strong>in</strong>gton et al. 1999). However,the power of the microarray technology allowed for theidentification of genes that are <strong>in</strong>volved <strong>in</strong> cell adhesion(connective tissue growth factor – CTGF, glycosylphosphatidyl<strong>in</strong>ositolspecific phospholipase D1 – GPLD1,milk fat globule-EGF factor 8 prote<strong>in</strong> – MFGE8) aswell as genes <strong>in</strong>volved <strong>in</strong> endometrial remodell<strong>in</strong>g(matrix metalloprote<strong>in</strong>ase 19 – MMP19, tissue <strong>in</strong>hibitorof metalloprote<strong>in</strong>ase 2 – TIMP2) that have not previouslybeen described as be<strong>in</strong>g differentially regulated <strong>in</strong>the endometrium (Kle<strong>in</strong> et al. 2006). The second modelutilized SSH technology also, but used <strong>in</strong> vivo-derivedembryos (Bauersachs et al. 2006). This study identified109 transcripts that were differentially regulated <strong>in</strong> day18 pregnant animals when compared with day 18 cycl<strong>in</strong>ganimals. Of these 109 transcripts, 34, 28 and 3 geneswere enriched <strong>in</strong> the GO terms for immune responsegenes, response to stimulus and antigen presentation,respectively.The earlier studies looked at differences <strong>in</strong> geneexpression <strong>in</strong> the endometrium associated with pregnancyon a day when maternal recognition had alreadyoccurred. However, <strong>in</strong> cattle, the majority of embryonicloss occurs prior to maternal recognition of pregnancy(Dunne et al. 2000; Humblot 2001). Retrospectivestudies have shown a correlation between concentrationsof progesterone and pregnancy rates both <strong>in</strong> beefand dairy cattle (Disk<strong>in</strong> et al. 2006). Other studies <strong>in</strong>cattle have shown that progesterone supplementation<strong>in</strong>creases embryo size on day 14 of pregnancy (Garrettet al. 1988). Recent studies <strong>in</strong> our laboratory (Carteret al. 2008), Forde et al. (2008b) have tried to addressthe issue of early embryo mortality by exam<strong>in</strong><strong>in</strong>g thedifferential effects of elevated progesterone <strong>in</strong> theimmediate post-conception period and pregnancy onendometrial gene expression <strong>in</strong> cycl<strong>in</strong>g and pregnantanimals at different physiological stages of the earlydevelopmental axis (i.e. days 5, 7, 13 and 16 postconception,correspond<strong>in</strong>g to the 16-cell ⁄ early morulaÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Use of Microarray Technology for <strong>Reproduction</strong> <strong>in</strong> Cattle 365stage, the blastocyst stage, the beg<strong>in</strong>n<strong>in</strong>g of elongationand the time of maternal recognition of pregnancy,respectively). In our model, no genes were differentiallyexpressed <strong>in</strong> pregnant compared with cycl<strong>in</strong>g animalsbefore day 16. Of the genes that were differentiallyexpressed between pregnant and cycl<strong>in</strong>g animals (on day16), GO analyses reveals that genes <strong>in</strong>volved <strong>in</strong> immuneresponse were significantly over-represented <strong>in</strong> pregnantvs cycl<strong>in</strong>g animals. Comparisons between our lists ofdifferentially expressed genes on day 16 of pregnancyand those from (Bauersachs et al. 2006) (day 18) showthat 32 genes are differentially regulated on both days <strong>in</strong>both the models. This suggests that these genes are<strong>in</strong>volved <strong>in</strong> very early maternal recognition of pregnancyand may be critical for the establishment of pregnancy.In the same study, we identified large numbers of genesthat were differentially expressed <strong>in</strong> the endometria ofanimals with high vs low concentrations of progesteroneon days 5, 7 and 13. GO analysis of differentiallyexpressed genes on day 5 <strong>in</strong>dicate that genes <strong>in</strong>volved <strong>in</strong>the positive regulation of transcription are progesteroneregulated and on day 7 there is a shift towards genes<strong>in</strong>volved <strong>in</strong> cell division. This <strong>in</strong>dicates a potentiallycritical role of progesterone <strong>in</strong> regulat<strong>in</strong>g endometrialfunction early <strong>in</strong> pregnancy (days 5–13) when theembryo is develop<strong>in</strong>g prior to its signall<strong>in</strong>g for thematernal recognition of pregnancy. Further work needsto be done to ascerta<strong>in</strong> a functional role for these newlydescribed genes <strong>in</strong>volved <strong>in</strong> differences <strong>in</strong> endometrialgene expression associated with pregnancy.ConclusionsKnowledge of the global pattern of gene expression isimportant for understand<strong>in</strong>g critical regulatory pathwaysthat are necessary for successful tissue developmentand <strong>in</strong> recent years there has been a big <strong>in</strong>crease <strong>in</strong>the number of publications report<strong>in</strong>g the use of microarraysto study reproduction <strong>in</strong> cattle. These publicationsconclude that genes <strong>in</strong>volved with cellsurvival ⁄ death, <strong>in</strong>tracellular signall<strong>in</strong>g, transcriptionand translation, cell division and proliferation andcellular metabolism are the ma<strong>in</strong> cellular processes thatcontrol the development of reproductive tissues. In thiscontext, microarrays have proved themselves to be apowerful tool. However, a note of caution rem<strong>in</strong>ds usthat our understand<strong>in</strong>g of these cellular events will likelyimprove further <strong>in</strong> the future with the development ofbetter tools to analyse these data (bio<strong>in</strong>formatics). Inaddition, <strong>in</strong>formation provided us<strong>in</strong>g microarrays islimited by the number and nature of the spots on eacharray and new sequenc<strong>in</strong>g technologies now have thepromise to overcome this problem and have thepotential to replace microarrays as the method of choicefor global gene expression studies (Fields 2007).AcknowledgementsThis research, as well as similar ongo<strong>in</strong>g studies, is funded by ScienceFoundation Ireland (grant numbers: 02 ⁄ IN1 ⁄ B78, PICA award,06 ⁄ INI ⁄ B62, 07 ⁄ SRC ⁄ B1156). The op<strong>in</strong>ions, f<strong>in</strong>d<strong>in</strong>gs and conclusionsor recommendations expressed <strong>in</strong> this material are those of the authorsand do not necessarily reflect the views of the Science FoundationIreland.ReferencesAdjaye J, Herwig R, Herrmann D, Wruck W, Benkahla A,Br<strong>in</strong>k TC, Nowak M, Carnwath JW, Hultschig C, NiemannH, Lehrach H, 2004: Cross-species hybridisation of humanand bov<strong>in</strong>e orthologous genes on high density cDNAmicroarrays. BMC Genomics 5, 83.Adjaye J, Herwig R, Br<strong>in</strong>k TC, Herrmann D, Greber B,Sudheer S, Groth D, Carnwath JW, Lehrach H, NiemannH, 2007: Conserved molecular portraits of bov<strong>in</strong>e andhuman blastocysts as a consequence of the transition frommaternal to embryonic control of gene expression. PhysiolGenomics 31, 315–327.Aust<strong>in</strong> KJ, Bany BM, Belden EL, Rempel LA, Cross JC,Hansen TR, 2003: Interferon-stimulated gene-15 (Isg15)expression is up-regulated <strong>in</strong> the mouse uterus <strong>in</strong> response tothe implant<strong>in</strong>g conceptus. Endocr<strong>in</strong>ology 144, 3107–3113.Aust<strong>in</strong> KJ, Carr AL, Pru JK, Hearne CE, George EL, BeldenEL, Hansen TR, 2004: Localization of ISG15 and conjugatedprote<strong>in</strong>s <strong>in</strong> bov<strong>in</strong>e endometrium us<strong>in</strong>g immunohistochemistryand electron microscopy. Endocr<strong>in</strong>ology 145, 967–975.Bauersachs S, Blum H, Mallok S, Wenigerk<strong>in</strong>d H, Rief S, Prelle K,Wolf E, 2003: Regulation of ipsilateral and contralateral bov<strong>in</strong>eoviduct epithelial cell function <strong>in</strong> the postovulation period:a transcriptomics approach. Biol Reprod 68, 1170–1177.Bauersachs S, Rehfeld S, Ulbrich SE, Mallok S, Prelle K,Wenigerk<strong>in</strong>d H, E<strong>in</strong>spanier R, Blum H, Wolf E, 2004:Monitor<strong>in</strong>g gene expression changes <strong>in</strong> bov<strong>in</strong>e oviductepithelial cells dur<strong>in</strong>g the oestrous cycle. J Mol Endocr<strong>in</strong>ol32, 449–466.Bauersachs S, Ulbrich SE, Gross K, Schmidt SE, Meyer HH,E<strong>in</strong>spanier R, Wenigerk<strong>in</strong>d H, Vermehren M, Blum H,S<strong>in</strong>owatz F, Wolf E, 2005: Gene expression profil<strong>in</strong>g ofbov<strong>in</strong>e endometrium dur<strong>in</strong>g the oestrous cycle: detection ofmolecular pathways <strong>in</strong>volved <strong>in</strong> functional changes. J MolEndocr<strong>in</strong>ol 34, 889–908.Bauersachs S, Ulbrich SE, Gross K, Schmidt SE, Meyer HH,Wenigerk<strong>in</strong>d H, Vermehren M, S<strong>in</strong>owatz F, Blum H, WolfE, 2006: Embryo-<strong>in</strong>duced transcriptome changes <strong>in</strong> bov<strong>in</strong>eendometrium reveal species-specific and common molecularmarkers of uter<strong>in</strong>e receptivity. <strong>Reproduction</strong> 132, 319–331.Beb<strong>in</strong>gton C, Bell SC, Doherty FJ, Fazleabas AT, Flem<strong>in</strong>gSD, 1999: Localization of ubiquit<strong>in</strong> and ubiquit<strong>in</strong> crossreactiveprote<strong>in</strong> <strong>in</strong> human and baboon endometrium anddecidua dur<strong>in</strong>g the menstrual cycle and early pregnancy.Biol Reprod 60, 920–928.Beyhan Z, Ross PJ, Iager AE, Kocabas AM, Cunniff K, RosaGJ, Cibelli JB, 2007: Transcriptional reprogramm<strong>in</strong>g ofsomatic cell nuclei dur<strong>in</strong>g preimplantation development ofcloned bov<strong>in</strong>e embryos. Dev Biol 305, 637–649.Bonnet A, Dalbies-Tran R, Sirard MA, 2008: Opportunitiesand challenges <strong>in</strong> apply<strong>in</strong>g genomics to the study ofoogenesis and folliculogenesis <strong>in</strong> farm animals. <strong>Reproduction</strong>135, 119–128.Bryant PA, Venter D, Rob<strong>in</strong>s-Browne R, Curtis N, 2004:Chips with everyth<strong>in</strong>g: DNA microarrays <strong>in</strong> <strong>in</strong>fectiousdiseases. Lancet Infect Dis 4, 100–111.Carter F, Fair T, Park S, Wade M, Evans ACO, Lonergan P,2007: Gene expression profil<strong>in</strong>g of immature and <strong>in</strong> vitromatured bov<strong>in</strong>e oocytes us<strong>in</strong>g Affymetrix Genechip technology.Reprod Fertil Dev 19, Abstract 263, 2480.Carter F, Forde N, Duffy P, Wade M, Fair T, Crowe MA,Evans ACO, Kenny DA, Roche JF, Lonergan P, 2008:Effect of <strong>in</strong>creas<strong>in</strong>g progesterone concentration from Day 3of pregnancy on subsequent embryo survival and development<strong>in</strong> beef heifers. Reprod Fertil Dev 20, 368–375.Chen Y, Antoniou E, Liu Z, Hearne LB, Roberts RM, 2007:A microarray analysis for genes regulated by <strong>in</strong>terferon-tau<strong>in</strong> ov<strong>in</strong>e lum<strong>in</strong>al epithelial cells. <strong>Reproduction</strong> 134, 123–135.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


366 ACO Evans, N Forde, GM O’Gorman, AE Zielak, P Lonergan and T FairCorcoran D, Fair T, Park S, Rizos D, Patel OV, Smith GW,Coussens PM, Ireland JJ, Boland MP, Evans ACO, LonerganP, 2006: Suppressed expression of genes <strong>in</strong>volved <strong>in</strong>transcription and translation <strong>in</strong> <strong>in</strong> vitro compared with <strong>in</strong>vivo cultured bov<strong>in</strong>e embryos. <strong>Reproduction</strong> 131, 651–660.Dalbies-Tran R, Mermillod P, 2003: Use of heterologouscomplementary DNA array screen<strong>in</strong>g to analyze bov<strong>in</strong>eoocyte transcriptome and its evolution dur<strong>in</strong>g <strong>in</strong> vitromaturation. Biol Reprod 68, 252–261.Dalma-Weiszhausz DD, Warr<strong>in</strong>gton J, Tanimoto EY, MiyadaCG, 2006: The affymetrix GeneChip platform: an overview.Methods Enzymol 410, 3–28.De Bruyne V, Al-Mulla F, Pot B, 2007: Methods formicroarray data analysis. Methods Mol Biol 382, 373–391.Dessie SW, R<strong>in</strong>gs F, Holker M, Gilles M, Jennen D, Tholen E,Havelicek V, Besenfelder U, Sukhorukov VL, ZimmermanU, Endter JM, Sirad MA, Schellander K, Tesfaye D, 2007:Dielectrophoretic behavior of <strong>in</strong> vitro-derived bov<strong>in</strong>e metaphaseII oocytes and zygotes and its relation to <strong>in</strong> vitroembryonic developmental competence and mRNA expressionpattern. <strong>Reproduction</strong> 133, 931–946.Disk<strong>in</strong> MG, Murphy JJ, Sreenan JM, 2006: Embryo survival<strong>in</strong> dairy cows managed under pastoral conditions. AnimReprod Sci 96, 297–311.Dunne LD, Disk<strong>in</strong> MG, Sreenan JM, 2000: Embryo and foetalloss <strong>in</strong> beef heifers between day 14 of gestation and full term.Anim Reprod Sci 58, 39–44.El-Sayed A, Hoelker M, R<strong>in</strong>gs F, Salilew D, Jennen D, TholenE, Sirard MA, Schellander K, Tesfaye D, 2006: Large-scaletranscriptional analysis of bov<strong>in</strong>e embryo biopsies <strong>in</strong> relationto pregnancy success after transfer to recipients. PhysiolGenomics 28, 84–96.Evans ACO, Ireland JLH, W<strong>in</strong>n ME, Lonergan P, Smith GW,Coussens PM, Ireland JJ, 2004: Identification of genes<strong>in</strong>volved <strong>in</strong> apoptosis and dom<strong>in</strong>ant follicle developmentdur<strong>in</strong>g follicular waves <strong>in</strong> cattle. Biol Reprod 70, 1475–1484.Everts RE, Band MR, Liu ZL, Kumar CG, Liu L, Loor JJ,Oliveira R, Lew<strong>in</strong> HA, 2005: A 7872 cDNA microarray andits use <strong>in</strong> bov<strong>in</strong>e functional genomics. Vet Immunol Immunopathol105, 235–245.Fair T, Murphy M, Rizos D, Moss C, Mart<strong>in</strong> F, Boland MP,Lonergan P, 2004: Analysis of differential maternal mRNAexpression <strong>in</strong> developmentally competent and <strong>in</strong>competentbov<strong>in</strong>e two-cell embryos. Mol Reprod Dev 67, 136–144.Fair T, Carter F, Park S, Evans ACO, Lonergan P, 2007:Global gene expression analysis dur<strong>in</strong>g bov<strong>in</strong>e oocyte <strong>in</strong>vitro maturation. Theriogenology 68(Suppl. 1), S91–S97.Fields S, 2007: Molecular biology. Site-see<strong>in</strong>g by sequenc<strong>in</strong>g.Science 316, 1441–1442.Forde N, Rogers M, Canty MJ, Lonergan P, Smith GW,Coussens PM, Ireland JJ, Evans ACO, 2007: Association ofthe prion prote<strong>in</strong> and its expression with ovarian follicledevelopment <strong>in</strong> cattle. Mol Reprod Dev 75, 248–249.Forde N, Mihm M, Canty M, Zielak A, Baker P, Park S,Lonergan P, Smith G, Coussens P, Ireland J, Evans A, 2008a:Differential expression of signal transduction factors <strong>in</strong>ovarian follicle development; a role for betaglycan andFIBP <strong>in</strong> granulosa cells <strong>in</strong> cattle. Physiol Genomics 33,193–204.Forde N, Carter F, Fair T, Crowe MA, Evans ACO, SpencerTE, Bazer FW, O’Gaora P, McBride R, Boland MP,Lonergan P, Roche JF, 2008b: Effect of pregnancy andprogesterone on gene expression <strong>in</strong> the uter<strong>in</strong>e endometriumof cattle. Annual Meet<strong>in</strong>g of the Society for the Study of<strong>Reproduction</strong>, Hawaii, USA (27 to 30 May 2008). Biologyof <strong>Reproduction</strong>, Special Issue, Abstract #37.Fortune JE, Rivera GM, Yang MY, 2004: Follicular development:the role of the follicular microenvironment <strong>in</strong> selectionof the dom<strong>in</strong>ant follicle. Anim Reprod Sci 82–83, 109–126.Garrett JE, Geisert RD, Zavy MT, Morgan GL, 1988:Evidence for maternal regulation of early conceptus growthand development <strong>in</strong> beef cattle. J Reprod Fertil 84, 437–446.Ghanem N, Holker M, R<strong>in</strong>gs F, Jennen D, Tholen E, SirardMA, Torner H, Kanitz W, Schellander K, Tesfaye D, 2007:Alterations <strong>in</strong> transcript abundance of bov<strong>in</strong>e oocytesrecovered at growth and dom<strong>in</strong>ance phases of the firstfollicular wave. BMC Dev Biol 7, 90.Grant GR, Manduchi E, Stoeckert CJ Jr, 2007: Analysis andmanagement of microarray gene expression data. CurrProtoc Mol Biol (Suppl. 77) Chapter 19.6, 1–30.Hager J, 2006: Mak<strong>in</strong>g and us<strong>in</strong>g spotted DNA microarrays <strong>in</strong>an academic core laboratory. Methods Enzymol 410, 135–168.Heyman Y, Chavatte-Palmer P, LeBourhis D, Camous S,Vignon X, Renard JP, 2002: Frequency and occurrence oflate-gestation losses from cattle cloned embryos. BiolReprod 66, 6–13.Hill JR, Burghardt RC, Jones K, Long CR, Looney CR, Sh<strong>in</strong>T, Spencer TE, Thompson JA, W<strong>in</strong>ger QA, Westhus<strong>in</strong> ME,2000: Evidence for placental abnormality as the major causeof mortality <strong>in</strong> first-trimester somatic cell cloned bov<strong>in</strong>efetuses. Biol Reprod 63, 1787–1794.Holloway AJ, van Laar RK, Tothill RW, Bowtell DD, 2002:Options available – from start to f<strong>in</strong>ish – for obta<strong>in</strong><strong>in</strong>g datafrom DNA microarrays II. Nat Genet 32(Suppl.), 481–489.Horcajadas JA, Sharkey AM, Catalano RD, Sherw<strong>in</strong> JR,Dom<strong>in</strong>guez F, Burgos LA, Castro A, Peraza MR, PellicerA, Simon C, 2006: Effect of an <strong>in</strong>trauter<strong>in</strong>e device on thegene expression profile of the endometrium. J Cl<strong>in</strong> Endocr<strong>in</strong>olMetab 91, 3199–3207.Hughes TR, Mao M, Jones AR, Burchard J, Marton MJ,Shannon KW, Lefkowitz SM, Ziman M, Schelter JM, MeyerMB, Kobayashi S, Davis C, Dai H, He YD, Stephaniants SB,Cavet G, Walker WL, West A, Coffey E, Shoemaker DD,Stoughton R, Blanchard AP, Friend SH, L<strong>in</strong>sley PS, 2001:Expression profil<strong>in</strong>g us<strong>in</strong>g microarrays fabricated by an <strong>in</strong>kjetoligonucleotide synthesizer. Nat Biotechnol 19, 342–347.Humblot P, 2001: Use of pregnancy specific prote<strong>in</strong>s andprogesterone assays to monitor pregnancy and determ<strong>in</strong>ethe tim<strong>in</strong>g, frequencies and sources of embryonic mortality<strong>in</strong> rum<strong>in</strong>ants. Theriogenology 56, 1417–1433.Johnson GA, Spencer TE, Burghardt RC, Joyce MM, BazerFW, 2000: Interferon-tau and progesterone regulate ubiquit<strong>in</strong>cross-reactive prote<strong>in</strong> expression <strong>in</strong> the ov<strong>in</strong>e uterus.Biol Reprod 62, 622–627.Joyce M, Burghardt R, Bazer F, Zaunbrecher G, Johnson G,2003: Interferon-stimulated genes (ISGs) are <strong>in</strong>duced <strong>in</strong> theendometrium of pregnant but not pseudopregnant pigs. BiolReprod 68 (Suppl. 1), Abstract 230, 206.Kashiwagi A, DiGirolamo CM, Kanda Y, Niikura Y, EsmonCT, Hansen TR, Shioda T, Pru JK, 2007: The postimplantationembryo differentially regulates endometrial gene expressionand decidualization. Endocr<strong>in</strong>ology 148, 4173–4184.Kle<strong>in</strong> C, Bauersachs S, Ulbrich SE, E<strong>in</strong>spanier R, Meyer HH,Schmidt SE, Reichenbach HD, Vermehren M, S<strong>in</strong>owatz F,Blum H, Wolf E, 2006: Monozygotic tw<strong>in</strong> model reveals novelembryo-<strong>in</strong>duced transcriptome changes of bov<strong>in</strong>e endometrium<strong>in</strong> the preattachment period. Biol Reprod 74, 253–264.Kreil DP, Russell RR, Russell S, 2006: Microarray oligonucleotideprobes. Methods Enzymol 410, 73–98.L<strong>in</strong>genfelter BM, Dailey RA, Inskeep EK, Vernon MW, PooleDH, Rh<strong>in</strong>ehart JD, Yao J, 2008: Microarray analysis ofgene expression <strong>in</strong> granulosal cells from persistent follicles <strong>in</strong>cattle. Anim Reprod Sci 104, 405–413.Lonergan P, Rizos D, Gutierrez-Adan A, Fair T, Boland MP,2003: Oocyte and embryo quality: effect of orig<strong>in</strong>, cultureconditions and gene expression patterns. Reprod DomestAnim 38, 259–267.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Use of Microarray Technology for <strong>Reproduction</strong> <strong>in</strong> Cattle 367Lonergan P, Pedersen HG, Rizos D, Greve T, Thomsen PD,Fair T, Evans A, Boland MP, 2004: Effect of the postfertilizationculture environment on the <strong>in</strong>cidence of chromosomeaberrations <strong>in</strong> bov<strong>in</strong>e blastocysts. Biol Reprod 71,1096–1100.Lonergan P, Fair T, Corcoran D, Evans ACO, 2006: Effect ofculture environment on gene expression and developmentalcharacteristics <strong>in</strong> IVF-derived embryos. Theriogenology 65,137–152.Lyons P, 2003: Advances <strong>in</strong> spotted microarray resources forexpression profil<strong>in</strong>g. Brief Funct Genomic Proteomic 2, 21–30.McGall GH, Christians FC, 2002: High-density geneChipoligonucleotide probe arrays. Adv Biochem Eng Biotechnol77, 21–42.McKusick VA, Ruddle FH, 1987: A new discipl<strong>in</strong>e, a newname, a new journal. Genomics 1, 1–2.Mihm M, Bleach EC, 2003: Endocr<strong>in</strong>e regulation of ovarianantral follicle development <strong>in</strong> cattle. Anim Reprod Sci 78,217–237.Mihm M, Evans ACO, 2008: Mechanisms for dom<strong>in</strong>ant follicleselection <strong>in</strong> monovulatory species: a comparison of morphological,endocr<strong>in</strong>e and <strong>in</strong>traovarian events <strong>in</strong> cows, maresand women. Reprod Domest Anim 43(Suppl. 2), 48–56.Mihm M, Baker PJ, Ireland JL, Smith GW, Coussens PM,Evans ACO, Ireland JJ, 2006: Molecular evidence that growthof dom<strong>in</strong>ant follicles <strong>in</strong>volves a reduction <strong>in</strong> follicle-stimulat<strong>in</strong>ghormone dependence and an <strong>in</strong>crease <strong>in</strong> lute<strong>in</strong>iz<strong>in</strong>ghormone dependence <strong>in</strong> cattle. Biol Reprod 74, 1051–1059.Mihm M, Baker PJ, Flem<strong>in</strong>g LM, Monteiro AM, O’ShaughnessyPJ, 2008: Differentiation of the bov<strong>in</strong>e dom<strong>in</strong>antfollicle from the cohort upregulates mRNA expression fornew tissue development genes. <strong>Reproduction</strong> 135, 253–265.Misirlioglu M, Page GP, Sagirkaya H, Kaya A, Parrish JJ,First NL, Memili E, 2006: Dynamics of global transcriptome<strong>in</strong> bov<strong>in</strong>e matured oocytes and preimplantationembryos. Proc Natl Acad Sci U S A 103, 18905–18910.Mitko K, Ulbrich SE, Wenigerk<strong>in</strong>d H, S<strong>in</strong>owatz F, Blum H,Wolf E, Bauersachs S, 2008: Dynamic changes <strong>in</strong> messengerRNA profiles of bov<strong>in</strong>e endometrium dur<strong>in</strong>g the oestrouscycle. <strong>Reproduction</strong> 135, 225–240.Mourot M, Dufort I, Gravel C, Algriany O, Dieleman S, SirardMA, 2006: The <strong>in</strong>fluence of follicle size, FSH-enrichedmaturation medium, and early cleavage on bov<strong>in</strong>e oocytematernal mRNA levels. Mol Reprod Dev 73, 1367–1379.Niemann H, Carnwath JW, Kues W, 2007: Application ofDNA array technology to mammalian embryos. Theriogenology68(Suppl. 1), S165–S177.Patel OV, Bettegowda A, Ireland JJ, Coussens PM, LonerganP, Smith GW, 2007: Functional genomics studies of oocytecompetence: evidence that reduced transcript abundance forfollistat<strong>in</strong> is associated with poor developmental competenceof bov<strong>in</strong>e oocytes. <strong>Reproduction</strong> 133, 95–106.Pfister-Genskow M, Myers C, Childs LA, Lacson JC, PattersonT, Betthauser JM, Goueleke PJ, Koppang RW, LangeG, Fisher P, Watt SR, Forsberg EJ, Zheng Y, Leno GH,Schultz RM, Liu B, Chetia C, Yang X, Hoeschele I,Eilertsen KJ, 2005: Identification of differentially expressedgenes <strong>in</strong> <strong>in</strong>dividual bov<strong>in</strong>e preimplantation embryos producedby nuclear transfer: improper reprogramm<strong>in</strong>g ofgenes required for development. Biol Reprod 72, 546–555.Quackenbush J, 2002: Microarray data normalization andtransformation. Nat Genet 32(Suppl.), 496–501.Rizos D, Ward F, Duffy P, Boland MP, Lonergan P, 2002:Consequences of bov<strong>in</strong>e oocyte maturation, fertilization orearly embryo development <strong>in</strong> vitro versus <strong>in</strong> vivo: implicationsfor blastocyst yield and blastocyst quality. MolReprod Dev 61, 234–248.Ross JW, Ashworth MD, White FJ, Johnson GA, Ayoubi PJ,DeSilva U, Whitworth KM, Prather RS, Geisert RD, 2007:Premature estrogen exposure alters endometrial gene expressionto disrupt pregnancy <strong>in</strong> the pig. Endocr<strong>in</strong>ology 148,4761–4773.Schena M, Shalon D, Davis RW, Brown PO, 1995: Quantitativemonitor<strong>in</strong>g of gene expression patterns with acomplementary DNA microarray. Science 270, 467–470.Schulze A, Downward J, 2001: Navigat<strong>in</strong>g gene expressionus<strong>in</strong>g microarrays – a technology review. Nat Cell Biol 3,E190–E195.Sirard MA, Richard F, Blond<strong>in</strong> P, Robert C, 2006: Contributionof the oocyte to embryo quality. Theriogenology 65,126–136.Sisco B, Hagemann LJ, Shell<strong>in</strong>g AN, Pfeffer PL, 2003:Isolation of genes differentially expressed <strong>in</strong> dom<strong>in</strong>ant andsubord<strong>in</strong>ate bov<strong>in</strong>e follicles. Endocr<strong>in</strong>ology 144, 3904–3913.Smith GW, Rosa GJ, 2007: Interpretation of microarray data:trudg<strong>in</strong>g out of the abyss towards elucidation of biologicalsignificance. J Anim Sci 85, E20–E23.Smith SL, Everts RE, Tian XC, Du F, Sung LY, Rodriguez-Zas SL, Jeong BS, Renard JP, Lew<strong>in</strong> HA, Yang X, 2005:Global gene expression profiles reveal significant nuclearreprogramm<strong>in</strong>g by the blastocyst stage after clon<strong>in</strong>g. ProcNatl Acad Sci U S A 102, 17582–17587.Somers J, Smith C, Donnison M, Wells DN, Henderson H,McLeay L, Pfeffer PL, 2006: Gene expression profil<strong>in</strong>g of<strong>in</strong>dividual bov<strong>in</strong>e nuclear transfer blastocysts. <strong>Reproduction</strong>131, 1073–1084.Suchyta SP, Sipkovsky S, Kruska R, Jeffers A, McNulty A,Coussens MJ, Tempelman RJ, Halgren RG, Saama PM,Bauman DE, Boisclair YR, Burton JL, Collier RJ, DePetersEJ, Ferris TA, Lucy MC, McGuire MA, Medrano JF,Overton TR, Smith TP, Smith GW, Sonstegard TS, Spa<strong>in</strong>JN, Spiers DE, Yao J, Coussens PM, 2003: Developmentand test<strong>in</strong>g of a high-density cDNA microarray resource forcattle. Physiol. Genomics 15, 158–164.Ushizawa K, Herath CB, Kaneyama K, Shiojima S, HirasawaA, Takahashi T, Imai K, Ochiai K, Tokunaga T, TsunodaY, Tsujimoto G, Hashizume K, 2004: cDNA microarrayanalysis of bov<strong>in</strong>e embryo gene expression profiles dur<strong>in</strong>gthe pre-implantation period. Reprod Biol Endocr<strong>in</strong>ol 2, 77.Webb R, Campbell BK, 2007: Development of the dom<strong>in</strong>antfollicle: mechanisms of selection and ma<strong>in</strong>tenance of oocytequality. Soc Reprod Fertil Suppl 64, 141–163.Wrenzycki C, Herrmann D, Lucas-Hahn A, Korsawe K,Lemme E, Niemann H, 2005: Messenger RNA expressionpatterns <strong>in</strong> bov<strong>in</strong>e embryos derived from <strong>in</strong> vitro proceduresand their implications for development. Reprod Fertil Dev17, 23–35.Wrenzycki C, Herrmann D, Niemann H, 2007: MessengerRNA <strong>in</strong> oocytes and embryos <strong>in</strong> relation to embryoviability. Theriogenology 68(Suppl. 1), S77–S83.Yang X, Smith SL, Tian XC, Lew<strong>in</strong> HA, Renard JP,Wakayama T, 2007: Nuclear reprogramm<strong>in</strong>g of clonedembryos and its implications for therapeutic clon<strong>in</strong>g. NatGenet 39, 295–302.Zielak AE, Canty MJ, Forde N, Coussens PM, Smith GW,Lonergan P, Ireland JJ, Evans ACO, 2007a: Differentialexpression of genes for transcription factors <strong>in</strong> theca andgranulosa cells follow<strong>in</strong>g selection of a dom<strong>in</strong>ant follicle <strong>in</strong>cattle. Mol Reprod Dev 19, 967–975.Zielak AE, Forde N, Park SDE, Doohan F, Coussens PM,Smith GW, Ireland JJ, Lonergan P, Evans ACO, 2007b:Identification of novel genes associated with dom<strong>in</strong>antfollicle development <strong>in</strong> cattle. Reprod Fertil Dev 19, 967–975.Author’s address (for correspondence): ACO Evans, UCD Veter<strong>in</strong>arySciences Centre, Belfield, Dubl<strong>in</strong> 4, Ireland. E-mail: alex.evans@ucd.ieConflict of <strong>in</strong>terest: All authors declare no conflict of <strong>in</strong>terests.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 368–373 (2008); doi: 10.1111/j.1439-0531.2008.01186.xISSN 0936-6768Breed<strong>in</strong>g Soundness Evaluation and Semen Analysis for Predict<strong>in</strong>g Bull FertilityJP Kastelic 1,2 and JC Thundathil 21 Agriculture and Agri-Food Canada, Lethbridge Research Centre, Lethbridge; 2 Faculty of Veter<strong>in</strong>ary Medic<strong>in</strong>e, University of Calgary, Calgary, AB,CanadaContentsBull fertility is <strong>in</strong>fluenced by numerous factors. Although 20–40% of bulls <strong>in</strong> an unselected population may have reducedfertility, few are completely sterile. Breed<strong>in</strong>g soundness refersto a bull’s ability to get cows pregnant. A standard breed<strong>in</strong>gsoundness evaluation identifies bulls with substantial deficits <strong>in</strong>fertility, but does not consistently identify sub-fertile bulls.In this regard, the use of frozen-thawed semen (from bulls <strong>in</strong>commercial AI centres) that meets m<strong>in</strong>imum quality standardscan result <strong>in</strong> pregnancy rates that differ by 20–25 percentagepo<strong>in</strong>ts. Although no s<strong>in</strong>gle diagnostic test can accuratelypredict variations <strong>in</strong> fertility among bulls that are produc<strong>in</strong>gapparently normal semen, recent studies suggested that acomb<strong>in</strong>ation of laboratory tests were predictive of fertility.This review is focused on recent developments <strong>in</strong> prediction ofbull fertility, based on assessments at the molecular, cellularand whole-animal levels.IntroductionBreed<strong>in</strong>g soundness refers to a bull’s ability to get cowspregnant. Although 20–40% of bulls may have reducedfertility, few are completely sterile (Kastelic et al. 2000).Sub-fertile bulls delay conception, prolong the calv<strong>in</strong>gseason, reduce calf wean<strong>in</strong>g weights and <strong>in</strong>crease femaleculls. Multiple sire breed<strong>in</strong>g groups and low breed<strong>in</strong>gpressure may mask sub-fertile bulls, but s<strong>in</strong>gle-siremat<strong>in</strong>g groups and AI <strong>in</strong>crease the importance of bullfertility.Traditional Breed<strong>in</strong>g Soundness EvaluationIn general, bull breed<strong>in</strong>g soundness potential is eitherdeterm<strong>in</strong>ed by breed<strong>in</strong>g many normal, fertile females(and assess<strong>in</strong>g pregnancy rates) or conduct<strong>in</strong>g a breed<strong>in</strong>gsoundness evaluation. No s<strong>in</strong>gle measurement orcriterion reliably predicts fertility and therefore, severalcriteria are usually evaluated. The standards of theSociety for Theriogenology (http://www.therio.org) are<strong>in</strong>tended to assess the likelihood of a bull establish<strong>in</strong>gpregnancy <strong>in</strong> ‡25 healthy, cycl<strong>in</strong>g females <strong>in</strong> a 65–70 daybreed<strong>in</strong>g season. The classification is based on a physicalevaluation and acceptable thresholds for testiculardevelopment, sperm motility and normal sperm morphology(libido and tests for venereal diseases are notrout<strong>in</strong>ely evaluated). A breed<strong>in</strong>g soundness evaluation<strong>in</strong>cludes assessment of size, muscl<strong>in</strong>g, body conditionand freedom from disease and physical defects. Scrotalcircumference (SC) is highly correlated with pairedtestes weight, which is correlated with daily spermproduction and semen quality (reviewed by Barth 2007).Bulls with large SC have half-sib heifers and daughterswith earlier puberty and greater fertility (reviewed byBarth 2007). As the heritability of SC <strong>in</strong> young bulls(1–2 years) is 0.5, it responds well to selection(reviewed by Barth 2007). Coe and Gibson (1993)evaluated 264 beef bulls (13 breeds), and at 200 daysof age, calves with SC >23 cm had a 95% probability ofSC >34 cm by 365 days of age, whereas calves with SC34 cm by365 days.Scrotal circumference is measured by forc<strong>in</strong>g the testesto the bottom of the scrotum and us<strong>in</strong>g a flexible tape toapply moderate tension at the largest circumference. Thescrotum should have a dist<strong>in</strong>ct neck, testes should befreely moveable, similar <strong>in</strong> size, firm and resilient, withnormal epididymides and spermatic cords. The sheathand penis should be of appropriate size and free ofdefects. The most common abnormality of the accessorysex glands is vesicular adenitis (usually unilateral), and itoccurs <strong>in</strong> 2–4% of bulls 1–2 years of age, but is uncommon(30% morphologicallyabnormal sperm or >20% head defects (http://www.therio.org; reviewed by Barth 2007).A bull that is healthy and sound, with adequate SC,‡70% morphologically normal sperm, and ‡30% progressivemotility, is designated Satisfactory PotentialBreeder (http://www.therio.org; Barth 2007). A bullwith temporary conditions (likely to resolve) is designatedClassification Deferred. This typically <strong>in</strong>cludesrecent puberty, an <strong>in</strong>jury or lameness that is likely toresolve, or temporary testicular degeneration (e.g.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Breed<strong>in</strong>g Soundness and Semen Analysis <strong>in</strong> Bulls 369because of hot weather). A bull with undesirableheritable defects, small SC, debilitat<strong>in</strong>g <strong>in</strong>jury or disease,or permanent testicular degeneration, is designatedUnsatisfactory Potential Breeder.Bulls <strong>in</strong> AI CentresBulls <strong>in</strong> commercial AI centres often have substantialvariations <strong>in</strong> fertility, and frozen semen that meetsm<strong>in</strong>imum quality standards can yield pregnancy ratesthat differ by 20–25% (Larson and Miller 2000).Standard semen end po<strong>in</strong>ts identify grossly abnormalsemen, but do not consistently identify sub-fertile bullswith apparently normal semen (Gadea et al. 2004). Asbull fertility is <strong>in</strong>fluenced by a wide range of factors, nos<strong>in</strong>gle diagnostic test can accurately predict fertility(Petrunk<strong>in</strong>a et al. 2007; Rodriguez-Mart<strong>in</strong>ez 2007).Sperm Plasma Membrane ViabilityAs a functional sperm plasma membrane is essential forfertilization, its viability (<strong>in</strong>tegrity) should be assessed.Eos<strong>in</strong> permeates non-viable sperm (p<strong>in</strong>k to red spermheads), whereas viable sperm appear white. Comb<strong>in</strong>ationsof various fluorescent probes, specific for viable ornon-viable sperm, are also used, and the most commonis Syber-14 and propidium iodide (Gillan et al. 2005).Syber-14 penetrates ‘viable’ sperm (with an <strong>in</strong>tactplasma membrane), mak<strong>in</strong>g the DNA bright green(Gillan et al. 2005), whereas propidium iodide penetratesdead and non-viable sperm and renders them red.The proportion of live and dead sperm (green and red,respectively) is determ<strong>in</strong>ed with fluorescent microscopyor flow cytometry.Brito et al. (2003) studied several methods for evaluat<strong>in</strong>gsperm membrane viability (eos<strong>in</strong> ⁄ nigros<strong>in</strong>, Trypanblue, fluorescent probes and the response of sperm to theexposure of a hypoosmotic solution) and their relationshipto cleavage (IVF). Although these sta<strong>in</strong>s evaluate thephysical <strong>in</strong>tegrity of sperm membrane, the hypoosmoticswell<strong>in</strong>g test (HOST; Jeyendran et al. 1984) evaluates itsfunctional competence. This was the only plasmalemmaevaluation method that contributed to conventionalsperm quality tests <strong>in</strong> predict<strong>in</strong>g the success of <strong>in</strong> vitrofertilization (based on cleavage rate; Brito et al. 2003).An Annex<strong>in</strong>-V b<strong>in</strong>d<strong>in</strong>g assay us<strong>in</strong>g fluorescence-activatedcell sort<strong>in</strong>g (FACS) assesses externalization ofphosphatidyl ser<strong>in</strong>e of cryopreserved bov<strong>in</strong>e semen (Anzaret al. 2002; Januskauskas et al. 2003). This test detectstranslocation of membrane phosphatidyl ser<strong>in</strong>e, whichprecedes loss of membrane <strong>in</strong>tegrity dur<strong>in</strong>g cryopreservation-<strong>in</strong>duced,apoptosis-like cell degradation. However,its correlation with fertility of frozen-thawed bullsemen varied among studies (Rodriguez-Mart<strong>in</strong>ez 2003).Sperm MotilitySperm motility (total or progressive) was traditionallyevaluated subjectively with light microscopy. Thismethod detects gross abnormalities, but not subtlevariations <strong>in</strong> motility that can affect fertility. However,computer-assisted sperm analysis (CASA), a much moreobjective method, measures specific motion characteristicsassociated with functional status. For example,capacitated sperm are hyperactivated (high amplitudeand low frequency; Yanagimachi 1994). Furthermore,comb<strong>in</strong>ations of sperm motion characteristics assessedby CASA (beat cross frequency, l<strong>in</strong>earity, average pathvelocity, straightness and curvil<strong>in</strong>ear velocity) weresignificantly correlated with bull fertility (Farrell et al.1998), as were average path velocity, total motility,l<strong>in</strong>early motile sperm and total number of motile sperm<strong>in</strong> swim-up preparations (Hallap et al. 2006). However,comparisons of CASA results are only reliable whenidentical sperm evaluation sett<strong>in</strong>gs are used.Sperm–Oviduct InteractionBillions of sperm are ejaculated, but


370 JP Kastelic and JC Thundathilsperm–oocyte <strong>in</strong>teractions. Follow<strong>in</strong>g capacitation,sperm <strong>in</strong>teract with the zona pellucida (ZP). For this<strong>in</strong>teraction, galactosyl transferase (Larson and Miller2000), p47 (Enssl<strong>in</strong> et al. 1998), sp56 (Cheng et al. 1994)and zonadhes<strong>in</strong> (Hardy and Garbers 1995) are apparentlysperm receptors, whereas zona glycoprote<strong>in</strong> (ZP3)is the ligand (Yanagimachi 1994). This <strong>in</strong>teraction leadsto an acrosome reaction (Yanagimachi 1994); the <strong>in</strong>neracrosomal membrane <strong>in</strong>teracts with a second glycoprote<strong>in</strong>,ZP2, facilitat<strong>in</strong>g the secondary b<strong>in</strong>d<strong>in</strong>g of sperm tothe zona matrix dur<strong>in</strong>g penetration (Yanagimachi 1994;Aitken 2006).There are several reports regard<strong>in</strong>g the associationbetween sperm–zona b<strong>in</strong>d<strong>in</strong>g and fertility. There weredifferences among bulls (with known fertility) <strong>in</strong> therelative number of sperm bound to bov<strong>in</strong>e oocytes(Fazeli et al. 1993). However, large numbers of oocytesare needed and differences among bulls are detectableonly if they exceed differences among oocytes (Zhanget al. 1995). Alternatively, a hemizona assay (Fazeliet al. 1997) uses both halves of a ZP to compare b<strong>in</strong>d<strong>in</strong>gability of control sperm (known fertility) and test sperm(unknown fertility). Zona b<strong>in</strong>d<strong>in</strong>g and hemizona b<strong>in</strong>d<strong>in</strong>gassays and non-return rates were significantlycorrelated. In vitro zona penetration assays have beenused to predict <strong>in</strong> vivo fertility (Puglisi et al. 2004).However, sperm penetration varies accord<strong>in</strong>g to sperm–oocyte ratio, duration of <strong>in</strong>cubation and hepar<strong>in</strong> concentration,limit<strong>in</strong>g the value of this test.Sperm Oolemma Fusion and Sperm DNADecondensationHenault et al. (1995) used a homologous zona-freeoocyte penetration assay to demonstrate the effects ofaccessory gland fluid from low- vs high-fertility bulls.Moreover, competitive penetration of zona-free bov<strong>in</strong>eoocytes by fluorochrome-labelled bull sperm was relatedto <strong>in</strong> vivo fertility (Henault and Killian 1995) andevaluated the ability of sperm to undergo chromat<strong>in</strong>decondensation and pronucleus formation. In theabsence of a ZP, there is an equal opportunity for allsperm to fuse with the oocyte membrane, undergo DNAdecondensation and pronuclei formation (Thundathilet al. 2001a).Sperm chromat<strong>in</strong> <strong>in</strong>tegrity is an important determ<strong>in</strong>antof the ability of sperm to form normal pronuclei.Sperm DNA is associated with histone nucleoprote<strong>in</strong>sand organized <strong>in</strong>to classical nucleosome core particlesdur<strong>in</strong>g early spermatogenesis. However, these histonenucleoprote<strong>in</strong>s are replaced by transition prote<strong>in</strong>s,which are subsequently replaced by protam<strong>in</strong>es (Yanagimachi1994). Ultimately, chromat<strong>in</strong> of mature spermhas a compact toroidal structure that resists denaturation.Lewis and Aitken (2005) suggested oxidativestress was a major cause of sperm DNA damage, withreduced pre-implantation embryo development andpregnancy rates. Increased testicular temperature reducedthe stability of sperm DNA (Karab<strong>in</strong>us et al.1997) and impaired the ability of these sperm to undergoDNA decondensation and pronuclei formation (Walterset al. 2006). However, l<strong>in</strong>ks among elevated testiculartemperature, oxidative stress and sperm DNA damage<strong>in</strong> bulls rema<strong>in</strong> unclear. The sperm cell structure assay(SCSA) uses flow cytometry to determ<strong>in</strong>e chromat<strong>in</strong><strong>in</strong>tegrity, based on resistance to acid denaturation.Sperm are exposed to low pH and sta<strong>in</strong>ed with acrid<strong>in</strong>eorange, which emits green or red fluorescence when itb<strong>in</strong>ds to double- (<strong>in</strong>tact) or s<strong>in</strong>gle-stranded DNA(denatured), respectively. The ratio of red to (red + -green) fluorescence measures chromat<strong>in</strong> denaturation,which is significantly correlated with fertility (Ballacheyet al. 1987, 1988; Januskauskas et al. 2001; Waterhouseet al. 2006). In brief, flow cytometry-based approachesprovide a quantitative measure of the structural <strong>in</strong>tegrityof sperm chromat<strong>in</strong>, based on a large number ofsperm, whereas IVF-based tests evaluate the ability ofsperm to undergo DNA decondensation and pronuclearformation dur<strong>in</strong>g fertilization.Association Between In Vitro Fertilization andFertilityMarquant-Le Guienne et al. (1990) reported that pronuclearformation can be used to predict field fertility ofbulls. Zhang et al. (1997) reported that fertilizationbased on cleavage rate was highly correlated with nonreturnrates, but blastocyst production varied amongtest dates. However, fertility predictions were moreaccurate when based on several laboratory assays vs as<strong>in</strong>gle assay (Truelson et al. 1996). In this regard, aseven-variable model (post-thaw total motility, postthawsperm with a l<strong>in</strong>ear motile pattern, sperm concentration,concentration of motile sperm after swim-up,sperm ZP-b<strong>in</strong>d<strong>in</strong>g, cleavage rate of total oocytes andblastocyst rate of total oocytes) accounted for 84.6% ofthe variation <strong>in</strong> non-return rates (Zhang et al. 1999).However, this approach may not be sensitive enough todiscrim<strong>in</strong>ate among highly fertile bulls. Similarly, amodel with 30 post-thaw sperm characteristics (<strong>in</strong>clud<strong>in</strong>gcleavage rate) accurately predicted fertility (based onconception and non-return rates) of both high- and lowfertilitybulls (Phillips et al. 2004).Future DirectionsThe cell biology approaches described earlier may serveas supplementary tests to a standard breed<strong>in</strong>g soundnessevaluation and improve the reliability of fertility predictions.However, a better understand<strong>in</strong>g of the regulationof sperm function and its contributions to earlyembryo development at the molecular level may lead toreliable molecular markers of fertility, with implicationsfor predict<strong>in</strong>g fertility variations <strong>in</strong> bulls used for AI.As DNA is transcriptionally <strong>in</strong>active <strong>in</strong> ejaculatedsperm, physiological functions are regulated by structuraland functional prote<strong>in</strong>s and identify<strong>in</strong>g theseprote<strong>in</strong>s may have implications for predict<strong>in</strong>g fertility.The role of heat-shock prote<strong>in</strong>s <strong>in</strong> sperm capacitation(Kamarudd<strong>in</strong> et al. 2004) and their potential role <strong>in</strong>sperm–oocyte b<strong>in</strong>d<strong>in</strong>g have been reported (Matweeet al. 2001). Specific sperm prote<strong>in</strong>s <strong>in</strong>volved <strong>in</strong> the<strong>in</strong>teraction between sperm and ZP (Larson and Miller2000; Inoue et al. 2005) and sperm–oocyte adhesion andegg activation (Evans and Florman 2002) have beenidentified, and the role of angiotens<strong>in</strong> convert<strong>in</strong>g enzymeÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Breed<strong>in</strong>g Soundness and Semen Analysis <strong>in</strong> Bulls 371(ACE) <strong>in</strong> fertilization (Kondoh et al. 2005) has beendescribed. In addition, PAWP, a sperm-specific WWdoma<strong>in</strong>-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>, promotes meiotic resumptionand pronuclear development dur<strong>in</strong>g fertilization (Wuet al. 2007).We recently used scrotal <strong>in</strong>sulation to <strong>in</strong>duce abnormalspermatogenesis (unpublished) and identified spermprote<strong>in</strong>s associated with abnormal spermatogenesis as aresult of elevated testicular temperature. There wasdifferential expression (between normal and abnormalsperm of the same bull) of the alpha 4 subunit ofNa + ⁄ K + ATPase, tissue <strong>in</strong>hibitor of metalloprote<strong>in</strong>ase-2 (TIMP-2), ACE and hexok<strong>in</strong>ase-1. Further studies areneeded to elucidate the specific role of these spermprote<strong>in</strong>s <strong>in</strong> fertilization and early development.Although the sperm prote<strong>in</strong>s described earlier areassociated with the regulation of specific sperm functions,prote<strong>in</strong>s with changes <strong>in</strong> expression patterns <strong>in</strong>response to variations <strong>in</strong> fertility rema<strong>in</strong> unknown, buthave great potential as fertility markers. In this regard,hepar<strong>in</strong>-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s (24–31 kDa) were proposed asa genetic marker for fertility differences <strong>in</strong> bullsproduc<strong>in</strong>g normal semen (Bell<strong>in</strong> et al. 1998; McCauleyet al. 1999). Furthermore, there are associations betweenspecific sem<strong>in</strong>al plasma prote<strong>in</strong>s and fertility(Killian et al. 1993; Moura et al. 2006). Sperm prote<strong>in</strong>sfrom low- vs high-fertility Nelore bulls with acceptablesemen had differential expression (two-dimensional gelelectrophoresis) of sperm membrane prote<strong>in</strong>s (Roncolettaet al. 2006). Similarly, accessory gland fluids fromhigh-fertility Holste<strong>in</strong> bulls had more bov<strong>in</strong>e sem<strong>in</strong>alplasma prote<strong>in</strong> (BSP) 30 kDa and phospholipase A2,whereas osteopont<strong>in</strong> appeared to improve the ability ofepididymal sperm (from low-fertility bulls) to penetrateoocytes <strong>in</strong> vitro (Moura et al. 2007). Therefore, identify<strong>in</strong>gfertility-associated sperm or sem<strong>in</strong>al plasma prote<strong>in</strong>sby compar<strong>in</strong>g low- vs high-fertility bulls, and<strong>in</strong>vestigat<strong>in</strong>g the role of these prote<strong>in</strong>s <strong>in</strong> the regulationof sperm function, fertilization or embryo development,may identify markers that predict fertility.Morphologically abnormal sperm failed dur<strong>in</strong>g gamete<strong>in</strong>teraction or pre-implantation development (Thundathilet al. 2000, 2001b; Walters et al. 2005). Inaddition, embryos result<strong>in</strong>g from the fertilization ofoocytes by morphologically normal sperm, coexist<strong>in</strong>g <strong>in</strong>the ejaculate along with abnormal sperm, had reduceddevelopmental competence, suggest<strong>in</strong>g that these spermwere functionally impaired. Therefore, <strong>in</strong>creas<strong>in</strong>g the<strong>in</strong>sem<strong>in</strong>ation dose to compensate for <strong>in</strong>fertility as aresult of compensable factors (Saacke et al. 2000)requires further <strong>in</strong>vestigation. Data from commercialembryo production units suggested that bulls differ <strong>in</strong>their ability to produce pre-implantation embryos <strong>in</strong> vivo(Thundathil and Mapletoft, unpublished data). In thisregard, damage to sperm DNA because of oxidativestress, chromosome anomalies and environmental effects,<strong>in</strong>clud<strong>in</strong>g elevated testicular temperature, time ofAI relative to oestrus, duration of semen storage,duration of sperm–oocyte <strong>in</strong>teraction, age of malesand <strong>in</strong>fectious agents <strong>in</strong> semen <strong>in</strong>fluence quality ofembryos (reviewed by Chenoweth 2007). However, morestudies are needed to elucidate the effects of paternalgenes (Brow<strong>in</strong>g and Strome 1996), sperm RNA(Krawetz 2005; Miller et al. 2005; Miller and Ostermeier2006; Boerke et al. 2007), specific sperm and sem<strong>in</strong>alplasma prote<strong>in</strong>s (Cancel et al. 1997; McCauley et al.2001; Roncoletta et al. 2006) and evolutionarily conservedfactors associated with sperm DNA (Wu andChu 2008) on fertilization and early embryo development.As fertility is affected by numerous factors, thesearch for fertility markers should <strong>in</strong>clude the wholeanimallevel. Bov<strong>in</strong>e testes must be 4–5°C below bodycoretemperature (38°C) for normal spermatogenesis(Setchell 1978). Assessment of scrotal surface temperatureswith <strong>in</strong>frared thermography (scrotal thermogram)provided detailed <strong>in</strong>formation regard<strong>in</strong>g a bull’sability to regulate testicular temperature (Coulter 1988;Kastelic et al. 2000). Beef bulls with abnormal scrotalthermograms had lower fertility to natural service(Lunstra and Coulter 1997). Furthermore, ultrasonographicevaluations of the testicular vascular cone andits fat cover were <strong>in</strong>dicative of thermoregulatorycapability, and associated with semen productionpotential and semen quality (Kastelic et al. 2001;Arteaga et al. 2005).ConclusionA traditional breed<strong>in</strong>g soundness exam<strong>in</strong>ation willusually identify bulls that are grossly abnormal. However,a comprehensive approach, <strong>in</strong>clud<strong>in</strong>g assess<strong>in</strong>gsperm function and fertility at the molecular, cellularand whole-animal levels, is needed to predict fertility ofbulls that are produc<strong>in</strong>g apparently normal sperm.ReferencesAitken RJ, 2006: Sperm function tests and fertility. Int JAndrol 29, 69–75.Anzar M, He L, Buhr MM, Kroetsch TG, Pauls KP, 2002:Sperm apoptosis <strong>in</strong> fresh and cryopreserved bull semendetected by flow cytometry and its relationship with fertility.Biol Reprod 66, 354–360.Arteaga AA, Barth AD, Brito LF, 2005: Relationship betweensemen quality and pixel-<strong>in</strong>tensity of testicular ultrasonogramsafter scrotal <strong>in</strong>sulation <strong>in</strong> beef bulls. Theriogenology64, 408–415.Ballachey BE, Hohenboken WD, Evenson DP, 1987: Heterogeneityof sperm nuclear chromat<strong>in</strong> structure and itsrelationship to bull fertility. Biol Reprod 36, 915–925.Ballachey BE, Evenson DP, Saacke RG, 1988: The spermchromat<strong>in</strong> structure assay. Relationship with alternate testsof semen quality and heterospermic performance of bulls.J Androl 9, 109–115.Barth AD, 2007: Evaluation of potential breed<strong>in</strong>g soundnessof the bull. <strong>in</strong>: Youngquist RS, Threlfall WR (eds), CurrentTherapy <strong>in</strong> Large Animal Theriogenology 2. SaundersElsevier, Philadelphia, pp. 228–240.Bell<strong>in</strong> ME, Oyarzo JN, Hawk<strong>in</strong>s HE, Zhang H, Smith RG,Forrest DW, Sprott LR, Ax RL, 1998: Fertility-associatedantigen on bull sperm <strong>in</strong>dicates fertility potential. J Anim Sci76, 2032–2039.Boerke A, Dieleman SJ, Gadella BM, 2007: A possible role forsperm RNA <strong>in</strong> early embryo development. Theriogenology68(Suppl. 1), S147–S155.Boilard M, Bailey J, Coll<strong>in</strong> S, Dufour M, Sirard M-A, 2002:Effect of bov<strong>in</strong>e oviduct epithelial cell apical plasmaÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


372 JP Kastelic and JC Thundathilmembranes on sperm function assessed by a novel flowcytometric approach. Biol Reprod 67, 1125–1132.Brito LFC, Barth AD, Bilodeau-Goeseels S, Panich PL,Kastelic JP, 2003: Comparison of methods to evaluate theplasmalemma of bov<strong>in</strong>e sperm and their relationship with <strong>in</strong>vitro fertilization rate. Theriogenology 60, 1539–1551.Brow<strong>in</strong>g H, Strome S, 1996: A sperm-supplied factor requiredfor embryogenesis <strong>in</strong> C. elegans. Development 122, 391–404.Cancel AM, Chapman DA, Killian GJ, 1997: Osteopont<strong>in</strong> isthe 55-kilodalton fertility-associated prote<strong>in</strong> <strong>in</strong> Holste<strong>in</strong> bullsem<strong>in</strong>al plasma. Biol Reprod 57, 1293–1301.Cheng A, Le T, Palacios M, Bookb<strong>in</strong>der LH, Wassarman PM,Suzuki F, Bleil JD, 1994: Sperm-egg recognition <strong>in</strong> themouse: characterization of sp56, a sperm prote<strong>in</strong> hav<strong>in</strong>gspecific aff<strong>in</strong>ity for ZP3. J Cell Biol 125, 867–878.Chenoweth PJ, 2007: Influence of the male on embryo quality.Theriogenology 68, 308–315.Coe PH, Gibson CD, 1993: Adjusted 200-day scrotal size as apredictor of 365-day scrotal circumference. Theriogenology40, 1065–1072.Cormier N, Sirard MA, Bailey JL, 1997: Premature capacitationof bov<strong>in</strong>e spermatozoa is <strong>in</strong>itiated by cryopreservation.J Androl 18, 461–468.Coulter GH, 1988: Thermography of bull testes. In: Proceed<strong>in</strong>gsof the 12th Technical Conference on Artificial Insem<strong>in</strong>ationand <strong>Reproduction</strong>, National Association of AnimalBreeders, pp. 58–63.Enssl<strong>in</strong> M, Vogel T, Calvete JJ, Thole HH, Schmidtke J,Matsuda T, To¨pfer-Petersen E, 1998: Molecular clon<strong>in</strong>gand characterization of P47, a novel boar sperm-associatedzona pellucida-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> homologous to afamily of mammalian secretory prote<strong>in</strong>s. Biol Reprod 58,1057–1064.Evans JP, Florman HM, 2002: The state of the union: the cellbiology of fertilization. Nat Cell Biol 4, S57–S63.Farrell PB, Presicce DA, Brockett CC, Foote RH, 1998:Quantification of bull sperm characteristics measured bycomputer assisted sperm analysis (CASA) and the relationshipto fertility. Theriogenology 49, 871–879.Fazeli AR, Steenweg W, Bevers MM, de Loos FA, van denBroek J, Colenbrander B, 1993: Development of a spermzona pellucida b<strong>in</strong>d<strong>in</strong>g assay for bull semen. Vet Rec 2, 14–16.Fazeli AR, Zhang BR, Steenweg W, Larsson B, Bevers MM,van den Broek J, Rodriguez-Mart<strong>in</strong>ez H, Colenbrander B,1997: Relationship between sperm-zona pellucida b<strong>in</strong>d<strong>in</strong>gassays and the 56-day nonreturn rate of cattle <strong>in</strong>sem<strong>in</strong>atedwith frozen-thawed bull semen. Theriogenology 48, 853–863.Gadea J, Selles E, Marco MA, 2004: The predictive value ofporc<strong>in</strong>e sem<strong>in</strong>al parameters on fertility outcome undercommercial conditions. Reprod Dom Anim 39, 303–308.Gillan L, Evans G, Maxwell WMC, 2005: Flow cytometricevaluation of sperm parameters <strong>in</strong> relation to fertilitypotential. Theriogenology 63, 445–457.Gwathmey TM, Ignotz GG, Mueller JL, Manjunath P, SuarezSS, 2006: Bov<strong>in</strong>e sem<strong>in</strong>al plasma prote<strong>in</strong>s PDC-109, BSP-A3, and BSP-30-kDa share functional roles <strong>in</strong> stor<strong>in</strong>g sperm<strong>in</strong> the oviduct. Biol Reprod 75, 501–507.Hallap T, Jaakma U, Rodriguez-Mart<strong>in</strong>ez H, 2006: Changes <strong>in</strong>semen quality <strong>in</strong> Estonian Holste<strong>in</strong> AI bulls at 3, 5 and7 years of age. Reprod Dom Anim 41, 214–218.Hardy DM, Garbers DL, 1995: A sperm membrane prote<strong>in</strong>that b<strong>in</strong>ds <strong>in</strong> a species-specific manner to the egg extracellularmatrix is homologous to von Willebrand factor. J BiolChem 270, 26025–26028.Henault MA, Killian GJ, 1995: Effects of sperm preparationand bull fertility on <strong>in</strong> vitro penetration of zona-free bov<strong>in</strong>eoocytes. Theriogenology 43, 739–749.Henault MA, Killian GJ, Kavanaugh JF, Griel LC Jr, 1995:Effect of accessory sex gland fluid from bulls of differ<strong>in</strong>gfertilities on the ability of cauda epididymal sperm topenetrate zona-free bov<strong>in</strong>e oocytes. Biol Reprod 52, 390–397.Hunter RHF, 2008: Sperm release from oviduct epithelialb<strong>in</strong>d<strong>in</strong>g is controlled hormonally by peri-ovulatory graafianfollicles. Mol Reprod Dev 75, 167–174.Hunter RHF, Fléchon B, Fléchon JE, 1987: Pre- and periovulatorydistribution of viable spermatozoa <strong>in</strong> the pigoviduct: Scann<strong>in</strong>g electron microscope study. Tissue Cell 19,423–436.Hunter RHF, Fle´chon B, Fle´chon JE, 1991: Distribution,morphology and epithelial <strong>in</strong>teractions of bov<strong>in</strong>e spermatozoa<strong>in</strong> the oviduct before and after ovulation: A scann<strong>in</strong>gelectron microscope study. Tissue Cell 23, 641–656.Ignotz GG, Cho MY, Suarez SS, 2007: Annex<strong>in</strong>s are candidateoviductal receptors for bov<strong>in</strong>e sperm surface prote<strong>in</strong>s andthus may serve to hold bov<strong>in</strong>e sperm <strong>in</strong> the oviductalreservoir. Biol Reprod 77, 906–913.Inoue N, Ikawa M, Isotani A, Okabe M, 2005: The immunoglobul<strong>in</strong>superfamily prote<strong>in</strong> Izumo is required for sperm tofuse with eggs. Nature 434, 152–153.Januskauskas A, Johannisson A, So¨derquist L, Rodriguez-Mart<strong>in</strong>ez H, 2000: Assessment of the fertiliz<strong>in</strong>g potential offrozen-thawed bov<strong>in</strong>e spermatozoa by calcium ionophoreA23187-<strong>in</strong>duced acrosome reaction <strong>in</strong> vitro. Theriogenology53, 859–875.Januskauskas A, Johanmsson A, Rodriguez-Mart<strong>in</strong>ez H,2001: Assessment of sperm quality through fluorometryand sperm chromat<strong>in</strong> structure assay <strong>in</strong> relation to fieldfertility of frozen thawed semen from Swedish AI bulls.Theriogenology 55, 947–961.Januskauskas A, Johannisson A, Rodriguez-Mart<strong>in</strong>ez H,2003: Subtle membrane changes <strong>in</strong> cryopreserved bull semen<strong>in</strong> relation with sperm viability, chromat<strong>in</strong> structure, andfield fertility. Theriogenology 60, 743–758.Jeyendran RS, Van der Ven HH, Perez-Pelaez M, Crabo BG,Zaneveld LJD, 1984: Development of an assay to assess thefunctional <strong>in</strong>tegrity of the human sperm membrane and itsrelationship to other semen characteristics. J Reprod Fertil70, 219–228.Kamarudd<strong>in</strong> M, Kroetsch T, Basrur PK, Hansen PJ, K<strong>in</strong>gWA, 2004: Immunolocalization of heat shock prote<strong>in</strong> 70 <strong>in</strong>bov<strong>in</strong>e spermatozoa. Andrologia 36, 327–334.Karab<strong>in</strong>us DS, Vogler CJ, Saacke RG, Evenson DP, 1997:Chromat<strong>in</strong> structural changes <strong>in</strong> sperm after scrotal <strong>in</strong>sulationof Holste<strong>in</strong> bulls. J Androl 18, 549–555.Kastelic JP, Cook RB, Coulter GH, 2000: Scrotal ⁄ testicularthermoregulation <strong>in</strong> bulls. <strong>in</strong>: Chenoweth PJ (ed.), Topics <strong>in</strong>Bull Fertility. International Veter<strong>in</strong>ary Information Service.Available from http://www.ivis.org.Kastelic JP, Cook RB, Pierson RA, Coulter GH, 2001:Relationships among scrotal and testicular characteristics,sperm production, and sem<strong>in</strong>al quality <strong>in</strong> 129 beef bulls.Can J Vet Res 65, 111–115.Killian GJ, Chapman DA, Rogowski LA, 1993: Fertilityassociatedprote<strong>in</strong>s <strong>in</strong> Holste<strong>in</strong> bull sem<strong>in</strong>al plasma. BiolReprod 49, 1202–1207.Kondoh G, Tojo H, Nakatani Y, Komazawa N, Murata C,Yamagata K, Maeda Y, K<strong>in</strong>oshita T, Okabe M, Taguchi R,Takeda J, 2005: Angiotens<strong>in</strong>-convert<strong>in</strong>g enzyme is a GPIanchoredprote<strong>in</strong> releas<strong>in</strong>g factor crucial for fertilization.Nat Med 11, 160–166.Krawetz SA, 2005: Paternal contribution: new <strong>in</strong>sights andfuture challenges. Nat Rev Genet 6, 633–642.Larson JL, Miller DJ, 2000: Can relative spermatozoalgalactosyltransferase activity be predictive of dairy bullfertility? J Dairy Sci 83, 2473–2479.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Breed<strong>in</strong>g Soundness and Semen Analysis <strong>in</strong> Bulls 373Lewis SE, Aitken RJ, 2005: DNA damage to spermatozoa hasimpacts on fertilization and pregnancy. Cell Tissue Res 322,33–41.Lunstra DD, Coulter GH, 1997: Relationship between scrotal<strong>in</strong>frared temperature patterns and natural-mat<strong>in</strong>g fertility <strong>in</strong>beef bulls. J Anim Sci 75, 767–774.Marquant-Le Guienne B, Humblot P, Thibier M, Thibault C,1990: Evaluation of bull semen fertility by homologous <strong>in</strong>vitro fertilization tests. Reprod Nutr Dev 30, 259–266.Matwee C, Kamarudd<strong>in</strong> M, Betts DH, Basrur PK, K<strong>in</strong>g WA,2001: The effects of antibodies to heat shock prote<strong>in</strong> 70 <strong>in</strong>fertilization and embryo development. Mol Hum Reprod 7,829–837.McCauley TC, Zhang H, Bell<strong>in</strong> ME, Ax RL, 1999: Purificationand characterization of fertility-associated antigen(FAA) <strong>in</strong> bov<strong>in</strong>e sem<strong>in</strong>al fluid. Mol Reprod Dev 54, 145–153.McCauley TC, Zhang HM, Bell<strong>in</strong> ME, Ax RL, 2001:Identification of a hepar<strong>in</strong>-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> <strong>in</strong> bov<strong>in</strong>e sem<strong>in</strong>alfluid as tissue <strong>in</strong>hibitor of metalloprote<strong>in</strong>ases-2. Mol ReprodDev 58, 336–341.Miller D, Ostermeier GC, 2006: Spermatozoal RNA: Why is itthere and what does it do? Gynecol Obstet Fertil 34, 840–846.Miller D, Ostermeier GC, Krawetz SA, 2005: The controversy,potential and roles of spermatozoal RNA. Trends Mol Med11, 156–163.Moura AA, Koc H, Chapman DA, Killian GJ, 2006:Identification of prote<strong>in</strong>s <strong>in</strong> the accessory sex gland fluidassociated with fertility <strong>in</strong>dexes of dairy bulls: a proteomicapproach. J Androl 27, 201–211.Moura AA, Chapman DA, Killian GJ, 2007: Prote<strong>in</strong>s of theaccessory sex glands associated with the oocyte-penetrat<strong>in</strong>gcapacity of cauda epididymal sperm from Holste<strong>in</strong> bulls ofdocumented fertility. Mol Reprod Dev 74, 214–222.Petrunk<strong>in</strong>a AM, Waberski D, Gunzel-Apel AR, Topfer-Petersen E, 2007: Determ<strong>in</strong>ants of sperm quality and fertility<strong>in</strong> domestic species. <strong>Reproduction</strong> 134, 3–17.Phillips NJ, Mcgowan MR, Johnston SD, Mayer DG, 2004:Relationship between thirty post-thaw spermatozoal characteristicsand the field fertility of 11 high-use Australiandairy AI sires. Anim Reprod Sci 81, 47–61.Puglisi R, Balduzzi D, Galli A, 2004: In vitro sperm penetrationspeed and its relationship with <strong>in</strong> vivo bull fertility.Reprod Dom Anim 39, 424–428.Rodriguez-Mart<strong>in</strong>ez H, 2003: Laboratory semen assessmentand prediction of fertility: still utopia? Reprod Dom Anim38, 312–318.Rodriguez-Mart<strong>in</strong>ez H, 2007: State of the art <strong>in</strong> farm animalsperm evaluation. Reprod Fertil Dev 19, 91–101.Roncoletta M, Morani Eda S, Esper CR, Barnabe VH,Francesch<strong>in</strong>i PH, 2006: Fertility-associated prote<strong>in</strong>s <strong>in</strong>Nelore bull sperm membranes. Anim Reprod Sci 91, 77–87.Saacke RG, Dalton JC, Nadir S, Nebel RL, Bame JH, 2000:Relationship of sem<strong>in</strong>al traits and <strong>in</strong>sem<strong>in</strong>ation time tofertilization rate and embryo quality. Anim Reprod Sci 2,663–677.Setchell BP, 1978: The scrotum and thermoregulation. In:Setchell BP (ed), The Mammalian Testis. Cornell UniversityPress, Ithaca, pp. 90–108.Suarez SS, 2002: Formation of a reservoir of sperm <strong>in</strong> theoviduct. Reprod Domest Anim 37, 140–143.Thundathil J, Gil J, Januskauskas A, Larsson B, So¨derquist L,Mapletoft R, Rodrıguez-Martınez H, 1999: Relationshipbetween the proportion of capacitated spermatozoa present<strong>in</strong> frozen-thawed semen and fertility with artificial <strong>in</strong>sem<strong>in</strong>ation.Int J Androl 22, 366–373.Thundathil J, Meyer R, Palasz AT, Barth AD, Mapletoft RJ,2000: Effect of the knobbed acrosome defect <strong>in</strong> bov<strong>in</strong>esperm on IVF and embryo production. Theriogenology 54,921–934.Thundathil J, Palom<strong>in</strong>o J, Barth A, Mapletoft R, Barros C,2001a: Fertiliz<strong>in</strong>g characteristics of bov<strong>in</strong>e sperm with flattenedor <strong>in</strong>dented acrosomes. Anim Reprod Sci 67, 231–243.Thundathil J, Palasz AT, Barth AD, Mapletoft RJ, 2001b: Theuse of <strong>in</strong> vitro fertilization techniques to <strong>in</strong>vestigate thefertiliz<strong>in</strong>g ability of bov<strong>in</strong>e sperm with proximal cytoplasmicdroplets. Anim Reprod Sci 65, 181–192.Thundathil JC, Anzar M, Buhr MM, 2006: Na + ⁄ K + ATPaseas a signal<strong>in</strong>g molecule dur<strong>in</strong>g bov<strong>in</strong>e sperm capacitation.Biol Reprod 75, 308–317.Truelson SL, Graham JK, Mortimer RG, Field TG, 1996: Invitro penetration <strong>in</strong>to bov<strong>in</strong>e oocytes and zona-free hamsteroocytes by bull spermatozoa treated with liposomes. J DairySci 79, 991–999.Walters AH, Eyestone WE, Saacke RG, Pearson RE, GwazdauskasFC, 2005: Bov<strong>in</strong>e embryo development after IVFwith spermatozoa hav<strong>in</strong>g abnormal morphology. Theriogenology63, 1925–1937.Walters AH, Saacke RG, Pearson RE, Gwazdauskas FC,2006: Assessment of pronuclear formation follow<strong>in</strong>g <strong>in</strong> vitrofertilization with bov<strong>in</strong>e spermatozoa obta<strong>in</strong>ed after thermal<strong>in</strong>sulation of the testis. Theriogenology 65, 1016–1028.Waterhouse KE, Haugan T, Kommisrud E, Tverdal A,Flatberg G, Farstad W, Evenson DP, De Angelis PM,2006: Sperm DNA damage is related to field fertility ofsemen from young Norwegian Red bulls. Reprod Fertil Dev18, 781–788.Watson PF, 2000: The causes of reduced fertility withcryopreserved semen. Anim Reprod Sci 60–61, 481–492.Wu TF, Chu DS, 2008: Epigenetic processes implementeddur<strong>in</strong>g spermatogenesis dist<strong>in</strong>guish the paternal pronucleus<strong>in</strong> the embryo. Reprod Biomed Onl<strong>in</strong>e 16, 13–22.Wu AT, Sutovsky P, Manandhar G, Xu W, Katayama M,Day BN, Park KW, Yi YJ, Xi YW, Prather RS, Oko R,2007: PAWP, a sperm-specific WW doma<strong>in</strong>-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>,promotes meiotic resumption and pronuclear developmentdur<strong>in</strong>g fertilization. J Biol Chem 282, 12164–12175.Yanagimachi R, 1994: Mammalian fertilization. In: Knobil E,Neill JD (eds), The Physiology of <strong>Reproduction</strong>. RavenPress, New York, pp. 189–317.Zhang BR, Larsson B, Rodriguez-Mart<strong>in</strong>ez H, 1995: Influenceof batches of bov<strong>in</strong>e oocytes on the outcome of an <strong>in</strong>tactzona pellucida b<strong>in</strong>d<strong>in</strong>g assay and <strong>in</strong> vitro fertilization. Int JAndrol 18, 213–220.Zhang BR, Larsson B, Lundeheim N, Rodriguez-Mart<strong>in</strong>ez H,1997: Relationship between embryo development <strong>in</strong> vitroand 56-day non return rates of cows <strong>in</strong>sem<strong>in</strong>ated withfrozen-thawed semen from dairy bulls. Theriogenology 48,221–231.Zhang BR, Larsson B, Lundeheim N, Håård MG, Rodriguez-Mart<strong>in</strong>ez H, 1999: Prediction of bull fertility by comb<strong>in</strong>ed <strong>in</strong>vitro assessments of frozen-thawed semen from young dairybulls enter<strong>in</strong>g an AI-programme. Int J Androl 22, 253–260.Zhao Y, Buhr MM, 1996: Localization of various ATPases <strong>in</strong>fresh and cryopreserved bov<strong>in</strong>e spermatozoa. Anim ReprodSci 44, 139–148.Author’s address (for correspondence): John Kastelic, LethbridgeResearch Centre, 5403-1st Ave South, P.O. Box 3000, Lethbridge, AB,Canada T1J 4B1. E-mail: kastelicj@agr.gc.caConflict of <strong>in</strong>terest: J Kastelic is the Co-Editor-<strong>in</strong>-Chief of the journalTheriogenology, published by Elsevier Science, Inc; J Thundthil hasreceived fund<strong>in</strong>g support from the University of Calgary; Agricultureand Food Council of Alberta and Alberta Livestock IndustryDevelopment Fund; Westgen Endowment Fund; and L’AllianceBoviteq Inc.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 374–378 (2008); doi: 10.1111/j.1439-0531.2008.01187.xISSN 0936-6768Sanitary Procedures for the Production of Extended SemenGC AlthouseDepartment of Cl<strong>in</strong>ical Studies, New Bolton Center, University of Pennsylvania, Kennett Square, PA, USAContentsSemen is collected and processed from a variety of animalspecies for use <strong>in</strong> artificial <strong>in</strong>sem<strong>in</strong>ation breed<strong>in</strong>g programmes.Because of the <strong>in</strong>herent nature of the semen collection process,bacterial contam<strong>in</strong>ation of the ejaculate is a common occurrence.Additionally, manipulation of the ejaculate dur<strong>in</strong>gprocess<strong>in</strong>g <strong>in</strong> the laboratory can expose the sample to possible<strong>in</strong>troduction of bacterial contam<strong>in</strong>ation. If preventative measuresat the stud fail to adequately control these risks,decreases <strong>in</strong> semen quality, dose longevity and fertility mayoccur. Multiple mammalian and non-mammalian sources havebeen identified as orig<strong>in</strong>s of contam<strong>in</strong>ation <strong>in</strong> the stud.Knowledge of these sources has aided the <strong>in</strong>dustries <strong>in</strong>develop<strong>in</strong>g strategies that help <strong>in</strong> controll<strong>in</strong>g the <strong>in</strong>troductionof contam<strong>in</strong>ant bacteria <strong>in</strong> extended semen. A primary step <strong>in</strong>m<strong>in</strong>imiz<strong>in</strong>g contam<strong>in</strong>ation is <strong>in</strong> the practice of good hygiene bystud personnel. Prudent general sanitation protocols shouldalso be followed <strong>in</strong> the laboratory, animal hous<strong>in</strong>g and semencollection areas. Cleanl<strong>in</strong>ess and attention to the actual semencollection process can also aid <strong>in</strong> reduc<strong>in</strong>g bacterial loadorig<strong>in</strong>at<strong>in</strong>g from the stud semen donor. Attentiveness to all ofthese steps significantly contributes to an overall reduction <strong>in</strong>the type and amount of bacterial contam<strong>in</strong>ation. However,their complete elim<strong>in</strong>ation stills rema<strong>in</strong>s unavoidable. Toaddress residual bacteria load <strong>in</strong> the sample, antimicrobialsare commonly used <strong>in</strong> semen extenders <strong>in</strong>tended to promote<strong>in</strong> vitro sperm longevity beyond that of a few hours. Currentresearch by the animal <strong>in</strong>dustries cont<strong>in</strong>ues <strong>in</strong> the selection andprudent use of antimicrobials that will lead to the successand susta<strong>in</strong>ability of this modality <strong>in</strong> controll<strong>in</strong>g bacterialcontam<strong>in</strong>ation.IntroductionMany animal <strong>in</strong>dustries have come to embrace certa<strong>in</strong>assisted reproductive technologies (ART). Of particular<strong>in</strong>terest is the technique of artificial <strong>in</strong>sem<strong>in</strong>ation (AI).Not only was it one of the earliest ART’s applied at acommercial level, but it rema<strong>in</strong>s the most common andwidespread ART used today (Althouse 2007). Bothdomestic food (i.e. beef, dairy, poultry, sw<strong>in</strong>e) andcompanion (i.e. dogs, horses) animal <strong>in</strong>dustries havereadily <strong>in</strong>corporated AI as a viable option <strong>in</strong> theirbreed<strong>in</strong>g management programmes. Underly<strong>in</strong>g thereproductive success of any of these programmes,however, is the need for and availability of a goodquality extended semen product.Driven <strong>in</strong> part by scales of efficiency and everprevalent biosecurity concerns, many animal <strong>in</strong>dustrieshave evolved to the po<strong>in</strong>t of hav<strong>in</strong>g dedicated farms (i.e.studs) which house the stud animals from which semenis collected, analysed and processed. The end result ofthis process<strong>in</strong>g is the development of semen doses, whichare then fresh-extended ⁄ chilled or frozen and subsequentlydelivered by ground or mail courier to thebreed<strong>in</strong>g farm. In those <strong>in</strong>dustries which utilizefresh-extended ⁄ chilled semen, an evolution <strong>in</strong> extendertechnology has and cont<strong>in</strong>ues to occur, which lengthensthe number of days that sperm viability can be ma<strong>in</strong>ta<strong>in</strong>ed<strong>in</strong> this liquid, chilled state. An undesirable sideeffect of this process, however, has been a concomitant<strong>in</strong>crease <strong>in</strong> the <strong>in</strong>cidence of extended semen doses whichconta<strong>in</strong> spermicidal levels of bacterial contam<strong>in</strong>ation(Althouse et al. 2000). These spermicidal effectsare direct and are bacterial concentration-dependent(Teague et al. 1971; Auroux et al. 1991; Wolff et al.1993; Monga and Roberts 1994; Diemer et al. 1996).The purpose of this paper is to present the generalconcepts and current understand<strong>in</strong>g of bacterial dynamics<strong>in</strong> extended semen, and to discuss methods thereofto reduce and control bacterial contam<strong>in</strong>ation.Bacterial Growth Dynamics <strong>in</strong> Semen ExtendersGiven the <strong>in</strong>herent nature and impact of those bacteriawhich exhibit pathogenesis, much work has beenperformed over the years <strong>in</strong>vestigat<strong>in</strong>g bacterial growthand the identification of factors which regulate suchgrowth. This available body of work <strong>in</strong> addition to morerecent works <strong>in</strong>vestigat<strong>in</strong>g bacteria growth dynamics <strong>in</strong>select semen extenders (Varner et al. 1998; Althouse andLu 2005; Althouse et al. 2008) provides us with a goodunderstand<strong>in</strong>g of bacterial behaviour under variousconditions, and of which much of the follow<strong>in</strong>g discussionis based.Most bacteria genera today exhibit the qualities ofbe<strong>in</strong>g phenomenally hardy organisms with a survivalistability to adapt to a wide variation of conditions. Withextenders used for stor<strong>in</strong>g semen, a primary purpose ofthese diluents is to provide the necessary nutrients andmilieu to prolong <strong>in</strong> vitro sperm cell viability. Unfortunately,these same attributes make for a potential mediathat contam<strong>in</strong>ant bacteria can flourish <strong>in</strong> if thesecontam<strong>in</strong>ants exhibit resistance to the <strong>in</strong>clusion antibiotics.Growth patterns of bacteria are similar acrossmany genera. A typical bacterial growth curve ispresented <strong>in</strong> Fig. 1.After <strong>in</strong>troduction of bacteria to a novel environment(e.g. semen extender), a period of negligible or laggrowth occurs. The length of time for this lag phase isdependent upon such th<strong>in</strong>gs as the genera <strong>in</strong>oculated,level of <strong>in</strong>oculums, time needed for recovery from thephysical damage or shock because of the transfer,environmental conditions, and the <strong>in</strong>nate temporalrequirements of the bacterium to synthesize essentialcomponents necessary to metabolize the available substrate(s).Once adaptation is met, a period of rapidgrowth ensues. Dur<strong>in</strong>g this log or exponential phase,cells are doubl<strong>in</strong>g on a regular basis, lead<strong>in</strong>g to aÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Sanitary Procedures for Extended Semen 375Fig. 1. Phases of bacterial growth (adapted from Banwart 1979)geometric progression <strong>in</strong> cell number. Essential componentsof this growth process may <strong>in</strong>clude an <strong>in</strong>crease <strong>in</strong>cell mass and ribosome number, duplication of thebacterial chromosome, cell wall and plasma membranesynthesis, chromosomal partition<strong>in</strong>g with septum formation,and f<strong>in</strong>ally cell division. This rapid growthphase eventually reaches a po<strong>in</strong>t at which it plateaus,transition<strong>in</strong>g <strong>in</strong>to the stationary phase. The bacterialconcentration and length of time of the stationary phasehas traditionally been thought to be controlled bynutrient exhaustion, physical space limitation, and ⁄ oraccumulation of <strong>in</strong>hibitory levels of metabolites, <strong>in</strong>clud<strong>in</strong>gend products. Recent work has also alluded to thesecretion of quorum-sens<strong>in</strong>g compounds, which allowfor <strong>in</strong>terbacterial communication <strong>in</strong> the gaug<strong>in</strong>g ofpopulation density (Carbonell et al. 2002; Gonzalez andKeshavan 2006; Fulghesu et al. 2007). If no change ismade <strong>in</strong> the present conditions, a death phase eventuallycommences, which is driven by the decl<strong>in</strong>e <strong>in</strong> the viablecell population.As with spermatozoa, bacteria are unable to regulatetheir temperature. Spermatozoa are dependent upon theenvironment to provide this critical component toprolonged viability. Likewise, this component is ofequal importance to the viability and growth of bacteria.All bacteria have an optimum temperature (T o ) – thetemperature at which maximum growth rate is atta<strong>in</strong>ed.If the bacteria are exposed to environmental temperaturesabove their T o , plasma membrane fluidity<strong>in</strong>creases, with concomitant alteration of cell functionand decreased growth. At some temperature po<strong>in</strong>t, nofurther compensation can occur, lead<strong>in</strong>g to perturbationof the cell and death. Spermatozoa are much moresensitive to <strong>in</strong>creased temperature change than manyother cell types, <strong>in</strong>clud<strong>in</strong>g bacteria. Unlike bacteria,sperm do not have <strong>in</strong>tracellular organelles to aid <strong>in</strong>modify<strong>in</strong>g plasma membrane fluidity for survivability.Consequently, exposure to a matter of a few degreesabove body temperature is all that is necessary for spermdeath to occur.As environmental temperature decreases (e.g. chill<strong>in</strong>gfresh-extended semen), cell plasma membrane fluiditychanges <strong>in</strong> conjunction with decreased growth rate andmetabolism. At some po<strong>in</strong>t, growth and metabolismcease, and the cells become dormant. This phenomenonis employed to reduce metabolism and <strong>in</strong>duce dormancyof sperm <strong>in</strong> extended semen. Although this is beneficialto sperm longevity, it can benefit contam<strong>in</strong>ant bacteriaeven more, as they have the <strong>in</strong>tracellular mach<strong>in</strong>ery toadapt to lower environmental temperatures to the po<strong>in</strong>tthat some genera may aga<strong>in</strong> thrive giv<strong>in</strong>g way to theirprofuse overgrowth <strong>in</strong> the chilled extender.From prior studies <strong>in</strong> sw<strong>in</strong>e, the majority of contam<strong>in</strong>antsidentified <strong>in</strong> extended semen are Gram-negativebacteria, with a majority of the contam<strong>in</strong>ant bacteriabelong<strong>in</strong>g to the Family Enterobacteriaceae (Althouseet al. 2000, 2008; Althouse and Lu 2005). Our laboratoryhas seen similar types of contam<strong>in</strong>ants <strong>in</strong> extendedsemen from other species as well. In these and otherstudies, bacterial contam<strong>in</strong>ant overgrowth of certa<strong>in</strong>genera has been associated with deleterious effects onsemen quality and longevity (Sone et al. 1989; Althouseet al. 2000; Akhter et al. 2008; Aurich and Spergser2007). Spermatozoal agglut<strong>in</strong>ation, decreased spermatozoalmotility and viability are potential end effects tobacteriospermic overgrowth. When such doses are used<strong>in</strong> an AI programme, <strong>in</strong>creased regular returns tooestrus, post-<strong>in</strong>sem<strong>in</strong>ation vulvar discharges andreduced herd reproductive performance have beenreported <strong>in</strong> some species (Sone et al. 1989; Althouseet al. 2000; Bukhar<strong>in</strong> et al. 2000). Depend<strong>in</strong>g uponbacterial type and species along with species orig<strong>in</strong> ofthe sperm, an actual sperm to bacteria (CFU) ratio ofapproximately 1:1 up to 100:1 <strong>in</strong> a sample appearssufficient to generate the undesired effects on the semendose and fertility (Tamuli et al. 1984; Auroux et al.1991; Althouse et al. 2000), with these effects be<strong>in</strong>g of atime-dependent nature. Hence, extenders which providethe ability to extend storage life of sperm also providethe opportunity for certa<strong>in</strong> contam<strong>in</strong>ant bacteria toreach their necessary threshold for elicit<strong>in</strong>g a negativeeffect on fertility potential.Sources and Control of Contam<strong>in</strong>ationUs<strong>in</strong>g pigs as a model, multiple contam<strong>in</strong>ation po<strong>in</strong>tshave been identified <strong>in</strong> the production of extendedsemen (Althouse et al. 2000; Althouse and Lu 2005).From this work, contam<strong>in</strong>ation sources have beenclassified <strong>in</strong>to either be<strong>in</strong>g of mammalian or nonmammalianorig<strong>in</strong>, and are usually found to orig<strong>in</strong>atefrom a s<strong>in</strong>gle source on a stud ⁄ farm. These s<strong>in</strong>glesources have been found to yield the same or differentbacterial stra<strong>in</strong>s at different times at the stud ⁄ farm.Likewise, different sources of contam<strong>in</strong>ation have beenfound at different times on the same stud ⁄ farm. Commonsources of bacterial contam<strong>in</strong>ation of extendedsemen are outl<strong>in</strong>ed <strong>in</strong> Table 1. Many of these sources,once contam<strong>in</strong>ated, act as seed<strong>in</strong>g po<strong>in</strong>ts for furtherejaculates which come <strong>in</strong>to contact with this source.From our laboratories work performed to date,a multitude of contam<strong>in</strong>ation sources appear to exist.The first step <strong>in</strong> m<strong>in</strong>imiz<strong>in</strong>g bacterial contam<strong>in</strong>ationof extended semen is the practice of good hygiene andgeneral sanitation by personnel. Personnel which come<strong>in</strong> contact with any materials which are subsequentlyused <strong>in</strong> the collection and process<strong>in</strong>g of semen, or whoÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


376 GC AlthouseTable 1. Potential sources of bacterial contam<strong>in</strong>ation of extendedsemenMammalianFaecalPreputial cavity fluidsSk<strong>in</strong> ⁄ hairRespiratory secretionsPersonnelNon-mammalianWater (e.g. tap, purified)Plant matter (i.e. feed, bedd<strong>in</strong>g)S<strong>in</strong>ks ⁄ dra<strong>in</strong>sAir ⁄ ventilation systemsInanimate objectsare <strong>in</strong>volved <strong>in</strong> the actual semen collection process, needto be aware that their person can be a source ofcontam<strong>in</strong>ation or act as a fomite <strong>in</strong> the transfer ofcontam<strong>in</strong>ation. In these situations, prudent hand wash<strong>in</strong>gand ⁄ or use of protective gloves should be encouraged.Personnel should avoid any contact of their barehands with materials which can later come <strong>in</strong>to contactwith the semen or extender. Personnel with upperrespiratory <strong>in</strong>fections should be cognizant of and avoidcontam<strong>in</strong>ation of materials, semen or extender throughaerosolization of the contam<strong>in</strong>ant dur<strong>in</strong>g sneez<strong>in</strong>g orcough<strong>in</strong>g. Caps and hair nets can be of value if worn bypersonnel perform<strong>in</strong>g the semen collection process andby laboratory personnel as an aid <strong>in</strong> m<strong>in</strong>imiz<strong>in</strong>g hairand dander as a contam<strong>in</strong>ation source. Clean protectivegarments and shoes ⁄ boots, provided on site by the stud,should be available for use by all stud personnel.Prudent general sanitation protocols should be <strong>in</strong>place <strong>in</strong> the laboratory, animal hous<strong>in</strong>g and semencollection areas. In the barn, regular removal of organicmaterial, a byproduct of normal husbandry, followed bya thorough clean<strong>in</strong>g with a broad-spectrum dis<strong>in</strong>fectantis recommended. Use of sanitizers and dis<strong>in</strong>fectantsshould be used prudently <strong>in</strong> the laboratory, but productsand protocols need to be specific to avoid residues,which may be detrimental to semen quality upon latercontact.To m<strong>in</strong>imize bacterial load orig<strong>in</strong>at<strong>in</strong>g from the studsemen donor, the ventral abdomen may need to becleaned and dried prior to commenc<strong>in</strong>g with semencollection. Trimm<strong>in</strong>g of hair surround<strong>in</strong>g the preputialorifice should occur on an as needed basis to elim<strong>in</strong>atethe accumulation of organic matter at this site and its<strong>in</strong>advertent <strong>in</strong>troduction <strong>in</strong>to the ejaculate dur<strong>in</strong>g semencollection. Clean<strong>in</strong>g of the preputial open<strong>in</strong>g andsurround<strong>in</strong>g area with a s<strong>in</strong>gle-use disposable wipeshould be considered if the area is wet and ⁄ or hasorganic material present. In some species, preputialfluids can conta<strong>in</strong> high numbers of bacteria (Aamdalet al. 1958); therefore, these fluids should be evacuatedimmediately prior to the semen collection process. Whencollect<strong>in</strong>g semen us<strong>in</strong>g an artificial vag<strong>in</strong>a or glovedhand, the collector should position the penis <strong>in</strong> a mannerconsistent with m<strong>in</strong>imiz<strong>in</strong>g gravitational contam<strong>in</strong>ationof the semen collection vessel with preputial fluids.Lastly, if perform<strong>in</strong>g gloved hand semen collection,divert<strong>in</strong>g the pre-sperm fraction from the semen collectionvessel may help <strong>in</strong> reduc<strong>in</strong>g ejaculate bacterial load.A synopsis outl<strong>in</strong><strong>in</strong>g general stud hygiene and sanitationprocedures is provided <strong>in</strong> Table 2.Table 2. General stud hygiene and sanitation recommendationsStud personnel1. Application of good hand hygiene, <strong>in</strong>clud<strong>in</strong>g appropriate wash<strong>in</strong>g and use of protective gloves, should be practiced throughout all areas of the stud2. Personnel should avoid any contact of bare hands with materials which can later come <strong>in</strong>to contact with the semen or extender3. Personnel with upper respiratory <strong>in</strong>fections should be cognizant of and avoid contam<strong>in</strong>ation of materials, semen or extender through aerosolization dur<strong>in</strong>gsneez<strong>in</strong>g or cough<strong>in</strong>g4. Caps and hair nets can be of value if worn by personnel perform<strong>in</strong>g the semen collection process and by laboratory personnel as an aid <strong>in</strong> m<strong>in</strong>imiz<strong>in</strong>g hairand dander as a contam<strong>in</strong>ation source5. Clean protective garments and shoes ⁄ boots, provided on site by the stud, should be available for use by all stud personnelAnimal hous<strong>in</strong>g ⁄ handl<strong>in</strong>g1. Animal hous<strong>in</strong>g should be put on a regular sanitary ma<strong>in</strong>tenance schedule, <strong>in</strong>clud<strong>in</strong>g removal of organic material and application of a broad-spectrumdis<strong>in</strong>fectant2. Trimm<strong>in</strong>g of hair surround<strong>in</strong>g the preputial orifice should occur on an as needed basis to elim<strong>in</strong>ate the accumulation of organic matter at this site and its<strong>in</strong>advertent <strong>in</strong>troduction <strong>in</strong>to the ejaculate dur<strong>in</strong>g semen collection3. The ventral abdomen should be clean and dry prior to commenc<strong>in</strong>g with semen collection4. Clean<strong>in</strong>g of the preputial open<strong>in</strong>g and surround<strong>in</strong>g area with a s<strong>in</strong>gle-use disposable wipe should be considered if the area is wet and ⁄ or has organic materialpresent5. In some species, preputial fluids can conta<strong>in</strong> high numbers of bacteria, therefore, these fluids should be evacuated immediately prior to the semen collectionprocess6. When collect<strong>in</strong>g semen us<strong>in</strong>g an artificial vag<strong>in</strong>a or gloved hand, the collector should position the penis <strong>in</strong> such a way as to m<strong>in</strong>imize gravitationalcontam<strong>in</strong>ation of the semen collection vessel with preputial fluids7. If perform<strong>in</strong>g gloved hand semen collection, divert<strong>in</strong>g the pre-sperm fraction from the semen collection vessel may aid <strong>in</strong> reduc<strong>in</strong>g ejaculate bacterial load8. The semen collection area and any collection equipment should be thoroughly cleaned and dis<strong>in</strong>fected at the end of each collection dayLaboratory1. Encourage s<strong>in</strong>gle-use disposable products when economically feasible to m<strong>in</strong>imize cross-contam<strong>in</strong>ation2. When us<strong>in</strong>g reusable laboratory materials (i.e. glassware, plasticware, plastic tub<strong>in</strong>g, conta<strong>in</strong>ers, etc.) which cannot be heat ⁄ gas sterilized or boiled, cleanthese reusable’s <strong>in</strong>itially us<strong>in</strong>g a laboratory-grade detergent (residue-free) with water, followed by a distilled water r<strong>in</strong>se, and lastly through a 70% alcohol(non-denatured) r<strong>in</strong>se. Allow sufficient time and proper ventilation for complete evaporation of residual alcohol on the reusable. R<strong>in</strong>se reusable with semenextender prior to their first use of the day3. Laboratory purified water should be checked on a m<strong>in</strong>imum quarterly basis if <strong>in</strong>-house, and by lot if outsourced. Any bacterial growth should be consideredsignificant and appropriate action taken to identify and elim<strong>in</strong>ate the contam<strong>in</strong>ant source4. Dis<strong>in</strong>fect countertops and contam<strong>in</strong>ated lab equipment at end of process<strong>in</strong>g day with a residue-free detergent and r<strong>in</strong>se5. Floor should be mopped at end of day with a dis<strong>in</strong>fectant6. Break down bulk products <strong>in</strong>to smaller, daily use quantities immediately after open<strong>in</strong>g7. Ultraviolet light<strong>in</strong>g can be <strong>in</strong>stalled to aid <strong>in</strong> sanitiz<strong>in</strong>g reusable’s and laboratory surfaces; however, safety precautions should be <strong>in</strong>tegrated to preventexposure to personnelÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Sanitary Procedures for Extended Semen 377Because of the level of frequency <strong>in</strong> which watersources have been identified as a source of contam<strong>in</strong>ation,regular monitor<strong>in</strong>g of these sources is an importantcomponent <strong>in</strong> a control programme. Well or municipalwater should be checked on a m<strong>in</strong>imum quarterly basisfor coliform load, which should meet or exceed localpublic health ord<strong>in</strong>ances. Purified water systems shouldbe checked on a m<strong>in</strong>imum quarterly basis; any bacterialgrowth should be considered significant and appropriateaction taken to elim<strong>in</strong>ate the contam<strong>in</strong>ant from thesystem. Facilities us<strong>in</strong>g purchased purified water shouldhave each lot checked for bacterial growth; once aga<strong>in</strong>,any bacterial growth is significant, and another watersource should be considered.F<strong>in</strong>al quality assurance for bacterial control should beascerta<strong>in</strong>ed by regular monitor<strong>in</strong>g of the extended semensamples. Depend<strong>in</strong>g upon the desired level of qualitycontrol, whole system or po<strong>in</strong>t dose screen<strong>in</strong>g formicrobial contam<strong>in</strong>ation can be performed on randomlyselected <strong>in</strong>dividual or pooled semen which are at least48 h of age post-process<strong>in</strong>g (Althouse et al. 2003). Ourown preference is to perform this monitor<strong>in</strong>g on theextended semen product on its day of expiration. As ageneral rule, recommended number of doses screenedper month should be 1% of total monthly collections orfour samples ⁄ week, whichever is greater (Althouse et al.2003). Any positive contam<strong>in</strong>ant growth should becritically assessed and a follow-up performed.In Vitro Control of Contam<strong>in</strong>ation <strong>in</strong> ExtendedSemenBecause of the unavoidable presence of bacteria <strong>in</strong> theejaculate, even under the best hygiene and sanitaryconditions, antimicrobials are a necessary component ofsemen extenders which are <strong>in</strong>tended to promote <strong>in</strong> vitrolongevity of sperm beyond that of a few hours. Whenlook<strong>in</strong>g across our common domestic animal species, themost common classes of antimicrobials used today <strong>in</strong>semen extenders are the am<strong>in</strong>oglycosides, am<strong>in</strong>ocyclitols,b-lactams, l<strong>in</strong>cosamides, macrolides, polypeptides andqu<strong>in</strong>olones (Table 3). In general, these antimicrobialsTable 3. Common antimicrobials currently used <strong>in</strong> semen extendersBov<strong>in</strong>e Equ<strong>in</strong>e Porc<strong>in</strong>e PoultryAm<strong>in</strong>ocyclitolsSpect<strong>in</strong>omyc<strong>in</strong> d dAm<strong>in</strong>oglycosidesAmakac<strong>in</strong>dGentamic<strong>in</strong> d d d dNeomyc<strong>in</strong>dStreptomyc<strong>in</strong> d d dBeta-lactamsAmoxicill<strong>in</strong>dPenicill<strong>in</strong> d d dTicarcill<strong>in</strong>dL<strong>in</strong>cosamidesL<strong>in</strong>comyc<strong>in</strong> d dMacrolidesTylos<strong>in</strong> d d dPolypeptidesPolymix<strong>in</strong> d dQu<strong>in</strong>olonesEnrofloxac<strong>in</strong> d dwork on susceptible bacteria by block<strong>in</strong>g or <strong>in</strong>terfer<strong>in</strong>gwith a biochemical reaction essential to the life cycle.Several thresholds must be obta<strong>in</strong>ed for antimicrobialsto work <strong>in</strong> bacterially contam<strong>in</strong>ated extended semen<strong>in</strong> vitro. The selected antimicrobial(s) must be at sufficientconcentrations to provide adequate readily-availableactive product, the compound must be <strong>in</strong> a state tobe able to permeate (active and ⁄ or passive mechanisms)the bacteria <strong>in</strong> sufficient quantities, and the compoundmust occupy a sufficient number of active sites over aperiod of time with<strong>in</strong> the bacteria to elicit their detrimentaleffect. Interference <strong>in</strong> this process at any po<strong>in</strong>t canlead to antimicrobial resistance of the bacteria (forreview, see Althouse and Lu 2005). Recent work hasbegun to elucidate the thermotemporal dynamics ofcontam<strong>in</strong>ant bacteria and antimicrobials <strong>in</strong> semenextenders (Althouse et al. 2008). Through this typework, it is hoped that prudent antimicrobial selectionand use by the <strong>in</strong>dustries will lead to the success andsusta<strong>in</strong>ability of this modality <strong>in</strong> our efforts to controlcontam<strong>in</strong>ation.ConclusionsThe current body of research substantiates the risk thatexists of contam<strong>in</strong>at<strong>in</strong>g semen with bacteria dur<strong>in</strong>g thecollection and process<strong>in</strong>g of an ejaculate. If preventativemeasures at the stud fail to adequately control this risk,decreases <strong>in</strong> semen quality, dose longevity and fertilitymay occur. To date, multiple mammalian and nonmammaliansources have been identified as orig<strong>in</strong>s ofthis contam<strong>in</strong>ation at a stud. In addition to generalcleanl<strong>in</strong>ess, targeted hygiene and sanitation procedurescan greatly aid <strong>in</strong> controll<strong>in</strong>g the <strong>in</strong>troduction ofcontam<strong>in</strong>ant bacteria <strong>in</strong> the extended semen product.Even with these measures <strong>in</strong> place to reduce <strong>in</strong>com<strong>in</strong>gbacterial load, it is unrealistic to expect that an extendedsemen product will be devoid of bacterial contam<strong>in</strong>ation.Hence, prudent use of antimicrobials <strong>in</strong> the semendiluents’ is an essential component to the overall controlof contam<strong>in</strong>ant bacteria <strong>in</strong> extended semen used<strong>in</strong> programmes <strong>in</strong>corporat<strong>in</strong>g assisted reproductivetechnologies.ReferencesAamdal J, Hogset I, Filseth O, 1958: Extirpation of thepreputial diverticulum of boars used for artificial <strong>in</strong>sem<strong>in</strong>ation.J Am Vet Med Assoc 132, 522–524.Akhter S, Ansari MS, Andrabi SM, Ullah N, Qayyum M,2008: Effects of antibiotics <strong>in</strong> extender on bacterial andspermatozoal quality of cooled buffalo (Bubalus bubalis)bull semen. Reprod Domest Anim 43, 272–278.Althouse GC, 2007: Artificial <strong>in</strong>sem<strong>in</strong>ation. In: Schatten H,Constant<strong>in</strong>escu GM (eds), Comparative Reproductive Biology.Blackwell Publish<strong>in</strong>g, Ames, IA, USA, pp. 159–169.Althouse GC, Lu KG, 2005: Bacteriospermia <strong>in</strong> extendedporc<strong>in</strong>e semen. Theriogenology 63, 573–584.Althouse GC, Kuster CE, Clark SG, Weisiger RM, 2000:Field <strong>in</strong>vestigations of bacterial contam<strong>in</strong>ants and theireffects on extended porc<strong>in</strong>e semen. Theriogenology 53,1167–1176.Althouse GC, Reicks D, Spronk GD, Trayer TP, 2003: Health,hygiene, and sanitation guidel<strong>in</strong>es for boar studs provid<strong>in</strong>gÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


378 GC Althousesemen to the domestic market. J Sw<strong>in</strong>e Health Prod 11, 204–206.Althouse GC, Pierdon MS, Lu KG, 2008: Thermotemporaldynamics of contam<strong>in</strong>ant bacteria and antimicrobials <strong>in</strong>extended porc<strong>in</strong>e semen. Theriogenology, <strong>in</strong> press.Aurich C, Spergser J, 2007: Influence of bacteria and gentamic<strong>in</strong>on cold-stored stallion spermatozoa. Theriogenology67, 912–918.Auroux MR, Jacques L, Mathieu D, Auer J, 1991: Is thesperm bacterial ratio a determ<strong>in</strong><strong>in</strong>g factor <strong>in</strong> impairment ofsperm motility: an <strong>in</strong>-vitro study <strong>in</strong> man with Escherichiacoli. Int J Androl 14, 264–270.Banwart GJ, 1979: Basic Food Microbiology. AVI Publish<strong>in</strong>gCo., Westport, i pp.Bukhar<strong>in</strong> OV, Kuz’m<strong>in</strong> MD, Ivanov IuB, 2000: [The role ofthe microbial factor <strong>in</strong> the pathogenesis of male <strong>in</strong>fertility].Zh Mikrobiol Epidemiol Immunobiol 2, 106–110.Carbonell X, Chorchero JL, Cubarsi R, Vila P, Villaverde A,2002: Control of Escherichia coli growth rate through celldensity. Microbiol Res 157, 257–265.Diemer T, Weidner W, Michelmann HW, Schiefer HG, RovanE, Mayer F, 1996: Influence of Escherichia coli on motilityparameters of human spermatozoa <strong>in</strong> vitro. Int J Androl 19,271–277.Fulghesu L, Giallorenzo C, Savoia D, 2007: Evaluation ofdifferent compounds as quorum sens<strong>in</strong>g <strong>in</strong>hibitors <strong>in</strong> Pseudomonasaerug<strong>in</strong>osa. J Chemother 19, 388–391.Gonzalez JE, Keshavan ND, 2006: Mess<strong>in</strong>g with bacterialquorum sens<strong>in</strong>g. Microbiol Mol Biol Rev 70, 859–875.Monga M, Roberts JA, 1994: Sperm agglut<strong>in</strong>ation by bacteria:receptor-specific <strong>in</strong>teractions. J Androl 15, 151–156.Sone M, Kawarasaki T, Ogasa A, Nakahara T, 1989: Effectsof bacteria-contam<strong>in</strong>ated boar semen on the reproductiveperformance. Jpn J Anim Reprod 35, 159–164.Tamuli MK, Sharma DK, Rajkonwar CK, 1984: Studies onthe microbial flora of boar semen. Indian Vet J 61, 858–861.Teague NS, Bogarsky S, Glenn JF, 1971: Interference ofhuman spermatozoa motility by Escherichia coli. Fertil Steril22, 281–285.Varner DD, Scanlan CM, Thompson JA, Brumbaugh GW,Blanchard TL, Carlton CM, Johnson L, 1998: Bacteriologyof preserved stallion semen and antibiotics <strong>in</strong> semenextenders. Theriogenology 50, 559–573.Wolff H, Panhans A, Stolz W, Meurer M, 1993: Adherence ofEscherichia coli to sperm: a mannose mediated phenomenonlead<strong>in</strong>g to agglut<strong>in</strong>ation of sperm and E. coli. Fertil Steril 60,154–158.Author’s address (for correspondence): GC Althouse, Department ofCl<strong>in</strong>ical Studies, New Bolton Center, University of Pennsylvania, 382West Street Road, Kennett Square, Pennsylvania 19348-1692, USA.E-mail: gca@vet.upenn.eduConflict of <strong>in</strong>terest: The author declares no conflict of <strong>in</strong>terest.Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 379–385 (2008); doi: 10.1111/j.1439-0531.2008.01188.xISSN 0936-6768Management of Goat <strong>Reproduction</strong> and Insem<strong>in</strong>ation for Genetic Improvement <strong>in</strong>FranceB Leboeuf 1 , JA Delgadillo 2 , E Manfredi 3 , A Piace` re 4 , V Cle´ment 4 , P. Mart<strong>in</strong> 5 , M Pellicer 6 , P Boue´5 and R de Cremoux 41 INRA-UEICP, Venours, Rouille´, France; 2 CIRCA, Universidad Autonoma Agraria Antonio Narro, Torreon, Coahuila, Mexico; 3 INRA SAGA;4 Institut de l’Elevage, Castanet-Tolosan cedex; 5 Caprige`ne-France, Agropole, Mignaloux-Beauvoir; 6 Physiologie de la <strong>Reproduction</strong> et desComportements, UMR 6175 INRA-CNRS-Universite´ de Tours-Haras Nationaux, Nouzilly, FranceContentsReproductive seasonality observed <strong>in</strong> all breeds of goatsorig<strong>in</strong>at<strong>in</strong>g from temperate latitudes and <strong>in</strong> some breeds fromsubtropical latitudes can now be controlled by artificialchanges <strong>in</strong> photoperiod. Short days stimulate sexual activity,while long days <strong>in</strong>hibit it. This knowledge has allowed thedevelopment of photoperiodic treatments to control sexualactivity <strong>in</strong> goats, for both the buck and doe. In the French<strong>in</strong>tensive milk production system, goat AI plays an importantrole to control reproduction and, <strong>in</strong> conjunction with progenytest<strong>in</strong>g, to improve milk production. Most dairy goats are<strong>in</strong>sem<strong>in</strong>ated out of the breed<strong>in</strong>g season with deep frozensemen, after <strong>in</strong>duction of oestrus and ovulation by hormonaltreatments. This protocol provides a kidd<strong>in</strong>g rate of approximately65%. New breed<strong>in</strong>g strategies have been developed,based on the buck effect associated with AI, to reduce the useof hormones. With the development of <strong>in</strong>sem<strong>in</strong>ation withfrozen semen, a classical selection programme was set up,<strong>in</strong>clud<strong>in</strong>g planned mat<strong>in</strong>g, progeny test<strong>in</strong>g and the diffusion ofproved sires by <strong>in</strong>sem<strong>in</strong>ations <strong>in</strong> herds. Functional traits havebecome important for efficient breed<strong>in</strong>g schemes <strong>in</strong> the dairygoat <strong>in</strong>dustries. Based on knowledge ga<strong>in</strong>ed over the pastdecade, the emphasis <strong>in</strong> selective breed<strong>in</strong>g has been placed onfunctional traits related to udder morphology and health. Neww<strong>in</strong>dows have been opened based on new molecular tools,allow<strong>in</strong>g the detection and mapp<strong>in</strong>g of genes of economicimportance.IntroductionGoats and sheep exhibit seasonal changes <strong>in</strong> reproductiveactivity, accompanied by variations <strong>in</strong> the availabilityof products over the year. Out-of-breed<strong>in</strong>g-seasonreproduction allows milk and meat production throughoutthe year <strong>in</strong> accordance with commercial requirementsand consumer expectations. Artificial<strong>in</strong>sem<strong>in</strong>ation has an important role <strong>in</strong> goat breed<strong>in</strong>g,especially <strong>in</strong> <strong>in</strong>tensive systems of production, to controlreproduction and, <strong>in</strong> conjunction with accurate progenytest<strong>in</strong>g, to improve the production of milk, hair andmeat. At the farm level, the control of reproductionallows kidd<strong>in</strong>g at a precise season of the year,a synchronization of kidd<strong>in</strong>g over a limited period oftime, and facilitates supplementary feed<strong>in</strong>g to meet thedemands of lactation. Most goat milk is processed <strong>in</strong>tohigh-quality cheese. As a result of the evolution ofdemand by consumers for typically regional cheese andfor more quality and safety, dairy goats have to beimproved for production (milk yield) and milk composition(fat and prote<strong>in</strong>s) to rema<strong>in</strong> competitive. Newfunctional traits must also be considered so as to reduceproduction costs and <strong>in</strong>crease product quality and safety.Reproductive Seasonality and Control of theTim<strong>in</strong>g of <strong>Reproduction</strong>Seasonal variations <strong>in</strong> female and male sexual activityA reproductive seasonality is observed <strong>in</strong> most breeds ofgoat orig<strong>in</strong>at<strong>in</strong>g from high latitudes (>35°) and <strong>in</strong> somelocal breeds from subtropical latitudes (25°–35°). In theAlp<strong>in</strong>e breed, as <strong>in</strong> the local does from subtropicalMexico, the breed<strong>in</strong>g season beg<strong>in</strong>s <strong>in</strong> the early autumnand ends <strong>in</strong> the late w<strong>in</strong>ter (Chem<strong>in</strong>eau et al. 1992;Delgadillo et al. 2004a). Males of these breeds alsodisplay wide changes <strong>in</strong> their sexual activity. In Alp<strong>in</strong>ebucks, the breed<strong>in</strong>g season beg<strong>in</strong>s <strong>in</strong> the early autumnand ends <strong>in</strong> late w<strong>in</strong>ter, while <strong>in</strong> local bucks fromsubtropical Mexico, it beg<strong>in</strong>s <strong>in</strong> the late spr<strong>in</strong>g and ends<strong>in</strong> the early w<strong>in</strong>ter (Delgadillo et al. 1991, 1999). In theseseasonal breeds, sexual behaviour, testicular size, an<strong>in</strong>dex of the spermatogenetic activity, and the quantitativeand qualitative sperm production of bucks decreasesdramatically dur<strong>in</strong>g the non-breed<strong>in</strong>g season (Delgadilloet al. 1991, 1999).Treatments to control the tim<strong>in</strong>g of reproductionPhotoperiodic control of reproductionThe annual reproductive rhythm <strong>in</strong> breeds of goats andsheep from temperate latitudes and <strong>in</strong> some local breedsfrom the subtropics is controlled by photoperiod(Delgadillo et al. 2004b; Chem<strong>in</strong>eau et al. 2006). Inartificial conditions, plasma testosterone levels <strong>in</strong>creasedur<strong>in</strong>g short days and decrease dur<strong>in</strong>g long days(Delgadillo et al. 2004b). In females, ovulations startdur<strong>in</strong>g short days and end dur<strong>in</strong>g long days (Gebbieet al. 1999). In order to manipulate sexual activity, theanimals must perceive alternations between long andshort days to prevent the establishment of sexualrefractor<strong>in</strong>ess.Control of male sexual activity by abolition of seasonalvariations or <strong>in</strong>duction of sexual activity <strong>in</strong> thenon-breed<strong>in</strong>g seasonIn Alp<strong>in</strong>e and Saanen male goats subjected to alternationsof 1 or 2 months of long days (16 h of light ⁄ day:LD) and 1 or 2 months of short days (8 h of light ⁄ day:SD), marked seasonality of sexual behaviour, testis sizeand quantitative and qualitative sperm production wereabolished or reduced (Delgadillo et al. 1991, 1992).When bucks were collected twice a week dur<strong>in</strong>g twoconsecutive years, these light-treated males producedÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


380 B Leboeuf, JA Delgadillo, E Manfredi, A Piace` re, V Cle´ment, P Mart<strong>in</strong>, M Pellicer, P Boue´, and R de Cremoux50% more AI doses than a control group under naturallight (Delgadillo et al. 1991). Fertility of deep-frozensemen was <strong>in</strong> the same range as that obta<strong>in</strong>ed <strong>in</strong> thecontrol animals dur<strong>in</strong>g the natural sexual season. Theseresults were improved us<strong>in</strong>g alternations of 1.5 monthsof LD and 1.5 months of SD (Leboeuf et al. 2004a).This photoperiodic treatment is now used <strong>in</strong> the Frenchnational selection programme.In male goats, sexual activity can be stimulated dur<strong>in</strong>gthe non-breed<strong>in</strong>g season us<strong>in</strong>g artificial long daysfollowed by melaton<strong>in</strong> or natural photoperiod(Delgadillo et al. 2004a; Pellicer-Rubio et al. 2007). Insubtropical Mexican male goats, 2.5 months of longdays (16 h of light ⁄ day) from 1st of November followedby natural photoperiod, <strong>in</strong>creased testosterone secretionfrom February to April, dur<strong>in</strong>g the non-breed<strong>in</strong>gseason. These long-day treated animals displayed an<strong>in</strong>tense sexual behaviour when exposed to anoestrusdoes. In the control groups, testosterone secretion andsexual behaviour were lower than <strong>in</strong> the animals treatedwith artificial photoperiod (Delgadillo et al. 2004a).Control of female sexual activity <strong>in</strong> the non-breed<strong>in</strong>gseason by the male effectThe so-called male effect is a technique to stimulate thesexual activity <strong>in</strong> seasonally anovulatory goats (Pellicer-Rubio et al. 2007). Most female goats have a shortovarian cycle of 5–7 days, followed by a second ovulationassociated with oestrous behaviour and a normalluteal phase (Chem<strong>in</strong>eau et al. 2006). The response ofdoes to the male effect can be improved by the use ofmales rendered sexually active dur<strong>in</strong>g the non-breed<strong>in</strong>gseason by photoperiodic treatments. In subtropicalMexican goats, males treated with 2.5 months of longdays were very effective at <strong>in</strong>duc<strong>in</strong>g fertile ovulatoryactivity <strong>in</strong> anovulatory goats kept under natural photoperiodbut not untreated bucks (80% vs 10%, respectively;Delgadillo et al. 2004a). In more seasonal breeds(Alp<strong>in</strong>e and Saanen), the treatment of females withartificial photoperiod is also necessary to improve theresponse to the male effect. Under these conditions,most does exposed to males ovulated (99%) and 81%kidded (Pellicer-Rubio et al. 2007).Production and Storage of Semen for ArtificialInsem<strong>in</strong>ationManagement of semen production and storageThe major problem for goat semen preservation is thebulbo-urethral gland secretion (BUS) responsible fordeterioration of spermatozoa diluted <strong>in</strong> skimmed milkor <strong>in</strong> media conta<strong>in</strong><strong>in</strong>g egg yolk when stored at 37°C ordeep frozen. The component from BUS responsible forthis effect was identified as a 55- to 60-kDa glycoprote<strong>in</strong>lipase (Busgp60) belong<strong>in</strong>g to the pancreatic lipaserelatedprote<strong>in</strong> 2 family (GoPLRP2; Sias et al. 2005).This enzyme was able to hydrolyse both triole<strong>in</strong> andmilk triglycerides <strong>in</strong>to free fatty acids, which strongly<strong>in</strong>hibit the motility and damage the membranes of buckspermatozoa (Pellicer-Rubio and Combarnous 1998).Techniques of production and storage of semen basedon cryopreservation have been proposed by Corteel(1974). After separation of sem<strong>in</strong>al plasma from spermcells to avoid deterioration of spermatozoa, sperm cellsare diluted <strong>in</strong> a skimmed milk-based glucose (0.5 M) andglycerol 7%; Corteel 1974). Semen is then stored <strong>in</strong>0.2 ml straws conta<strong>in</strong><strong>in</strong>g 1 · 10 8 sperm cells and deepfrozen progressively <strong>in</strong> three steps <strong>in</strong>to liquid nitrogen()196°C).For liquid preservation, goat semen is usually stockedat 4°C. Many extenders were tested for liquid preservationbut the most efficient one is a skimmed milk-basedmedia (Dauzier 1966), or Native Phosphocase<strong>in</strong>ate(PPCN) (Leboeuf et al. 2004b). However, sperm fertilityafter AI is reta<strong>in</strong>ed for only 12–24 h and decreasesthereafter with the duration of liquid preservation.Furthermore, there was no significant difference offertility after AI between semen ‘washed’ before dilutionand semen ‘unwashed’. So, it seems that the deleteriouseffect of sem<strong>in</strong>al lipase is m<strong>in</strong>imized at 4°C whencompared with its effect at 37°C or after freez<strong>in</strong>g ⁄ thaw<strong>in</strong>g.Use of stored semen by artificial <strong>in</strong>sem<strong>in</strong>ationInsem<strong>in</strong>ation of cycl<strong>in</strong>g goats <strong>in</strong> the breed<strong>in</strong>g season afternatural oestrusDifferent practical methods of oestrous detection areused, <strong>in</strong>clud<strong>in</strong>g entire males fitted with an apron,vasectomized males or androgenized castrated malesor females fitted with a harness and mark<strong>in</strong>g crayon.Fertility of goats <strong>in</strong>sem<strong>in</strong>ated once <strong>in</strong> the 24 h follow<strong>in</strong>gthe beg<strong>in</strong>n<strong>in</strong>g of oestrus with deep-frozen-thawed spermatozoaranges from 60% to 65% (Corteel 1977).Insem<strong>in</strong>ation after synchronized and <strong>in</strong>duced oestrus byhormonal treatmentIn France, the synchronization treatment most commonlyused for dairy goats consists of the <strong>in</strong>sertion of avag<strong>in</strong>al sponge impregnated with a synthetic analogue ofprogesterone (45 mg fluorogestone acetate) for 11 days,together with an <strong>in</strong>tramuscular <strong>in</strong>jection of pregnantmore serum gonadotroph<strong>in</strong> (PMSG) (400–600 IUaccord<strong>in</strong>g to the milk production and season of treatment)and 50 lg of cloprostenol, both adm<strong>in</strong>istered 48 hbefore sponge removal. Treated goats are <strong>in</strong>sem<strong>in</strong>atedonce with a frozen-thawed dose of 1 · 10 8 spermatozoa,at 43–45 h after sponge removal (Corteel et al. 1988). Thefertility rate obta<strong>in</strong>ed <strong>in</strong> dairy goats given one <strong>in</strong>sem<strong>in</strong>ationis 60–65% (Boué and Sigwald 2001).Insem<strong>in</strong>ation after male effect <strong>in</strong> anoestrusThe new societal trends and European legislation, whichare opposed to the use of hormones and syntheticsubstances <strong>in</strong> the animal <strong>in</strong>dustries, have encouragedproducers to adopt practices that m<strong>in</strong>imize or completelyavoid the use of synthetic chemicals andhormone treatments (Mart<strong>in</strong> et al. 2004). In this regard,the male effect may be an efficient non-pharmacologicalalternative to hormones for <strong>in</strong>duc<strong>in</strong>g and synchroniz<strong>in</strong>goestrus <strong>in</strong> AI programmes. Artificial <strong>in</strong>sem<strong>in</strong>ation protocols<strong>in</strong>clud<strong>in</strong>g one or two <strong>in</strong>sem<strong>in</strong>ations over a 24-hperiod determ<strong>in</strong>ed by the occurrence of oestrus or by the<strong>in</strong>troduction of the buck have been validated. In theÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Management of Goat <strong>Reproduction</strong> and Insem<strong>in</strong>ation 381middle of seasonal anoestrus (May), a high level offertility (71–78% of kidd<strong>in</strong>g) was obta<strong>in</strong>ed us<strong>in</strong>g frozensemen on goats subjected to treatment with artificiallong days and progestagen followed by one AI 52 hafter <strong>in</strong>troduction of the buck equipped with an apron(Pellicer-Rubio et al. 2008).Genetic Variability of Selected TraitsProduction traitsGenetic parametersGenetic parameters of milk production traits [milk, fatand prote<strong>in</strong> yields (PYs), fat and PCs] have beenestimated for Alp<strong>in</strong>e and Sannen goats from differentcountries (Iloeje et al. 1981; Boichard et al. 1989;Belichon et al. 1998; Muller et al. 2002) but also onother breeds (Mavrogenis et al. 1984; Analla et al.1996). The same trends have been found for theseestimates: for yields, reported values of heritability varyfrom 0.30 to 0.40. Heritability of contents is higher,from 0.50 to 0.60, but with a genetic coefficient ofvariation lower than that for yields. The correlationbetween fat and PC is approximately 0.50 and thesetraits are <strong>in</strong> opposition with milk yield, <strong>in</strong> particular PC.A strong positive correlation (approximately 0.80–0.90)is usually found between milk, fat and PYs.Case<strong>in</strong> polymorphismCompared with the sheep and cow, the types of case<strong>in</strong> <strong>in</strong>goat milk are extremely variable, each hav<strong>in</strong>g severalvariants, and case<strong>in</strong> polymorphisms are associated withthe rate of case<strong>in</strong> synthesis. Variants can be classifiedaccord<strong>in</strong>g to their synthesis rate of as1-cas (Grosclaudeand Mart<strong>in</strong> 1997): A, B and C (strong alleles associatedwith high synthesis rates), E (<strong>in</strong>termediate rate), G andF (weak rates) and O (absence or only traces of as1-cas).Mahé et al. (1994) and Barbieri et al. (1995) found thatthe difference between extreme genotypes (AA and FF)for PC is approximately 4.5 g ⁄ kg which representsapproximately three genetic standard deviations, asestimated under the classical polygenic model for fielddata (r = 1.7 g ⁄ kg; Belichon et al. 1998).Allele frequencies at the as1-cas locus vary accord<strong>in</strong>gto breed (Jordana et al. 1995). Variation also existswith<strong>in</strong>-breed accord<strong>in</strong>g to the subpopulations sampled.Manfredi et al. (1998) showed, for the Alp<strong>in</strong>e breedwhich is selected for PY and PC, that the frequency ofstrong alleles <strong>in</strong> the dams-of-AI bucks subpopulationwas almost twice the correspond<strong>in</strong>g frequency <strong>in</strong> theunselected female subpopulation (0.46 vs 0.25). Therefore,allele frequencies at the as1-cas locus changed overtime <strong>in</strong> this population under selection pressure.Functional traitsType appraisalApart from <strong>in</strong>fluenc<strong>in</strong>g longevity by reduc<strong>in</strong>g thefrequency of cull<strong>in</strong>g for type traits, selection onmorphology, <strong>in</strong> particular udder morphology, willfacilitate the labour of the breeder by reduc<strong>in</strong>g milk<strong>in</strong>gtime. Another advantage is the benefit to udder health,for example a reduction of cl<strong>in</strong>ical and subcl<strong>in</strong>icalmastitis. Genetic variability has been found for morphologytraits (Luo et al. 1997; Piace` re et al. 1998;Manfredi et al. 2001; Cle´ment et al. 2006). Heritabilityestimates for traits related to the udder vary from 0.20 to0.35. Genetic correlations with dairy traits can be lowfor some traits, whereas others, <strong>in</strong> particular udder floor(UF) morphology, show a significant genetic oppositionwith milk, fat and PYs.Resistance to diseaseUdder health, namely cl<strong>in</strong>ical and subcl<strong>in</strong>ical mastitis,<strong>in</strong>fluences animal welfare and the quantity and quality ofmilk. Bacterial <strong>in</strong>fections <strong>in</strong> goats are measured by directbacterial count<strong>in</strong>g or by <strong>in</strong>direct measurements such asthe California mastitis test (CMT) and somatic cell counts(SCC). The reasons for the high SCC usually found <strong>in</strong>goat milk are unclear. Current research is focused on theadequate <strong>in</strong>terpretation of SCC (Sierra et al. 1998) andthe def<strong>in</strong>ition of SCC thresholds for detection of mammary<strong>in</strong>fections <strong>in</strong> goats (Baudry et al. 1998). Geneticparameters for SCC have been estimated (Rupp et al.2004). The heritability was approximately 0.20 and thegenetic correlation with milk production close to 0.Besides mastitis, other diseases have an impact onproduction costs, quality of products and animalwelfare. The capr<strong>in</strong>e arthritis-encephalitis virus (CAEV)provokes arthritis but it may also affect the mammarygland and aggravate bacterial mastitis. Susceptibility toCAEV <strong>in</strong>fection might be associated to polymorphismsat the MHC (Ruff and Lazary 1988; Amills et al. 1999).Among goat diseases, scrapie also has been considered.The <strong>in</strong>cubation period of this lethal spongiform encephalopathyis associated with a polymorphism of the PrPgene (Goldmann et al. 1996). As far as resistance toparasitism is concerned, genetic variability also existsand moderate heritabilities are exhibited (Baker et al.1999; Mandonnet et al. 1999).Milk<strong>in</strong>g abilityEfficient milk<strong>in</strong>g results from the <strong>in</strong>teraction betweenthe milk<strong>in</strong>g conditions, <strong>in</strong>clud<strong>in</strong>g mach<strong>in</strong>e parameters(vacuum level, pulsation rate, pulsation ratio; Lu et al.1991), and the udder anatomy and physiology(Bruckmaier et al. 1994). The existence of a major gene<strong>in</strong>fluenc<strong>in</strong>g milk<strong>in</strong>g ability (volume collected after 1 m<strong>in</strong>of milk<strong>in</strong>g) was postulated and confirmed by segregationanalyses (Ricordeau et al. 1990; Ilahi et al. 2000),<strong>in</strong>dicat<strong>in</strong>g that the major gene expla<strong>in</strong>s approximatelyhalf of the total genetic variation (total heritability of 0.5and polygenic heritability of 0.3). Phenotypic associationsbetween udder characteristics and milk<strong>in</strong>g abilityhave been reported (Montaldo and Mart<strong>in</strong>ez-Lozano1993; Bruckmaier et al. 1994) but genetic correlationsare unknown. Genetic correlations between milk<strong>in</strong>gspeed (volume collected after 1 m<strong>in</strong> of milk<strong>in</strong>g) andlactation yield were found to be low (rg = 0.10) by Ilahiet al. (2000) while phenotypic correlations between dailyyield and milk<strong>in</strong>g speed were over 0.25 (Peris et al. 1996;Ilahi et al. 1999). Phenotypic correlations betweenÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


382 B Leboeuf, JA Delgadillo, E Manfredi, A Piace` re, V Cle´ment, P Mart<strong>in</strong>, M Pellicer, P Boue´, and R de Cremouxmilk<strong>in</strong>g ability and mastitis, measured as CMT scores orSCC, are low accord<strong>in</strong>g to Montaldo and Mart<strong>in</strong>ez-Lozano (1993) and Ilahi (1999). Genetic correlationsbetween milk<strong>in</strong>g ability and udder health rema<strong>in</strong>unknown <strong>in</strong> goats.Contribution of AI to Genetic ImprovementProgrammes: The French Selection ProgrammeProduction from French goats, as measured by milkrecord<strong>in</strong>g on more than 100 000 animals <strong>in</strong> each breed,is on average 800 kg of milk for a lactation length of270 days. Fat and PC are higher <strong>in</strong> the Alp<strong>in</strong>e than <strong>in</strong>the Saanen breed: 3.7% and 3.2%, respectively, vs 3.5%and 3.1%. These results have been obta<strong>in</strong>ed afterapproximately 20 years of genetic improvement, with aselection scheme based on the use of AI.Determ<strong>in</strong>ation of the selection objectivesThe objectives of the selection programme are revisedperiodically <strong>in</strong> order to adapt the genetic goals tomarket demands, and to take <strong>in</strong>to account the genetictrends of the populations under selection (Piace` re et al.2002). S<strong>in</strong>ce 1970, the objective of the French programmewas the improvement of PY and PC withselection thresholds def<strong>in</strong>ed for each trait. However, asproduction traits are either negatively or positivelycorrelated, threshold selection on several traits is notefficient. Thus, <strong>in</strong> 1995, the former <strong>in</strong>dependent levels ofselection were replaced by a s<strong>in</strong>gle selection thresholddeterm<strong>in</strong>ed for a comb<strong>in</strong>ed <strong>in</strong>dex ICC = PY + 0.4 PC(Piace` re et al. 1997). Selection pressure on prote<strong>in</strong>resulted <strong>in</strong> positive trends for prote<strong>in</strong> and for fat content(FC) because of the positive correlation between bothtraits. However, ga<strong>in</strong>s on PC were higher than those forFC, thus <strong>in</strong>duc<strong>in</strong>g a slight negative trend <strong>in</strong> the ratioFC ⁄ PC. In 1999, the ICC was modified to stabilize theFC ⁄ PC ratio, by giv<strong>in</strong>g a slight positive weight<strong>in</strong>g to fatyield and FC. As Alp<strong>in</strong>e and Saanen goats have similarmatter yields, the production selection objective was thesame for both breeds.The same procedure was undertaken for type <strong>in</strong>dicesto determ<strong>in</strong>e a s<strong>in</strong>gle type objective. The type comb<strong>in</strong>ed<strong>in</strong>dex, named IMC (Index Morphologique Capr<strong>in</strong>, goattype <strong>in</strong>dex) (Cle´ment et al. 2006) comb<strong>in</strong>es most relevanttype <strong>in</strong>dices for breeders: udder profile (UP), udderfloor (UF) position, rear udder (RU) attachment andrear udder shape (RS). These four traits are responsiblefor 80% of the whole udder variability. However, typephenotypic and genetic differences between Alp<strong>in</strong>e andSaanen goats were observed. So the IMC was not thesame for both breeds:Alp<strong>in</strong>e IMC ¼ 1:5 UP þ UF þ RU þ RS;Saanen IMC ¼ UP þ UF þ RU þ 0:5 RSAs breeders wanted to obta<strong>in</strong> genetic progress for uddertype, as well as improv<strong>in</strong>g the milk production, they hadto def<strong>in</strong>e the respective weight<strong>in</strong>gs for both criteria <strong>in</strong> aglobal comb<strong>in</strong>ed <strong>in</strong>dex. Accord<strong>in</strong>g to the geneticparameters of different traits, which were slightlydifferent accord<strong>in</strong>g to the breed, they f<strong>in</strong>ally decided togive a weight of 44% and 33% on type <strong>in</strong> the Saanenand Alp<strong>in</strong>e breeds, respectively.Technical structures and general organizationSelection programmes for dairy rum<strong>in</strong>ants benefit fromthe cooperative action of farmers, milk plants, AIcentres and milk record<strong>in</strong>g organizations, with thetechnical and scientific support of specialized <strong>in</strong>stitutions.The breed<strong>in</strong>g law specifies the missions of eachorganization participat<strong>in</strong>g to the selection programme.There are seven organizations concerned with thebreed<strong>in</strong>g of dairy goats:1. The EDE (Regional Breed<strong>in</strong>g Bureaux) monitorsanimal identification. Accord<strong>in</strong>g to the Europeanrules, all French goats are officially identified with eartags and registered <strong>in</strong> a national database;2. France Controˆ le Laitier is responsible for milkrecord<strong>in</strong>g and provid<strong>in</strong>g technical advice to breederson breed<strong>in</strong>g and feed<strong>in</strong>g. Selection on productiontraits needs exhaustive records. In France, 340 000goats <strong>in</strong> 2200 herds are officially milk recorded andany breeder can access the milk record<strong>in</strong>g services.Records are registered <strong>in</strong> a national database;3. The Centres Re´gionaux Informatiques (CRI,Regional Comput<strong>in</strong>g Centers) are responsible forthe regional databases and all regional data are thencentralized <strong>in</strong> a national database: Le Centre deTraitement de l’Information Ge´ne´tique (CTIG,National Comput<strong>in</strong>g Center for Genetics);4. Capri-IA is the national semen production centreand the national union of AI cooperatives for goats.Capri-IA produces frozen semen, which is sent to AIcooperatives responsible for on-farm <strong>in</strong>sem<strong>in</strong>ations;5. The Institut de l’Elevage (IE, Breed<strong>in</strong>g Institute)gives technical support and coord<strong>in</strong>ates data collection,validation and storage <strong>in</strong> the national database;6. The Institut National de la Recherche Agronomique(INRA) gives scientific support for the def<strong>in</strong>itionof selection objectives, the genetic evaluation (<strong>in</strong>dexcalculation) and the design of the selection programme;7. Caprige` ne France is the goat breeders association.It groups 800 breeders and 160 000 Alp<strong>in</strong>e andSaanen goats. The Caprige` ne committee gathersrepresentatives of breeders, cheese producers, milkplants, M<strong>in</strong>istry of Agriculture and the technicalorganizations previously described. It is <strong>in</strong> charge ofthe general management of the selection programmeand the committee determ<strong>in</strong>es the selection objectives.Furthermore, Caprige` ne is responsible for the typerecord<strong>in</strong>g and the pedigree file management.Genetic evaluation of animalsThe statistical method used for genetic evaluation is aBLUP applied to an animal model. The advantage ofthis approach is that it provides unbiased breed<strong>in</strong>gvalues: first, all available <strong>in</strong>formation is taken <strong>in</strong>toaccount and second, it allows the dissociation of geneticand environmental effects. However, to separate part ofthe variance as a result of genetic effects from those ofÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Management of Goat <strong>Reproduction</strong> and Insem<strong>in</strong>ation 383the husbandry system, the method assumes that thereare some genetic l<strong>in</strong>ks between herds, all these l<strong>in</strong>ksbe<strong>in</strong>g called genetic connectedness. The ma<strong>in</strong> source ofconnectedness is AI.Genetic evaluation of French dairy goats for productiontraits is computed for all flocks recorded for milkproduction, but breed<strong>in</strong>g values are published only forconnected herds, that is, for herds which participate <strong>in</strong>the selection programme. Methods used to evaluateconnectedness are based on the percentage of goats withAI sires, but also on other source of connectedness, <strong>in</strong>particular, the use of goats with an AI paternal grandsire(Fouilloux et al. 2007), as the majority of naturalmat<strong>in</strong>gbucks are sired by AI.For genetic evaluation of type traits, connectedness isnot a problem, because approximately two-thirds ofscored goats are daughters of AI bucks and type scor<strong>in</strong>gis performed only <strong>in</strong> flocks us<strong>in</strong>g AI. Thus, breed<strong>in</strong>gvalues are calculated and published for a subpopulationof connected herds.Creation of genetic improvementA major step <strong>in</strong> the selection programme conducted <strong>in</strong>1992 with the creation of a structured selection nucleusformed with goat breeders highly <strong>in</strong>terested <strong>in</strong> selectionand genetic improvement. These breeders must <strong>in</strong>sem<strong>in</strong>ate30% of their goats, at least, and use 30% ofprogeny tested semen for their <strong>in</strong>sem<strong>in</strong>ations. In return,breeders enjoy technical advice for selection and get thelargest access to the best AI semen. In 2006, the selectionprogramme <strong>in</strong>volved 160 000 goats owned by 800breeders.Planned mat<strong>in</strong>gThe most important step <strong>in</strong> the genetic improvementprogramme is to breed AI males of high genetic level.They will come from planned mat<strong>in</strong>gs between the bestfemales and the best males <strong>in</strong> the population. Therefore,elite females selected to be dams of the next AI buckgeneration are <strong>in</strong>sem<strong>in</strong>ated with frozen semen from thebest AI males, named Sires. Dams and Sires are selectedaccord<strong>in</strong>g to their breed<strong>in</strong>g values on production andtype traits, comb<strong>in</strong>ed <strong>in</strong> a global <strong>in</strong>dex. Planned mat<strong>in</strong>gis determ<strong>in</strong>ed to avoid <strong>in</strong>breed<strong>in</strong>g and to preservegenetic variability.Sanitary and <strong>in</strong>dividual controlEvery year, 350 males born from planned mat<strong>in</strong>g are<strong>in</strong>spected on farm by Capri-IA <strong>in</strong> order to select the bestfor the semen production centre. They are exam<strong>in</strong>ed forpedigree (DNA validation), breed standard and sanitarystandard. The sanitary standard of each dam and eachherd is also tested. Accord<strong>in</strong>g to the tests’ results, 200males are selected for the quarant<strong>in</strong>e of the semenproduction centre, where they are controlled on thebasis of sanitary standards and growth. At the end ofthe quarant<strong>in</strong>e, only 120 males enter the semen productioncentre, where they are controlled on the basis ofsexual behaviour (libido and ability to give semen <strong>in</strong> anartificial vag<strong>in</strong>a), semen production (semen quantity andquality) and sperm survival after deep freez<strong>in</strong>g-thaw<strong>in</strong>g.After these controls, rema<strong>in</strong><strong>in</strong>g 70 males are progenytested to determ<strong>in</strong>e their genetic level. All elim<strong>in</strong>atedmales are slaughtered.Progeny test<strong>in</strong>gFor progeny test<strong>in</strong>g, 200 <strong>in</strong>sem<strong>in</strong>ations are performedper male across milk-recorded herds. The goal is toobta<strong>in</strong> a m<strong>in</strong>imum of 30 milk-recorded daughters pertested male (the average number is 80 daughters permale). Milk records and type appraisal of the daughtersare used to compute the breed<strong>in</strong>g value of each male.The 35 best males are selected and the other ones areslaughtered.ConclusionThis review has ma<strong>in</strong>ly addressed the methods and thetechniques that have provided improvements to rout<strong>in</strong>epractical AI programmes and genetic improvementprogrammes. An important consideration is the tim<strong>in</strong>gof the AI, which should be performed close to the timeof ovulation <strong>in</strong> the females. Accurate and carefuldetection of oestrus or control of oestrus and ovulationare necessary to reach a satisfactory fertility level.Validation of new procedures such as the use of themale effect <strong>in</strong> highly seasonal goats will be required tomeet to the new expectations of the animal <strong>in</strong>dustriesand consumers which are opposed to the use of syntheticchemicals and hormonal treatments. The efficiency ofthe French selection programme is now widely provedand recognized. It fits with most dairy goat breeders’objectives, but further studies are still <strong>in</strong> progress to<strong>in</strong>tegrate new selection criteria and to adapt selectionobjectives to new needs and diversified market demands.The goat genetic map is be<strong>in</strong>g constructed, thus giv<strong>in</strong>gthe basis for QTL (Quantitative Trait Loci) detection.Molecular <strong>in</strong>formation will be particularly useful forgenetic improvement of dairy traits.ReferencesAmills M, Sanchez A, Manfredi E, 1999: Allelic frequencies ofthe Mhc class II DRB gene <strong>in</strong> capr<strong>in</strong>e arthritis-encephalitisvirus <strong>in</strong>fected goats. 2nd Annual Conference on new andRe-Emerg<strong>in</strong>g Infectious Diseases, University of Ill<strong>in</strong>ois,Urbana-Champaign, pp. 23.Analla M, Jime´nez-Gamero I, Mun˜ oz Serrano A, SerradillaJM, Falagan A, 1996: Estimation of genetic parameters formilk yield and fat and prote<strong>in</strong> contents of milk fromMurciano-Granad<strong>in</strong>a goats. J Dairy Sci 79, 1895–1898.Baker RL, Audho JO, Aduda EO, Thorpe W, 1999: GeneticResistance to Gastro<strong>in</strong>test<strong>in</strong>al Nematode Parasites <strong>in</strong> some<strong>in</strong>digenous breeds of sheep and goats <strong>in</strong> Kenya. In: FromJay L. Lush to Genomics: Visions for Animal Breed<strong>in</strong>g andGenetics. University Press, Ames, p. 180.Barbieri ME, Manfredi E, Elsen JM, Ricordeau G, Bouillon J,Grosclaude F, Mahé MF, Bibe´ B, 1995: Influence du locusde la case´<strong>in</strong>e as1 sur les performances laitie` res et lesparame` tres géne´tiques des che` vres de race Alp<strong>in</strong>e. GenetSel Evol 27, 437–450.Baudry C, Mercier P, Mallereau MP, Lenfant D, 1998:Utilisation des numérations cellulaires <strong>in</strong>dividuelles pour laÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


384 B Leboeuf, JA Delgadillo, E Manfredi, A Piace` re, V Cle´ment, P Mart<strong>in</strong>, M Pellicer, P Boue´, and R de Cremouxde´tection des <strong>in</strong>fections mammaires subcl<strong>in</strong>iques de laChe` vre: de´f<strong>in</strong>ition de seuils. In: Milk<strong>in</strong>g and Milk Productionof Dairy Sheep and Goats, EAAP Publication No. 95,Proceed<strong>in</strong>gs of the 6th International Symposium on themilk<strong>in</strong>g of Small Rum<strong>in</strong>ants, Athens, Greece, 26th September–1stOctober, pp. 119–123.Belichon S, Manfredi M, Piacère A, 1998: Genetic parametersof dairy traits <strong>in</strong> the Alp<strong>in</strong>e and Saanen goat breeds. GenetSel Evol 30, 529–534.Boichard D, Bouloc N, Ricordeau G, Piacere A, Barillet F,1989: Genetic parameters for first lactation dairy traits <strong>in</strong>the Alp<strong>in</strong>e and Saanen goat breeds. Genet Sel Evol 21,201–215.Boue´ P, Sigwald JP, 2001: Statistiques et bilan ge´ne´tique del’IA en espe` ce capr<strong>in</strong>e. CR No. 3309, November 2002, 34pp.Capri-IA et I.E. (Ed), 86800 Mignaloux Beauvoir, France.Bruckmaier R, Ritter C, Schams D, Blun J, 1994: Mach<strong>in</strong>emilk<strong>in</strong>g of dairy goats dur<strong>in</strong>g lactation: udder anatomy,milk<strong>in</strong>g characteristics, and blood concentrations of oxytoc<strong>in</strong>and prolact<strong>in</strong>. J Dairy Res 61, 457–466.Chem<strong>in</strong>eau P, Daveau A, Maurice F, Delgadillo JA, 1992:Seasonality of oestrus and ovulation is not modified bysubject<strong>in</strong>g female Alp<strong>in</strong>e goats to a tropical photoperiod.Small Rum<strong>in</strong> Res 8, 299–312.Chem<strong>in</strong>eau P, Pellicer-Rubio MT, Lassoued N, Khaldi G,Monniaux D, 2006: Male <strong>in</strong>duced short oestrous andovarian cycles <strong>in</strong> sheep and goats: a work<strong>in</strong>g hypothesis.Reprod Nutr Dev 46, 417–429.Cle´ment V, Mart<strong>in</strong> P, Barillet F, 2006: Elaboration d’un <strong>in</strong>dexsynthe´tique capr<strong>in</strong> comb<strong>in</strong>ant les caracte` res laitiers et descaracte` res de morphologie mammaire. Renc Rech Rum<strong>in</strong>13, 209–212.Corteel JM, 1974: Viabilite´ des spermatozoı¨des de boucconserve´s et congelés avec ou sans leur plasma sem<strong>in</strong>al:Effet du glucose. Ann Biol Anim Biochim Biophys 14, 741–745.Corteel JM, 1977: Production, storage and artificial <strong>in</strong>sem<strong>in</strong>ationof goat semen. In: Proceed<strong>in</strong>g of the Symposium‘‘Management of <strong>Reproduction</strong> <strong>in</strong> Sheep and Goats’’,Madison, WI, USA, 24–25 July 1977, pp. 41–57.Corteel JM, Leboeuf B, Baril G, 1988: Artificial breed<strong>in</strong>g ofadult goats and kids <strong>in</strong>duced with hormones to ovulateoutside the breed<strong>in</strong>g season. Small Rum<strong>in</strong> Res 1, 19–33.Dauzier L, 1966: Artificial <strong>in</strong>sem<strong>in</strong>ation <strong>in</strong> the goat. In:Dall<strong>in</strong>g (ed), Encyclopedia of Veter<strong>in</strong>ary Medic<strong>in</strong>e. W. Greenand Son, pp. 269–271.Delgadillo JA, Leboeuf B, Chem<strong>in</strong>eau P, 1991: Decrease <strong>in</strong> theseasonality of sexual behaviour and sperm production <strong>in</strong>bucks by exposure to short photoperiodic cycles. Theriogenology36, 755–770.Delgadillo JA, Leboeuf B, Chem<strong>in</strong>eau P, 1992: Abolition ofseasonal variations <strong>in</strong> semen quality and ma<strong>in</strong>tenance ofsperm fertiliz<strong>in</strong>g ability by short photoperiodic cycles <strong>in</strong> goatbucks. Small Rum<strong>in</strong> Res 9, 47–59.Delgadillo JA, Canedo GA, Chem<strong>in</strong>eau P, Guillaume D,Malpaux B, 1999: Evidence for an annual reproductiverhythm <strong>in</strong>dependent of food availability <strong>in</strong> male Creolegoats <strong>in</strong> subtropical northern Mexico. Theriogenology 52,727–737.Delgadillo JA, Fitz-Gonza´lez G, Duarte G, Véliz FG, Carrillo E,Flores JA, Vielma J, Herna´ndez H, Malpaux B, 2004a:Management of photoperiod to control capr<strong>in</strong>e reproduction<strong>in</strong> the subtropics. Reprod Fertil Dev 16, 471–478.Delgadillo JA, Cortez ME, Duarte G, Chem<strong>in</strong>eau P, Malpaux B,2004b: Evidence that photoperiod controls annual changes <strong>in</strong>testosterone secretion and body weight <strong>in</strong> subtropical malegoats. Reprod Nutr Dev 44, 183–193.Fouilloux MN, Cle´ment V, Laloe¨ D, 2008: Measur<strong>in</strong>gconnectedness among herds <strong>in</strong> mixed l<strong>in</strong>ear models: fromtheory to practice <strong>in</strong> large-size genetic evaluations. Acceptedfor publication <strong>in</strong> Genet Sel Evol.Gebbie FE, Forsyth IA, Arendt J, 1999: Effects of ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>gsolstice light and temperature on reproductive activity, coatgrowth, plasma prolact<strong>in</strong> and melaton<strong>in</strong> <strong>in</strong> goats. J ReprodFertil 116, 25–35.Goldmann W, Mart<strong>in</strong> T, Foster J, Hughes S, Smith G,Hughes K, Dawson M, Hunter N, 1996: Novel polymorphisms<strong>in</strong> the capr<strong>in</strong>e PrP gene: a codon 142 mutation associatedwith scrapie <strong>in</strong>cubation period. J Gen Virol 77, 2885–2891.Grosclaude F, Mart<strong>in</strong> P, 1997: Milk prote<strong>in</strong> polymorphism.In: Proceed<strong>in</strong>gs of the International Dairy FederationSem<strong>in</strong>ar, Palmerston North, New Zealand, pp. 241–253.Ilahi H1999: Variabilite´ ge´nétique du de´bit de traite chez lescapr<strong>in</strong>s laitiers. Ph.D. thesis, ENSAR, Rennes, France.Ilahi H, Chast<strong>in</strong> P, Bouvier F, Arha<strong>in</strong>x J, Ricard E, ManfrediE, 1999: Milk<strong>in</strong>g characteristics of dairy goats. Small Rum<strong>in</strong>Res 34, 97–102.Ilahi H, Manfredi E, Chast<strong>in</strong> P, Monod F, Elsen JM, Le RoyP, 2000: Genetic variability <strong>in</strong> milk<strong>in</strong>g speed of dairy goats.Genet Res 75, 315–319.Iloeje MU, Van Vleck LD, Wiggans GR, 1981: Components ofvariance for milk and fat yields <strong>in</strong> dairy goats. J Dairy Sci64, 2290–2293.Jordana J, Amills M, Diaz E, Angulo C, Serradilla JM,Sanchez A, 1995: Gene frequencies of capr<strong>in</strong>e as1-case<strong>in</strong>polymorphism <strong>in</strong> Spanish goat breeds. Small Rum<strong>in</strong> Res 20,215–221.Leboeuf B, Furstoss V, Guillouet P, Boue´ P, 2004a: Productionof artificial <strong>in</strong>sem<strong>in</strong>ation doses from Alp<strong>in</strong>e and Saanenbucks under various photoperiodic cycles. S Afr J Anim Sci34, 230–232.Leboeuf B, Guillouet P, Bonne´ JL, Forgerit Y, Magistr<strong>in</strong>i M,2004b: Goat semen preserved at 4°C until 76 hours beforeartificial <strong>in</strong>sem<strong>in</strong>ation: different attempts to ma<strong>in</strong>ta<strong>in</strong> thefertility. S Afr J Anim Sci 34, 233–235.Lu CD, Potchoiba MJ, Loetz ER, 1991: Influence ofvacuum level, pulsation ratio and rate on milk<strong>in</strong>g performanceand udder health <strong>in</strong> dairy goats. Small Rum<strong>in</strong> Res5, 1–8.Luo MF, Wiggans GR, Hubbard SM, 1997: Variance componentestimation and multitrait genetic evaluation for typetraits of dairy goats. J Dairy Sci 80, 594–600.Mahe´ MF, Manfredi E, Ricordeau G, Piacere A, Grosclaude F,1994: Effets du polymorphisme de la case´<strong>in</strong>e as1 sur lesperformances laitie` res: analyse <strong>in</strong>tra-descendance de boucsde race Alp<strong>in</strong>e. Genet Sel Evol 26, 151–157.Mandonnet N, Aumont G, Gruner L, Bouix J, Vu Tien Khang J,Arquet R, Varo H, 1999: Direct and maternal genetic effectsfor resistance to strongyles <strong>in</strong> Creole goats. In: 17th InternationalConference of the WAAP, Copenhagen, 15–19 August1999. Abst.Manfredi E, Leroux C, Piace` re A, Mart<strong>in</strong> P, Elsen JM,Grosclaude F, 1998: Use of a major gene <strong>in</strong> a dairy goatselection scheme. In: 49th Annual Meet<strong>in</strong>g of the EAAP,Warsaw, Poland, 24–27 August, pp. 1–4.Manfredi E, Piace` re A, Lahaye P, Ducrocq V, 2001: Geneticparameters of type appraisal <strong>in</strong> Saanen and Alp<strong>in</strong>e goats.Livest Prod Sci 70, 183–189.Mart<strong>in</strong> GB, Milton JTB, Davison RH, Hunzicker GEB,L<strong>in</strong>dsay DR, Blache D, 2004: Natural methods for <strong>in</strong>creas<strong>in</strong>greproductive efficiency <strong>in</strong> small rum<strong>in</strong>ants. Anim ReprodSci 82–83, 231–246.Mavrogenis AP, Constant<strong>in</strong>uou A, Louca A, 1984: Environmentaland genetic causes of variation <strong>in</strong> production traitsof Damascus goats. II. Goat productivity. Anim Prod 38,99–104.Montaldo H, Mart<strong>in</strong>ez-Lozano FJ, 1993: Phenotypic relationshipsbetween udder and milk<strong>in</strong>g characteristics, milkÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Management of Goat <strong>Reproduction</strong> and Insem<strong>in</strong>ation 385production and California mastitis test <strong>in</strong> goats. SmallRum<strong>in</strong> Res 12, 329–337.Muller CJC, Cloet SWP, Schoeman SJ, 2002: Estimation ofgenetic parameters for milk yield and milk composition ofSouth African Saanen goats. In: Proceed<strong>in</strong>gs of the 7thWorld Congress on Genetics Applied to Livestock Production,Montpellier, France, pp. 259–262.Pellicer-Rubio M, Combarnous Y, 1998: Deterioration of goatspermatozoa <strong>in</strong> skimmed milk-based extenders as a result ofoleic acid released by the bulbourethral lipase BUSgp60. JReprod Fertil 112, 95–105.Pellicer-Rubio MT, Leboeuf B, Bernelas D, Forgerit Y,Pougnard JL, Bonne´ JL, Senty E, Chem<strong>in</strong>eau P, 2007:Highly synchronous and fertile reproductive activity<strong>in</strong>duced by the male effect dur<strong>in</strong>g deep anoestrus <strong>in</strong> lactat<strong>in</strong>ggoats subjected to treatment with artificially long daysfollowed by a natural photoperiod. Anim Reprod Sci 98,241–258.Pellicer-Rubio MT, Leboeuf B, Bernelas D, Forgerit Y,Pougnard JL, Bonne´ JL, Senty E, Breton S, Brun F,Chem<strong>in</strong>eau P, 2008: High fertility us<strong>in</strong>g artificial <strong>in</strong>sem<strong>in</strong>ationdur<strong>in</strong>g deep anoestrus after <strong>in</strong>duction and synchronisationof ovulatory activity by the ‘‘male effect’’ <strong>in</strong> lactat<strong>in</strong>ggoats subjected to treatment with artificial long days andprogestagens. Accepted for publication <strong>in</strong> Anim Reprod Sci.Peris S, Such X, Caja G, 1996: Milkability of Murc<strong>in</strong>ao-Granad<strong>in</strong>a dairy goats: milk partition<strong>in</strong>g and flow ratedur<strong>in</strong>g milk<strong>in</strong>g accord<strong>in</strong>g to parity, prolificacy and mode ofsuckl<strong>in</strong>g. J Dairy Res 63, 1–9.Piace` re A, Bouloc-Duval N, Sigwald JP, Larzul C, Manfredi E,1997: Utilisation de l’<strong>in</strong>dex comb<strong>in</strong>e´ capr<strong>in</strong> et du polymorphismede la case´<strong>in</strong>e alpha s1 dans le sche´ma de sélectioncapr<strong>in</strong>. Renc Rech Rum<strong>in</strong> 4, 187–190.Piace` re A, Manfredi E, Lahaye P, 1998: Analyse ge´nétique dela morphologie des che` vres Saanen et Alp<strong>in</strong>e françaises. In:Proceed<strong>in</strong>gs of the 6th International Symposium on themilk<strong>in</strong>g of Small Rum<strong>in</strong>ants, Athens, Greece, 26th September–1stOctober, pp. 178–180.Ricordeau G, Bouillon J, Le Roy P, Elsen JM, 1990:De´term<strong>in</strong>isme géne´tique du de´bit de traite au cours de latraite des che` vres. INRA Prod Anim 3, 121–126.Ruff G, Lazary S, 1988: Evidence for l<strong>in</strong>kage between theCapr<strong>in</strong>e leucocyte antigen (CLA) system and susceptibilityto CAE virus-<strong>in</strong>duced arthritis <strong>in</strong> goats. Immunogenetics 28,303–309.Rupp R, Cle´ment V, Piace` re A, Manfredi E, 2004: Goat milksomatic cell count is a heritable trait. In: 55th AnnualMeet<strong>in</strong>g of the EAAP, Bled, Slovenia, 3–8 September 2004.Sias B, Ferrato F, Pellicer-Rubio M, Forgerit Y, Guillouet P,Leboeuf B, Carrie` re F, 2005: Clon<strong>in</strong>g and seasonal secretionof the pancreatic lipase-related prote<strong>in</strong>2 present <strong>in</strong> goatsem<strong>in</strong>al plasma, 2004. Biochim Biophys Acta 1686, 169–180.Sierra D, Sanchez A, Corrales JC, Contreras A, 1998: In:Proceed<strong>in</strong>gs of the 6th International Symposium on themilk<strong>in</strong>g of Small Rum<strong>in</strong>ants, Athens, Greece, 26th September–1stOctober, pp. 178–180.Author’s address (for correspondence): B Leboeuf, INRA-UEICP,Venours, 86480 Rouillé, France. E-mail: leboeuf@lusignan.<strong>in</strong>ra.frConflict of <strong>in</strong>terest: All authors declare no conflict of <strong>in</strong>terests.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 386–392 (2008); doi: 10.1111/j.1439-0531.2008.01189.xISSN 0936-6768Regulation of the Spermatogonial Stem Cell NicheN Kostereva 1 and M-C Hofmann 1,21 Department of Veter<strong>in</strong>ary Biosciences; 2 Institute for Genomic Biology, University of Ill<strong>in</strong>ois at Urbana-Champaign, Urbana, IL, USAContentsSpermatogonial stem cells (SSCs) reside with<strong>in</strong> specializedmicroenvironments called ‘niches’, which are essential for theirma<strong>in</strong>tenance and self-renewal. In the mammalian testis, thema<strong>in</strong> components of the niche <strong>in</strong>clude the Sertoli cell, thegrowth factors that this nurs<strong>in</strong>g cell produces, the basementmembrane, and stimuli from the vascular network between thesem<strong>in</strong>iferous tubules. This review focuses on signall<strong>in</strong>g pathwaysma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g SSCs self-renewal and differentiation anddescribes potential mechanisms of regulation of the spermatogonialstem cell niche.Stem Cells and their NicheStem cells are required for the growth, ma<strong>in</strong>tenanceand repair of many tissues. They are characterized bytheir abilities to self-renew and to produce numerousdifferentiated daughter cells, enabl<strong>in</strong>g them to play acentral role <strong>in</strong> tissue homeostasis. In order for theirma<strong>in</strong>tenance and self-renewal to be ensured, theyreceive essential signals from their microenvironment,which is called the stem cell ‘niche’. The concept ofstem cell niche was first suggested by Schofield <strong>in</strong> 1978,to describe a microenvironment that supports stemcells <strong>in</strong> the mammalian hematopoietic system (Schofield1978). Other stem cell niches have now beenidentified <strong>in</strong> most tissues of model organisms, <strong>in</strong>clud<strong>in</strong>gthe <strong>in</strong>test<strong>in</strong>e, sk<strong>in</strong>, bra<strong>in</strong> and testis. The niche regulatesspecific properties of the stem cell, <strong>in</strong>clud<strong>in</strong>g selfrenewal,pluripotency, quiescence and the ability todifferentiate <strong>in</strong>to s<strong>in</strong>gle or multiple l<strong>in</strong>eages (Adamsand Scadden 2006). To this effect, the niche can bedef<strong>in</strong>ed as a complex <strong>in</strong>terplay of short- and long-rangestimuli between the stem cells, their differentiat<strong>in</strong>gdaughters, neighbour<strong>in</strong>g cells, and the extracellularmatrix, collectively mak<strong>in</strong>g up a microenvironmentthat controls stem cell behaviour. Ultimately, thisbehaviour will depend on cellular <strong>in</strong>tr<strong>in</strong>sic factors thatare modulated by these signals (Watt and Hogan2000).The Spermatogonial Stem Cell NicheIn the mammalian testis, the stem cells that are at theorig<strong>in</strong> of spermatogenesis are called spermatogonialstem cells (SSCs). Spermatogonial stem cells reside <strong>in</strong>the basal part of the sem<strong>in</strong>iferous epithelium. These cellsare the only stem cells <strong>in</strong> the body that undergo selfrenewalthroughout life and transmit genetic <strong>in</strong>formationto the offspr<strong>in</strong>g (De Rooij and Russell 2000).Spermatogonial stem cells are morphologically undifferentiateds<strong>in</strong>gle cells that are not connected by<strong>in</strong>tercellular bridges like the more advanced germ cells(Dym and Fawcett 1971; Huck<strong>in</strong>s 1971; Oakberg 1971).They reside on the basement membrane and are also <strong>in</strong><strong>in</strong>timate contact with the Sertoli cells, the only somaticcells present with<strong>in</strong> the sem<strong>in</strong>iferous epithelium. Spermatogonialstem cells self-renew <strong>in</strong> order to ma<strong>in</strong>ta<strong>in</strong>spermatogenesis throughout the life of adult maleanimals. These cells also differentiate to produceApaired, Aaligned, A1-A4 spermatogonia and type Bspermatogonia, through a series of steps that willamplify the number of germ cells. F<strong>in</strong>ally, type Bspermatogonia will differentiate <strong>in</strong>to spermatocytes thatwill translocate to the <strong>in</strong>ner layer of the sem<strong>in</strong>iferousepithelium, undergo meiosis and give rise to haploidspermatids that will differentiate <strong>in</strong>to spermatozoa.Although some advances have recently been made, themolecular mechanisms underly<strong>in</strong>g the ma<strong>in</strong>tenance andself-renewal of SSCs only beg<strong>in</strong> to be elucidated. Inaddition, the signal that mediates the decision of SSCsto differentiate rather than self-renew is still unknown.This slow progress is due to the fact that these cells exist<strong>in</strong> low numbers (0.03% of the total number of germcells <strong>in</strong> an adult testis; Meistrich and Van Beek 1993)and that no specific membrane marker is available toisolate them unequivocally. However, enrichment techniquesare now available, that allow significant advances<strong>in</strong> our understand<strong>in</strong>g of the behaviour of SSCs<strong>in</strong> vitro (Kubota et al. 2003; Buageaw et al. 2005;Hofmann et al. 2005b; Braydich-Stolle et al. 2007). Inaddition, the transplantation technique established bythe group of R. Br<strong>in</strong>ster a decade ago providedresearchers with an <strong>in</strong> vivo functional assay to assessself-renewal and germl<strong>in</strong>e transmission after geneticmanipulations of these cells (Nagano et al. 2001;Br<strong>in</strong>ster 2002).In the mammalian testis, the somatic Sertoli cell, thebasement membrane and the cellular components of the<strong>in</strong>terstitial space between the sem<strong>in</strong>iferous tubules(Shetty and Meistrich 2007) are crucial components ofthe niche. The Sertoli cell provides growth factorsnecessary for self-renewal such as glial cell l<strong>in</strong>e-derivedneurotrophic factor (GDNF) and basic fibroblastgrowth factor (bFGF) (Meng et al. 2000; Kubota et al.2004; Hofmann et al. 2005b), the basement membraneand <strong>in</strong>tegr<strong>in</strong>s provide for anchorage (Sh<strong>in</strong>ohara et al.1999), and stimuli from the vascular network and<strong>in</strong>terstitial cells are crucial for the localization ofundifferentiated spermatogonia along specific portionsof the basement membrane (Yoshida et al. 2007). The<strong>in</strong>tegration of these signals provides the cues necessaryfor self-renewal and retention of the SSCs <strong>in</strong> theirundifferentiated state. These extr<strong>in</strong>sic signals will modulateSSC <strong>in</strong>tr<strong>in</strong>sic signals such as k<strong>in</strong>ases, secondmessengers and transcription factors to ensure homeostasis.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Regulation of the Spermatogonial Stem Cell Niche 387Cues for Self-RenewalOne factor that is essential for SSC self-renewal isGDNF. It is a prote<strong>in</strong> secreted by Sertoli cells afterbirth, and is specifically responsible for the ma<strong>in</strong>tenanceand proliferation of SSCs <strong>in</strong> vivo and <strong>in</strong> vitro (Menget al. 2000; Kubota et al. 2004; Hofmann et al. 2005b).Glial cell l<strong>in</strong>e-derived neurotrophic factor belongs to thetransform<strong>in</strong>g growth factor-beta superfamily, and signalsthrough a multi-component receptor formed by theRet tyros<strong>in</strong>e k<strong>in</strong>ase transmembrane receptor and its coreceptorglial cell l<strong>in</strong>e-derived neurotrophic factor familyreceptor alpha-1 (GFRa-1). The b<strong>in</strong>d<strong>in</strong>g of GDNFtriggers the activation of multiple signall<strong>in</strong>g pathways <strong>in</strong>responsive cells (Airaks<strong>in</strong>en and Saarma 2002).Mice lack<strong>in</strong>g GDNF or its receptors die with<strong>in</strong> thefirst day of birth with renal and neuronal abnormalities(Schuchardt et al. 1994; Moore et al. 1996; Pichel et al.1996; Sanchez et al. 1996). The problem of neonatalmortality can be overcome by graft<strong>in</strong>g GDNF, GFRa-1or Ret deficient neonatal testes under the back sk<strong>in</strong> ofimmunodeficient mice, and subsequently monitor<strong>in</strong>g thedevelopment of the grafted testes (Naughton et al.2006). This strategy revealed that any disruption ofGDNF-mediated Ret signall<strong>in</strong>g results <strong>in</strong> a lack of SSCself-renewal and <strong>in</strong>duces the progressive loss of spermatogenesisby germ cell depletion. In comparison,normal spermatogenesis and ma<strong>in</strong>tenance of SSC populationswas observed <strong>in</strong> the grafted wild type (WT)testes. Thus, GDNF, Ret and GFRa-1 are all crucial forSSC ma<strong>in</strong>tenance, emphasiz<strong>in</strong>g the essential role of theGDNF ⁄ Ret ⁄ GFRa1 signall<strong>in</strong>g pathway <strong>in</strong> SSCs.Spermatogonial stem cells have been difficult toisolate because of their low numbers and the lack ofsurface markers specific enough for immunosort<strong>in</strong>g.Some laboratories, <strong>in</strong>clud<strong>in</strong>g ours, have isolated SSCsus<strong>in</strong>g antibodies aga<strong>in</strong>st the GFRa-1 receptor (Buageawet al. 2005; Hofmann et al. 2005b). Although expressedby both the stem cell and its direct progeny, the Apairedspermatogonia, GFRa-1 is an adequate marker forpurify<strong>in</strong>g SSCs from testes us<strong>in</strong>g antibody selection.Us<strong>in</strong>g gravity sedimentation on a 2–4% bov<strong>in</strong>e serumalbum<strong>in</strong> (BSA) gradient followed by magnetic beadsisolation with a GFRa-1 antibody, we obta<strong>in</strong>ed a cellpurity of up to 98%. However, while the purity is high,the number of cells recovered is low. Other <strong>in</strong>vestigatorshave been successful at enrich<strong>in</strong>g SSCs us<strong>in</strong>g antibodiesto thymus cell antigen 1 (Thy-1) and fluorescenceactivatedcell sort<strong>in</strong>g, produc<strong>in</strong>g an enrichment that wassufficient for most of their studies (Kubota et al. 2003).Glial cell l<strong>in</strong>e-derived neurotrophic factor, together withother growth factors, has recently been used to expandthe number of SSCs <strong>in</strong> long-term cultures provided thatthey are grown onto feeder layers (Kanatsu-Sh<strong>in</strong>oharaet al. 2003; Ogawa et al. 2003; Kubota et al. 2004).We recently elucidated some of the pathways <strong>in</strong>ducedby GDNF <strong>in</strong> SSCs by us<strong>in</strong>g serum-free short-termcultures of SSCs and a spermatogonial stem cell l<strong>in</strong>e thatwe established (Hofmann et al. 2005a). After stimulationby GDNF, we observed that several k<strong>in</strong>ases fromthe Src family co-precipitate with the Ret receptor<strong>in</strong> SSCs (Braydich-Stolle et al. 2007). Further, wedemonstrated that Src activates a PI3K ⁄ Akt signall<strong>in</strong>gFig. 1. Glial cell l<strong>in</strong>e-derived neurotrophic factor can signal throughactivation of Src k<strong>in</strong>ases <strong>in</strong> SSCs. The GDNF can promote cell cycleprogression via activation of Src family k<strong>in</strong>ases (SFKs) such as Src,Yes, Fyn and Lyn. The SFKs further activate a PI3K ⁄ Akt pathway,which ultimately leads to an <strong>in</strong>crease <strong>in</strong> N-myc gene expression (fromBraydich-Stolle et al. 2007)pathway, which ultimately leads to N-Myc expressionand promotes SSC proliferation (Fig. 1). Subsequently,the groups of T. Sh<strong>in</strong>ohara and R. Br<strong>in</strong>ster establishedthe <strong>in</strong> vivo relevance of this signall<strong>in</strong>g axis for SSC selfrenewalby us<strong>in</strong>g germ cell transplantations after downregulat<strong>in</strong>gAkt and Src expression by RNA <strong>in</strong>terferenceor pharmacological <strong>in</strong>hibitors (Lee et al. 2007; Oatleyet al. 2007).Ret activation by the b<strong>in</strong>d<strong>in</strong>g of GDNF also <strong>in</strong>ducesthe anchor<strong>in</strong>g of the prote<strong>in</strong> adaptors Shc and Grb2 andthe activation of Ras <strong>in</strong> SSCs (He et al. 2008). Ras thenactivates ERK1 ⁄ 2, which ultimately leads to the phosphorylationand activation of transcription factors suchas CREB-1, ATF-1 and CREM-1. F<strong>in</strong>ally, theGDNF ⁄ Ret ⁄ Ras axis up-regulates the transcription ofthe immediate-early gene c-fos, the cell cycle activatorcycl<strong>in</strong> A, as well as CDK2 (Fig. 2). This scenario issimilar to what is observed <strong>in</strong> other cell types, whereCREB and c-fos enhance the expression of cycl<strong>in</strong> A,favour<strong>in</strong>g the G1 ⁄ S cell cycle transition (Desdouetset al. 1995). Therefore, our studies suggest that GDNF<strong>in</strong>duces SSC self-renewal through multiple signall<strong>in</strong>gpathways.Other transcription factors have been recently identified,that are crucial for SSC ma<strong>in</strong>tenance orself-renewal. These <strong>in</strong>clude B cell CLL ⁄ lymphoma 6,member b (Bcl6b) (Oatley et al. 2006), TATA boxb<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> (TBP)-associated factor 4b (Taf4b)(Falender et al. 2005) and promyelocytic leukaemiaÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


388 N Kostereva and M-C HofmannFig. 2. Glial cell l<strong>in</strong>e-derived neurotrophic factor can signal through aRas-<strong>in</strong>duced pathway <strong>in</strong> SSCs. The GDNF can promote cell cycleprogression via activation of the Ras ⁄ ERK1 ⁄ 2 pathway. Downstreamevents are also depicted. ‘P’ <strong>in</strong>dicates ‘phosphorylate’, and ‘A’ denotes‘activate’ (from He et al. 2008)z<strong>in</strong>c-f<strong>in</strong>ger (Plzf) (Buaas et al. 2004; Costoya et al.2004). The exact function of these molecules is notelucidated yet. However, there is evidence that Bcl6b is atarget of GDNF and plays a role <strong>in</strong> stem cell ma<strong>in</strong>tenance,as mice with a targeted disruption of Bcl6b havean <strong>in</strong>creased <strong>in</strong>cidence of Sertoli cell-only sem<strong>in</strong>iferoustubules (Oatley et al. 2006). In addition, it seems clearthat Plzf <strong>in</strong> WT animals represses SSC differentiation,while its loss <strong>in</strong> mutant or knockout mice shifts thebalance towards differentiation at the cost of selfrenewal.In addition, Plzf seems to directly repress thetranscription of the receptor c-kit, which is importantfor spermatogonial differentiation (Filipponi et al.2007), and undifferentiated spermatogonia isolated fromPlzf - ⁄ - mice exhibit a marked <strong>in</strong>crease <strong>in</strong> c-kit expression.Therefore, Plzf might ma<strong>in</strong>ta<strong>in</strong> the pool of SSCsthrough direct repression of c-kit expression.Regulation of DifferentiationWhile the exact determ<strong>in</strong>ant of the self-renewal ⁄ differentiationswitch has not been elucidated yet, progresshas also been made <strong>in</strong> our understand<strong>in</strong>g of the controland ma<strong>in</strong>tenance of SSCs differentiation. As mentionedabove, differentiat<strong>in</strong>g spermatogonia express the c-kitreceptor, and Kit ligand, produced by Sertoli cells,stimulates their DNA synthesis (Dolci et al. 2001). Inaddition, a recent study demonstrated that Kit ligandup-regulates the expression of early meiotic genes <strong>in</strong>these cells (Rossi et al. 2008), emphasiz<strong>in</strong>g the importanceof this growth factor for germ cell differentiation.In 2001, two groups of researchers described theexpression of Notch receptors and their ligands, Jagged-1and Jagged-2, <strong>in</strong> the mammalian testis (Diramiet al. 2001; Hayashi et al. 2001). In mammals, the Notchsignall<strong>in</strong>g pathway is <strong>in</strong>volved <strong>in</strong> cell fate decisionsdur<strong>in</strong>g various cellular and developmental processesFig. 3. The Notch signall<strong>in</strong>g pathway. After the b<strong>in</strong>d<strong>in</strong>g of Jagged 1 ⁄ 2to the Notch receptor, the <strong>in</strong>tracellular portion of the Notch receptor iscleaved and becomes Notch <strong>in</strong>tracellular doma<strong>in</strong> (NICD). The NICDis a transcription factor that translocates <strong>in</strong>to the nucleus to activatethe expression of target genes (adapted from E. Lai, Memorial Sloan-Ketter<strong>in</strong>g Cancer Center, Sloan-Ketter<strong>in</strong>g Institute, New York, NY,USA)(We<strong>in</strong>master 1997). Notch is a transmembrane receptorof approximately 300 kDa <strong>in</strong> size. Upon b<strong>in</strong>d<strong>in</strong>g to itsligand, Notch is proteolytically processed to generate a180 kDa fragment conta<strong>in</strong><strong>in</strong>g most of the extracellulardoma<strong>in</strong> and a 120 kDa fragment conta<strong>in</strong><strong>in</strong>g the transmembraneand cytoplasmic doma<strong>in</strong>s. This latter fragmentis <strong>in</strong> turn cleaved, releas<strong>in</strong>g a shorter 80 kDAfragment, called Notch <strong>in</strong>tracytoplamic doma<strong>in</strong>(NICD), which migrates <strong>in</strong>to the cell nucleus andfunctions as a transcription factor (Fig. 3). In thenucleus, activated Notch (NICD) <strong>in</strong>teracts with othertranscription factors to regulate cell fate decision.Because both ligand and receptor are localized <strong>in</strong> theplasma membrane of the effector and the target cell,respectively, a close cell–cell contact is necessary for theactivation of this pathway. In mammals, four Notchgenes (Notch1–4) have been isolated (We<strong>in</strong>master et al.1991; Lardelli et al. 1994; Uyttendaele et al. 1996). AllNotch receptors show complementary and comb<strong>in</strong>atorialexpression patterns dur<strong>in</strong>g the development ofvarious tissues, <strong>in</strong>clud<strong>in</strong>g the testis (Williams et al.1995; Mori et al. 2003). Immunocytochemistry revealedthat the Notch family prote<strong>in</strong>s are activated <strong>in</strong> specificgerm cell types <strong>in</strong> the sem<strong>in</strong>iferous tubules dur<strong>in</strong>g germcell development, and that their ligands Jagged-1 andJagged-2 are expressed by Sertoli cells (Dirami et al.2001). We observed that the expression of the Notch-1<strong>in</strong>tracytoplasmic doma<strong>in</strong> (N1-ICD) starts before birth <strong>in</strong>gonocytes, <strong>in</strong>creases as the germ cells proliferate andÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Regulation of the Spermatogonial Stem Cell Niche 389(a)(b)Fig. 4. Influence of GDNF andJagged-1 on SSCs <strong>in</strong> vitro. (a)Example of a colony of SSCsgrown <strong>in</strong> vitro (from Braydich-Stolle et al. 2007). (b) Example of acha<strong>in</strong> of differentiat<strong>in</strong>g spermatogoniagrown <strong>in</strong> vitro (from Hofmannet al. 2006). (c) Number ofcolonies and cha<strong>in</strong>s of spermatogoniaobta<strong>in</strong>ed after cultur<strong>in</strong>gSSCs with 100 ng ⁄ ml GDNF for 3and 6 days. (d) Number of coloniesand cha<strong>in</strong>s of spermatogonia obta<strong>in</strong>edafter cultur<strong>in</strong>g SSCs with10 lg ⁄ ml Jagged-1 for 3 and6 days. A significant <strong>in</strong>crease <strong>in</strong> thenumber of cha<strong>in</strong>s can be observed,<strong>in</strong>dicat<strong>in</strong>g that Jagged-1 mediatesSSC differentiation (*p < 0.05,Student’s t-test)(c)(d)differentiate <strong>in</strong>to type A and B spermatogonia, andpeaks <strong>in</strong> spermatocytes <strong>in</strong>dicat<strong>in</strong>g that activated Notch-1 might function to ma<strong>in</strong>ta<strong>in</strong> the proliferation of germcells as they differentiate. Further, addition of Jagged-1<strong>in</strong> the culture media of freshly isolated SSCs <strong>in</strong>creasedtheir differentiation, as the number of Aaligned cellssignificantly <strong>in</strong>creased <strong>in</strong> these cultures <strong>in</strong> comparisonwith the controls (Fig. 4). Suppression of Notch-1signall<strong>in</strong>g by us<strong>in</strong>g antibodies aga<strong>in</strong>st Notch-1 orJagged-2 led to maturation arrest <strong>in</strong> the rat testis(Hayashi et al. 2001). To evaluate a possible relationshipbetween an alteration of Notch signall<strong>in</strong>g and thepathogenesis of maturation arrest, Hayashi and colleaguesexam<strong>in</strong>ed the expression of Notch-1 and itsligand Jagged-2 <strong>in</strong> the testis of non-obstructive <strong>in</strong>fertilepatients (Hayashi et al. 2004). Patients showed maturationarrest at the spermatogonia, spermatocytes orspermatids stage. Patients with only spermatogonia <strong>in</strong>their sem<strong>in</strong>iferous tubules lacked expression of bothNotch-1 and Jagged-2 surface prote<strong>in</strong>s. Patients exhibit<strong>in</strong>ggerm cells that reached the spermatocyte andspermatid stage lacked either Notch-1 or Jagged-2.None of the patients affected with maturation arrestexpressed both Notch-1 and Jagged-2. These results<strong>in</strong>dicate that Notch-1 is specifically <strong>in</strong>volved <strong>in</strong> germ celldifferentiation <strong>in</strong> humans, and that some compensationmechanisms by other Notch family members or Jagged-1 can occur.An <strong>in</strong>terest<strong>in</strong>g possibility is that factors <strong>in</strong>duced byGDNF <strong>in</strong> SSCs would down-regulate Notch-1, and thusfavour self-renewal at the expense of differentiation. Forexample, microarray analysis of GDNF-regulated genes<strong>in</strong>dicated that Numb, an antagonist of Notch signall<strong>in</strong>g,is up-regulated (Braydich-Stolle et al. 2005; Hofmannet al. 2005b). In vitro, we observed that Numb promotesthe degradation of Notch-1 at the prote<strong>in</strong> level <strong>in</strong> germl<strong>in</strong>estem cells. Immunocytochemistry of Numb expression<strong>in</strong> sections of sem<strong>in</strong>iferous tubules parallels theexpression of Notch; it is found <strong>in</strong> type A spermatogoniaand peaks <strong>in</strong> spermatocytes, <strong>in</strong>dicat<strong>in</strong>g that Notch-1is tightly regulated dur<strong>in</strong>g the first step of spermatogenesis(Corall<strong>in</strong>i et al. 2006).Regulation of the Spermatogonial Stem CellNicheRecent studies have demonstrated that GDNF productionby Sertoli cells is regulated by FGF2 (Simon et al.2007). However, FGF2 knockout mice are viable andfertile, <strong>in</strong>dicat<strong>in</strong>g that other factors are necessary. Thesefactors <strong>in</strong>clude <strong>in</strong>terleuk<strong>in</strong>-1 beta (IL-1b) and tumournecrosis factor alpha (TNFa), which are producedessentially by monocytes, macrophages and dendriticcells (Simon et al. 2007). It is known that macrophagesconstitute approximately 20% of the <strong>in</strong>terstitial tissue ofthe testis, where they are a major source of cytok<strong>in</strong>es(Kern et al. 1995). In addition, GDNF production bySertoli cells is also regulated by FSH (Tadokoro et al.2002). Therefore, these data confirm that the stem cellniche is regulated at least <strong>in</strong> part by the vasculature andcellular components of the <strong>in</strong>terstitium between thesem<strong>in</strong>iferous tubules. The regulation of Jagged-1 ⁄ 2<strong>in</strong>Sertoli cells is not well understood, but studies havesuggested that EGF might up-regulate Jagged-2 through<strong>in</strong>teraction with the Sonic Hedgehog ⁄ Gli signall<strong>in</strong>gpathway (Kasper et al. 2006).Another molecule essential for the ma<strong>in</strong>tenance of theSSC niche and spermatogenesis is the Ets-related moleculeEtv5 (Chen et al. 2005; Hess et al. 2006). The Etv5is a transcription factor expressed by Sertoli cells andgerm cells. While Etv5 expression is under control ofGDNF <strong>in</strong> germ cells, it is driven by FGF2 and EGF <strong>in</strong>Sertoli cells (Oatley et al. 2006; Simon et al. 2007). TheEtv5 b<strong>in</strong>ds to other transcription factors to up-regulatethe expression of target genes (Gutierrez-Hartmannet al. 2007), and it is likely that its b<strong>in</strong>d<strong>in</strong>g partners andfunction <strong>in</strong> Sertoli cells and germ cells are different. Inmice with a targeted deletion of etv5 (etv5- ⁄ -), SSCs failto renew, and these cells are lost through differentiation.Microarray analysis revealed that Etv-5 expression wasessential for the production of several chemok<strong>in</strong>es,<strong>in</strong>clud<strong>in</strong>g Stromal cell-derived factor (SDF-1, CXCL-12), CXCL5 and CCL7 (Chen et al. 2005). In addition,Etv5 seems important for the expression of the matrixmetalloprote<strong>in</strong>ase-12 (MMP-12). As chemok<strong>in</strong>es andÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


390 N Kostereva and M-C HofmannMMPs are well known for their <strong>in</strong>volvement <strong>in</strong> stem cellrecruitment, migration and hom<strong>in</strong>g (Heissig et al. 2002;Von Luttichau et al. 2005), it appears that <strong>in</strong> Sertolicells, Etv5 is responsible for the production of factorsthat reta<strong>in</strong> stem cells <strong>in</strong>side their niches. Additionalstudies also suggest that the blood–testis barrier (Sertoli–Sertolitight junctional complex) is abnormal <strong>in</strong> theEtv5(- ⁄ -) mice, and several chemok<strong>in</strong>es and MMPs havebeen shown to be critical <strong>in</strong> modulat<strong>in</strong>g various componentsthat contribute to blood-testes barrier function(Morrow et al. 2008).Alterations of the spermatogonial stem cell niche willoccur with ag<strong>in</strong>g, and these changes contribute todeficient stem cell number and activity. This wasrecently demonstrated by transplant<strong>in</strong>g SSCs fromyoung, fertile male mice <strong>in</strong>to the 1-year and 2-yearatrophied testes of old males (Zhang et al. 2006). Theresults showed that 1-year-old testes are permissive forregeneration of spermatogenesis, while the 2-year-oldtestes are not, suggest<strong>in</strong>g a gradual change <strong>in</strong> the SSCmicroenvironment. Further studies demonstrated asignificant decrease of the production of GDNF bySertoli cells with age, which could <strong>in</strong> part expla<strong>in</strong> thedecl<strong>in</strong>e of stem cell numbers (Ryu et al. 2006). However,<strong>in</strong> reciprocal transplantation experiments, colonizationof young testes by 2-year-old SSCs was not as efficient ascolonization by 1-year-old SSCs, <strong>in</strong>dicat<strong>in</strong>g that stemcell <strong>in</strong>tr<strong>in</strong>sic factors are also altered as the animal ages(Zhang et al. 2006). Age-related decl<strong>in</strong>e of the spermatogonialstem cell niche occurs as well <strong>in</strong> theDrosophila testis, where the decrease of expression of akey self-renewal signal, Unpaired (Upd), correlates witha decrease <strong>in</strong> SSCs with ag<strong>in</strong>g (Boyle et al. 2007).Conversely, forced expression of Upd by the somaticcells of the niche ma<strong>in</strong>ta<strong>in</strong>s SSCs <strong>in</strong> older males.Therefore, similar molecular mechanisms with<strong>in</strong> thetesticular niche ma<strong>in</strong>ta<strong>in</strong> self-renewal across species, andtheir alterations contribute to a decl<strong>in</strong>e of the stem cellpool and spermatogenesis.ConclusionSpermatogonial stem cells are at the orig<strong>in</strong> of spermatogenesis.Their fate is regulated by a complex <strong>in</strong>terplay ofgrowth factors produced by Sertoli cells and other germcells, the extracellular matrix and vasculature components.The ensemble of these factors is called thespermatogonial stem cell niche. The behaviour of theSSCs is the result of <strong>in</strong>teractions between the signalsgiven from the niche and stem cell <strong>in</strong>tr<strong>in</strong>sic factors suchas k<strong>in</strong>ases, phosphatases and transcription factors.Growth factors provided by the niche that regulate thefate of SSCs <strong>in</strong>clude GDNF, stem cell factor (SCF) andJagged-1 ⁄ 2. There is little known about the mechanismsthat regulate the production of these factors by Sertolicells, but FGF2, FSH and cytok<strong>in</strong>es produced by<strong>in</strong>terstitial cells seem <strong>in</strong>volved. Another molecule thatregulates the function of the niche is the Sertoli celltranscription factor Etv5. By ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g the blood–testis barrier, and by regulat<strong>in</strong>g the production ofchemok<strong>in</strong>es, and metalloprote<strong>in</strong>ases by Sertoli cells,Etv5 contributes to the hom<strong>in</strong>g and physical ma<strong>in</strong>tenanceof SSCs with<strong>in</strong> the niche.AcknowledgementsFunded by NIH grants R01-HD044543 and K02-HD054607.ReferencesAdams G, Scadden D, 2006: The hematopoietic stem cell <strong>in</strong> itsplace. Nat Immunol 7, 333–337.Airaks<strong>in</strong>en M, Saarma M, 2002: The GDNF family: signall<strong>in</strong>g,biological functions and therapeutic value. Nat RevNeurosci 3, 383–394.Boyle M, Wong C, Rocha M, Jones D, 2007: Decl<strong>in</strong>e <strong>in</strong> selfrenewalfactors contributes to ag<strong>in</strong>g of the stem cell niche <strong>in</strong>the Drosophila testis. Cell Stem Cell 1, 470–478.Braydich-Stolle L, Nolan C, Dym M, Hofmann M, 2005: Roleof glial cell l<strong>in</strong>e-derived neurotrophic factor <strong>in</strong> germ-l<strong>in</strong>estem cell fate. Ann N Y Acad Sci 1061, 94–99.Braydich-Stolle L, Kostereva N, Dym M, Hofmann M, 2007:Role of Src family k<strong>in</strong>ases and N-Myc <strong>in</strong> spermatogonialstem cell proliferation. Dev Biol 304, 34–45.Br<strong>in</strong>ster R, 2002: Germl<strong>in</strong>e stem cell transplantation andtransgenesis. Science 296, 2174–2176.Buaas F, Kirsh A, Sharma M, Mclean DJ, Morris J, GriswoldM, De Rooij DG, Braun R, 2004: Plzf is required <strong>in</strong> adultmale germ cells for stem cell self-renewal. Nat Genet 36,647–652.Buageaw A, Sukhwani M, Ben-Yehudah A, Ehmcke J, RaweV, Pholpramool C, Orwig K, Schlatt S, 2005: GDNFfamily receptor alpha1 phenotype of spermatogonial stemcells <strong>in</strong> immature mouse testes. Biol Reprod 73, 1011–1016.Chen C, Ouyang W, Grigura V, Zhou Q, Carnes K, LimH, Zhao G, Arber S, Kurpios N, Murphy TL, Cheng A,Hassell J, Chandrashekar V, Hofmann MC, Hess R, MurphyKM, 2005: ERM is required for transcriptionalcontrol of the spermatogonial stem cell niche. Nature436, 1030–1034.Corall<strong>in</strong>i S, Fera S, Grisanti L, Falciatori I, Muciaccia B,Stefan<strong>in</strong>i M, Vic<strong>in</strong>i E, 2006: Expression of the adaptorprote<strong>in</strong> m-Numb <strong>in</strong> mouse male germ cells. <strong>Reproduction</strong>132, 887–897.Costoya JA, Hobbs R, Barna M, Cattoretti G, Manova K,Sukhwani M, Orwig K, Wolgemuth D, Pandolfi P, 2004:Essential role of Plzf <strong>in</strong> ma<strong>in</strong>tenance of spermatogonial stemcells. Nat Genet 36, 653–659.De Rooij DG, Russell L, 2000: All you wanted to knowabout spermatogonia but were afraid to ask. J Androl 21,776–798.Desdouets C, Matesic G, Mol<strong>in</strong>a CA, Foulkes N, Sassone-Corsi P, Brechot C, Sobczak-Thepot J, 1995: Cell cycleregulation of cycl<strong>in</strong> A gene expression by the cyclic AMPresponsivetranscription factors CREB and CREM. MolCell Biol 15, 3301–3309.Dirami G, Rav<strong>in</strong>dranath N, Achi M, Dym M, 2001:Expression of Notch pathway components <strong>in</strong> spermatogoniaand Sertoli cells of neonatal mice. J Androl 22, 944–952.Dolci S, Pellegr<strong>in</strong>i M, Di AS, Geremia R, Rossi P, 2001:Signal<strong>in</strong>g through extracellular signal-regulated k<strong>in</strong>ase isrequired for spermatogonial proliferative response to stemcell factor. J Biol Chem 276, 40225–40233.Dym M, Fawcett D, 1971: Further observations on thenumbers of spermatogonia, spermatocytes, and spermatidsconnected by <strong>in</strong>tercellular bridges <strong>in</strong> the mammalian testis.Biol Reprod 4, 195–215.Falender A, Freiman R, Geles K, Lo K, Hwang K, Lamb D,Morris PL, Tjian R, Richards J, 2005: Ma<strong>in</strong>tenance ofspermatogenesis requires TAF4b, a gonad-specific subunitof TFIID. Genes Dev 19, 794–803.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Regulation of the Spermatogonial Stem Cell Niche 391Filipponi D, Hobbs R, Ottolenghi S, Rossi P, Jann<strong>in</strong>i E,Pandolfi P, Dolci S, 2007: Repression of kit expression byPlzf <strong>in</strong> germ cells. Mol Cell Biol 27, 6770–6781.Gutierrez-Hartmann A, Duval D, Bradford A, 2007: ETStranscription factors <strong>in</strong> endocr<strong>in</strong>e systems. Trends Endocr<strong>in</strong>olMetab 18, 150–158.Hayashi T, Kageyama Y, Ishizaka K, Xia G, Kihara K,Oshima H, 2001: Requirement of Notch 1 and its ligandjagged 2 expressions for spermatogenesis <strong>in</strong> rat and humantestes. J Androl 22, 999–1011.Hayashi T, Yamada T, Kageyama Y, Negishi T, Kihara K,2004: Expression failure of the Notch signal<strong>in</strong>g system isassociated with the pathogenesis of maturation arrest <strong>in</strong>male <strong>in</strong>fertility patients. Fertil Steril 81, 697–699.He Z, Jiang J, Kokk<strong>in</strong>aki M, Golestaneh N, Hofmann M,Dym M, 2008: GDNF up-regulates c-fos transcription viathe Ras ⁄ ERK1 ⁄ 2 pathway to promote mouse spermatogonialstem cell proliferation. Stem Cells 26, 266–278.Heissig B, Hattori K, Dias S, Friedrich M, Ferris B, HackettN, Crystal RG, Besmer P, Lyden D, Moore M, Werb Z,Rafii S, 2002: Recruitment of stem and progenitor cells fromthe bone marrow niche requires MMP-9 mediated release ofkit-ligand. Cell 109, 625–637.Hess R, Cooke P, Hofmann M, Murphy K, 2006: Mechanistic<strong>in</strong>sights <strong>in</strong>to the regulation of the spermatogonial stem cellniche. Cell Cycle 5, 1164–1170.Hofmann M, Braydich-Stolle L, Dett<strong>in</strong> L, Johnson E, Dym M,2005a: Immortalization of mouse germ l<strong>in</strong>e stem cells. StemCells 23, 200–210.Hofmann M, Braydich-Stolle L, Dym M, 2005b: Isolation ofmale germ-l<strong>in</strong>e stem cells; <strong>in</strong>fluence of GDNF. Dev Biol 279,114–124.Huck<strong>in</strong>s C, 1971: The spermatogonial stem cell population <strong>in</strong>adult rats. I. Their morphology, proliferation and maturation.Anat Rec 169, 533–557.Kanatsu-Sh<strong>in</strong>ohara M, Ogonuki N, Inoue K, Miki H, OguraA, Toyokuni S, Sh<strong>in</strong>ohara T, 2003: Long-term proliferation<strong>in</strong> culture and germl<strong>in</strong>e transmission of mouse male germl<strong>in</strong>estem cells. Biol Reprod 69, 612–616.Kasper M, Schnidar H, Neill G, Hanneder M, Kl<strong>in</strong>gler S, BlaasL, Schmid C, Hauser-Kronberger C, Regl G, Philpott M,Aberger F, 2006: Selective modulation of Hedgehog ⁄ GLItarget gene expression by epidermal growth factor signal<strong>in</strong>g<strong>in</strong> human kerat<strong>in</strong>ocytes. Mol Cell Biol 26, 6283–6298.Kern S, Robertson S, Mau V, Maddocks S, 1995: Cytok<strong>in</strong>esecretion by macrophages <strong>in</strong> the rat testis. Biol Reprod 53,1407–1416.Kubota H, Avarbock M, Br<strong>in</strong>ster R, 2003: Spermatogonialstem cells share some, but not all, phenotypic and functionalcharacteristics with other stem cells. Proc Natl Acad SciUSA 100, 6487–6492.Kubota H, Avarbock M, Br<strong>in</strong>ster R, 2004: Growth factorsessential for self-renewal and expansion of mouse spermatogonialstem cells. Proc Natl Acad Sci USA 101, 16489–16494.Lardelli M, Dahlstrand J, Lendahl U, 1994: The novel Notchhomologue mouse Notch 3 lacks specific epidermal growthfactor-repeats and is expressed <strong>in</strong> proliferat<strong>in</strong>g neuroepithelium.Mech Dev 46, 123–136.Lee J, Kanatsu-Sh<strong>in</strong>ohara M, Inoue K, Ogonuki N, Miki H,Toyokuni S, Kimura T, Nakano T, Ogura A, Sh<strong>in</strong>ohara T,2007: Akt mediates self-renewal division of mouse spermatogonialstem cells. Development 134, 1853–1859.Meistrich M, Van Beek M, 1993: Spermatogonial stem cells.In: Desjard<strong>in</strong>s C, Ew<strong>in</strong>g LL (eds), Spermatogonial StemCells. Oxford University Press, New York, pp. 266–295.Meng X, L<strong>in</strong>dahl M, Hyvonen M, Parv<strong>in</strong>en M, De Rooij DG,Hess M, Raatika<strong>in</strong>en-Ahokas A, Sa<strong>in</strong>io K, Rauvala H,Lakso M, Pichel JG, Westphal H, Saarma M, Sariola H,2000: Regulation of cell fate decision of undifferentiatedspermatogonia by GDNF. Science 287, 1489–1493.Moore M, Kle<strong>in</strong> R, Far<strong>in</strong>as I, Sauer H, Arman<strong>in</strong>i M, PhillipsH, Reichardt L, Ryan A, Carver-Moore K, Rosenthal A,1996: Renal and neuronal abnormalities <strong>in</strong> mice lack<strong>in</strong>gGDNF. Nature 382, 76–79.Mori S, Kadokawa Y, Hosh<strong>in</strong>aga K, Marunouchi T, 2003:Sequential activation of Notch family receptors dur<strong>in</strong>gmouse spermatogenesis. Dev Growth Differ 45, 7–13.Morrow C, Hostetler C, Griswold M, Hofmann MC, MurphyK, Cooke P, Hess R, 2008: ETV5 is required for cont<strong>in</strong>uousspermatogenesis <strong>in</strong> adult mice and may mediate blood testesbarrier function and testicular immune privilege. Ann N YAcad Sci 1120, 144–151.Nagano M, Br<strong>in</strong>ster C, Orwig K, Ryu B, Avarbock M,Br<strong>in</strong>ster R, 2001: Transgenic mice produced by retroviraltransduction of male germ-l<strong>in</strong>e stem cells. Proc Natl AcadSci USA 98, 13090–13095.Naughton C, Ja<strong>in</strong> S, Strickland A, Gupta A, Milbrandt J,2006: Glial cell-l<strong>in</strong>e derived neurotrophic factor (GDNF)-mediated RET signal<strong>in</strong>g regulates spermatogonial stem cellfate. Biol Reprod 74, 314–321.Oakberg E, 1971: Spermatogonial stem-cell renewal <strong>in</strong> themouse. Anat Rec 169, 515–531.Oatley JM, Avarbock M, Telaranta A, Fearon DT, Br<strong>in</strong>ster R,2006: Identify<strong>in</strong>g genes important for spermatogonial stemcell self-renewal and survival. Proc Natl Acad Sci USA 103,9524–9529.Oatley JM, Avarbock M, Br<strong>in</strong>ster R, 2007: Glial cell l<strong>in</strong>ederivedneurotrophic factor regulation of genes essential forself-renewal of mouse spermatogonial stem cells is dependenton Src family k<strong>in</strong>ase signal<strong>in</strong>g. J Biol Chem 282, 25842–25851.Ogawa T, Ohmura M, Yumura Y, Sawada H, Kubota Y,2003: Expansion of mur<strong>in</strong>e spermatogonial stem cellsthrough serial transplantation. Biol Reprod 68, 316–322.Pichel J, Shen L, Sheng H, Granholm A, Drago J, Gr<strong>in</strong>berg A,Lee E, Huang S, Saarma M, Hoffer B, Sariola H, WestphalH, 1996: Defects <strong>in</strong> enteric <strong>in</strong>nervation and kidney development<strong>in</strong> mice lack<strong>in</strong>g GDNF. Nature 382, 73–76.Rossi P, Lolicato F, Grimaldi P, Dolci S, Di Sauro A,Filipponi D, Geremia R, 2008: Transcriptome analysis ofdifferentiat<strong>in</strong>g spermatogonia stimulated with kit ligand.Gene Expr Patterns 8, 58–70.Ryu B, Orwig K, Oatley JM, Avarbock M, Br<strong>in</strong>ster R, 2006:Effects of ag<strong>in</strong>g and niche microenvironment on spermatogonialstem cell self-renewal. Stem Cells 24, 1505–1511.Sanchez MP, Silos-Santiago I, Frisen J, He B, Lira S, BarbacidM, 1996: Renal agenesis and the absence of enteric neurons<strong>in</strong> mice lack<strong>in</strong>g GDNF. Nature 382, 70–73.Schofield R, 1978: The relationship between the spleen colonyform<strong>in</strong>gcell and the haemopoietic stem cell. Blood Cells 4,7–25.Schuchardt A, D’agati V, Larsson-Blomberg L, Costant<strong>in</strong>i F,Pachnis V, 1994: Defects <strong>in</strong> the kidney and enteric nervoussystem of mice lack<strong>in</strong>g the tyros<strong>in</strong>e k<strong>in</strong>ase receptor Ret.Nature 367, 380–383.Shetty G, Meistrich M, 2007: The miss<strong>in</strong>g niche for spermatogonialstem cells: do blood vessels po<strong>in</strong>t the way? Cell StemCell 1, 361–363.Sh<strong>in</strong>ohara T, Avarbock M, Br<strong>in</strong>ster R, 1999: Beta1- andalpha6-<strong>in</strong>tegr<strong>in</strong> are surface markers on mouse spermatogonialstem cells. Proc Natl Acad Sci USA 96, 5504–5509.Simon L, Ekman G, Tyagi G, Hess R, Murphy KM, Cooke P,2007: Common and dist<strong>in</strong>ct factors regulate expression ofmRNA for ETV5 and GDNF, Sertoli cell prote<strong>in</strong>s essentialfor spermatogonial stem cell ma<strong>in</strong>tenance. Exp Cell Res 313,3090–3099.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


392 N Kostereva and M-C HofmannTadokoro Y, Yomogida K, Ohta H, Tohda A, Nishimune Y,2002: Homeostatic regulation of germ<strong>in</strong>al stem cell proliferationby the GDNF ⁄ FSH pathway. Mech Dev 113, 29–39.Uyttendaele H, Marazzi G, Wu G, Yan Q, Sassoon D,Kitajewski J, 1996: Notch4 ⁄ <strong>in</strong>t-3, a mammary proto-oncogene,is an endothelial cell-specific mammalian Notch gene.Development 122, 2251–2259.Von Luttichau I, Notohamiprodjo M, Wechselberger A,Peters C, Henger A, Seliger C, Djafarzadeh R, Huss R,Nelson PJ, 2005: Human adult CD34-progenitor cellsfunctionally express the chemok<strong>in</strong>e receptors CCR1,CCR4, CCR7, CXCR5, and CCR10 but not CXCR4. StemCells Dev 14, 329–336.Watt F, Hogan B, 2000: Out of Eden: stem cells and theirniches. Science 287, 1427–1430.We<strong>in</strong>master G, 1997: The <strong>in</strong>s and outs of notch signal<strong>in</strong>g. MolCell Neurosci 9, 91–102.We<strong>in</strong>master G, Roberts VJ, Lemke G, 1991: A homolog ofDrosophila Notch expressed dur<strong>in</strong>g mammalian development.Development 113, 199–205.Williams R, Lendahl U, Lardelli M, 1995: Complementaryand comb<strong>in</strong>atorial patterns of Notch gene family expressiondur<strong>in</strong>g early mouse development. Mech Dev 53, 357–368.Yoshida S, Sukeno M, Nabeshima Y, 2007: A vasculatureassociatedniche for undifferentiated spermatogonia <strong>in</strong> themouse testis. Science 317, 1722–1726.Zhang X, Ebata K, Robaire B, Nagano M, 2006: Ag<strong>in</strong>gof male germ l<strong>in</strong>e stem cells <strong>in</strong> mice. Biol Reprod 74, 119–124.Author’s address (for correspondence): Marie-Claude Hofmann,Department of Veter<strong>in</strong>ary Biosciences and Institute for GenomicBiology, University of Ill<strong>in</strong>ois at Urbana-Champaign, 2001 SouthL<strong>in</strong>coln Avenue, Urbana, IL 61802, USA. E-mail: mhofmann@uiuc.eduConflict of <strong>in</strong>terest: The authors declare no conflict of <strong>in</strong>terests.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 393–400 (2008); doi: 10.1111/j.1439-0531.2008.01190.xISSN 0936-6768Factors Affect<strong>in</strong>g Oocyte Quality: Who is Driv<strong>in</strong>g the Follicle?P Mermillod, R Dalbie` s-Tran, S Uzbekova, A The´lie, J-M Traverso, C Perreau, P Papillier and P MongetPhysiologie de la <strong>Reproduction</strong> et des Comportements, UMR 6175 INRA, CNRS, Universite´ de Tours, Haras Nationaux, Nouzilly, FranceContentsMammalian ovaries conta<strong>in</strong> a large stock of oocytes enclosed<strong>in</strong> primordial follicles. Ovarian cyclic activity <strong>in</strong>duces some ofthese follicles to <strong>in</strong>itiate growth towards a possible ovulation.However, most of these follicles term<strong>in</strong>ate their growth at anymoment and degenerate through atresia. In grow<strong>in</strong>g follicles,only a subset of oocytes are capable to support meiosis,fertilization and early embryo development to the blastocyststage, as shown through embryo <strong>in</strong> vitro production experiments.This proportion of competent oocytes is <strong>in</strong>creas<strong>in</strong>galong with follicular size. Grow<strong>in</strong>g l<strong>in</strong>es of evidence suggestthat oocyte competence relies on the storage of gene products(messenger RNA or prote<strong>in</strong>) that will be determ<strong>in</strong>ant tosupport early stages of embryo development, before fullactivation of embryonic genome. Given these facts, thequestion is: are these gene products stored <strong>in</strong> oocytes dur<strong>in</strong>glate folliculogenesis, allow<strong>in</strong>g an <strong>in</strong>creas<strong>in</strong>g proportion of themto become competent? Alternatively, these transcripts may bestored dur<strong>in</strong>g early folliculogenesis as the oocyte grows anddisplays high transcription activity. Several arguments supportthis latter hypothesis and are discussed <strong>in</strong> this review: (i) manyattempts at prolonged culture of oocytes from antral follicleshave failed to <strong>in</strong>crease developmental competence, suggest<strong>in</strong>gthat developmental competence may be acquired before antralformation; (ii) the recent discovery of oocyte secreted factorsand of their ability to regulate many parameters of surround<strong>in</strong>gsomatic cells, possibly <strong>in</strong>fluenc<strong>in</strong>g the fate of folliclesbetween ovulation or atresia, suggests a central role of oocytequality <strong>in</strong> the success of folliculogenesis. F<strong>in</strong>ally, <strong>in</strong> addition totheir role <strong>in</strong> <strong>in</strong>terfollicular regulation of ovulation rate, latefolliculogenesis regulation and atresia could also be seen as aselective process aimed at the elim<strong>in</strong>ation through follicularatresia of oocytes that did not succeed to store proper geneproducts set dur<strong>in</strong>g their growth.IntroductionIn mammals, the ovulation delivers a very special cell:the oocyte. This cell has encountered a long andcomplex process of differentiation, lead<strong>in</strong>g to thereduction of DNA complement from 2n to n and tothe preparation of the cytoplasm to orchestrate thefusion of the male and female genome complements andthe remodell<strong>in</strong>g of the result<strong>in</strong>g complete genomecapable to ensure the early development and cell l<strong>in</strong>eagedifferentiation.These particularities place the mammalian oocyte atthe centre of the procreation process. In addition, therecent emergence of embryo-based technologies [<strong>in</strong> vitrofertilization (IVF), transgenesis and clon<strong>in</strong>g] has re<strong>in</strong>forcedthe <strong>in</strong>terest of the scientific community <strong>in</strong>to theoocyte. Beyond these applications, the study of oocytefunctions and specificities provides <strong>in</strong>terest<strong>in</strong>g clues forthe better comprehension of several basic biologicalprocesses (cell cycle regulation, genome silenc<strong>in</strong>g, posttranscriptionalregulation of gene expression, etc.).The oocyte enters the meiotic division cycle <strong>in</strong> thefoetal gonad and stops the meiotic progression at thelate prophase stage (diplotene stage, germ<strong>in</strong>al vesicle)around the time of birth, depend<strong>in</strong>g on the species. Itrema<strong>in</strong>s at this meiotic stage for the complete durationof oocyte growth <strong>in</strong> the ovarian follicle. Dur<strong>in</strong>g folliculogenesis,the oocyte grows, undergoes modifications ofits ultrastructure and stores the RNA and prote<strong>in</strong>materials necessary to f<strong>in</strong>ally become competent toresume and complete maturation, support fertilizationand <strong>in</strong>itiate chromat<strong>in</strong> remodell<strong>in</strong>g and embryo development.These different competencies are sequentialacquisitions dur<strong>in</strong>g oocyte differentiation (Sirard et al.2006). The ability to resume meiosis [germ<strong>in</strong>al vesiclebreakdown (GVBD)], to progress to metaphase I and tometaphase II may appear sequentially <strong>in</strong> some specieslike sheep and goats (Mermillod et al. 1999). In cattle,one can consider that all oocytes are meioticallycompetent soon after antrum formation. Indeed,oocytes from antral follicles spontaneously resumemeiosis when placed <strong>in</strong> culture (Edwards 1965).At this stage of meiotic competence, not all oocytesare able to be fertilized and to <strong>in</strong>itiate early embryodevelopment. These acquisitions seem to be progressive<strong>in</strong> the course of follicular growth. It means that theproportion of competent oocytes <strong>in</strong>creases dur<strong>in</strong>g folliculogenesis,but at the end very few oocytes reach thef<strong>in</strong>al stage of full competence because of the loss of mostfollicles by physiological atresia occurr<strong>in</strong>g at any time oftheir growth. Therefore, the progression of the proportionof competent oocytes with<strong>in</strong> the population ofgrow<strong>in</strong>g follicles may be the result of late oocytedifferentiation through accumulation of factors (messengers,prote<strong>in</strong>s), which will be <strong>in</strong>volved <strong>in</strong> the successof early developmental steps or the result of the selectionof follicles conta<strong>in</strong><strong>in</strong>g the more fit oocytes, <strong>in</strong> an<strong>in</strong>creas<strong>in</strong>gly selective hormonal environment and challeng<strong>in</strong>g<strong>in</strong>terfollicular regulation.The scope of this short review will be to def<strong>in</strong>e oocytecompetence, to describe experimental models designed<strong>in</strong> domestic species for the study of this competence andto discuss respective <strong>in</strong>puts of oocyte and surround<strong>in</strong>gsomatic cells <strong>in</strong> driv<strong>in</strong>g an ovarian follicle to ovulationand delivery of a fully competent oocyte.Oocyte Competence Def<strong>in</strong>ition and EvaluationDevelopmental competence of the oocyte (or oocytequality) may be def<strong>in</strong>ed as its ability to mature, befertilized and give rise to normal and fertile offspr<strong>in</strong>gafter normal gestation (Duranthon and Renard 2001).Although the ability of oocytes to reach theblastocyst stage <strong>in</strong> culture is not a perfect reflection ofÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


394 P Mermillod, R Dalbie` s-Tran, S Uzbekova, A The´lie, J-M Traverso, C Perreau, P Papillier and P Mongetdevelopmental competence, it could be used as a filter todiscrim<strong>in</strong>ate between oocyte populations, which aremore or less advanced on the way to competence. In thisview, <strong>in</strong> vitro production (IVP) of embryos is commonlyused as a means to diagnose the competence of oocytebatches represent<strong>in</strong>g different physio-pathological situations.Additional criteria may be used to <strong>in</strong>vestigate thequality of result<strong>in</strong>g embryos such as the evaluation ofthe level of expression of specific gene sets (Corcoranet al. 2005; Knijn et al. 2005), embryo metabolism, or<strong>in</strong> vitro survival of embryos after freez<strong>in</strong>g or vitrification(Rizos et al. 2001). Nevertheless, embryo transfer,pregnancy results and offspr<strong>in</strong>g viability rema<strong>in</strong> theultimate way to f<strong>in</strong>ally conclude about oocyte quality.In vitro production experiments provide <strong>in</strong>formationabout the proportion of competent oocytes <strong>in</strong> a batch.In addition to this global evaluation, experimentalprotocols have been developed to analyze the competenceof <strong>in</strong>dividual oocytes (Carolan et al. 1996; Fenget al. 2007). In these conditions, it becomes possible tocorrelate oocyte competence to some environmentalparameters (i.e. follicular characteristics). This approachallows for a more precise evaluation of the l<strong>in</strong>ks betweencumulus oocyte complex (COC) morphology or otherfollicular parameters and quality of the enclosed oocyte.Several morphological or molecular markers ofoocyte quality have been proposed (Wang and Sun2007). Because the presence of cumulus cells is requiredfor the success of <strong>in</strong> vitro maturation (IVM) and IVF,morphological evaluation is usually applied to the wholeCOC. Compact, cont<strong>in</strong>uous multilayer cumulus <strong>in</strong>vestmentand bright, homogeneous ooplasm are consideredas a sign of immature oocyte quality, whereas <strong>in</strong>complete,expanded, granulated or too th<strong>in</strong> (less than threelayers) cumulus or dark, heterogeneous oocyte cytoplasmare related to lower quality immature oocytes(Blond<strong>in</strong> and Sirard 1995).Different <strong>in</strong>tr<strong>in</strong>sic oocyte parameters may be measuredsuch as mitochondrial activity, calcium, ATP orglutathione contents, phosphodiesterase activity, baxtranscript expression, etc. (Wang and Sun 2007). Despitethe fact that the evaluation of these parameters is<strong>in</strong>vasive and requires oocyte destruction, they could beused to evaluate global quality of oocytes from different<strong>in</strong> vivo or <strong>in</strong> vitro treatments, but they cannot beconsidered as predictive parameters. Brilliant cresyl blue(BCB) sta<strong>in</strong><strong>in</strong>g has been proposed as an easy and vitalway to evaluate glucose 6-phosphate dehydrogenase(G6PDH) activity <strong>in</strong> oocytes (Alm et al. 2005). The blueBCB sta<strong>in</strong> is converted to colourless by G6PDHenzymatic activity. BCB sta<strong>in</strong>ed immature oocytes (i.e.oocytes with low G6PDH activity, rema<strong>in</strong><strong>in</strong>g blue afterBCB treatment) provided higher blastocyst rate ascompared with BCB negative oocytes or control ones,<strong>in</strong>dicat<strong>in</strong>g a negative correlation between G6PDHactivity level and oocyte quality. In addition, BCBsta<strong>in</strong><strong>in</strong>g before IVM did not affect IVP results, mak<strong>in</strong>gthis method an appropriate way of predictive oocytequality evaluation.Molecular markers of oocyte quality could also beidentified <strong>in</strong> surround<strong>in</strong>g tissues, like cumulus cells, or <strong>in</strong>follicular fluid. For example, the rate of apoptosis <strong>in</strong>bov<strong>in</strong>e cumulus cells before (Zeuner et al. 2003; Fenget al. 2007) or dur<strong>in</strong>g (Ikeda et al. 2003) IVM isnegatively correlated to oocyte quality. This is ofparticular <strong>in</strong>terest because, whereas apoptosis evaluationrequires the destruction of the cumulus cells, thismeasure could be performed on cumulus biopsy <strong>in</strong> thecourse of an IVF process or on cells removed frommature oocytes <strong>in</strong> an <strong>in</strong>tracytoplasmic sperm <strong>in</strong>jectionprocess.Oocyte Differentiation In vivoIt has been well established <strong>in</strong> several domestic andmodel species that the proportion of competent oocytes<strong>in</strong>creases with follicular size (cattle: Lonergan et al.1994; Lequarre et al. 2005; pig: Marchal et al. 2002;mouse: Eppig et al. 1992). This is represented <strong>in</strong> Fig. 1for cattle oocytes from follicles of less vs more than4 mm <strong>in</strong> diameter. The current hypothesis to expla<strong>in</strong> thisobservation is that germ<strong>in</strong>al vesicle arrested oocytescont<strong>in</strong>ue their functional differentiation dur<strong>in</strong>g thewhole course of follicular growth to f<strong>in</strong>ally reach fullcompetence (Mermillod et al. 1999). However, this also<strong>in</strong>dicates that even small classes of antral folliclesalready conta<strong>in</strong> a small proportion of fully competentoocytes. Therefore, the k<strong>in</strong>etics of oocyte differentiationalong the course of follicular growth is different fromone follicle to another.Several physiological factors are known to <strong>in</strong>terferewith the k<strong>in</strong>etics of evolution of the ovarian oocytepopulation towards competence. For example, <strong>in</strong> sheep,it has been shown that oocytes from ewes heterozygousfor the booroola fecundity gene were larger <strong>in</strong> diameterand more competent for development as compared withoocytes collected from follicles of the same size class <strong>in</strong>wild genotype ewes (Cognie et al. 1998). Interest<strong>in</strong>gly,the booroola mutation has been identified as a s<strong>in</strong>glenucleotide <strong>in</strong>activat<strong>in</strong>g mutation of the BMPRIb receptorof TGF-b (Transform<strong>in</strong>g Growth Factor-beta)superfamily members (Mulsant et al. 2001), which<strong>in</strong>cludes several oocyte secreted factors (OSF).In cattle, the age of the donor is <strong>in</strong>fluenc<strong>in</strong>g oocytequality. Oocyte collected from pre-pubertal calves havebeen shown to be less competent as compared withtheir adult counterparts (Khatir et al. 1996). TheFig. 1. Rate of cleavage 48 h post-<strong>in</strong>sem<strong>in</strong>ation (open bars) anddevelopment to the blastocyst stage on Day 7 post-<strong>in</strong>sem<strong>in</strong>ation (blackbars) for cattle oocytes collected from follicles of less than 4 mm <strong>in</strong>diameter (n = 203), from 4 to 5 mm (n = 260) and more than 6 mm(n = 109). Percentage of total oocytes, mean from five replicates± SEM (a, b: p < 0.05). Adapted from (Lequarre et al. 2005)Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Factors Affect<strong>in</strong>g Oocyte Quality 395physiological status of the donor cow also affects oocytequality. For example, oocytes from small follicles aremore competent dur<strong>in</strong>g follicular growth phase ascompared with oocyte collected dur<strong>in</strong>g the dom<strong>in</strong>antphase (Machatkova et al. 2004). In addition, <strong>in</strong>dividualdifferences between donor cows <strong>in</strong> the developmentalcompetence of oocytes collected by ovum pick up may<strong>in</strong>dicate genetic control of oocyte competence (Tamassiaet al. 2003).In mammals, atresia is observed at any stage offollicular growth (Cahill et al. 1979). One of the earliestsigns of follicular atresia is the drop of estradiolproduction by granulosa cells. We measured aromataseactivity <strong>in</strong> granulosa cells of dissected bov<strong>in</strong>e follicleswhile the correspond<strong>in</strong>g oocytes were submitted <strong>in</strong>dividuallyto IVP and we observed that oocytes able todevelop to the blastocyst stage were orig<strong>in</strong>at<strong>in</strong>g fromfollicles display<strong>in</strong>g high aromatase activity (Mermillodet al. 1999). Interest<strong>in</strong>gly, when atresia was <strong>in</strong>duced byprolonged follicular phase under gonadotrop<strong>in</strong> suppression<strong>in</strong> superovulated heifers, oocyte quality was notaffected (Oussaid et al. 2000). This contrast may <strong>in</strong>dicatethat grow<strong>in</strong>g follicles conta<strong>in</strong><strong>in</strong>g low-quality oocyteare <strong>in</strong>duced <strong>in</strong>to atresia, whereas artificially <strong>in</strong>duc<strong>in</strong>gatresia of large follicles does not affect oocyte quality.Therefore, atresia appears a consequence of oocyte lowquality rather than a cause of oocyte degradation.Modify<strong>in</strong>g hormonal environment by exogenous FSH<strong>in</strong>jection allows more follicles to reach ovulation. Thisprocedure is widely used <strong>in</strong> multiple ovulation andembryo transfer schemes <strong>in</strong> cattle (Lonergan 2007) andsmall rum<strong>in</strong>ant (Cognie et al. 2003) species. Optimizedtreatment <strong>in</strong>volv<strong>in</strong>g FSH stimulation followed by a 48-hcoast<strong>in</strong>g period even allowed to produce quite homogeneouspopulations of competent oocytes, reach<strong>in</strong>g up to80% of development to the blastocyst stage (Blond<strong>in</strong>et al. 2002). Modify<strong>in</strong>g the hormonal environmentreduces the selection pressure on follicles, result<strong>in</strong>g <strong>in</strong>a higher proportion of follicles progress<strong>in</strong>g to the preovulatorystage. However, some ultrastructure abnormalitieshave been reported <strong>in</strong> sheep superovulatedoocytes (O’Callaghan et al. 2000). This may be the resultof artificially driv<strong>in</strong>g to ovulation follicles conta<strong>in</strong><strong>in</strong>gpoorly fitted oocytes.Oocyte Differentiation In vitroBased on the hypothesis that developmental competenceis progressively acquired by an <strong>in</strong>creas<strong>in</strong>g proportion ofoocytes dur<strong>in</strong>g follicular growth, it would appear<strong>in</strong>terest<strong>in</strong>g to mimic this differentiation <strong>in</strong> vitro to allowmore oocytes to become competent. In vitro cont<strong>in</strong>uationof oocyte differentiation implies that culturedoocytes are ma<strong>in</strong>ta<strong>in</strong>ed at the germ<strong>in</strong>al vesicle stage toallow ongo<strong>in</strong>g regulation of expression of relevant genesdur<strong>in</strong>g a prematuration period. Several methods havebeen proposed to ma<strong>in</strong>ta<strong>in</strong> oocytes <strong>in</strong> meiotic arrest<strong>in</strong> vitro, <strong>in</strong>clud<strong>in</strong>g the use of chemicals aimed atma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g high cAMP level <strong>in</strong> oocytes such asforskol<strong>in</strong>, isobutyl methylxanth<strong>in</strong>e or cAMP stableanalogues (Sirard et al. 1998). Other drugs <strong>in</strong>hibit<strong>in</strong>gprote<strong>in</strong> synthesis (cycloheximide) or phosphorylation (6-dimethyl am<strong>in</strong>opur<strong>in</strong>e) were also tested (Lonergan et al.1997). All of these drugs were able to ma<strong>in</strong>ta<strong>in</strong> a more orless reversible <strong>in</strong>hibition of meiotic resumption <strong>in</strong>cultured cattle oocytes. However, no improvement ofoocyte quality was obta<strong>in</strong>ed after such <strong>in</strong>hibitions.More recently, drugs directly act<strong>in</strong>g by <strong>in</strong>hibition ofenzymatic activity of M-phase promot<strong>in</strong>g factor (MPF),the key element of G2 ⁄ M transition <strong>in</strong> eukaryotic cellcycle, were used to ma<strong>in</strong>ta<strong>in</strong> oocytes at the germ<strong>in</strong>alvesicle stage. MPF is a heterodimer of CDK1 catalyticsubunit and cycl<strong>in</strong> B regulatory subunit. Interest<strong>in</strong>gresults have been obta<strong>in</strong>ed by us<strong>in</strong>g puric moleculescompet<strong>in</strong>g with ATP for b<strong>in</strong>d<strong>in</strong>g the catalytic pocket ofMPF heterodimer. Butyrolactone I (Lonergan et al.2000) and roscovit<strong>in</strong>e (Mermillod et al. 2000) weresuccessfully used <strong>in</strong> this view and provided an efficientand reversible way to ma<strong>in</strong>ta<strong>in</strong> cattle oocytes at thegerm<strong>in</strong>al vesicle stage <strong>in</strong> culture for 24–48 h. However,as shown <strong>in</strong> Fig. 2, 24 h culture of germ<strong>in</strong>al vesicleoocyte did not result <strong>in</strong> improvement of developmentpotential even under different stimulation (Ponderatoet al. 2002). However, although these molecules allowedan efficient block of meiotic progression, several meiosisrelated cytoplasmic events did occur under <strong>in</strong>hibition,such as modifications of prote<strong>in</strong> synthesis andphosphorylation patterns, <strong>in</strong>dicat<strong>in</strong>g the <strong>in</strong>itiation ofcytoplasmic maturation (Vigneron et al. 2004a). Interest<strong>in</strong>gly,this approach allowed to po<strong>in</strong>t out severalMPF-<strong>in</strong>dependent signall<strong>in</strong>g pathways (Akt, JNK andAurora A) activated dur<strong>in</strong>g meiotic resumption (Vigneronet al. 2004b) and probably <strong>in</strong>volved <strong>in</strong> the regulationof some aspects of cytoplasmic maturation.Amongst these pathways, Aurora A, a ser<strong>in</strong>e ⁄ threon<strong>in</strong>eprote<strong>in</strong> k<strong>in</strong>ase, seems to be a major player <strong>in</strong> thephosphorylation cascade lead<strong>in</strong>g to MPF activation,meiotic resumption and post-transcriptional control ofgene expression (Uzbekova et al. 2008). Fail<strong>in</strong>g to<strong>in</strong>crease the quality of immature oocytes by allow<strong>in</strong>gthem to cont<strong>in</strong>ue their late differentiation dur<strong>in</strong>g aprematuration culture under chemical meiotic <strong>in</strong>hibitionmay be attributed to the diversity of the signall<strong>in</strong>gmechanisms <strong>in</strong>volved <strong>in</strong> the regulation of differentmaturation aspects. Alternatively, it may be due to anearlier determ<strong>in</strong>ation of oocyte quality, by the time ofhigher transcriptional activity <strong>in</strong> grow<strong>in</strong>g oocytes.Oocyte Gene ExpressionThe current hypothesis to expla<strong>in</strong> differential ability ofoocytes to develop after fertilization is that the oocyteshould store the appropriate set of mRNA and prote<strong>in</strong>sthat will be required dur<strong>in</strong>g the period of genomicsilenc<strong>in</strong>g from GVBD to major zygotic transition(MZT) and for the onset of MZT. Transcriptionalactivity is high <strong>in</strong> grow<strong>in</strong>g oocytes up to early antralstages of follicular growth and decreases to a basallevel thereafter, as follicular diameter reaches 3 mmand the diameter of the oocyte becomes 110 lm <strong>in</strong>bov<strong>in</strong>e (Fair et al. 1997). A transcription burst issuspected just before GVBD (Rodriguez and Far<strong>in</strong>2004a,b). Genes expressed at that time are supposed tobe <strong>in</strong>volved <strong>in</strong> meiotic resumption and progression butmay also have a role dur<strong>in</strong>g early development(Rodriguez et al. 2006).Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


396 P Mermillod, R Dalbie` s-Tran, S Uzbekova, A The´lie, J-M Traverso, C Perreau, P Papillier and P MongetEGF (n = 195)Rescovit<strong>in</strong>e (n = 202)Rescovit<strong>in</strong>e = EGF (n = 208)Fig. 2. Cleavage and rate of blastocystsat 7, 8 and 9 days post-<strong>in</strong>sem<strong>in</strong>ationfor oocytes matured <strong>in</strong>199 medium supplemented with10 ng ⁄ ml of epidermal growthfactor (EGF) either immediatelyafter collection (EGF) or after aprematuration treatment underroscovit<strong>in</strong>e <strong>in</strong>hibition <strong>in</strong> mediumalone (roscovit<strong>in</strong>e) or <strong>in</strong> the presenceof EGF. Means of three replicates.Inhibition of meioticresumption did not affect embryodevelopment, even under EGF stimulationNevertheless, if oocyte quality relies on mRNAstorage, these mRNAs could be stored dur<strong>in</strong>g ma<strong>in</strong>transcriptional activity <strong>in</strong> oocytes from pre-antral folliclesand kept <strong>in</strong> a stable form for future use dur<strong>in</strong>gmaturation or after fertilization. Indeed, whereas <strong>in</strong>somatic tissues, mRNA content directly reflects contemporaryprote<strong>in</strong> expression pattern, post-transcriptionalgene expression regulation is slightly different <strong>in</strong>oocyte where messengers could be masked and ma<strong>in</strong>ta<strong>in</strong>edfor long periods of time and then be processed fortranslation or destruction through polyadenylation regulation(Bettegowda and Smith 2007) or RNA <strong>in</strong>terferencemechanisms (Murchison et al. 2007).However, if it is admitted that oocyte developmentalcompetence may be acquired dur<strong>in</strong>g late folliculargrowth, one should admit that determ<strong>in</strong>ant messengersmay be produced <strong>in</strong> oocytes from late antral follicles.This transcription activity could be quantitatively lowbut functionally determ<strong>in</strong>ant. Much research has beendevoted towards identification of these critical mRNAdur<strong>in</strong>g the past 10 years.The development of highly sensitive molecular biologymethods has allowed to identify a panel of genesthat are preferentially expressed <strong>in</strong> oocytes of mammals(Cui and Kim 2007). The list of these genes is currentlyactively expand<strong>in</strong>g and will probably cont<strong>in</strong>ue to growdur<strong>in</strong>g the forthcom<strong>in</strong>g years, because of active researchwork and <strong>in</strong>creas<strong>in</strong>g sensitivity of molecular techniques.In particular, subtractive suppressive hybridization(SSH) approach allowed to evidence hundreds of genesoverexpressed <strong>in</strong> cattle oocytes as compared withsomatic tissues (Pennetier et al. 2005; Vallee et al. 2005).In mouse oocytes, knockout experiments allowed topo<strong>in</strong>t out several genes, which should be expressed <strong>in</strong>oocyte to allow embryo development after fertilization.These genes are referred to as ‘maternal effect genes’such as maternal antigen that embryo require (Mater) orzygotic arrest-1 (Zar-1). These genes may be <strong>in</strong>volved <strong>in</strong>the onset of transcriptional activity <strong>in</strong> zygote nucleusdur<strong>in</strong>g MZT occurr<strong>in</strong>g at the two-cell stage <strong>in</strong> mice(M<strong>in</strong>ami et al. 2007) and at the eight-cell stage <strong>in</strong>rum<strong>in</strong>ant species.Some orthologous genes of mouse maternal-effectgenes have been cloned <strong>in</strong> domestic species. For example,we cloned bov<strong>in</strong>e Mater and Zar-1 orthologousgenes <strong>in</strong> cattle (Pennetier et al. 2004; Uzbekova et al.2006). However, the study of bov<strong>in</strong>e Mater expressionprofile (Pennetier et al. 2006) <strong>in</strong>dicated that this genewas transcribed and translated at early stages offollicular growth, long before the suspected time ofdevelopmental competence acquisition. Although nomaternal effect gene has been shown to be translatedspecifically dur<strong>in</strong>g late folliculogenesis, it could beproposed that some determ<strong>in</strong>ant genes are progressivelyaccumulated dur<strong>in</strong>g this period and that full developmentalcompetence is acquired as a threshold of mRNAor prote<strong>in</strong> from these genes are reached <strong>in</strong> the oocyte.However, given that a significant population offollicles degenerate by atresia at different stages of theirgrowth, another explanation may be that folliclesconta<strong>in</strong><strong>in</strong>g competent oocytes are more able to survivethe <strong>in</strong>creas<strong>in</strong>gly challeng<strong>in</strong>g hormonal environmentthrough a better coord<strong>in</strong>ation of follicular somatic cellsproliferation and differentiation.Oocyte Regulation of Follicular GrowthThe role of somatic follicular cells <strong>in</strong> the establishmentof oocyte competence has been well established. Cumuluscells are participat<strong>in</strong>g to oocyte meiotic arrest andresumption and oocyte metabolism (glutathione storage,glucose metabolism), they also participate to oocyteÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Factors Affect<strong>in</strong>g Oocyte Quality 397capture by <strong>in</strong>fundibulum after ovulation and to fertilizationregulation (Van Soom et al. 2002).Dur<strong>in</strong>g the last few years, <strong>in</strong>creas<strong>in</strong>g l<strong>in</strong>es of evidenceshave suggested that the oocyte itself also regulates manyparameters of follicular somatic cells proliferation anddifferentiation. The TGF-b superfamily gathers morethan 35 members <strong>in</strong>volved <strong>in</strong> the regulation of severalphysiological processes <strong>in</strong> mammals. In particular,TGF-b, members are strongly <strong>in</strong>volved <strong>in</strong> many aspectsof ovarian function regulation (Knight and Glister2006). This superfamily <strong>in</strong>cludes different families ofgrowth factors such as TGF-b family, bone morphogeneticprote<strong>in</strong>s (BMP), growth and differentiation factors(GDF), activ<strong>in</strong> ⁄ <strong>in</strong>hib<strong>in</strong> and anti-mullerian hormone. Italso <strong>in</strong>cludes type I and type II receptors act<strong>in</strong>g throughSmad signall<strong>in</strong>g to modulate target genes expression, aswell as non-signall<strong>in</strong>g secreted or membrane-boundb<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s that may regulate TGF-b familymembers’ biologic activity. TGF-b family membersorig<strong>in</strong>ated from somatic follicular cells are <strong>in</strong>volved <strong>in</strong>the regulation of several aspects of follicular growth<strong>in</strong>itiation and progression (Knight and Glister 2006). Inaddition, GDF-9, BMP-15 (also called GDF-9B) andBMP-6 have been shown to be produced by oocytesfrom the primordial follicle stage (McNatty et al. 2001).In mouse, GDF-9 knockout <strong>in</strong>duces arrest of folliculardevelopment at the primary stage and sterility (Donget al. 1996) and GDF-9 may also be <strong>in</strong>volved <strong>in</strong>primordial to primary follicle transition <strong>in</strong> the rat ovary(Vitt et al. 2000). Oocyte secreted factors are necessaryto <strong>in</strong>duce the differentiation of pre-antral mouse granulosacells to cumulus cells [ability to expand uponepidermal growth factor (EGF) stimulation (Diaz et al.2007)]. In the sheep, naturally occurr<strong>in</strong>g <strong>in</strong>activat<strong>in</strong>gmutations of GDF-9 or BMP-15 <strong>in</strong>crease ovulation rate<strong>in</strong> heterozygous ewes while homozygous females are<strong>in</strong>fertile and active immunization aga<strong>in</strong>st GDF-9 andBMP-15 blocks folliculogenesis at the primary stage <strong>in</strong>ewe (McNatty et al. 2005). Dur<strong>in</strong>g antral folliculargrowth, GDF-9, BMP-15 and BMP-6 promote granulosacells proliferation and modulate their sensitivity toFSH, thus participat<strong>in</strong>g to the selection of the dom<strong>in</strong>antfollicle (Knight and Glister 2006). In addition, it hasbeen shown recently <strong>in</strong> bov<strong>in</strong>e that oocyte-secretedBMP-6 and BMP-15 are protect<strong>in</strong>g cumulus cellsaga<strong>in</strong>st apoptosis occurr<strong>in</strong>g dur<strong>in</strong>g culture of cattleCOC (Husse<strong>in</strong> et al. 2005) and GDF-9 preventspremature differentiation (lute<strong>in</strong>ization) of bov<strong>in</strong>e granulosacells (Spicer et al. 2006). Recently, JY-1 has beendiscovered as another oocyte-specific secreted factormodulat<strong>in</strong>g granulosa cell functions (Bettegowda et al.2007), highlight<strong>in</strong>g aga<strong>in</strong> the central role of oocyte andOSF <strong>in</strong> the regulation of follicular growth.In addition, the oocyte is able to regulate surround<strong>in</strong>gsomatic cells function through the modulation of theirmetabolism. Subtractive suppressive hybridizationallowed to identify six genes <strong>in</strong>volved <strong>in</strong> glycolysis thatwere overexpressed <strong>in</strong> mouse cumulus cells as comparedwith mural granulosa cells of antral follicles and thisoverexpression was dependant on the presence of theoocyte or of OSF (Sugiura et al. 2005). More recently, ithas been established that this oocyte regulation ofcumulus cells glycolysis is mediated by GDF-9 andFGF-8 OSF cooperation (Sugiura et al. 2007). In thesame way, GDF-9 and BMP-15 promote cholesterolsynthesis by cumulus cells (Su et al. 2008). This cholesterolmay be useful to oocyte that is unable to produceit. Interest<strong>in</strong>g oocyte graft<strong>in</strong>g experiments <strong>in</strong> mice(Eppig et al. 2002) showed that, placed <strong>in</strong> similarenvironment, oocytes collected from secondary follicleswere more able to promote follicular growth with appropriatemural granulosa ⁄ cumulus cell differentiation (LHFig. 3. Two hypotheses to expla<strong>in</strong> the <strong>in</strong>creas<strong>in</strong>g ratio of competent oocytes <strong>in</strong> grow<strong>in</strong>g follicular populations. (a) Primordial follicles entergrowth phase, oocytes are not yet competent (open circles). By the time of antrum formation, all oocytes become meiotically competent (grey),few are already developmentally competent (black). Dur<strong>in</strong>g further growth, some follicles are lost <strong>in</strong> atresia, whatever the status of their oocyte.Dur<strong>in</strong>g further follicular growth, the proportion of competent oocytes <strong>in</strong>creases because of ongo<strong>in</strong>g oocyte differentiation. More follicles are lostdur<strong>in</strong>g term<strong>in</strong>al growth. (b) Early follicular growth is similar. Because their oocytes are less able to drive follicular growth, follicles enclos<strong>in</strong>g lessdevelopmentally competent oocytes (grey) are lost <strong>in</strong> atresia, whereas follicles conta<strong>in</strong><strong>in</strong>g developmentally competent oocytes (black) cont<strong>in</strong>uetheir growth. F<strong>in</strong>ally, only follicles conta<strong>in</strong><strong>in</strong>g fully competent oocytes reach ovulationÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


398 P Mermillod, R Dalbie` s-Tran, S Uzbekova, A The´lie, J-M Traverso, C Perreau, P Papillier and P Mongetreceptor expression) as compared with oocytes fromprimary follicles.It appears f<strong>in</strong>ally that oocyte is able to regulate manyaspects of follicle somatic cells proliferation, differentiationand metabolic activities <strong>in</strong>volved <strong>in</strong> late folliculargrowth regulation. This opens the way to a newhypothesis, i.e. that oocytes <strong>in</strong> early antral folliclesalready display <strong>in</strong>tr<strong>in</strong>sic developmental competence.This competence may translate <strong>in</strong>to differential abilityto drive proper follicular differentiation <strong>in</strong> a decreas<strong>in</strong>gFSH support context and <strong>in</strong>creas<strong>in</strong>g <strong>in</strong>terfollicularcompetition. Poorly differentiated oocytes (oocytesbear<strong>in</strong>g some transcriptome abnormalities) fail <strong>in</strong> ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>gproper somatic cells differentiation status or toprotect these cells aga<strong>in</strong>st apoptosis <strong>in</strong>duced by <strong>in</strong>creas<strong>in</strong>glychalleng<strong>in</strong>g hormonal environment and f<strong>in</strong>allyleave their follicle degenerate on the way to atresia(Fig. 3). Hormonal stimulation of follicular growthpartly alleviates the selection pressure imposed onfollicles, and allows lower quality oocytes to drive theirfollicles to ovulation <strong>in</strong> these permissive conditions.The expression of many oocyte-specific genes dur<strong>in</strong>goocyte growth <strong>in</strong> pre-antral follicles is coord<strong>in</strong>ated bygerml<strong>in</strong>e-specific transcription factors such as FIGLAor NOBOX (Pangas and Rajkovic 2006). Interest<strong>in</strong>gly,NOBOX regulates oocyte genes <strong>in</strong>volved both <strong>in</strong> developmentalcompetence (Mater, Zar-1 and Mos) and <strong>in</strong>control of follicular function by oocyte (GDF-9 andBMP-15). This observation <strong>in</strong>dicates that oocytes deficient<strong>in</strong> terms of competence may also be deficient <strong>in</strong>their ability to properly drive their follicle to ovulation.ConclusionsDur<strong>in</strong>g its early growth <strong>in</strong> pre-antral follicles, the oocytestores specific gene products that will be determ<strong>in</strong>ant forits ability to coord<strong>in</strong>ate follicular growth and that willbe required dur<strong>in</strong>g early embryo development afterfertilization. Increas<strong>in</strong>g l<strong>in</strong>es of evidence suggest that bythe time of antrum formation, the quality of oocyte isalready determ<strong>in</strong>ed. This quality will first be expressedthrough the ability of the oocyte to drive folliculargrowth under an <strong>in</strong>creas<strong>in</strong>gly challeng<strong>in</strong>g hormonalenvironment up to ovulation, avoid<strong>in</strong>g atresia. Thisquality will be then expressed after fertilization throughthe ability to support early embryo development and to<strong>in</strong>itiate embryonic genome transcriptional activity. Ofcourse, further experimental data are required to supportthis hypothesis.ReferencesAlm H, Torner H, Lohrke B, Viergutz T, Ghoneim IM, KanitzW, 2005: Bov<strong>in</strong>e blastocyst development rate <strong>in</strong> vitro is<strong>in</strong>fluenced by selection of oocytes by brilliant cresyl bluesta<strong>in</strong><strong>in</strong>g before IVM as <strong>in</strong>dicator for glucose-6-phosphatedehydrogenase activity. Theriogenology 63, 2194–2205.Bettegowda A, Smith GW, 2007: Mechanisms of maternalmRNA regulation: implications for mammalian earlyembryonic development. Front Biosci 12, 3713–3726.Bettegowda A, Yao J, Sen A, Li Q, Lee K-B, Kobayashi Y,Patel OV, Coussens PM, Ireland JJ, Smith GW, 2007: JY-1,an oocyte-specific gene, regulates granulosa cell functionand early embryonic development <strong>in</strong> cattle. In: Proceed<strong>in</strong>gsof the National Academy of Sciences, %R 10.1073/pnas.0706383104, 0706383104.Blond<strong>in</strong> P, Sirard MA, 1995: Oocyte and follicularmorphology as determ<strong>in</strong><strong>in</strong>g characteristics for developmentalcompetence <strong>in</strong> bov<strong>in</strong>e oocytes. Mol Reprod Dev41, 54–62.Blond<strong>in</strong> P, Bousquet D, Twagiramungu H, Barnes F, SirardMA, 2002: Manipulation of follicular development toproduce developmentally competent bov<strong>in</strong>e oocytes. BiolReprod 66, 38–43.Cahill LP, Mariana JC, Mauleon P, 1979: Total follicularpopulations <strong>in</strong> ewes of high and low ovulation rates.J Reprod Fertil 55, 27–36.Carolan C, Lonergan P, Khatir H, Mermillod P, 1996: In vitroproduction of bov<strong>in</strong>e embryos us<strong>in</strong>g <strong>in</strong>dividual oocytes. MolReprod Dev 45, 145–150.Cognie Y, Benoit F, Poul<strong>in</strong> N, Khatir H, Driancourt MA,1998: Effect of follicle size and of the FecB Booroola gene onoocyte function <strong>in</strong> sheep. J Reprod Fertil 112, 379–386.Cognie Y, Baril G, Poul<strong>in</strong> N, Mermillod P, 2003: Currentstatus of embryo technologies <strong>in</strong> sheep and goat. Theriogenology59, 171–188.Corcoran D, Fair T, Lonergan P, 2005: Predict<strong>in</strong>g embryoquality: mRNA expression and the preimplantation embryo.Reprod Biomed Onl<strong>in</strong>e 11, 340–348.Cui XS, Kim NH, 2007: Maternally derived transcripts:identification and characterisation dur<strong>in</strong>g oocyte maturationand early cleavage. Reprod Fertil Dev 19, 25–34.Diaz FJ, Wigglesworth K, Eppig JJ, 2007: Oocytes determ<strong>in</strong>ecumulus cell l<strong>in</strong>eage <strong>in</strong> mouse ovarian follicles. J Cell Sci120, 1330–1340.Dong J, Albert<strong>in</strong>i DF, Nishimori K, Kumar TR, Lu N,Matzuk MM, 1996: Growth differentiation factor-9 isrequired dur<strong>in</strong>g early ovarian folliculogenesis. Nature 383,531–535.Duranthon V, Renard JP, 2001: The developmental competenceof mammalian oocytes: a convenient but biologicallyfuzzy concept. Theriogenology 55, 1277–1289.Edwards RG, 1965: Maturation <strong>in</strong> vitro of mouse, sheep, cow,pig, rhesus monkey and human ovarian oocytes. Nature208, 349–351.Eppig JJ, Schroeder AC, O’Brien MJ, 1992: Developmentalcapacity of mouse oocytes matured <strong>in</strong> vitro: effects ofgonadotrophic stimulation, follicular orig<strong>in</strong> and oocyte size.J Reprod Fertil 95, 119–127.Eppig JJ, Wigglesworth K, Pendola FL, 2002: The mammalianoocyte orchestrates the rate of ovarian follicular development.Proc Natl Acad Sci U S A 99, 2890–2894.Fair T, Hulshof SC, Hyttel P, Greve T, Boland M, 1997:Nucleus ultrastructure and transcriptional activity of bov<strong>in</strong>eoocytes <strong>in</strong> preantral and early antral follicles. Mol ReprodDev 46, 208–215.Feng WG, Sui HS, Han ZB, Chang ZL, Zhou P, Liu DJ, BaoS, Tan JH, 2007: Effects of follicular atresia and size on thedevelopmental competence of bov<strong>in</strong>e oocytes: a study us<strong>in</strong>gthe well-<strong>in</strong>-drop culture system. Theriogenology 67, 1339–1350.Husse<strong>in</strong> TS, Froiland DA, Amato F, Thompson JG,Gilchrist RB, 2005: Oocytes prevent cumulus cellapoptosis by ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g a morphogenic paracr<strong>in</strong>e gradientof bone morphogenetic prote<strong>in</strong>s. J Cell Sci 118, 5257–5268.Ikeda S, Imai H, Yamada M, 2003: Apoptosis <strong>in</strong> cumulus cellsdur<strong>in</strong>g <strong>in</strong> vitro maturation of bov<strong>in</strong>e cumulus-enclosedoocytes. <strong>Reproduction</strong> 125, 369–376.Khatir H, Lonergan P, Carolan C, Mermillod P, 1996:Prepubertal bov<strong>in</strong>e oocyte: a negative model for study<strong>in</strong>goocyte developmental competence. Mol Reprod Dev 45,231–239.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Factors Affect<strong>in</strong>g Oocyte Quality 399Knight PG, Glister C, 2006: TGF-beta superfamily membersand ovarian follicle development. <strong>Reproduction</strong> 132, 191–206.Knijn HM, Wrenzycki C, Hendriksen PJ, Vos PL, Ze<strong>in</strong>straEC, van der Weijden GC, Niemann H, Dieleman SJ, 2005:In vitro and <strong>in</strong> vivo culture effects on mRNA expression ofgenes <strong>in</strong>volved <strong>in</strong> metabolism and apoptosis <strong>in</strong> bov<strong>in</strong>eembryos. Reprod Fertil Dev 17, 775–784.Lequarre AS, Vigneron C, Ribaucour F, Holm P, Donnay I,Dalbies-Tran R, Callesen H, Mermillod P, 2005: Influenceof antral follicle size on oocyte characteristics and embryodevelopment <strong>in</strong> the bov<strong>in</strong>e. Theriogenology 63, 841–859.Lonergan P, 2007: State-of-the-art embryo technologies <strong>in</strong>cattle. Soc Reprod Fertil Suppl 64, 315–325.Lonergan P, Monaghan P, Rizos D, Boland MP, Gordon I,1994: Effect of follicle size on bov<strong>in</strong>e oocyte quality anddevelopmental competence follow<strong>in</strong>g maturation, fertilization,and culture <strong>in</strong> vitro. Mol Reprod Dev 37, 48–53.Lonergan P, Khatir H, Carolan C, Mermillod P, 1997: Bov<strong>in</strong>eblastocyst production <strong>in</strong> vitro after <strong>in</strong>hibition of oocytemeiotic resumption for 24 h. J Reprod Fertil 109, 355–365.Lonergan P, D<strong>in</strong>nyes A, Fair T, Yang X, Boland M, 2000:Bov<strong>in</strong>e oocyte and embryo development follow<strong>in</strong>g meiotic<strong>in</strong>hibition with butyrolactone I. Mol Reprod Dev 57, 204–209.Machatkova M, Krausova K, Jokesova E, Tomanek M, 2004:Developmental competence of bov<strong>in</strong>e oocytes: effects offollicle size and the phase of follicular wave on <strong>in</strong> vitroembryo production. Theriogenology 61, 329–335.Marchal R, Vigneron C, Perreau C, Bali-Papp A, MermillodP, 2002: Effect of follicular size on meiotic and developmentalcompetence of porc<strong>in</strong>e oocytes. Theriogenology 57,1523–1532.McNatty KP, Juengel JL, Wilson T, Galloway SM, Davis GH,2001: Genetic mutations <strong>in</strong>fluenc<strong>in</strong>g ovulation rate <strong>in</strong> sheep.Reprod Fertil Dev 13, 549–555.McNatty KP, Smith P, Moore LG, Reader K, Lun S,Hanrahan JP, Groome NP, Lait<strong>in</strong>en M, Ritvos O, JuengelJL, 2005: Oocyte-expressed genes affect<strong>in</strong>g ovulation rate.Mol Cell Endocr<strong>in</strong>ol 234, 57–66.Mermillod P, Oussaid B, Cognie Y, 1999: Aspects of follicularand oocyte maturation that affect the developmentalpotential of embryos. J Reprod Fertil Suppl 54, 449–460.Mermillod P, Tomanek M, Marchal R, Meijer L, 2000: Highdevelopmental competence of cattle oocytes ma<strong>in</strong>ta<strong>in</strong>ed atthe germ<strong>in</strong>al vesicle stage for 24 hours <strong>in</strong> culture by specific<strong>in</strong>hibition of MPF k<strong>in</strong>ase activity. Mol Reprod Dev 55, 89–95.M<strong>in</strong>ami N, Suzuki T, Tsukamoto S, 2007: Zygotic geneactivation and maternal factors <strong>in</strong> mammals. J Reprod Dev53, 707–715.Mulsant P, Lecerf F, Fabre S, Schibler L, Monget P, LannelucI, Pisselet C, Riquet J, Monniaux D, Callebaut I, Cribiu E,Thimonier J, Teyssier J, Bod<strong>in</strong> L, Cognie Y, Chitour N,Elsen JM, 2001: Mutation <strong>in</strong> bone morphogenetic prote<strong>in</strong>receptor-IB is associated with <strong>in</strong>creased ovulation rate <strong>in</strong>Booroola Mer<strong>in</strong>o ewes. Proc Natl Acad Sci U S A 98, 5104–5109.Murchison EP, Ste<strong>in</strong> P, Xuan Z, Pan H, Zhang MQ, SchultzRM, Hannon GJ, 2007: Critical roles for Dicer <strong>in</strong> the femalegerml<strong>in</strong>e. Genes Dev 21, 682–693.O’Callaghan D, Yaakub H, Hyttel P, Spicer LJ, Boland MP,2000: Effect of nutrition and superovulation on oocytemorphology, follicular fluid composition and systemichormone concentrations <strong>in</strong> ewes. J Reprod Fertil 118,303–313.Oussaid B, Lonergan P, Khatir H, Guler A, Monniaux D,Touze JL, Beckers JF, Cognie Y, Mermillod P, 2000: Effectof GnRH antagonist-<strong>in</strong>duced prolonged follicular phase onfollicular atresia and oocyte developmental competence<strong>in</strong> vitro <strong>in</strong> superovulated heifers. J Reprod Fertil 118, 137–144.Pangas SA, Rajkovic A, 2006: Transcriptional regulation ofearly oogenesis: <strong>in</strong> search of masters. Hum Reprod Update,12, 65–76.Pennetier S, Uzbekova S, Perreau C, Papillier P, Mermillod P,Dalbies-Tran R, 2004: Spatio-temporal expression of thegerm cell marker genes MATER, ZAR1, GDF9, BMP15,andVASA <strong>in</strong> adult bov<strong>in</strong>e tissues, oocytes, and preimplantationembryos. Biol Reprod 71, 1359–1366.Pennetier S, Uzbekova S, Guyader-Joly C, Humblot P,Mermillod P, Dalbies-Tran R, 2005: Genes preferentiallyexpressed <strong>in</strong> bov<strong>in</strong>e oocytes revealed by subtractiveand suppressive hybridization. Biol Reprod 73, 713–720.Pennetier S, Perreau C, Uzbekova S, Thelie A, Delaleu B,Mermillod P, Dalbies-Tran R, 2006: MATER prote<strong>in</strong>expression and <strong>in</strong>tracellular localization throughout folliculogenesisand preimplantation embryo development <strong>in</strong> thebov<strong>in</strong>e. BMC Dev Biol 6, 26.Ponderato N, Crotti G, Tur<strong>in</strong>i P, Duchi R, Galli C, Lazzari G,2002: Embryonic and foetal development of bov<strong>in</strong>e oocytestreated with a comb<strong>in</strong>ation of butyrolactone I and roscovit<strong>in</strong>e<strong>in</strong> an enriched medium prior to IVM and IVF. MolReprod Dev 62, 513–518.Rizos D, Ward F, Boland MP, Lonergan P, 2001: Effect ofculture system on the yield and quality of bov<strong>in</strong>e blastocystsas assessed by survival after vitrification. Theriogenology 56,1–16.Rodriguez KF, Far<strong>in</strong> CE, 2004a: Developmental capacity ofbov<strong>in</strong>e cumulus oocyte complexes after transcriptional<strong>in</strong>hibition of germ<strong>in</strong>al vesicle breakdown. Theriogenology61, 1499–1511.Rodriguez KF, Far<strong>in</strong> CE, 2004b: Gene transcription andregulation of oocyte maturation. Reprod Fertil Dev 16, 55–67.Rodriguez KF, Blomberg LA, Zuelke KA, Miles JR, AlexanderJE, Far<strong>in</strong> CE, 2006: Identification of candidate mRNAsassociated with gonadotrop<strong>in</strong>-<strong>in</strong>duced maturation of mur<strong>in</strong>ecumulus oocyte complexes us<strong>in</strong>g serial analysis of geneexpression. Physiol Genomics 27, 318–327.Sirard MA, Richard F, Mayes M, 1998: Controll<strong>in</strong>g meioticresumption <strong>in</strong> bov<strong>in</strong>e oocytes: a review. Theriogenology 49,483–497.Sirard MA, Richard F, Blond<strong>in</strong> P, Robert C, 2006: Contributionof the oocyte to embryo quality. Theriogenology 65,126–136.Spicer LJ, Aad PY, Allen D, Mazerbourg S, Hsueh AJ, 2006:Growth differentiation factor-9 has divergent effects onproliferation and steroidogenesis of bov<strong>in</strong>e granulosa cells.J Endocr<strong>in</strong>ol 189, 329–339.Su YQ, Sugiura K, Wigglesworth K, O’Brien MJ, Affourtit JP,Pangas SA, Matzuk MM, Eppig JJ, 2008: Oocyte regulationof metabolic cooperativity between mouse cumulus cells andoocytes: BMP15 and GDF9 control cholesterol biosynthesis<strong>in</strong> cumulus cells. Development 135, 111–121.Sugiura K, Pendola FL, Eppig JJ, 2005: Oocyte control ofmetabolic cooperativity between oocytes and companiongranulosa cells: energy metabolism. Dev Biol279, 20–30.Sugiura K, Su YQ, Diaz FJ, Pangas SA, Sharma S, WigglesworthK, O’Brien MJ, Matzuk MM, Shimasaki S, Eppig JJ,2007: Oocyte-derived BMP15 and FGFs cooperate topromote glycolysis <strong>in</strong> cumulus cells. Development 134,2593–2603.Tamassia M, Heyman Y, Lavergne Y, Richard C, Gel<strong>in</strong> V,Renard JP, Chastant-Maillard S, 2003: Evidence of oocytedonor cow effect over oocyte production and embryodevelopment <strong>in</strong> vitro. <strong>Reproduction</strong> 126, 629–637.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


400 P Mermillod, R Dalbie` s-Tran, S Uzbekova, A The´lie, J-M Traverso, C Perreau, P Papillier and P MongetUzbekova S, Roy-Sabau M, Dalbies-Tran R, Perreau C,Papillier P, Mompart F, Thelie A, Pennetier S, Cognie J,Cadoret V, Royere D, Monget P, Mermillod P, 2006:Zygote arrest 1 gene <strong>in</strong> pig, cattle and human: evidence ofdifferent transcript variants <strong>in</strong> male and female germ cells.Reprod Biol Endocr<strong>in</strong>ol 4, 12.Uzbekova S, Arlot-Bonnema<strong>in</strong>s Y, Dupont J, Dalbies-Tran R,Papillier P, Pennetier S, Thelie A, Perreau C, Mermillod P,Prigent C, Uzbekov R, 2008: Spatio-temporal expressionpatterns of Aurora k<strong>in</strong>ases A, B, and C and cytoplasmicpolyadenylation-element-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> <strong>in</strong> bov<strong>in</strong>e oocytesdur<strong>in</strong>g meiotic maturation. Biol Reprod 78, 218–233.Vallee M, Gravel C, Pal<strong>in</strong> MF, Reghenas H, Stothard P,Wishart DS, Sirard MA, 2005: Identification of novel andknown oocyte-specific genes us<strong>in</strong>g complementary DNAsubtraction and microarray analysis <strong>in</strong> three differentspecies. Biol Reprod 73, 63–71.Van Soom A, Tanghe S, De Pauw I, Maes D, de Kruif A,2002: Function of the cumulus oophorus before and dur<strong>in</strong>gmammalian fertilization. Reprod Domest Anim 37, 144–151.Vigneron C, Perreau C, Dalbies-Tran R, Joly C, Humblot P,Uzbekova S, Mermillod P, 2004a: Prote<strong>in</strong> synthesis andmRNA storage <strong>in</strong> cattle oocytes ma<strong>in</strong>ta<strong>in</strong>ed under meioticblock by roscovit<strong>in</strong>e <strong>in</strong>hibition of MPF activity. MolReprod Dev 69, 457–465.Vigneron C, Perreau C, Dupont J, Uzbekova S, Prigent C,Mermillod P, 2004b: Several signal<strong>in</strong>g pathways are<strong>in</strong>volved <strong>in</strong> the control of cattle oocyte maturation. MolReprod Dev 69, 466–474.Vitt UA, McGee EA, Hayashi M, Hsueh AJ, 2000: In vivotreatment with GDF-9 stimulates primordial and primaryfollicle progression and theca cell marker CYP17 <strong>in</strong> ovariesof immature rats. Endocr<strong>in</strong>ology 141, 3814–3820.Wang Q, Sun QY, 2007: Evaluation of oocyte quality:morphological, cellular and molecular predictors. ReprodFertil Dev 19, 1–12.Zeuner A, Muller K, Reguszynski K, Jewgenow K, 2003:Apoptosis with<strong>in</strong> bov<strong>in</strong>e follicular cells and its effect onoocyte development dur<strong>in</strong>g <strong>in</strong> vitro maturation. Theriogenology59, 1421–1433.Author’s address (for correspondence): P Mermillod, INRA, Physiologiede la <strong>Reproduction</strong> et des Comportements, UMR6175 INRA,CNRS, Universite´ de Tours, Haras Nationaux, 37380 Nouzilly,France. E-mail: mermillo@tours.<strong>in</strong>ra.frConflict of <strong>in</strong>terest: All authors declare no conflict of <strong>in</strong>terests.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 401–406 (2008); doi: 10.1111/j.1439-0531.2008.01191.xISSN 0936-6768Selected Aspects of Advanced Porc<strong>in</strong>e Reproductive TechnologyK Kikuchi 1 , N Kashiwazaki 2 , T Nagai 3 , M Nakai 1 , T Somfai 1 , J Noguchi 1 and H Kaneko 11 Division of Animal Sciences, National Institute of Agrobiological Sciences, Tsukuba, Ibaraki; 2 Laboratory of Animal <strong>Reproduction</strong>, School ofVeter<strong>in</strong>ary Medic<strong>in</strong>e, Azabu University, Sagamihara, Kanagawa; 3 National Institute of Livestock and Grassland Science, Tsukuba, Ibaraki, JapanContentsIn vitro fertilization (IVF) of <strong>in</strong> vitro matured (IVM) oocytes<strong>in</strong> pigs has become the most popular method of study<strong>in</strong>ggametogenesis and embryogenesis <strong>in</strong> this species. Furthermore,because of recent advances <strong>in</strong> <strong>in</strong> vitro culture (IVC) ofIVM–IVF embryos, <strong>in</strong> vitro production (IVP) of embryosnow enables us to generate viable embryos as successfully asfor <strong>in</strong> vivo-derived embryos and with less cost and <strong>in</strong> lesstime. These technologies contribute not only to developments<strong>in</strong> reproductive physiology and agriculture but also tothe conservation of porc<strong>in</strong>e genetic resources and theproduction of cloned or genetically modified pigs. However,<strong>in</strong> IVP, there still rema<strong>in</strong>s the problem of abnormal ploidy,which is caused by perform<strong>in</strong>g procedures under nonphysiologicalconditions. In recent years, unique technologiessuch as <strong>in</strong>tracytoplasmic sperm <strong>in</strong>jection (ICSI) or xenograft<strong>in</strong>gof gonadal tissue <strong>in</strong>to immunodeficient experimentalanimals have been developed to help conserve gameteresources. These technologies comb<strong>in</strong>ed with IVP areexpected to be useful for the conservation of gametes fromimportant genetic resources. Here, we discuss the developmentalability and normality of porc<strong>in</strong>e IVP embryos andalso the utilization of ICSI and xenograft<strong>in</strong>g <strong>in</strong> advanc<strong>in</strong>gbiotechnology <strong>in</strong> pigs.IVF and the Normality of Porc<strong>in</strong>e ZygotesThe <strong>in</strong> vitro developmental competence or viability ofporc<strong>in</strong>e <strong>in</strong> vitro matured (IVM)–<strong>in</strong> vitro fertilized (IVF)oocytes to the blastocyst stage was first confirmed andreported by Mattioli et al. (1989). Furthermore, pigletshave been born from IVM–IVF embryos after <strong>in</strong> vitroculture (IVC) to the 2- to 4-cell stages (Mattioli et al.1989; Yoshida et al. 1993). S<strong>in</strong>ce then, some laboratorieshave succeeded <strong>in</strong> produc<strong>in</strong>g piglets from embryoscleaved at the 2- to 4-cell stages after IVM–IVF andIVC for 24–36 h (Funahashi et al. 1996, 1997). Viablepiglets were also generated after transfer of <strong>in</strong> vitroproduced (IVP) embryos at the blastocyst stage (Marchalet al. 2001; Kikuchi et al. 2002). Over several recentyears, IVC procedures have been improved, but IVMand IVF systems still have unsolved problems, <strong>in</strong>clud<strong>in</strong>g(1) imbalance of nuclear and cytoplasmic maturationand (2) polyspermy. Both these phenomena causeabnormal ploidy <strong>in</strong> IVP embryos, potentially result<strong>in</strong>g<strong>in</strong> loss of embryos after their transfer to recipients. Weneed to make every effort to achieve normality <strong>in</strong> IVPembryos after IVM and IVF.Fertilization at the immature stage, before nuclearmaturationImmature oocytes before nuclear maturation, such asthose at the metaphase-I stage, acquire fertilization oractivation potential (Kikuchi et al. 1999) and developmentalpotential, at least to the blastocyst stage (Somfaiet al. 2005). Our recent data suggest that treatment ofimmature oocytes with cytochalas<strong>in</strong> B, a drug that<strong>in</strong>hibits act<strong>in</strong> filament polymerization, results <strong>in</strong> thefailure of extrusion of the first polar body dur<strong>in</strong>g thefirst meiosis (Somfai et al. 2006). This failure, whichmay also be caused under physiological conditions,leads to lack of completion of meiotic maturation <strong>in</strong>oocytes (Somfai et al., unpublished). The developmentalcompetence of these immature oocytes is poor, result<strong>in</strong>g<strong>in</strong> low rates of blastocyst formation and low cellnumbers <strong>in</strong> the blastocysts. Chromosomal analysissuggests that these immature porc<strong>in</strong>e oocytes fertilized<strong>in</strong> vitro develop abnormally <strong>in</strong> terms of ploidy andespecially show <strong>in</strong>creased rates of triploidy (Somfaiet al. 2005).Polyspermic fertilization of matured oocytesThe best-known problem that occurs dur<strong>in</strong>g porc<strong>in</strong>eIVF is polyspermy (Nagai 1996). Many researchershave been try<strong>in</strong>g to solve this problem by differentapproaches, but so far no one has succeeded.Although it has been suggested that polyspermicoocytes have the ability to extrude the extra spermchromosomes <strong>in</strong> the early embryonic stage, beforeblastulation (Funahashi and Day 1997), the abnormalploidies are reportedly ma<strong>in</strong>ta<strong>in</strong>ed until the blastocyststage (Han et al. 1999; McCauley et al. 2003; Somfaiet al. <strong>in</strong> press).Utilization of Intracytoplasmic Sperm Injectionfor PigsIntracytoplasmic sperm <strong>in</strong>jection (ICSI) was first performedto study the early events of fertilization <strong>in</strong>hamsters (Uehara and Yanagimachi 1976, 1977). Fromthe 1990s, ICSI has been used for assisted reproductivetechnology <strong>in</strong> humans. Also <strong>in</strong> animals, viable offspr<strong>in</strong>gcan be generated by us<strong>in</strong>g sperm with poor motility orviability (Table 1). These successful reports confirm theooplasmic capacity for fertilization and development toterm after ICSI <strong>in</strong> both <strong>in</strong> vivo matured and IVMoocytes <strong>in</strong> mammals. If the spermatozoa have lost theirmotility, ICSI is <strong>in</strong>dispensable for fertilization to producethe next generation. In pigs, the use of <strong>in</strong> vivomatured oocytes led to the first successful production ofpiglets after ICSI (Kolbe and Holtz 2000; Mart<strong>in</strong> 2000);the use of ICSI <strong>in</strong> porc<strong>in</strong>e IVM oocytes for successfulpiglet production was first confirmed <strong>in</strong> our laboratory(Nakai et al. 2003).Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


402 K Kikuchi, N Kashiwazaki, T Nagai, M Nakai, T Somfai, J Noguchi and H KanekoTable 1. First successful productions of offspr<strong>in</strong>g after ICSI <strong>in</strong>different speciesSpeciesIn vivo maturedOocyte sourceIn vitro maturedHuman Palermo et al. (1992) Nagy et al. (1996)Mouse Ahmadi et al. (1995)Kimura and Yanagimachi (1995a)Lacham-Kaplan and Trounson (1995)Cow Goto et al. (1991)Sheep Catt et al. (1996)Rabbit Li et al. (2001)Monkey Hewitson et al. (1999)Cat Gomez et al. (2000)Horse Cochran et al. (1998)RatHirabayashi et al. (2002a)Hamster Yamauchi et al. (2002)Pig Kolbe and Holtz (2000) Nakai et al. (2003)Mart<strong>in</strong> (2000)Generation of zygotes from immotile spermatozoa orimmature sperm cellsOffspr<strong>in</strong>g have also been obta<strong>in</strong>ed by us<strong>in</strong>g immaturesperm cells such as round spermatids <strong>in</strong> mice (Oguraet al. 1994) and rats (Hirabayashi et al. 2002b), orelongated spermatids <strong>in</strong> monkeys (Hewitson et al. 2002).In mice, both primary (Kimura et al. 1998; Ogura et al.1998; Sasagawa et al. 1998) and secondary (Kimura andYanagimachi 1995b) spermatocytes could be <strong>in</strong>jected,and the procedure yielded liv<strong>in</strong>g pups. In pigs, however,when round spermatids were <strong>in</strong>jected <strong>in</strong>to IVM oocytes(Kim et al. 1999), no piglets were obta<strong>in</strong>ed aftertransfer. No other immature sperm cells are useful forembryonic development <strong>in</strong> pigs.Avoid<strong>in</strong>g polyspermyConsider<strong>in</strong>g the fact that the ultimate procedure toblock polyspermy has not yet been established <strong>in</strong>porc<strong>in</strong>e IVF, s<strong>in</strong>gle sperm <strong>in</strong>jection can be a mostwelcome procedure to generate normal (monospermic)embryos and piglets. However, the efficacy of ICSI <strong>in</strong>pigs rema<strong>in</strong>s low (Nakai et al. 2003). In this report, wehave transferred 598 oocytes after ICSI <strong>in</strong>to total sevenrecipients; however, only three live piglets obta<strong>in</strong>ed froma s<strong>in</strong>gle recipient. To accelerate the acrosome reaction,we have pre-treated spermatozoa with calcium ionophorebefore sperm <strong>in</strong>jection (Nakai et al. 2003), or withdithiothreitol to <strong>in</strong>duce <strong>in</strong> vitro decondensation of thesperm nuclei (Nakai et al. 2006). However, no <strong>in</strong>creasedefficacy of embryo production was confirmed <strong>in</strong> ourlaboratory.Freeze-dry<strong>in</strong>g of spermatozoaNew technologies for the conservation of spermatozoaother than by cryopreservation <strong>in</strong> liquid nitrogen havebeen anticipated. One of the most welcome procedures isfreeze-dry<strong>in</strong>g of spermatozoa, because the spermatozoacan be stored at less cost, and without the need for liquidnitrogen, at 5°C or room temperature. After freezedry<strong>in</strong>g,the spermatozoa lose their motility and shouldbe fertilized by ICSI. Successful offspr<strong>in</strong>g productionhas been reported <strong>in</strong> mice (Wakayama and Yanagimachi1998; Kaneko et al. 2003a; Ward et al. 2003; Kanekoand Nakagata 2006; Kawase et al. 2007), rabbits (Liuet al. 2004), and rats (Hirabayashi et al. 2005a; Hochiet al., 2008). In mice, DNA damage was <strong>in</strong>duced dur<strong>in</strong>gthe hold<strong>in</strong>g period after rehydration before sperm<strong>in</strong>jection (Wakayama and Yanagimachi 1998). Therelationship between the duration of rehydration offreeze-dried sperm and the <strong>in</strong> vitro development offreeze-dried sperm-<strong>in</strong>jected oocytes was exam<strong>in</strong>ed <strong>in</strong>pigs (Nakai et al. 2007). The results suggested thatembryos obta<strong>in</strong>ed after ICSI with a s<strong>in</strong>gle freeze-driedsperm head have the competence to be implanted and todevelop to the early fetal stage, and also that theduration of rehydration does not <strong>in</strong>fluence the <strong>in</strong> vitrodevelopmental ability of the embryos <strong>in</strong> pigs.Xenograft<strong>in</strong>g of testicular tissues <strong>in</strong>to immunodeficientmiceIn mice, germ cells used for transplantation <strong>in</strong>to thetestis can be homolog taken from an unrelated male(Br<strong>in</strong>ster and Avarbock 1994; Br<strong>in</strong>ster and Zimmermann1994). The possibility of spermatogenesis by thismethod has been also suggested <strong>in</strong> pigs (Honaramoozet al. 2002a). In these cases, germ cells, <strong>in</strong>clud<strong>in</strong>gspermatogonial stem cells, were <strong>in</strong>jected directly <strong>in</strong>tothe sem<strong>in</strong>iferous tubules, and it was expected thatspermatozoa from the <strong>in</strong>troduced cells would be produced<strong>in</strong> the ejaculate. However, this technique requiresspecial skills for the <strong>in</strong>jection and also accurate separationprocedures to select the spermatozoa derived fromthe <strong>in</strong>troduced cells. Germ cell transplantation <strong>in</strong>to micehas produced complete donor-derived spermatogenesis<strong>in</strong> rodents, but unfortunately it did not succeed <strong>in</strong> othermammalian species.Xenograft<strong>in</strong>g of testicular tissues can be performed<strong>in</strong>to immunodeficient mice heterotopically, such asunder the sk<strong>in</strong> of the back. The report by Honaramoozet al. (2002b) suggested first the possibility of productionof mammalian sperm <strong>in</strong> testicular tissues grafted<strong>in</strong>to nude mice. However, successful embryo productionby us<strong>in</strong>g the sperm cells from xenografted testiculartissues has been limited to rhesus monkeys (Honaramoozet al. 2004). In our study (Kikuchi et al. 2006a),testicular tissues from male piglets 6–15 days old weretransplanted under the sk<strong>in</strong> of the backs of castratednude mice 5–8 weeks old. When porc<strong>in</strong>e testes werexenografted <strong>in</strong>to mice, spermatids and spermatozoawere obta<strong>in</strong>ed <strong>in</strong> 70.4% (19 ⁄ 27) of the mice but showedonly fa<strong>in</strong>t motility, suggest<strong>in</strong>g the need for ICSI to givetheir ability to fertilize oocytes. When a total of 253oocytes (four replications) were <strong>in</strong>jected with sperm, 63oocytes (24.9% ± 7.1%) developed to the blastocyststage. Their total mean number of cells was 41.9 ± 3.9.These values were comparable to those from <strong>in</strong> vitrofertilization with frozen-thawed spermatozoa [25.3%and 48.7 cells, respectively (Kikuchi et al. 2002)], show<strong>in</strong>gthe promis<strong>in</strong>g ability of the embryos to develop <strong>in</strong>topiglets after embryo transfer. The results suggest thepossibility of produc<strong>in</strong>g embryos with developmentalpotential by us<strong>in</strong>g porc<strong>in</strong>e spermatozoa differentiatedfrom the gonocytes with<strong>in</strong> the xenografts.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Advanced Porc<strong>in</strong>e Reproductive Technology 403Other technologies related to ICSIThe establishment of a porc<strong>in</strong>e IVM–ICSI proceduremay help us to generate transgenic pigs by spermmediatedgene transfer. The possibility of mammalianspermatozoa be<strong>in</strong>g able to act as vectors for foreignDNA has been reported by Brackett et al. (1971), andsuccessful fertilization by gene-mediated spermatozoahas been reported <strong>in</strong> mice (Lavitrano et al. 1989) andpigs (Lavitrano et al. 1997, 1999; Lazzereschi et al.2000). This method comb<strong>in</strong>ed with ICSI has succeeded<strong>in</strong> mice (Perry et al. 1999) and rats (Hirabayashi et al.2005b), and is now expected to be used <strong>in</strong> othermammalian species, <strong>in</strong>clud<strong>in</strong>g pigs. In general, theefficacy of the sperm-mediated gene transfer method,as compared with micro<strong>in</strong>jection method <strong>in</strong>to thepronuclei, was reviewed as excellent <strong>in</strong> the productionof transgenic animals (Smith 1999; Wall 2002). Theefficacy of transgenic pig production may be improvedby us<strong>in</strong>g a comb<strong>in</strong>ation of IVM oocytes and spermmediatedgene transfer by ICSI, as has been suggestedby Kurome et al. (2006).Conservation of Ovarian Resources byXenograft<strong>in</strong>g <strong>in</strong>to MicePrimordial follicles act as stores of ovarian follicles andare potential resources of oocytes for medical, agriculturaland zoological purposes. Ovarian graft<strong>in</strong>g is apotential method of matur<strong>in</strong>g the oocytes <strong>in</strong> theprimordial follicles (primordial oocytes) of large mammals.Graft<strong>in</strong>g of ovarian tissues to another site <strong>in</strong> thebody (autograft<strong>in</strong>g) has been successful <strong>in</strong> produc<strong>in</strong>gviable offspr<strong>in</strong>g <strong>in</strong> humans (Donnez et al. 2004) andother primates (Lee et al. 2004). Cross-species ovariangraft<strong>in</strong>g (xenograft<strong>in</strong>g) seems to be more advantageousfor the multiplication and conservation of domestic orendangered animals. Mouse oocytes that grow with<strong>in</strong>ovarian tissue xenografted to nude rats acquire theability to generate pups (Snow et al. 2002). To date,ovarian tissues have been prepared from species phylogeneticallydistant from mice, <strong>in</strong>clud<strong>in</strong>g humans (Oktayet al. 1998; Weissman et al. 1999; Kim et al. 2002; Gooket al. 2003), dogs (Metcalfe et al. 2001), monkeys(Candy et al. 1995), sheep (Gosden et al. 1994), cows(Senbon et al. 2003), pigs (Kaneko et al. 2003b; Kagawaet al. 2005), tammar wallabies (Mattiske et al. 2002) andcommon wombats (Cleary et al. 2003, 2004), and thenxenografted <strong>in</strong>to immunodeficient mice. To our knowledge,only our previous study (Kaneko et al. 2003b), <strong>in</strong>which neonatal pig ovarian tissues were xenografted,had proven that primordial oocytes can develop <strong>in</strong> thehost mice and acquire fertiliz<strong>in</strong>g ability.In our series of studies (Kaneko et al. 2003b, 2006;Kikuchi et al. 2006b), we have generated viable embryosfrom porc<strong>in</strong>e primordial oocytes xenografted <strong>in</strong>to nudemice. Here, we summarize the recent data and discuss thepossibility of further improvement of this technology.IVM–IVF of porc<strong>in</strong>e oocytes grown <strong>in</strong> host miceOvarian tissues from 20-day-old piglets, <strong>in</strong> which mostof the follicles were primordial upon histological evaluation,were transplanted under the capsules of bothkidneys of ovariectomized nude mice (Kaneko et al.2003b). Forty-five to 70 days after graft<strong>in</strong>g, the hostmice <strong>in</strong> all groups for the first time showed vag<strong>in</strong>alcornification. They were treated with 5 IU of equ<strong>in</strong>echorionic gonadotrop<strong>in</strong> (eCG) 10 days (eCG-10),30 days (eCG-30), or 60 days (eCG-60) after the detectionof cornified epithelial cells <strong>in</strong> their vag<strong>in</strong>al smears.Ovarian grafts and blood samples were obta<strong>in</strong>ed 48 hafter eCG treatment. The mice <strong>in</strong> all groups formedsmall numbers of antral follicles <strong>in</strong> the grafts. However,we recovered large numbers of full-sized oocytes(‡115 lm <strong>in</strong> diameter) only from mice <strong>in</strong> the eCG-60group; the numbers of full-sized oocytes <strong>in</strong> the othergroups were low. Peripheral levels of total <strong>in</strong>hib<strong>in</strong> werehighest <strong>in</strong> the eCG-60 group; this supports our f<strong>in</strong>d<strong>in</strong>gthat the most enhanced growth of antral folliclesoccurred <strong>in</strong> this eCG-60 group. When the oocytesobta<strong>in</strong>ed from the eCG-60 group were matured <strong>in</strong> vitro,17% were found to be at metaphase II. Moreover, 55%of IVM oocytes with a first polar body were fertilized <strong>in</strong>vitro, result<strong>in</strong>g <strong>in</strong> both male and female pronuclearformation. These results clearly demonstrate that fertilizationof oocytes from porc<strong>in</strong>e primordial follicles isachievable by a comb<strong>in</strong>ation of xenograft<strong>in</strong>g and <strong>in</strong> vitroculture.Developmental ability <strong>in</strong> vitro and <strong>in</strong> vivo <strong>in</strong> recipient miceafter IVF of porc<strong>in</strong>e oocytes grown <strong>in</strong> most miceIn the next step, we evaluated the developmental abilityof the <strong>in</strong> vitro fertilized oocytes after transfer torecipients or <strong>in</strong> vitro culture (Kikuchi et al. 2006b). Asdeterm<strong>in</strong>ed from the <strong>in</strong>cidence of the first polar body,the maturation rates of the oocytes collected from thehost mice 48 h after treatment with eCG ranged from25.1% to 42.5%. The oocytes were fertilized <strong>in</strong> vitro andcultured <strong>in</strong> vitro for 6 days, or transferred <strong>in</strong>to oestrussynchronizedrecipients and recovered after 6 days. Onday 6 of culture, 15.4% of the matured oocytes hadcleaved to the 2- to 8-cell stage. However, neither theembryos cultured <strong>in</strong> vitro nor those transferred andrecovered developed to advanced embryonic stages,such as morulae or blastocysts. These results suggestthat the developmental ability of xenografted oocytes is<strong>in</strong>sufficient, even after <strong>in</strong> vitro maturation.Improvement of developmental ability after treatment ofhost mice with gonadotrop<strong>in</strong>sTo improve the developmental ability of primordialoocytes xenografted to nude mice, we treated the hostmice with gonadotroph<strong>in</strong>s to accelerate folliculargrowth (Kaneko et al. 2006). Gonadotroph<strong>in</strong> treatmentswere commenced around 60 days after vag<strong>in</strong>alcornification. Ovarian grafts were obta<strong>in</strong>ed 2 or 3 daysafter treatment with eCG (eCG-2 and eCG-3 groups,respectively), after porc<strong>in</strong>e FSH <strong>in</strong>fusion by osmoticpump for 7 or 14 days, or after <strong>in</strong>fusion of porc<strong>in</strong>eFSH for 14 days with a s<strong>in</strong>gle <strong>in</strong>jection of estradiolantiserum (FSH-7, FSH-14 and FSH-14EA groups,respectively). Gonadotrop<strong>in</strong> treatments accelerated folliculargrowth with<strong>in</strong> the xenografts compared withÓ 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


404 K Kikuchi, N Kashiwazaki, T Nagai, M Nakai, T Somfai, J Noguchi and H Kanekothat <strong>in</strong> control mice given no gonadotrop<strong>in</strong>s, consistentwith markedly higher circulat<strong>in</strong>g <strong>in</strong>hib<strong>in</strong> levels <strong>in</strong> thegonadotrop<strong>in</strong>-treated mice. In contrast, circulat<strong>in</strong>gmouse FSH levels were depressed. We recovered largenumbers of full-sized oocytes with meiotic competenceto the mature stage from the eCG-3, FSH-7 and FSH-14EA groups, unlike <strong>in</strong> the control group. Moreover,56% of IVM oocytes with a first polar body werefertilized <strong>in</strong> vitro <strong>in</strong> the FSH-14EA group. After IVFand subsequent IVC for 7 days, one blastocyst wasobta<strong>in</strong>ed from each of the eCG-3, FSH-7 and FSH-14EA groups, whereas no blastocysts appeared <strong>in</strong> theother groups. These results suggest that exogenousgonadotrop<strong>in</strong>s – not mouse FSH – stimulated thegrow<strong>in</strong>g follicles that had developed from the primordialfollicles <strong>in</strong> the xenografts: the effects were <strong>in</strong>completebut improved the meiotic and developmentalabilities of the oocytes to some extent.Further improvementOur studies suggest that, <strong>in</strong> pigs, oocytes derived fromprimordial follicles, even after <strong>in</strong> vivo growth and IVM,seem to have difficulty achiev<strong>in</strong>g cytoplasmic maturation.One of the procedures to enhance embryonicdevelopment is the transfer of metaphase-II chromosomesto an enucleated cytoplast with full developmentalability; this has been successfully reported <strong>in</strong> mice(Wang et al. 2001). On the contrary, we recentlyreported the ‘Centri-Fusion’ method of nuclear transfer(Fahrud<strong>in</strong> et al. 2007; Nagai et al. 2007). This mayenable the modification of cytoplasmic maturationalability, even <strong>in</strong> oocytes from xenografted ovaries.ConclusionPorc<strong>in</strong>e IVP and related technologies are expected to beuseful for the conservation of genetic resources andother related technologies. To <strong>in</strong>crease the reliability ofthese technologies, improvements <strong>in</strong> the procedures usedare still required. We believe that more basic research toclarify the mechanisms of oocyte maturation, fertilizationand embryonic development will improve thesetechnologies <strong>in</strong> pigs.AcknowledgementsThis study was supported <strong>in</strong> part by a Grant-<strong>in</strong>-Aid for ScientificResearch (17380170) to H.K. from the Japanese Society for Promotionof Science.ReferencesAhmadi A, Ng SC, Liow SL, Ali J, Bongso A, Ratnam SS,1995: Intracytoplasmic sperm <strong>in</strong>jection of mouse oocyteswith 5 mM Ca 2+ at different <strong>in</strong>tervals. Hum Reprod 10,431–435.Brackett BG, Baranska W, Sawicki W, Koprowski H, 1971:Uptake of heterologous genome by mammalian spermatozoaand its transfer to ova through fertilization. Proc NatlAcad Sci U S A 68, 353–357.Br<strong>in</strong>ster RL, Avarbock MR, 1994: Germl<strong>in</strong>e transmission ofdonor haplotype follow<strong>in</strong>g spermatogonial transplantation.Proc Natl Acad Sci U S A 91, 11303–11307.Br<strong>in</strong>ster RL, Zimmermann JW, 1994: Spermatogenesis follow<strong>in</strong>gmale germ-cell transplantation. Proc Natl Acad SciUSA91, 11298–11302.Candy CJ, Wood MJ, Whitt<strong>in</strong>gham DG, 1995: Folliculardevelopment <strong>in</strong> cryopreserved marmoset ovarian tissue aftertransplantation. Hum Reprod 10, 2334–2338.Catt SL, Catt JW, Gomez MC, Maxwell WM, Evans G, 1996:Birth of a male lamb derived from an <strong>in</strong> vitro maturedoocyte fertilised by <strong>in</strong>tracytoplasmic <strong>in</strong>jection of a s<strong>in</strong>glepresumptive male sperm. Vet Rec 139, 494–495.Cleary M, Paris MC, Shaw J, Jenk<strong>in</strong> G, Trounson A, 2003:Effect of ovariectomy and graft position on cryopreservedcommon wombat (Vombatus urs<strong>in</strong>us) ovarian tissue follow<strong>in</strong>gxenograft<strong>in</strong>g to nude mice. Reprod Fertil Dev 15, 333–342.Cleary M, Shaw JM, Jenk<strong>in</strong> G, Trounson AO, 2004: Influenceof hormone environment and donor age on cryopreservedcommon wombat (Vombatus urs<strong>in</strong>us) ovarian tissue xenografted<strong>in</strong>to nude mice. Reprod Fertil Dev 16, 699–707.Cochran R, Me<strong>in</strong>tjes M, Reggio B, Hylan D, Carter J, P<strong>in</strong>toC, Paccamonti D, Godke RA, 1998: Live foals producedfrom sperm-<strong>in</strong>jected oocytes derived from pregnant mares.J Equ<strong>in</strong>e Vet Sci 18, 736–740.Donnez J, Dolmans MM, Demylle D, Jadoul P, Pirard C,Squifflet J, Mart<strong>in</strong>ez-Madrid B, van Langendonckt A, 2004:Live birth after orthotopic transplantation of cryopreservedovarian tissue. Lancet 364, 1405–1410.Fahrud<strong>in</strong> M, Kikuchi K, Karja NWK, Ozawa M, Somfai T,Ohnuma K, Noguchi J, Kaneko H, Nagai T, 2007:Development to the blastocyst stage of porc<strong>in</strong>e somatic cellnuclear transfer embryos reconstructed by the fusion ofcumulus cells and cytoplasts prepared by gradient centrifugation.Clon<strong>in</strong>g Stem Cells 9, 216–228.Funahashi H, Day BN, 1997: Advances <strong>in</strong> <strong>in</strong> vitro productionof pig embryos. J Reprod Fertil Suppl 52, 271–283.Funahashi H, Kim NH, Stumpf TT, Cantley TC, Day BN,1996: Presence of organic osmolytes <strong>in</strong> maturation mediumenhances cytoplasmic maturation of porc<strong>in</strong>e oocytes. BiolReprod 54, 1412–1419.Funahashi H, Cantley TC, Day BN, 1997: Synchronizationof meiosis <strong>in</strong> porc<strong>in</strong>e oocytes by exposure to dibutyrylcyclic adenos<strong>in</strong>e monophosphate improves developmentalcompetence follow<strong>in</strong>g <strong>in</strong> vitro fertilization. Biol Reprod 57,49–53.Gomez MC, Pope CE, Harris, Davis A, Mikota S, Dresser BL,2000: Births of kittens produced by <strong>in</strong>tracytoplasmic sperm<strong>in</strong>jection of domestic cat oocytes matured <strong>in</strong> vitro. ReprodFertil Dev 12, 423–433.Gook DA, Edgar DH, Borg J, Archer J, Lutjen PJ, McBa<strong>in</strong>JC, 2003: Oocyte maturation, follicle rupture and lute<strong>in</strong>ization<strong>in</strong> human cryopreserved ovarian tissue follow<strong>in</strong>gxenograft<strong>in</strong>g. Hum Reprod 18, 1772–1781.Gosden RG, Boulton MI, Grant K, Webb R, 1994: Folliculardevelopment from ovarian xenografts <strong>in</strong> SCID mice.J Reprod Fertil 101, 619–623.Goto K, K<strong>in</strong>oshita A, Takuma Y, Ogawa K, 1991: Birth ofcalves after the transfer of oocytes fertilized by sperm<strong>in</strong>jection. Theriogenology 35, 205.Han YM, Abeydeera LR, Kim JH, Moon HB, Cabot RA, DayBN, Prather RS, 1999: Growth retardation of <strong>in</strong>ner cellmass cells <strong>in</strong> polyspermic porc<strong>in</strong>e embryos produced <strong>in</strong>vitro. Biol Reprod 60, 1110–1113.Hewitson L, Dom<strong>in</strong>ko T, Takahashi D, Mart<strong>in</strong>ovich C,Ramalho-Santos J, Sutovsky P, Fanton J, Jacob D, MonteithD, Neur<strong>in</strong>ger M, Battaglia D, Simerly C, Schatten G,1999: Unique checkpo<strong>in</strong>ts dur<strong>in</strong>g the first cell cycle offertilization after <strong>in</strong>tracytoplasmic sperm <strong>in</strong>jection <strong>in</strong> rhesusmonkeys. Nat Med 5, 431–433.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Advanced Porc<strong>in</strong>e Reproductive Technology 405Hewitson L, Mart<strong>in</strong>ovich C, Simerly C, Takahashi D, SchattenG, 2002: Rhesus offspr<strong>in</strong>g produced by <strong>in</strong>tracytoplasmic<strong>in</strong>jection of testicular sperm and elongated spermatids.Fertil Steril 77, 794–801.Hirabayashi M, Koto M, Aoto T, Sekimoto A, Ueda M,Miyoshi I, Kasai N, Hochi S, 2002a: Offspr<strong>in</strong>g derived from<strong>in</strong>tracytoplasmic <strong>in</strong>jection of transgenic rat sperm. TransgenicRes 11, 221–228.Hirabayashi M, Kato M, Aoto T, Ueda M, Hochi S, 2002b:Rescue of <strong>in</strong>fertile transgenic rat l<strong>in</strong>es by <strong>in</strong>tracytoplasmic<strong>in</strong>jection of cryopreserved round spermatids. Mol ReprodDev 62, 295–299.Hirabayashi M, Kato M, Ito J, Hochi S, 2005a: Viable ratoffspr<strong>in</strong>g derived from oocytes <strong>in</strong>tracytoplasmically <strong>in</strong>jectedwith freeze-dried sperm heads. Zygote 13, 79–85.Hirabayashi M, Kato M, Ishikawa A, Kaneko R, Yagi T,Hochi S, 2005b: Factors affect<strong>in</strong>g production of transgenicrats by ICSI-mediated DNA transfer: effects of sonicationand freeze-thaw<strong>in</strong>g of spermatozoa, rat stra<strong>in</strong>s for spermand oocyte donors, and different constructs of exogenousDNA. Mol Reprod Dev 70, 422–428.Hochi S, Watanabe K, Kato M, Hirabayashi M, 2008: Liverats result<strong>in</strong>g from <strong>in</strong>jection of oocytes with spermatozoadried and stored for one year. Mol Reprod Dev 75, 890–894.Honaramooz A, Megee SO, Dobr<strong>in</strong>ski I, 2002a: Germ celltransplantation <strong>in</strong> pigs. Biol Reprod 66, 21–28.Honaramooz A, Snedaker A, Boiani M, Scholer H, Dobr<strong>in</strong>skiI, Schlatt S, 2002b: Sperm from neonatal mammalian testesgrafted <strong>in</strong> mice. Nature 418, 778–781.Honaramooz A, Li MW, Penedo MC, Meyers S, Dobr<strong>in</strong>ski I,2004: Accelerated maturation of primate testis by xenograft<strong>in</strong>g<strong>in</strong>to mice. Biol Reprod 70, 1500–1503.Kagawa N, Sakurai Y, Miyano T, Manabe N, 2005: Effects oflong-term graft<strong>in</strong>g on follicular growth <strong>in</strong> porc<strong>in</strong>e ovariancortical grafts xenoplanted to severe comb<strong>in</strong>ed immunodeficient(SCID) mice. J Reprod Dev 51, 77–85.Kaneko T, Nakagata N, 2006: Improvement <strong>in</strong> the long-termstability of freeze-dried mouse spermatozoa by add<strong>in</strong>g of achelat<strong>in</strong>g agent. Cryobiology 53, 279–282.Kaneko T, Whitt<strong>in</strong>gham DG, Overstreet JW, Yanagimachi R,2003a: Tolerance of the mouse sperm nuclei to freeze-dry<strong>in</strong>gdepends on their disulphide status. Biol Reprod 69, 1859–1862.Kaneko H, Kikuchi K, Noguchi J, Hosoe M, Akita T, 2003b:Maturation and fertilization of porc<strong>in</strong>e oocytes fromprimordial follicles by a comb<strong>in</strong>ation of xenograft<strong>in</strong>g and<strong>in</strong> vitro culture. Biol Reprod 69, 1488–1493.Kaneko H, Kikuchi K, Noguchi J, Ozawa M, Ohnuma K,Maedomari N, Kashiwazaki N, 2006: Effects of gonadotrop<strong>in</strong>treatments on meiotic and developmental competence ofoocytes <strong>in</strong> porc<strong>in</strong>e primordial follicles follow<strong>in</strong>g xenograft<strong>in</strong>gto nude mice. <strong>Reproduction</strong> 131, 279–288.Kawase Y, Hani T, Kamada N, Jishage K, Suzuki H, 2007:Effects of pressure at primary dry<strong>in</strong>g of freeze-dry<strong>in</strong>g mousereproduction ability and preservation potential. <strong>Reproduction</strong>133, 841–846.Kikuchi K, Nagai T, D<strong>in</strong>g J, Yamauchi N, Noguchi J, IzaikeY, 1999: Cytoplasmic maturation for activation of pigfollicular oocytes cultured and arrested at metaphase-I. JReprod Fertil 116, 143–156.Kikuchi K, Onishi A, Kashiwazaki N, Iwamoto M, Noguchi J,Kaneko H, Akita T, Nagai T, 2002: Successful pigletproduction after transfer of blastocysts produced by amodified <strong>in</strong> vitro system. Biol Reprod 66, 1033–1041.Kikuchi K, Nakai M, Kashiwazaki N, Ozawa M, MaedomariN, Noguchi J, Ohnuma K, Kaneko H, 2006a: Porc<strong>in</strong>eblastocysts derived from <strong>in</strong> vitro matured oocytes <strong>in</strong>jected<strong>in</strong>tracytoplasmically with a sperm from testicular tissuexenografted <strong>in</strong>to nude mice. Reprod Fertil Dev 18, 247.Kikuchi K, Kaneko H, Nakai M, Noguchi J, Ozawa M,Ohnuma K, Kashiwazaki N, 2006b: In vitro and <strong>in</strong> vivodevelopmental ability of oocytes derived from porc<strong>in</strong>eprimordial follicles xenografted <strong>in</strong>to nude mice. J ReprodDev 52, 51–57.Kim NH, Sh<strong>in</strong> JS, Kim C, Jun SH, Lee HT, Chung KS, 1999:Fertilization and <strong>in</strong> vitro development of porc<strong>in</strong>e oocytesfollow<strong>in</strong>g <strong>in</strong>tracytoplasmic <strong>in</strong>jection of round spermatid orround spermatid nuclei. Theriogenology 51, 1441–1449.Kim SS, Soules MR, Battaglia DE, 2002: Follicular development,ovulation, and corpus luteum formation <strong>in</strong> cryopreservedhuman ovarian tissue after xenotransplantation.Fertil Steril 78, 77–82.Kimura Y, Yanagimachi R, 1995a: Intracytoplasmic sperm<strong>in</strong>jection <strong>in</strong> the mouse. Biol Reprod 52, 709–720.Kimura Y, Yanagimachi R, 1995b: Development of normalmice from oocytes <strong>in</strong>jected with secondary spermatocytenuclei. Biol Reprod 53, 855–862.Kimura Y, Tateno H, Handel MA, Yanagimachi R, 1998:Factors affect<strong>in</strong>g meiotic and developmental competence ofprimary spermatocyte nuclei <strong>in</strong>jected <strong>in</strong>to mouse oocytes.Biol Reprod 59, 871–877.Kolbe T, Holtz W, 2000: Birth of a piglet derived from anoocyte fertilized by <strong>in</strong>tracytoplasmic sperm <strong>in</strong>jection (ICSI).Anim Reprod Sci 64, 97–101.Kurome M, Ueda H, Tomii R, Naruse K, Nagashima H, 2006:Production of transgenic-clone pigs by the comb<strong>in</strong>ation ofICSI-mediated gene transfer with somatic cell nucleartransfer. Transgenic Res 15, 229–240.Lacham-Kaplan O, Trounson A, 1995: Intracytoplasmicsperm <strong>in</strong>jection <strong>in</strong> mice: <strong>in</strong>creased fertilization and developmentto term after <strong>in</strong>duction of the acrosome reaction. HumReprod 10, 2462–2469.Lavitrano M, Camaioni A, Fazio VM, Dolci S, Farace MG,Spadafora C, 1989: Sperm cells as vectors for <strong>in</strong>troduc<strong>in</strong>gforeign DNA <strong>in</strong>to eggs: genetic transformation of mice. Cell57, 717–723.Lavitrano M, Forni M, Varzi V, Pucci L, Bacci ML, DiStefano C, Fioretti D, Zoraqi G, Moioli B, Rossi M,Lazzereschi D, Stoppacciaro A, Seren E, Alfani D, Cortes<strong>in</strong>iR, Frati L, 1997: Sperm mediated gene transfer: productionof pigs transgenic for a human regulator of complementactivation. Transplant Res 29, 3508–3509.Lavitrano M, Stoppacciaro A, Bacci ML, Forni M, Fioretti D,Pucci L, Di Stefano C, Lazzereschi D, Rughetti A, CerettaS, Zannoni A, Rahimi H, Moioli B, Rossi M, Nuti M, RossiG, Seren E, Alfani D, Cortes<strong>in</strong>i R, Frati L, 1999: Humandecay accelerat<strong>in</strong>g factor transgenic pigs for xenotransplantationobta<strong>in</strong>ed by sperm-mediated gene transfer. TransplantRes 31, 927–974.Lazzereschi D, Forni M, Cappello F, Bacci ML, Di Stefano C,Marfe G, Giancotti P, Renzi L, Wang HJ, Rossi M, DellaCasa G, Pretagost<strong>in</strong>i R, Frati G, Bruzzone P, Stassi G,Stoppacciaro A, Turchi V, Cortes<strong>in</strong>i R, S<strong>in</strong>ibaldi P, Frati L,Lavitrano M, 2000: Efficiency of transgenesis us<strong>in</strong>g spermmediatedgene transfer: generation of hDAF transgenic pigs.Transplant Res 32, 892–894.Lee DM, Yeoman RR, Battaglia DE, Stouffer RL, Zel<strong>in</strong>ski-Wooten MB, Fanton JW, Wolf DP, 2004: Live birth afterovarian tissue transplant. Nature 428, 137–138.Li GP, Chen DY, Lian L, Sun QY, Wang MK, Liu JL, Li JS,Han ZM, 2001: Viable rabbits derived from reconstructedoocytes by germ<strong>in</strong>al vesicle transfer after <strong>in</strong>tracytoplasmicsperm <strong>in</strong>jection (ICSI). Mol Reprod Dev 58, 180–185.Liu JL, Kusakabe H, Chang CC, Suzuki H, Schmidt DW,Julian M, Pfeffer R, Bormann CL, Tian XC, YanagimachiR, Yang X, 2004: Freeze-dried sperm fertilization leads tofull-term development <strong>in</strong> rabbits. Biol Reprod 70, 1776–1781.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


406 K Kikuchi, N Kashiwazaki, T Nagai, M Nakai, T Somfai, J Noguchi and H KanekoMarchal R, Feugang JM, Perreau C, Venturi E, Terqui M,Miemillod P, 2001: Meiotic and developmental competenceof prepubertal and adult sw<strong>in</strong>e oocytes. Theriogenology 56,17–29.Mart<strong>in</strong> MJ, 2000: Development of <strong>in</strong> vivo-matured porc<strong>in</strong>eoocytes follow<strong>in</strong>g <strong>in</strong>tracytoplasmic sperm <strong>in</strong>jection. BiolReprod 63, 109–112.Mattioli M, Bacci ML, Galeati G, Seren E, 1989: Developmentalcompetence of pig oocytes matured and fertilized <strong>in</strong>vitro. Theriogenology 39, 1201–1207.Mattiske D, Shaw G, Shaw JM, 2002: Influence of donor ageon development of gonadal tissue from pouch young of thetammar wallaby, Macropus eugenii, after cryopreservationand xenograft<strong>in</strong>g <strong>in</strong>to mice. <strong>Reproduction</strong> 123, 143–153.McCauley TC, Mazza MR, Didion BA, Mao J, Wu G,Coppola G, Coppola GF, Di Berard<strong>in</strong>o D, Day BN, 2003:Chromosomal abnormalities <strong>in</strong> Day-6, <strong>in</strong> vitro-produced pigembryos. Theriogenology 60, 1569–1580.Metcalfe SS, Shaw JM, Gunn IM, 2001: Xenograft<strong>in</strong>g ofcan<strong>in</strong>e ovarian tissue to ovariectomized severe comb<strong>in</strong>edimmunodeficient (SCID) mice. J Reprod Fertil Suppl 57,323–329.Nagai T, 1996: In vitro maturation and fertilization of pigoocytes. Anim Reprod Sci 42, 153–163.Nagai T, Kikuchi K, Imai K, Fahrud<strong>in</strong> M, 2007: Handmadesomatic cell clon<strong>in</strong>g and its related studies <strong>in</strong> farm animals. JMammal Ova Res 24, 99–106.Nagy ZP, Cecile J, Liu J, Loccufier A, Devroey P, VanSteirteghem A, 1996: Pregnancy and birth after <strong>in</strong>tracytoplasmicsperm <strong>in</strong>jection of <strong>in</strong> vitro matured germ<strong>in</strong>al-vesiclestage oocytes: case report. Fertil Steril 65, 1047–1050.Nakai M, Kashiwazaski N, Takizawa A, Hayashi Y, NakatsukasaE, Fuchimoto D, Noguchi J, Kaneko H, Sh<strong>in</strong>o M,Kikuchi K, 2003: Viable piglets generated from porc<strong>in</strong>eoocytes matured <strong>in</strong> vitro and fertilized by <strong>in</strong>tracytoplasmicsperm head <strong>in</strong>jection. Biol Reprod 68, 1003–1008.Nakai M, Kashiwazaki N, Takizawa A, Maedomari N, OzawaM, Noguchi J, Kaneko H, Sh<strong>in</strong>o M, Kikuchi K, 2006:Morphological changes <strong>in</strong> boar sperm nuclei with reduceddisulfide bonds <strong>in</strong> electrostimulated porc<strong>in</strong>e oocytes. <strong>Reproduction</strong>131, 603–611.Nakai M, Kashiwazaki N, Takizawa A, Maedomari N, OzawaO, Noguchi J, Kaneko H, Sh<strong>in</strong>o M, Kikuchi K, 2007:Effects of chelat<strong>in</strong>g agents dur<strong>in</strong>g freeze-dry<strong>in</strong>g of boarspermatozoa on DNA fragmentation and on developmentalability <strong>in</strong> vitro and <strong>in</strong> vivo after <strong>in</strong>tracytoplasmic sperm head<strong>in</strong>jection. Zygote 15, 15–24.Ogura A, Matsuda J, Yanagimachi R, 1994: Birth of normalyoung after electrofusion of mouse oocytes with roundspermatids. Proc Natl Acad Sci U S A 91, 7460–7462.Ogura A, Suzuki O, Tanemura K, Mochida K, Kobayashi Y,Matsuda J, 1998: Development of normal mice frommetaphase I oocytes fertilized with primary spermatocytes.Proc Natl Acad Sci U S A 95, 5611–5615.Oktay K, Newton H, Mullan J, Gosden RG, 1998: Developmentof human primordial follicles to antral stages <strong>in</strong>SCID ⁄ hpg mice stimulated with follicle stimulat<strong>in</strong>g hormone.Hum Reprod 13, 1133–1138.Palermo G, Joris H, Devroey P, Van Steirteghem AC, 1992:Pregnancies after <strong>in</strong>tracytoplasmic <strong>in</strong>jection of s<strong>in</strong>gle spermatozoon<strong>in</strong>to an oocyte. Lancet 340, 17–18.Perry AC, Wakayama T, Kishikawa H, Kasai T, Okabe M,Toyoda Y, Yanagimachi R, 1999: Mammalian transgenesisby <strong>in</strong>tracytoplasmic sperm <strong>in</strong>jection. Science 284, 1180–1183.Sasagawa I, Kuretake S, Eppig JJ, Yanagimachi R, 1998:Mouse primary spermatocytes can complete two meioticdivisions with<strong>in</strong> the oocyte cytoplasm. Biol Reprod 58, 248–254.Senbon S, Ota A, Tachibana M, Miyano T, 2003: Bov<strong>in</strong>eoocytes <strong>in</strong> secondary follicles grow and acquire meioticcompetence <strong>in</strong> severe comb<strong>in</strong>ed immunodeficient mice.Zygote 11, 139–149.Smith KR, 1999: Sperm cell mediated transgenesis: a review.Anim Biotechnol 10, 1–13.Snow M, Cox S-L, Jenk<strong>in</strong> G, Trounson A, Shaw J, 2002:Generation of live young from xenografted mouse ovaries.Science 297, 2227.Somfai T, Kikuchi K, Medvedev SU, Onishi A, Iwamoto M,Fuchimoto D, Ozawa M, Noguchi N, Kaneko H, OhnumaK, Sato E, Nagai T, 2005: Development to the blastocyststage of immature pig oocytes arrested before the metaphase-IIstage and fertilized <strong>in</strong> vitro. Anim Reprod Sci 90,307–328.Somfai T, Ozawa M, Noguchi J, Kaneko H, Ohnuma K,Karja NWK, Farhud<strong>in</strong> M, Maedomari N, D<strong>in</strong>nye´s A,Nagai T, Kikuchi K, 2006: Diploid porc<strong>in</strong>e parthenotesproduced by the <strong>in</strong>hibition of first polar body extrusiondur<strong>in</strong>g <strong>in</strong> vitro maturation of follicular oocytes. <strong>Reproduction</strong>132, 559–570.Somfai T, Ozawa M, Noguchi J, Kaneko H, Karja NWK,Fahrud<strong>in</strong> M, Nakai M, Maedomari N, D<strong>in</strong>nye´s A, Nagai T,Kikuchi K(<strong>in</strong> press): In vitro development of polyspermicporc<strong>in</strong>e oocytes: relationship between early fragmentationand excessive number of penetrat<strong>in</strong>g spermatozoa. AnimReprod Sci 117, 131–147.Uehara T, Yanagimachi R, 1976: Microsurgical <strong>in</strong>jection ofspermatozoa <strong>in</strong>to hamster eggs with subsequent transformationof sperm nuclei <strong>in</strong>to male pronuclei. Biol Reprod 15,467–470.Uehara T, Yanagimachi R, 1977: Behavior of nuclei oftesticular, caput and cauda epididymal spermatozoa <strong>in</strong>jected<strong>in</strong>to hamster eggs. Biol Reprod 16, 315–321.Wakayama T, Yanagimachi R, 1998: Development of normalmice from oocytes <strong>in</strong>jected with freeze-dried spermatozoa.Nat Biotechnol 16, 639–641.Wall RJ, 2002: New gene transfer methods. Theriogenology57, 189–201.Wang MK, Chen DY, Lui JL, Li GP, Sun QY, 2001: In vitrofertilisation of mouse oocytes reconstructed by transfer ofmetaphase-II chromosomes results <strong>in</strong> live births. Zygote 9,9–14.Ward MA, Kaneko T, Kusakabe H, Biggers JD, Whitt<strong>in</strong>ghamDG, Yanagimachi R, 2003: Long-term preservation ofmouse spermatozoa after freeze-dry<strong>in</strong>g and freez<strong>in</strong>g withoutcryoprotection. Biol Reprod 69, 2100–2108.Weissman A, Gotlieb L, Colgan T, Jurisicova A, GreenblattEM, Casper RF, 1999: Prelim<strong>in</strong>ary experience with subcutaneoushuman ovarian cortex transplantation <strong>in</strong> the NOD-SCID mouse. Biol Reprod 60, 1462–1467.Yamauchi Y, Yanagimachi R, Horiuchi T, 2002: Full-termdevelopment of golden hamster oocytes follow<strong>in</strong>g <strong>in</strong>tracytoplasmicsperm head <strong>in</strong>jection. Biol Reprod 67, 534–553.Yoshida M, Mizoguchi Y, Ishiaki K, Kojima T, Nagai T,1993: Birth of piglets derived from <strong>in</strong> vitro fertilization ofpig oocytes matured <strong>in</strong> vitro. Theriogenology 39, 1303–1311.Author’s address (for correspondence): Kazuhiro Kikuchi, Division ofAnimal Sciences, National Institute of Agrobiological Sciences,Kannondai 2-1-2, Tsukuba, Ibaraki 305-8602, Japan. E-mail:kiku@affrc.go.jpConflict of <strong>in</strong>terest: All authors declare no conflict of <strong>in</strong>terests.Ó 2008 The Authors. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 407–416 (2008); doi: 10.1111/j.1439-0531.2008.01192.xISSN 0936-6768Climb<strong>in</strong>g Mount Efficiency – Small Steps, Not Giant Leaps Towards Higher Clon<strong>in</strong>gSuccess <strong>in</strong> Farm <strong>Animals</strong>Bjo¨rn ObackAgResearch Ltd., Ruakura Research Centre, Hamilton, New ZealandContentsDespite more than a decade of research efforts, farm animalclon<strong>in</strong>g by somatic cell nuclear transfer (SCNT) is stillfrustrat<strong>in</strong>gly <strong>in</strong>efficient. Inefficiency manifests itself at differentlevels, which are currently not well <strong>in</strong>tegrated. At the molecularlevel, it leads to widespread genetic, epigenetic andtranscriptional aberrations <strong>in</strong> cloned embryos. At the organismallevel, these genome-wide abnormalities compromisedevelopment of cloned foetuses and offspr<strong>in</strong>g. Specific moleculardefects need to be causally l<strong>in</strong>ked to specific clonedphenotypes, <strong>in</strong> order to design specific treatments to correctthem. Clon<strong>in</strong>g efficiency depends on the ability of the nucleardonor cell to be fully reprogrammed <strong>in</strong>to an embryonic stateand the ability of the enucleated recipient cell to carry out thereprogramm<strong>in</strong>g reactions. It has been postulated that reprogrammabilityof the somatic donor cell epigenome is <strong>in</strong>fluencedby its differentiation status. However, directcomparisons between cells of divergent differentiation statuswith<strong>in</strong> several somatic l<strong>in</strong>eages have found no conclusiveevidence for this. Choos<strong>in</strong>g somatic stem cells as donors hasnot improved clon<strong>in</strong>g efficiency, <strong>in</strong>dicat<strong>in</strong>g that donor cell typemay be less critical for clon<strong>in</strong>g success. Different recipient cells,on the otherhand, vary <strong>in</strong> their reprogramm<strong>in</strong>g ability. Inbov<strong>in</strong>e, us<strong>in</strong>g zygotes <strong>in</strong>stead of oocytes has <strong>in</strong>creased clon<strong>in</strong>gsuccess. Other improvements <strong>in</strong> livestock clon<strong>in</strong>g efficiency<strong>in</strong>clude better coord<strong>in</strong>at<strong>in</strong>g donor cell type with cell cycle stageand aggregat<strong>in</strong>g cloned embryos. In the future, it will beimportant to demonstrate if these small <strong>in</strong>creases at every stepare cumulative, add<strong>in</strong>g up to an <strong>in</strong>tegrated clon<strong>in</strong>g protocolwith greatly improved efficiency.The Importance of Farm Animal Clon<strong>in</strong>gIn more than a decade s<strong>in</strong>ce the birth of Dolly the sheep,cloned offspr<strong>in</strong>g have been produced by somatic cellnuclear transfer (SCNT) <strong>in</strong> 18 mammalian species.Despite this ever grow<strong>in</strong>g list, SCNT rema<strong>in</strong>s very<strong>in</strong>efficient compared with other assisted reproductivetechnologies such as <strong>in</strong> vitro fertilization (IVF) orartificial <strong>in</strong>sem<strong>in</strong>ation. Typically, clon<strong>in</strong>g efficiency,quantified as the proportion of all embryos transferred<strong>in</strong>to surrogate mothers that develop <strong>in</strong>to viable offspr<strong>in</strong>g,is about 1%–5% (Oback and Wells 2007a). Overthree-quarters of all clon<strong>in</strong>g laboratories are work<strong>in</strong>g onfarm animals (cattle, pig, goat, sheep, buffalo and deer),illustrat<strong>in</strong>g that the ma<strong>in</strong> objective beh<strong>in</strong>d SCNT is stillcommercially driven – namely to multiply elite animalswith desired phenotypic traits and to produce geneticallymodified animals (Oback and Wells 2007a). As aconsequence of <strong>in</strong>creased research effort and fund<strong>in</strong>g<strong>in</strong> the area, the total number of clon<strong>in</strong>g publications has<strong>in</strong>creased by an order of magnitude <strong>in</strong> the past decade,and still cont<strong>in</strong>ues to grow (Fig. 1). Cattle SCNT haslong dom<strong>in</strong>ated the NT publication record, account<strong>in</strong>gfor an annual average of about 25% of PubMed-listedpapers s<strong>in</strong>ce 1994. Pig is the second most importantcloned farm animal by this measure (13% of NTpublications), followed by goat, sheep, buffalo and reddeer (altogether 6%). Overall, farm animal clon<strong>in</strong>g thusaccounts for 44% of clon<strong>in</strong>g publications, laboratoryanimals (mouse, rabbit, monkey and rat) for 22%, otherspecies (<strong>in</strong>clud<strong>in</strong>g human) for 16% and general reviewarticles, which are not species-specific, for the rema<strong>in</strong><strong>in</strong>g18%. Based solely on past research <strong>in</strong>vestment andoutput, i.e. the number of labs <strong>in</strong>volved and theirpublications, cattle is still the most important clonedlivestock species (Oback and Wells 2007a).Nuclear Reprogramm<strong>in</strong>gAfter NT of a fully differentiated donor cell <strong>in</strong>to acytoplast, the result<strong>in</strong>g reconstruct can develop <strong>in</strong>to anembryo and even a viable animal. The logical alternative,i.e. that the NT reconstruct cleaves <strong>in</strong>to fullydifferentiated donor cells, has never been observed.Eras<strong>in</strong>g transcriptional programme and epigenetic identityof the donor cell is referred to as nuclear reprogramm<strong>in</strong>g.The molecular dom<strong>in</strong>ance of the oocyte overany somatic cell type tested may simply be due to itbe<strong>in</strong>g a 1000-fold larger <strong>in</strong> volume and thus conta<strong>in</strong><strong>in</strong>g a1000-fold excess of oocyte-specific factors, <strong>in</strong> which casethe reprogramm<strong>in</strong>g dom<strong>in</strong>ance should disappear oncecell size differences are experimentally adjusted. This issupported by the observation that nuclear reprogramm<strong>in</strong>galso occurs <strong>in</strong> differentiated cells fused to nondivid<strong>in</strong>gmult<strong>in</strong>ucleate heterokaryons (Blau et al. 1983;Terranova et al. 2006) with the direction of reprogramm<strong>in</strong>gbe<strong>in</strong>g dictated by the ratio of the nuclei derivedfrom each cell type (Pavlath and Blau 1986). Thecapacity to reverse stable heritable epigenetic modifications,such as DNA-methylation, is not particular tooocytes, but also occurs <strong>in</strong> embryonic stem (ES) cells(Tada et al. 2003) and even fully differentiated skeletalmuscle cells (Zhang et al. 2007). However, so far onlyoocytes have been capable of reprogramm<strong>in</strong>g somaticcells to the extent of giv<strong>in</strong>g rise to a completely newcloned organism. Clone survival <strong>in</strong>to adulthood is thusthe most <strong>in</strong>formative and mean<strong>in</strong>gful measure of extensivedonor cell reprogramm<strong>in</strong>g. Reprogramm<strong>in</strong>g efficiencyafter NT critically depends on two processes: theability of the nuclear donor cell to be fully reprogrammedand the ability of the oocyte to carry out thereprogramm<strong>in</strong>g reactions. As it is currently unclearwhich process is more important for reprogramm<strong>in</strong>gsuccess, both will be discussed <strong>in</strong> this review.Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


408 B ObackNumber of publications200180160140120100806040200sheep cattle goatpigred deer,buffalo1994 1995 1996 1997 1998 1999 2000 2001 2002 2003 2004 2005 2006 2007Publication yearFig. 1. Increase <strong>in</strong> clon<strong>in</strong>g-relatedpublications over time. Arrowspo<strong>in</strong>t to the year when the firstclone of the <strong>in</strong>dicated farm animalwas published. The number ofpeer-reviewed publications wasbased on search<strong>in</strong>g PubMed, Webof Science Ò [v3.0], and conferenceproceed<strong>in</strong>gsReprogrammability of the Donor CellA hypothesis has been emerg<strong>in</strong>g that epigenetic reprogramm<strong>in</strong>gafter NT and clon<strong>in</strong>g efficiency are <strong>in</strong>verselycorrelated with donor cell differentiation (Oback andWells 2007b). This hypothesis is ma<strong>in</strong>ly supported bythree l<strong>in</strong>es of evidence from comparative mouse clon<strong>in</strong>gexperiments us<strong>in</strong>g: (i) progressively advanced blastomeredonor nuclei from early-cleavage stages, (ii) EScells and (iii) term<strong>in</strong>ally differentiated lymphocytes andneurons. These comparisons demonstrated that earlyblastomeres result <strong>in</strong> much higher clon<strong>in</strong>g efficiency thansomatic cells (Hiiragi and Solter 2005). However, theyfailed to conclusively determ<strong>in</strong>e whether differentiationstatus significantly affects clon<strong>in</strong>g efficiency with<strong>in</strong>somatic donor cell l<strong>in</strong>eages (Oback and Wells 2007b).Term<strong>in</strong>ally differentiated cells vs term<strong>in</strong>ally differentiatedcellsThe first study that compared NT clon<strong>in</strong>g efficiency oftwo epigenetically dist<strong>in</strong>ct donor cell subpopulationswith<strong>in</strong> the same somatic l<strong>in</strong>eage was performed <strong>in</strong>lymphocytes. It compared two fully differentiated T celltypes, namely natural killer T (NKT) lymphocytes andhelper T cells. The two lymphocyte populations wereisolated from the same stra<strong>in</strong> of male mice, purified to>98% us<strong>in</strong>g fluorescence-activated cell sort<strong>in</strong>g and usedfreshly for NT. As shown before (Hochedl<strong>in</strong>ger andJaenisch 2002), clon<strong>in</strong>g from T lymphocytes wasextremely <strong>in</strong>efficient. NKT cells supported early postimplantationdevelopment (60% vs 7% to implantation)much better than helper T cells; however, clon<strong>in</strong>gefficiency was not significantly different (1.5% vs 0%;Fig. 2). The lymphocyte data confirmed that term<strong>in</strong>allydifferentiated cells can be reprogrammed, but providedno comparison for the efficiency of this process with lessdifferentiated cells from the same l<strong>in</strong>eage, e.g. haematopoieticstem or precursor cells.Progenitor cells vs term<strong>in</strong>ally differentiated cellsCells of divergent differentiation status from the samesomatic l<strong>in</strong>eage were first compared <strong>in</strong> mouse, butaga<strong>in</strong> the results were not conclusive (Yamazaki et al.2001). Undifferentiated neural progenitors and moredifferentiated immature young neurons from the foetalcerebral cortex resulted <strong>in</strong> live offspr<strong>in</strong>g with differentefficiencies (12% vs 2%; Fig. 2); however, numberswere too small to be significant. In order to directly testthe hypothesis <strong>in</strong> farm animals, we used <strong>in</strong>creas<strong>in</strong>gly% clon<strong>in</strong>g efficiency20 antlerblood18muscle16neural14sk<strong>in</strong>121086420ASCadipocyteHSCgranulocyteNKT cellT cellmuscle fibroblastMPCmyotubeNSCNPCimmatureneuronmatureneuron 1KSC (male)KPC (male)Antler Blood Muscle Neural Sk<strong>in</strong>Increas<strong>in</strong>g differentiationKSC (female)KPC (female)Fig. 2. Somatic clon<strong>in</strong>g efficiencyand donor cell type. Cell typesfrom different l<strong>in</strong>eages are listed bydegree of differentiation. Clon<strong>in</strong>gefficiency is measured as theproportion of surviv<strong>in</strong>g offspr<strong>in</strong>gper number of embryos transferred;1 neuronal clones werescored for normal development atmid-gestation. ASC, antler stemcell; HSC, haematopoietic stemcell; NKT, natural killer T cell;MPC, muscle precursor cell; NSC,neural stem cell; KSC, kerat<strong>in</strong>ocytestem cell; KPC, kerat<strong>in</strong>ocyteprecursor cell. For mouse NT(blood, neural and sk<strong>in</strong> cells), datafrom different research groupsus<strong>in</strong>g the same cell type and cellcycle stage but different genotypeswere pooledÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Steps to Improve Farm Animal Clon<strong>in</strong>g Efficiency 409differentiated donors from the bov<strong>in</strong>e skeletal musclel<strong>in</strong>eage, namely foetal myogenic precursors and their<strong>in</strong> vitro differentiated progeny, post-mitotic mononucleatedmyotubes (Green et al. 2007). Several factorsserved as early (PAX7 and MYOD1) and late (MYOG,MYF6 and MHY) markers of myogenic determ<strong>in</strong>ationand differentiation, respectively. Rather than rely<strong>in</strong>g onantigen profil<strong>in</strong>g of bulk populations, we determ<strong>in</strong>edthe purity of <strong>in</strong>dividually size-selected cells actuallyprepared for NT. After selection, s<strong>in</strong>gle precursors wereessentially all MYOD1- and PAX7-positive as well asMYOG- and MYH-negative, whereas myotubes werePAX7-negative, weakly MYOD1- and stronglyMYOG-positive and already expressed the late differentiationmarker MHY. Follow<strong>in</strong>g NT, muscle-specificgenes PAX7, MYOD1, MYOG and MYF6, weresilenced and rema<strong>in</strong>ed undetectable up to the blastocyststage. Despite significant differences <strong>in</strong> development toblastocysts, <strong>in</strong> vivo viability of those cloned embryoswas not dependant on donor cell type or differentiationstatus.Adult stem cells vs term<strong>in</strong>ally differentiated cellsWe then compared cells from either end of the somaticdifferentiation cont<strong>in</strong>uum, namely undifferentiated antlerstem cells (ASCs) and their term<strong>in</strong>ally differentiatedprogeny, adipocytes (Berg et al. 2007). Antler stemcells are anatomically and histologically well def<strong>in</strong>edand capable of proliferation, lifelong self-renewal andproduction of differentiat<strong>in</strong>g daughter cells. In vivo,they give rise to all different antler l<strong>in</strong>eages (e.g.sk<strong>in</strong>, blood, nerve, cartilage, bone and connectivetissue), even after ectopic transplantation <strong>in</strong>to mice(Li et al. 2001). In vitro, ASCs are highly proliferative,giv<strong>in</strong>g rise to cell l<strong>in</strong>es that can be differentiated <strong>in</strong>toseveral mesodermal cell l<strong>in</strong>eages, such as bone, cartilageand even adipocytes, a cell type which they wouldnot normally form <strong>in</strong> vivo (Berg et al. 2007). Follow<strong>in</strong>gNT, antlerogenic (POU5F1 and PTHLH) as well asadipogenic markers (COL1A2, PPARG and GAPDH)were successfully reprogrammed <strong>in</strong> cloned blastocysts.Despite differences prior to NT, transcript abundanceof donor-specific markers did not differ significantly <strong>in</strong>cloned blastocysts from both cell types, <strong>in</strong>dicat<strong>in</strong>g thattranscriptional reprogramm<strong>in</strong>g was <strong>in</strong>dependent of thedonor cell source. In keep<strong>in</strong>g with the similar degree ofgene expression reprogramm<strong>in</strong>g for ASCs vs adipocytes,there were also no differences <strong>in</strong> development tothe blastocyst stage. Importantly, survival to wean<strong>in</strong>gwas not significantly different between ASCs andadipocytes. This f<strong>in</strong>d<strong>in</strong>g was consistent with previousresults <strong>in</strong> bov<strong>in</strong>e us<strong>in</strong>g mesenchymal stem cells withoutany obvious <strong>in</strong>crease <strong>in</strong> clon<strong>in</strong>g efficiency (Kato et al.2004). The farm animal data are also supported by anumber of comparative studies <strong>in</strong> mouse us<strong>in</strong>g stemcells isolated from haematopoietic (Inoue et al. 2005,2006), mesenchymal (Inoue et al. 2007), neuronal(Yamazaki et al. 2001) and sk<strong>in</strong> (Li et al. 2007)l<strong>in</strong>eages. None of these studies have found anysignificant evidence that adult stem cells result <strong>in</strong>higher clon<strong>in</strong>g efficiency than differentiated cells fromeither the same or some unrelated l<strong>in</strong>eage (Fig. 2). Infact, at least for haematopoietic and mesenchymal stemcells, clon<strong>in</strong>g efficiencies were among the lowestreported for any somatic donor cell type (Inoue et al.2006, 2007).Differentiated vs embryonic stem cellsEmbryonic stem cells are immortal pluripotent (able togive rise to all cell types of the embryo proper) cellsderived from plac<strong>in</strong>g blastomeres <strong>in</strong>to <strong>in</strong> vitro culture(Evans and Kaufman 1981; Mart<strong>in</strong> 1981). In mouse,high ES-cell clon<strong>in</strong>g efficiencies have been reported(Rideout et al. 2000); however, these are matched bycerta<strong>in</strong> somatic cell types, provided they share the samegenotype, sex and cell cycle stage (Oback and Wells2007b). All attempts to derive livestock bona fide EScells have failed so far. However, long-term ‘ES-like cell’cultures <strong>in</strong> farm animal species resemble mur<strong>in</strong>e ES cells<strong>in</strong> morphology, antigen profile, extensive <strong>in</strong> vitro differentiationability even after long-term culture andsomatic cell chimerism (Renard et al. 2007). Clon<strong>in</strong>gefficiency with ES-like cells appears to be relatively high(Saito et al. 2003). Because ES cells can be differentiated<strong>in</strong>to a variety of cell types, it would be <strong>in</strong>structive tocompare their clon<strong>in</strong>g efficiency with that of theirdifferentiated progeny. Given the limited availabilityof ES cells <strong>in</strong> livestock, this experiment would perhapsbest be conducted <strong>in</strong> mouse. Initial experiments alongthose l<strong>in</strong>es demonstrated that embryos cloned from EScellderived neural stem cells (NSCs) showed muchhigher clon<strong>in</strong>g efficiency than differentiated neurons asdonors (Blelloch et al. 2006). However, clon<strong>in</strong>g efficiencywas quantified by the frequency of deriv<strong>in</strong>g EScell l<strong>in</strong>es from NT blastocysts. Repeat<strong>in</strong>g such anexperiment and measur<strong>in</strong>g survival <strong>in</strong>to adulthoodmight clarify whether two epigenotypes from each endof the differentiation spectrum show any differences <strong>in</strong>reprogrammability. Early embryos are not the onlysource for derivation of pluripotent cells. Multipotentialspermatogonial stem cells, isolated from mouse testis,also result <strong>in</strong> germl<strong>in</strong>e transmission (Kanatsu-Sh<strong>in</strong>oharaet al. 2004) and would make <strong>in</strong>terest<strong>in</strong>g donor candidates.Induced pluripotent stem (iPS) cells, obta<strong>in</strong>edafter transduction of somatic cells with a core set oftranscription factors (Takahashi and Yamanaka 2006),may be even more similar to ES cells and theirapplicability for livestock clon<strong>in</strong>g will be very importantto evaluate.Cell cycle coord<strong>in</strong>ationCloned calves have been generated from serum-starved(Kato et al. 1998; Wells et al. 1999), proliferat<strong>in</strong>g(Cibelli et al. 1998; Kas<strong>in</strong>athan et al. 2001) and mitoticallyarrested somatic donor cells (Tani et al. 2001;Heyman et al. 2002b). For non-transgenic fibroblasts,the overall output of viable calves at wean<strong>in</strong>g wassignificantly higher with serum-starved than with G 1cells (Wells et al. 2003). For transgenic fibroblasts,however, the results were reversed (Wells et al. 2003).This suggests that it may be necessary to coord<strong>in</strong>atedonor cell type and cell cycle stage <strong>in</strong> order to maximizeoverall clon<strong>in</strong>g efficiency.Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


410 B ObackReprogramm<strong>in</strong>g Ability of the Recipient CellWhile there are over 200 donor cell types to choosefrom, choices for the recipient cell for NT are muchmore limited. The most commonly used type is harvestedas non-activated MI oocytes from follicles ofslaughtered adult animals. The maturation <strong>in</strong>to MIIoocytes can occur <strong>in</strong> vivo or <strong>in</strong> vitro and both types havebeen successfully used for clon<strong>in</strong>g. In mice, naturallyovulated oocytes improve clon<strong>in</strong>g efficiency comparedwith superovulated ones (Hiiragi and Solter 2005);however, us<strong>in</strong>g them <strong>in</strong> mono-ovulatory species, suchas cattle, is impractical. In contrast to the donor, geneticfactors should be less important for choos<strong>in</strong>g therecipient cell because its nuclear genome is removedprior to NT. However, genotype affects transcriptomeand proteome of the oocyte. The fact that hybrid vigouris beneficial for recipient cells (Gao et al. 2004) and thatearly transcriptional activation varies <strong>in</strong> clones reconstructedwith oocytes from different mouse stra<strong>in</strong>semphasizes the importance of the recipient cell’s genotypedur<strong>in</strong>g clon<strong>in</strong>g (Vassena et al. 2008). Furthercomparative studies are needed to fully evaluate the<strong>in</strong>fluence of oocyte source and maturation method onlivestock clon<strong>in</strong>g efficiency. Cytoplasts prepared fromartificially activated MII oocytes (early telophase or TII)have also sometimes been used to clone goats (Baguisiet al. 1999) and cattle (Bordignon and Smith 1998;Kurosaka et al. 2002; Bordignon et al. 2003). Presently,there is no evidence that MII and TII cytoplasm differ <strong>in</strong>their reprogramm<strong>in</strong>g ability.In early mammalian clon<strong>in</strong>g experiments, fertilizedoocytes (zygotes) were successfully used as recipientswith two-cell stage donor cells <strong>in</strong> mouse (McGrath andSolter 1984; Robl et al. 1986), cattle (Prather and First1990) and pig (Prather et al. 1989). Once more advancedembryonic or even somatic donors were used, enucleatedzygotes repeatedly failed to support early developmentof the reconstituted NT embryo (Wakayama et al.2000). This 20-year-old perception that SCNT successcritically depended on the use of unfertilized oocyteschanged radically when the first cloned cattle (Schurmannet al. 2006) and mice (Greda et al. 2006; Egli et al.2007) were born from NT with somatic and embryonicdonors, respectively, <strong>in</strong>to zygotes.Recipient zygotes can be either <strong>in</strong> <strong>in</strong>terphase or <strong>in</strong>mitosis. Interphase zygotes were first reproducibly usedfor clon<strong>in</strong>g mice <strong>in</strong> 2006 (Greda et al. 2006), perhapspartially confirm<strong>in</strong>g much earlier attempts (Illmenseeand Hoppe 1981). They succeeded by develop<strong>in</strong>g a newmicromanipulation technique for enucleation. The classicalway of enucleat<strong>in</strong>g (McGrath and Solter 1983),removes <strong>in</strong>tact pronuclei <strong>in</strong>clud<strong>in</strong>g all nuclear componentsand some surround<strong>in</strong>g cytoplasm. In contrast, thenew technique appears to more selectively remove onlythe pronuclear envelope and its attached chromat<strong>in</strong>,while leav<strong>in</strong>g other pronuclear components, such asnucleoli, beh<strong>in</strong>d. These zygotic cytoplasts supportedfull-term development after NT of eight-cell donor cells.It would be <strong>in</strong>formative to determ<strong>in</strong>e which factorsrema<strong>in</strong> <strong>in</strong> the zygotic cytoplasm after selective enucleationand if these factors would be sufficient to alsoreprogram somatic donor nuclei. However, the latterexperiment is technically challeng<strong>in</strong>g because of the cellcycle <strong>in</strong>compatibility between somatic donor andrecipient cell. Donor–recipient cell cycle coord<strong>in</strong>ationis important to ma<strong>in</strong>ta<strong>in</strong> normal ploidy and promotereprogramm<strong>in</strong>g, both of which are l<strong>in</strong>ked to the level ofcycl<strong>in</strong> B ⁄ Cdk 1 complex activity (M-Cdk, formerlymaturation-promot<strong>in</strong>g factor) <strong>in</strong> the recipient (Obackand Wells 2002). Interphase zygotes conta<strong>in</strong> lowM-Cdk-activity, thus an <strong>in</strong>troduced donor nucleus ofany cell cycle stage no longer undergoes efficientchromat<strong>in</strong> remodell<strong>in</strong>g, completely abolish<strong>in</strong>g developmentpast the eight-cell stage (Tani et al. 2001). In orderto circumvent this problem for SCNT, zygotes have tobe at a stage when they conta<strong>in</strong> enough M-Cdk-activityto disassemble the donor nuclear envelope and allowearly embryo development to proceed. This wasachieved by us<strong>in</strong>g zygotes which are either just exit<strong>in</strong>g(i.e. early telophase) or enter<strong>in</strong>g mitosis (i.e. metaphase),and thus conta<strong>in</strong> enough residual or newly synthesizedM-Cdk-activity, respectively. With early bov<strong>in</strong>e zygotes,enucleated just 4 h after <strong>in</strong>sem<strong>in</strong>ation, post-implantationdevelopment of SCNT embryos was improvedcompared with us<strong>in</strong>g chemically activated MII cytoplasts(Schurmann et al. 2006). Pregnancy establishmentwas significantly higher and this <strong>in</strong>itial difference translated<strong>in</strong>to about twofold higher development <strong>in</strong>to calvesat wean<strong>in</strong>g (Schurmann et al. 2006). There are severalreasons why sperm-activated oocytes might have beenbeneficial. Firstly, sperm is the natural agent of activation,<strong>in</strong>duc<strong>in</strong>g long-last<strong>in</strong>g Ca 2+ -oscillations that moreclosely resemble natural fertilization than the s<strong>in</strong>glelarge Ca 2+ -rise triggered by artificial activation (Fissoreet al. 1992). Secondly, sperm delivers factors to the eggthat may improve early embryonic development (Krawetz2005). These <strong>in</strong>clude: (i) the centrosome, which isusually removed together with its associated componentsdur<strong>in</strong>g enucleation; (ii) some 3000 differentmRNAs and (iii) micro-RNAs, which do not code forprote<strong>in</strong>s but play a role <strong>in</strong> controll<strong>in</strong>g gene activity.Thirdly, there may be early sperm-derived transcriptsaris<strong>in</strong>g <strong>in</strong> the time <strong>in</strong>terval between sperm–egg fusionand enucleation. Although there is no direct evidence forsuch early paternal transcription, it has been shown thatsome embryonic genome activation occurs shortly afterfertilization <strong>in</strong> cattle (Memili et al. 1998). The nature ofthese early transcripts and whether they come from thematernal and ⁄ or paternal genome is not known. Allthese molecules may participate <strong>in</strong> chromat<strong>in</strong> remodell<strong>in</strong>g,pronuclear formation, establishment of impr<strong>in</strong>ts orembryonic genome activation (Krawetz 2005). The<strong>in</strong>creased reprogramm<strong>in</strong>g ability of early zygotes clearlywarrants a more detailed analysis <strong>in</strong>to their RNA andprote<strong>in</strong> composition.Soon after the cattle study, the birth of cloned miceafter NT <strong>in</strong>to mitotically arrested late zygotes confirmedtheir suitability as NT recipients (Egli et al. 2007). Us<strong>in</strong>gmetaphase-arrested zygotes before their first cleavage,cloned offspr<strong>in</strong>g were obta<strong>in</strong>ed after NT with mitoticallyarrested zygotes, 2-, 8- and ES-cells, but not somaticdonors. Because a direct comparison between metaphase-arrestedoocyte and zygote recipients, us<strong>in</strong>g donorcells of the same type and cell cycle stage, has not yetÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Steps to Improve Farm Animal Clon<strong>in</strong>g Efficiency 411been reported <strong>in</strong> mouse, it is currently unknown ifzygotes can also improve clon<strong>in</strong>g efficiency <strong>in</strong> thisspecies. Taken together, these studies unequivocallydemonstrated that zygotes reta<strong>in</strong> the factors necessaryto completely reprogram embryonic and somatic genomes.These are likely to reside <strong>in</strong> the pronuclei dur<strong>in</strong>g<strong>in</strong>terphase, be<strong>in</strong>g redistributed throughout the cytoplasmdur<strong>in</strong>g mitosis.Improv<strong>in</strong>g Reprogramm<strong>in</strong>g In vitroA number of <strong>in</strong> vitro approaches have been devised to<strong>in</strong>crease somatic clon<strong>in</strong>g success. These <strong>in</strong>clude treat<strong>in</strong>gdonor cells with pharmacological agents to alter theirepigenetic marks (Enright et al. 2003; Shi et al. 2003),fus<strong>in</strong>g transiently permeabilized cells conta<strong>in</strong><strong>in</strong>g artificiallycondensed chromat<strong>in</strong> (Sullivan et al. 2004) orus<strong>in</strong>g serial NT (Ono et al. 2001). One of the mostpromis<strong>in</strong>g methods has been embryo aggregation. Inmouse, aggregation of two or three NT embryos led tonormalized gene expression and higher clon<strong>in</strong>g efficiency<strong>in</strong> some studies (Boiani et al. 2003) but not <strong>in</strong> others(Yabuuchi et al. 2002). In bov<strong>in</strong>e, we aggregated threeone-cell NT embryos dur<strong>in</strong>g <strong>in</strong> vitro culture (Obacket al. 2003; Misica-Turner et al. 2007). Aggregationaffected development of embryonic and somatic clonedembryos differently. In aggregates of embryonic clones,<strong>in</strong> vitro development was impaired, but the fewdevelop<strong>in</strong>g blastocysts appeared molecularly normal.Theoretically, one would expect an n-fold <strong>in</strong>crease <strong>in</strong>post-blastocyst survival us<strong>in</strong>g n numbers of aggregationpartners, provided there were no positive or negative<strong>in</strong>teractions with<strong>in</strong> the composite blastocyst. Embryoclones were <strong>in</strong> agreement with this prediction, show<strong>in</strong>gon average 2.5-fold higher clon<strong>in</strong>g efficiency. In otherwords, there was no evidence for complementary <strong>in</strong>teractionsresult<strong>in</strong>g <strong>in</strong> <strong>in</strong>creased survival beyond whatwould be expected as a direct numerical consequence ofaggregation. A similar <strong>in</strong>crease <strong>in</strong> term survival ofaggregated embryonic clones has been described before(Peura et al. 1998). In contrast, SCNT aggregatesdeveloped normally <strong>in</strong> vitro, but the result<strong>in</strong>g blastocystsshowed reduced POU5F1 expression and no effecton vivo survival, <strong>in</strong>dicat<strong>in</strong>g that they were <strong>in</strong> factcompromised compared with embryo clone aggregates.These differences <strong>in</strong> tim<strong>in</strong>g of developmental failurereveal strik<strong>in</strong>g biological differences between embryonicand somatic clones <strong>in</strong> response to aggregation.Measur<strong>in</strong>g Reprogramm<strong>in</strong>g Before EmbryoTransferCloned embryos before transfer display a number ofepigenetic abnormalities such as aberrant DNA-methylation(Kang et al. 2003) and post-translational histonemodifications (Santos et al. 2003; Wang et al. 2007;Yang et al. 2007a,b). It is likely that abnormal epigenotypewill result <strong>in</strong> aberrant gene expression. At leastfor one gene, a mechanistic l<strong>in</strong>k between DNA-methylationand transcriptional errors has <strong>in</strong>deed been established.This gene encodes the transcription factorPOU5F1, which is essential for regulat<strong>in</strong>g embryoniccell pluripotency and has often been used to monitorreprogramm<strong>in</strong>g success after NT (Boiani et al. 2002;Munsie et al. 2002; Bortv<strong>in</strong> et al. 2003; Misica-Turneret al. 2007; Wuensch et al. 2007). In <strong>in</strong>terspecies NTexperiments us<strong>in</strong>g Xenopus recipient oocytes and mousedonor thymocytes, selective POU5F1 promoter DNAdemethylationwas causally l<strong>in</strong>ked to <strong>in</strong>correctlyre-activat<strong>in</strong>g POU5F1 transcription (Simonsson andGurdon 2004). Because discrepancies between RNAand prote<strong>in</strong> levels exist (Tian et al. 2004), it rema<strong>in</strong>s tobe formally demonstrated that POU5F1 prote<strong>in</strong> expressionwas also aberrant and that this subsequentlycontributed to some of the aberrant organismal phenotypesobserved <strong>in</strong> clones. Prote<strong>in</strong>s ultimately carry outcellular functions, thus their expression profil<strong>in</strong>g representscellular phenotype better than RNA profil<strong>in</strong>g.However, the proteome with its many low-abundanceprote<strong>in</strong>s, often carry<strong>in</strong>g post-translational modifications,is more difficult to analyze and has consequentlynot been comprehensively profiled <strong>in</strong> clones (Fig. 3).RNA levels, on the contrary, can be amplified, mak<strong>in</strong>gthem easier and cheaper to quantify globally. NumerousNT ES cell-like cellsFig. 3. Genome-wide profil<strong>in</strong>g ofclones at different levels. Shown isnuclear transfer (NT) embryodevelopment from one-cell to blastocyst,which can either be used togenerate ES cell-like cells or clonedoffspr<strong>in</strong>g. Bars <strong>in</strong>dicate differentgenome-wide profil<strong>in</strong>g approachesto exam<strong>in</strong>e different levels ofcellular <strong>in</strong>formation (DNA, RNA,prote<strong>in</strong>, etc.) at different developmentaltime w<strong>in</strong>dows. Shad<strong>in</strong>gwith<strong>in</strong> bars <strong>in</strong>dicates to what degreeeach approach has been used for aparticular developmental <strong>in</strong>terval,rang<strong>in</strong>g from little use (white) toheavy use (dark shad<strong>in</strong>g). Arrowsbetween bars <strong>in</strong>dicate the flow of<strong>in</strong>formation from genotype tophenotype+NTrecipient donor NT 1-cell NT blastocystGenomeEpigenomeTranscriptomeProteomePhenomeNT animalÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


412 B Obackstudies have shown that the spatiotemporal regulationof gene expression is abnormal <strong>in</strong> cloned pre-implantationembryos. This applies to both term<strong>in</strong>ation of thesomatic and <strong>in</strong>itiation of the embryonic gene expressionprogramme. Analysis of transcriptional reprogramm<strong>in</strong>ghas focused on two developmental stages: early-cleavagestages at the time of embryonic genome activation andblastocysts.Repress<strong>in</strong>g donor genesTransient and reversible changes <strong>in</strong> donor cell-specificgene expression can occur simply as an artefact oftryps<strong>in</strong>ization (Green et al. 2007), emphasiz<strong>in</strong>g theimportance of analyz<strong>in</strong>g cells <strong>in</strong> the state they are <strong>in</strong>when used for NT. However, even if the <strong>in</strong>itial silenc<strong>in</strong>gof some genes reflects their manipulation for NT, manydonor cell-specific mRNAs rema<strong>in</strong> repressed until theblastocyst stage. Microarray data have shown that therecipient ooplasm, which is transcriptionally <strong>in</strong>active atthe time of NT (Latham et al. 1992), establishes atranscriptionally repressive state with<strong>in</strong> the donor cellgenome of the early NT reconstruct (Vassena et al.2007). As a result, the transcriptome of cloned mur<strong>in</strong>eone-cell embryos was very similar to that of controls.Only 259 (=1.6%) of total mRNAs were differentiallyexpressed between SCNT and fertilized embryos at theone-cell stage, with 53 (=20%) of those be<strong>in</strong>g newtranscripts and 206 (=80%) be<strong>in</strong>g maternally suppliedmRNAs, which were either stabilized or precociouslydegraded <strong>in</strong> clones. The few newly transcribed genesthat were overexpressed <strong>in</strong> SCNT embryos, comparedwith both fertilized and parthenogenetic controls,encoded mostly transcription factors that were alsohighly expressed <strong>in</strong> the donor cells.Reprogramm<strong>in</strong>g is a process that occurs over aprotracted period of time, and the next developmentalstage that was analyzed <strong>in</strong> great detail is the blastocyst.Expression of several donor-specific candidate genesfrom different l<strong>in</strong>eages, e.g. cumulus (Bortv<strong>in</strong> et al.2003; Inoue et al. 2006) and haematopoietic <strong>in</strong> mouse(Inoue et al. 2006), skeletal muscle <strong>in</strong> bov<strong>in</strong>e (Greenet al. 2007) and antler vs adipocyte <strong>in</strong> cerv<strong>in</strong>e NT (Berget al. 2007) was ext<strong>in</strong>guished by that stage. Likewise,global gene expression comparisons demonstrated thatthe majority of genes was differentially expressedbetween donors and their NT blastocyst derivatives,<strong>in</strong>dicat<strong>in</strong>g extensive donor transcriptome reprogramm<strong>in</strong>g(Smith et al. 2005). Even two female adult ear sk<strong>in</strong>fibroblast l<strong>in</strong>es that differed <strong>in</strong> the expression of over3000 transcripts prior to NT resulted <strong>in</strong> <strong>in</strong>dist<strong>in</strong>guishablegene expression profiles at the blastocyst stage(Smith et al. 2005). Despite their similar degree of geneexpression reprogramm<strong>in</strong>g, these two l<strong>in</strong>es still showedsignificantly different clon<strong>in</strong>g efficiency, suggest<strong>in</strong>g thatglobal transcriptional profil<strong>in</strong>g may be of limited valuewhen screen<strong>in</strong>g for ‘good’ vs ‘bad’ donor l<strong>in</strong>es. However,not every gene was properly repressed, leav<strong>in</strong>g asmall set of differentially expressed genes that ma<strong>in</strong>ta<strong>in</strong>edtheir expression levels <strong>in</strong> both donors and theircorrespond<strong>in</strong>g blastocysts. These fibroblast genes were<strong>in</strong>volved <strong>in</strong> various biological functions and it would be<strong>in</strong>formative to determ<strong>in</strong>e if the same genes, or theirassociated chromat<strong>in</strong> modifications, are generally resistantto silenc<strong>in</strong>g, <strong>in</strong>dependent of cell type. Other genesthat ma<strong>in</strong>ta<strong>in</strong> epigenetic donor cell memory have beendescribed <strong>in</strong> endoderm and neuroectoderm donors <strong>in</strong>frog (Ng and Gurdon 2005) and myogenic donors aftermouse (Gao et al. 2003) and frog NT (Ng and Gurdon2008). Apart from altered metabolic demands (Gaoet al. 2003), functional consequences of cont<strong>in</strong>ued donorcell gene expression <strong>in</strong> cloned embryos are not known.In summary, the donor cell genome was largelyrepressed by the late one-cell stage, but still detectablewith a small array of transcripts at both early and latepreimplantation stages. This raises the question if<strong>in</strong>duced donor genome silenc<strong>in</strong>g would improve development.Activat<strong>in</strong>g embryo genesFollow<strong>in</strong>g fertilization, the embryo undergoes drasticmorphological changes that culm<strong>in</strong>ate <strong>in</strong> the differentiationof two separate l<strong>in</strong>eages, trophectoderm and<strong>in</strong>ner cell mass. Dur<strong>in</strong>g this process, failure to correctlyactivate embryonic genes at the right time, place andabundance will have detrimental effects on development,as illustrated by a large number of s<strong>in</strong>gle genemutations that <strong>in</strong>terfere with normal embryogenesis <strong>in</strong>the mouse. Cloned embryos are clearly defective <strong>in</strong>recapitulat<strong>in</strong>g this correct stage-specific gene expression.In mouse, already the first transiently <strong>in</strong>ducedgenes (TIG) transcribed from the embryonic genomewere absent or greatly reduced <strong>in</strong> cloned two-cellembryos, especially if they were also absent from thedonor cell (Vassena et al. 2007, 2008). Even expressionof those few TIG transcripts already present <strong>in</strong> thedonor cell was reduced <strong>in</strong> clones, probably becausethey became silenced after NT and only partially reactivatedat the two-cell stage. Other studies, us<strong>in</strong>gdifferent sets of early markers, also observed suppressionof embryonically activated mRNAs <strong>in</strong> clonedtwo-cells; however, the donor cell expression was notanalyzed (Suzuki et al. 2006; Inoue et al. 2007). Microarrayanalysis confirmed substantial differences betweenthe transcriptomes of NT vs fertilized or artificiallyactivated embryos at the one-cell and, at an order ofmagnitude greater, the two-cell stages (Vassena et al.2007). At the one-cell stage, 53 (=0.3% of total) newtranscripts were at least twofold differentially expressed(8 down- and 45 upregulated), at the two-cell stage thatnumber <strong>in</strong>creased to 1539 (452 down- and 1087upregulated) of different transcripts relative to controlembryos. In addition, a large number of maternalmRNAs underwent either precocious or delayed degradation.The comb<strong>in</strong>ed effects of aberrant transcriptionand mRNA handl<strong>in</strong>g prom<strong>in</strong>ently affected genes<strong>in</strong>volved <strong>in</strong> transcriptional regulation. SCNT embryosmay be predisposed to aberrantly express genes encod<strong>in</strong>gprote<strong>in</strong>s that are responsible for establish<strong>in</strong>g andma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g a stable donor cell differentiation status.Their widespread dysregulation likely caused a ‘rippleeffect’ that alters the transcriptome of many otherfunctions <strong>in</strong>clud<strong>in</strong>g oxidative phosphorylation, membranetransport, and mRNA transport and process<strong>in</strong>g(Vassena et al. 2007).Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Steps to Improve Farm Animal Clon<strong>in</strong>g Efficiency 413In most farm animals, the transfer of early-cleavagestage embryos <strong>in</strong>to oviducts is technically difficult andrecipient animals are expensive. Because high embryowastage dur<strong>in</strong>g pre-implantation development wouldfurther <strong>in</strong>crease recipient cost, cloned embryos areusually transferred at the blastocyst stage. Be<strong>in</strong>g ableto select blastocysts with a high chance of develop<strong>in</strong>g<strong>in</strong>to viable offspr<strong>in</strong>g is thus a very important goal <strong>in</strong>clon<strong>in</strong>g research. This has been addressed by a numberof microarray studies compar<strong>in</strong>g bov<strong>in</strong>e NT and IVFblastocysts (Pfister-Genskow et al. 2005; Smith et al.2005; Somers et al. 2006; Beyhan et al. 2007; Long et al.2007; Zhou et al. 2007). Compar<strong>in</strong>g these studies, therewere three strik<strong>in</strong>g observations. Firstly, transcriptabundance for over 95% of genes analyzed was similarbetween NT and IVF embryos, suggest<strong>in</strong>g that mostgenes were reprogrammed. Secondly, no dist<strong>in</strong>ct set ofconsistently misexpressed genes was found <strong>in</strong> clonedembryos. Altogether, 281 genes were classified as differentiallyexpressed (169 down- and 112 upregulated).Only four of these genes, mostly housekeep<strong>in</strong>g ribosomaltranscripts, were found <strong>in</strong> two <strong>in</strong>dependentstudies and none of them were consistently up- ordownregulated. Thirdly, average changes <strong>in</strong> transcriptabundance were only twofold, with less than 3%show<strong>in</strong>g more than fourfold changes. The lack of acommon transcriptional signature can have either technicalor biological reasons. Technically, the studiesdiffered <strong>in</strong> microarrays and analytical algorithms used,add<strong>in</strong>g cross-platform to the <strong>in</strong>evitable cross-laboratoryvariation (Bammler et al. 2005). Moreover, transcriptomeprofil<strong>in</strong>g can only be as powerful as the biologicaldef<strong>in</strong>ition of the start<strong>in</strong>g cell material. In that respect,comparative analysis of the data sets was clearlyconfounded by multiple parameter changes, e.g. <strong>in</strong>genotype, donor and recipient cell source, activationmethod, culture protocol, morphological grade, developmentalstage and state of blastocysts (fresh vscryopreserved), to name just a few. Despite thesedifferences <strong>in</strong> experimental nuances, analysis of a totalof 130 NT and 121 IVF embryos across the six studies,either <strong>in</strong>dividually or <strong>in</strong> pools, should have revealedrobust ‘hotspots’ of preferentially affected transcripts ifthey <strong>in</strong>deed existed. Why have they not been found? Amajor problem is that the <strong>in</strong>tracellular range of transcriptabundance <strong>in</strong> blastocyst cell types is largelyunknown. In yeast, transcript abundance varies oversix orders of magnitude, rang<strong>in</strong>g from a few hundredcopies per cell for glycolytic mRNAs to one-thousandthper cell for transcripts encod<strong>in</strong>g transcription factorswith critical regulatory roles (Holland 2002). It isentirely plausible that ‘hotspot’ transcripts are expressedat low levels or <strong>in</strong> a small fraction of the cells studiedand microarrays are simply not sensitive enough todetect them (Evans et al. 2002). Alternatively, thesenegative results may argue for a model of randomizedreprogramm<strong>in</strong>g. At the s<strong>in</strong>gle-cell level, gene expressionlevels already fluctuate randomly under normal conditions(Kaern et al. 2005; Raser and O’Shea 2005).Nuclear reprogramm<strong>in</strong>g will add to this <strong>in</strong>tr<strong>in</strong>sicstochastic variation, <strong>in</strong>creas<strong>in</strong>g the noise and mask<strong>in</strong>gexpression differences at <strong>in</strong>dividual gene level. A stochasticmodel is supported by the f<strong>in</strong>d<strong>in</strong>g that transcriptlevels of small sets of candidate genes, represent<strong>in</strong>g 0.1%of the about 10 000 genes expressed <strong>in</strong> blastocysts (Zenget al. 2004), vary greatly between <strong>in</strong>dividual NTembryos (Bortv<strong>in</strong> et al. 2003) and cannot dist<strong>in</strong>guishbetween NT and IVF embryos (deCamargo et al. 2005;Smith et al. 2007). Carefully quantify<strong>in</strong>g abundance ofseveral functionally unrelated transcripts <strong>in</strong> s<strong>in</strong>gleembryos revealed that they are <strong>in</strong>dependently reprogrammed:one gene can be close to ‘normal’, i.e. close tothe average expression level for this gene across manyembryos, whereas another gene <strong>in</strong> the same embryowould be aberrantly expressed. These patterns of normalvs aberrantly expressed genes would be different fordifferent embryos, argu<strong>in</strong>g aga<strong>in</strong>st a determ<strong>in</strong>istic modelof reprogramm<strong>in</strong>g (C. Smith, personal communication).Further evidence comes from <strong>in</strong>duced reprogramm<strong>in</strong>g <strong>in</strong>iPS cells. Ectopic expression of key transcription factors<strong>in</strong>itiates a slow reprogramm<strong>in</strong>g process that depends onstochastic epigenetic events, which sequentially activateES-cell markers, eventually result<strong>in</strong>g <strong>in</strong> stable pluripotency(Meissner et al. 2007). However, if reprogramm<strong>in</strong>gwas completely random, it should also switch on(or off) genes that were silent (or active) <strong>in</strong> both thedonor cell and dur<strong>in</strong>g normal embryogenesis. Genesfall<strong>in</strong>g <strong>in</strong> these two categories have not yet beenanalyzed comprehensively.ConclusionsDespite more than a decade of research efforts, rout<strong>in</strong>efarm animal clon<strong>in</strong>g efficiency is still frustrat<strong>in</strong>gly low.The molecular cause for this low efficiency has not beenelucidated. There is a wealth of <strong>in</strong>formation describ<strong>in</strong>gwidespread genetic, epigenetic and transcriptional aberrations<strong>in</strong> cloned embryos; however, these genome-wideanalyses at different levels are currently not well<strong>in</strong>tegrated. Importantly, these observations have yet tobe causally connected with the observed phenotypes <strong>in</strong>cloned foetuses and offspr<strong>in</strong>g. Because some clonedphenotypes are highly reproducible <strong>in</strong> farm animals, e.g.hydroallantois <strong>in</strong> cattle (Heyman et al. 2002a), it shouldbe possible to establish their underly<strong>in</strong>g molecularcauses and design specific treatments to overcome them.Current methods to improve clon<strong>in</strong>g efficiency are stillfairly unspecific and have at best led to a slight <strong>in</strong>crease<strong>in</strong> success rates. Choos<strong>in</strong>g undifferentiated somatic stemcells as donors has not improved clon<strong>in</strong>g efficiency <strong>in</strong>five different l<strong>in</strong>eages, <strong>in</strong>dicat<strong>in</strong>g that donor cell typemay not be critical for clon<strong>in</strong>g success. It rema<strong>in</strong>s to beseen if the limited number of NT experiments reportedwas simply not sufficient to detect a hierarchical relationbetween cell differentiation and clon<strong>in</strong>g efficiency or ifsuch a relation is not universally true. In any case,vary<strong>in</strong>g just one parameter alone, e.g. somatic donor celldifferentiation status, is unlikely to lift clon<strong>in</strong>g success tothe efficiency level of other assisted reproductive technologies,such as IVF or artificial <strong>in</strong>sem<strong>in</strong>ation. So far,improvements resulted from better coord<strong>in</strong>ation betweendonor cell type and cell cycle stage, the use ofmitotic zygotes as NT recipients and, at least forembryonic clones, the aggregation of cloned embryos(Fig. 4). Each of these improvements optimized one stepof the clon<strong>in</strong>g procedure at a time. In the future, it willÓ 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


414 B ObackFig. 4. Climb<strong>in</strong>g mount efficiency: the path to clon<strong>in</strong>g success. Shownare key steps that improve clon<strong>in</strong>g efficiency, result<strong>in</strong>g <strong>in</strong> healthyclones. 1. Coord<strong>in</strong>at<strong>in</strong>g donor cell type and cell cycle stage. 2. Us<strong>in</strong>gzygotes rather than unfertilized oocytes as reprogramm<strong>in</strong>g reactors.3. Aggregat<strong>in</strong>g cloned embryos dur<strong>in</strong>g <strong>in</strong> vitro culturebe important to demonstrate if the small sequential<strong>in</strong>creases at every step are cumulative, add<strong>in</strong>g up to one<strong>in</strong>tegrated clon<strong>in</strong>g protocol with greatly improved efficiency.AcknowledgementsI would like to thank Dr. David Wells for critical read<strong>in</strong>g of themanuscript and all members of our clon<strong>in</strong>g team for excellent technicalsupport <strong>in</strong> the field and laboratory. This work was supported by theNew Zealand Foundation for Research, Science and Technology andAgResearch.ReferencesBaguisi A, Behboodi E, Melican DT, Pollock JS, DestrempesMM, Cammuso C, Williams JL, Nims SD, Porter CA,Midura P, Palacios MJ, Ayres SL, Denniston RS, HayesML, Ziomek CA, Meade HM, Godke RA, Gav<strong>in</strong> WG,Overstrom EW, Echelard Y, 1999: Production of goats bysomatic cell nuclear transfer. Nat Biotechnol 17, 456–461.Bammler T, Beyer RP, Bhattacharya S, Boorman GA, BoylesA, Bradford BU, Bumgarner RE, Bushel PR, Chaturvedi K,Choi D, Cunn<strong>in</strong>gham ML, Deng S, Dressman HK, Fann<strong>in</strong>RD, Far<strong>in</strong> FM, Freedman JH, Fry RC, Harper A, HumbleMC, Hurban P, Kavanagh TJ, Kaufmann WK, Kerr KF,J<strong>in</strong>g L, Lapidus JA, Lasarev MR, Li J, Li YJ, LobenhoferEK, Lu X, Malek RL, Milton S, Nagalla SR, O’Malley JP,Palmer VS, Pattee P, Paules RS, Perou CM, Phillips K, Q<strong>in</strong>LX, Qiu Y, Quigley SD, Rodland M, Rusyn I, Samson LD,Schwartz DA, Shi Y, Sh<strong>in</strong> JL, Sieber SO, Slifer S, SpeerMC, Spencer PS, Sproles DI, Swenberg JA, Suk WA,Sullivan RC, Tian R, Tennant RW, Todd SA, Tucker CJ,Van Houten B, Weis BK, Xuan S, Zarbl H, 2005:Standardiz<strong>in</strong>g global gene expression analysis betweenlaboratories and across platforms. Nat Methods 2, 351–356.Berg DK, Li C, Asher G, Wells DN, Oback B, 2007: Red deercloned from antler stem cells and their differentiatedprogeny. Biol Reprod 77, 384–394.Beyhan Z, Ross PJ, Iager AE, Kocabas AM, Cunniff K, RosaGJ, Cibelli JB, 2007: Transcriptional reprogramm<strong>in</strong>g ofsomatic cell nuclei dur<strong>in</strong>g preimplantation development ofcloned bov<strong>in</strong>e embryos. Dev Biol 305, 637–649.Blau HM, Chiu CP, Webster C, 1983: Cytoplasmic activationof human nuclear genes <strong>in</strong> stable heterocaryons. Cell 32,1171–1180.Blelloch R, Wang Z, Meissner A, Pollard S, Smith A, JaenischR, 2006: Reprogramm<strong>in</strong>g efficiency follow<strong>in</strong>g somatic cellnuclear transfer is <strong>in</strong>fluenced by the differentiation andmethylation state of the donor nucleus. Stem Cells 24, 2007–2013.Boiani M, Eckardt S, Leu NA, Scholer HR, McLaughl<strong>in</strong> KJ,2003: Pluripotency deficit <strong>in</strong> clones overcome by clone-cloneaggregation: epigenetic complementation? EMBO J 22,5304–5312.Boiani M, Eckardt S, Scholer HR, McLaughl<strong>in</strong> KJ, 2002: Oct4distribution and level <strong>in</strong> mouse clones: consequences forpluripotency. Genes Dev 16, 1209–1219.Bordignon V, Keyston R, Lazaris A, Bilodeau AS, Pontes JH,Arnold D, Fecteau G, Keefer C, Smith LC, 2003: Transgeneexpression of green fluorescent prote<strong>in</strong> and germ l<strong>in</strong>etransmission <strong>in</strong> cloned calves derived from <strong>in</strong> vitrotransfectedsomatic cells. Biol Reprod 68, 2013–2023.Bordignon V, Smith LC, 1998: Telophase enucleation: animproved method to prepare recipient cytoplasts for use <strong>in</strong>bov<strong>in</strong>e nuclear transfer. Mol Reprod Dev 49, 29–36.Bortv<strong>in</strong> A, Eggan K, Skaletsky H, Akutsu H, Berry DL,Yanagimachi R, Page DC, Jaenisch R, 2003: Incompletereactivation of Oct4-related genes <strong>in</strong> mouse embryos clonedfrom somatic nuclei. Development 130, 1673–1680.Cibelli JB, Stice SL, Golueke PJ, Kane JJ, Jerry J, Blackwell C,Ponce de Leon FA, Robl JM, 1998: Cloned transgeniccalves produced from nonquiescent fetal fibroblasts. Science280, 1256–1258.deCamargo ALS, Powell AM, do Vale Filho VR, Wall RJ,2005: Comparison of gene expression <strong>in</strong> <strong>in</strong>dividual preimplantationbov<strong>in</strong>e embryos produced by <strong>in</strong> vitro fertilisationor somatic cell nuclear transfer. Reprod Fertil Dev 17,487–496.Egli D, Rosa<strong>in</strong>s J, Birkhoff G, Eggan K, 2007: Developmentalreprogramm<strong>in</strong>g after chromosome transfer <strong>in</strong>to mitoticmouse zygotes. Nature 447, 679–685.Enright BP, Kubota C, Yang X, Tian XC, 2003: Epigeneticcharacteristics and development of embryos cloned fromdonor cells treated by trichostat<strong>in</strong> A or 5-aza-2¢-deoxycytid<strong>in</strong>e.Biol Reprod 69, 896–901.Evans MJ, Kaufman MH, 1981: Establishment <strong>in</strong> culture ofpluripotential cells from mouse embryos. Nature 292, 154–156.Evans SJ, Datson NA, Kabbaj M, Thompson RC, VreugdenhilE, De Kloet ER, Watson SJ, Akil H, 2002: Evaluation ofAffymetrix Gene Chip sensitivity <strong>in</strong> rat hippocampal tissueus<strong>in</strong>g SAGE analysis. Serial analysis of gene expression. EurJ Neurosci 16, 409–413.Fissore RA, Dobr<strong>in</strong>sky JR, Balise JJ, Duby RT, Robl JM,1992: Patterns of <strong>in</strong>tracellular Ca2 + concentrations <strong>in</strong>fertilized bov<strong>in</strong>e eggs. Biol Reprod 47, 960–969.Gao S, Chung YG, Williams JW, Riley J, Moley K, LathamKE, 2003: Somatic cell-like features of cloned mouseembryos prepared with cultured myoblast nuclei. BiolReprod 69, 48–56.Gao S, Czirr E, Chung YG, Han Z, Latham KE, 2004: Geneticvariation <strong>in</strong> oocyte phenotype revealed through parthenogenesisand clon<strong>in</strong>g: correlation with differences <strong>in</strong> pronuclearepigenetic modification. Biol Reprod 70, 1162–1170.Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Steps to Improve Farm Animal Clon<strong>in</strong>g Efficiency 415Greda P, Karasiewicz J, Modl<strong>in</strong>ski JA, 2006: Mouse zygotesas recipients <strong>in</strong> embryo clon<strong>in</strong>g. <strong>Reproduction</strong> 132, 741–748.Green AL, Wells DN, Oback B, 2007: Cattle cloned from<strong>in</strong>creas<strong>in</strong>gly differentiated muscle cells. Biol Reprod 77,395–406.Heyman Y, Chavatte-Palmer P, LeBourhis D, Camous S,Vignon X, Renard JP, 2002: Frequency and occurrence oflate-gestation losses from cattle cloned embryos. BiolReprod 66, 6–13.Heyman Y, Zhou Q, Lebourhis D, Chavatte-Palmer P, RenardJP, Vignon X, 2002: Novel approaches and hurdles tosomatic clon<strong>in</strong>g <strong>in</strong> cattle. Clon<strong>in</strong>g Stem Cells 4, 47–55.Hiiragi T, Solter D, 2005: Reprogramm<strong>in</strong>g is essential <strong>in</strong>nuclear transfer. Mol Reprod Dev 70, 417–421.Hochedl<strong>in</strong>ger K, Jaenisch R, 2002: Monoclonal mice generatedby nuclear transfer from mature B and T donor cells.Nature 415, 1035–1038.Holland MJ, 2002: Transcript abundance <strong>in</strong> yeast varies oversix orders of magnitude. J Biol Chem 277, 14363–14366.Illmensee K, Hoppe PC, 1981: Nuclear transplantation <strong>in</strong> Musmusculus: developmental potential of nuclei from preimplantationembryos. Cell 23, 9–18.Inoue K, Noda S, Ogonuki N, Miki H, Inoue S, Katayama K,Mekada K, Miyoshi H, Ogura A, 2007: Differentialdevelopmental ability of embryos cloned from tissue-specificstem cells. Stem Cells 25, 1279–1285.Inoue K, Ogonuki N, Miki H, Hirose M, Noda S, Kim JM,Aoki F, Miyoshi H, Ogura A, 2006: Inefficient reprogramm<strong>in</strong>gof the hematopoietic stem cell genome follow<strong>in</strong>gnuclear transfer. J Cell Sci 119, 1985–1991.Inoue K, Wakao H, Ogonuki N, Miki H, Se<strong>in</strong>o K, Nambu-Wakao R, Noda S, Miyoshi H, Koseki H, Taniguchi M,Ogura A, 2005: Generation of cloned mice by directnuclear transfer from natural killer T cells. Curr Biol 15,1114–1118.Kaern M, Elston TC, Blake WJ, Coll<strong>in</strong>s JJ, 2005: Stochasticity<strong>in</strong> gene expression: from theories to phenotypes. Nat RevGenet 6, 451–464.Kanatsu-Sh<strong>in</strong>ohara M, Inoue K, Lee J, Yoshimoto M,Ogonuki N, Miki H, Baba S, Kato T, Kazuki Y, ToyokuniS, Toyoshima M, Niwa O, Oshimura M, Heike T, NakahataT, Ish<strong>in</strong>o F, Ogura A, Sh<strong>in</strong>ohara T, 2004: Generation ofpluripotent stem cells from neonatal mouse testis. Cell 119,1001–1012.Kang YK, Lee KK, Han YM, 2003: Reprogramm<strong>in</strong>g DNAmethylation <strong>in</strong> the preimplantation stage: peep<strong>in</strong>g withDolly’s eyes. Curr Op<strong>in</strong> Cell Biol 15, 290–295.Kas<strong>in</strong>athan P, Knott JG, Wang Z, Jerry DJ, Robl JM, 2001:Production of calves from G1 fibroblasts. Nat Biotechnol19, 1176–1178.Kato Y, Imabayashi H, Mori T, Tani T, Taniguchi M, HigashiM, Matsumoto M, Umezawa A, Tsunoda Y, 2004: Nucleartransfer of adult bone marrow mesenchymal stem cells:developmental totipotency of tissue-specific stem cells froman adult mammal. Biol Reprod 70, 415–418.Kato Y, Tani T, Sotomaru Y, Kurokawa K, Kato J, DoguchiH, Yasue H, Tsunoda Y, 1998: Eight calves cloned fromsomatic cells of a s<strong>in</strong>gle adult. Science 282, 2095–2098.Krawetz SA, 2005: Paternal contribution: new <strong>in</strong>sights andfuture challenges. Nat Rev Genet 6, 633–642.Kurosaka S, Nagao Y, M<strong>in</strong>ami N, Yamada M, Imai H, 2002:Dependence of DNA synthesis and <strong>in</strong> vitro development ofbov<strong>in</strong>e nuclear transfer embryos on the stage of the cell cycleof donor cells and recipient cytoplasts. Biol Reprod 67, 643–647.Latham KE, Solter D, Schultz RM, 1992: Acquisition of atranscriptionally permissive state dur<strong>in</strong>g the 1-cell stage ofmouse embryogenesis. Dev Biol 149, 457–462.Li C, Harris AJ, Suttie JM, 2001: Tissue <strong>in</strong>teractions andantlerogenesis: new f<strong>in</strong>d<strong>in</strong>gs revealed by a xenograftapproach. J Exp Zool 290, 18–30.Li J, Greco V, Guasch G, Fuchs E, Mombaerts P, 2007: Micecloned from sk<strong>in</strong> cells. Proc Natl Acad Sci U S A. 104, 2738–2743.Long JE, Cai X, He LQ, 2007: Gene profil<strong>in</strong>g of cattleblastocysts derived from nuclear transfer, <strong>in</strong> vitro fertilizationand <strong>in</strong> vivo development based on cDNA library. AnimReprod Sci 100, 243–256.Mart<strong>in</strong> GR, 1981: Isolation of a pluripotent cell l<strong>in</strong>e from earlymouse embryos cultured <strong>in</strong> medium conditioned by teratocarc<strong>in</strong>omastem cells. Proc Natl Acad Sci U S A 78, 7634–7638.McGrath J, Solter D, 1983: Nuclear transplantation <strong>in</strong> themouse embryo by microsurgery and cell fusion. Science 220,1300–1302.McGrath J, Solter D, 1984: Inability of mouse blastomerenuclei transferred to enucleated zygotes to support development<strong>in</strong> vitro. Science 226, 1317–1319.Meissner A, Wernig M, Jaenisch R, 2007: Direct reprogramm<strong>in</strong>gof genetically unmodified fibroblasts <strong>in</strong>to pluripotentstem cells. Nat Biotechnol 25, 1177–1181.Memili E, Dom<strong>in</strong>ko T, First NL, 1998: Onset of transcription<strong>in</strong> bov<strong>in</strong>e oocytes and preimplantation embryos. MolReprod Dev 51, 36–41.Misica-Turner PM, Oback FC, Eichenlaub M, Wells DN,Oback B, 2007: Aggregat<strong>in</strong>g embryonic but not somaticnuclear transfer embryos <strong>in</strong>creases clon<strong>in</strong>g efficiency <strong>in</strong>cattle. Biol Reprod 76, 268–278.Munsie M, O’Brien C, Mountford P, 2002: Transgenic strategyfor demonstrat<strong>in</strong>g nuclear reprogramm<strong>in</strong>g <strong>in</strong> the mouse.Clon<strong>in</strong>g Stem Cells 4, 121–130.Ng RK, Gurdon JB, 2005: Epigenetic memory of active genetranscription is <strong>in</strong>herited through somatic cell nucleartransfer. Proc Natl Acad Sci U S A 102, 1957–1962.Ng RK, Gurdon JB, 2008: Epigenetic memory of an active genestate depends on histone H3.3 <strong>in</strong>corporation <strong>in</strong>to chromat<strong>in</strong><strong>in</strong> the absence of transcription. Nat Cell Biol 10, 102–109.Oback B, Wells D, 2002: Donor cells for nuclear clon<strong>in</strong>g: manyare called, but few are chosen. Clon<strong>in</strong>g Stem Cells 4, 147–168.Oback B, Wells DN, 2007a: Clon<strong>in</strong>g cattle: the methods <strong>in</strong> themadness. Adv Exp Med Biol 591, 30–57.Oback B, Wells DN, 2007b: Donor cell differentiation,reprogramm<strong>in</strong>g, and clon<strong>in</strong>g efficiency: elusive or illusivecorrelation? Mol Reprod Dev 74, 646–654.Ono Y, Shimozawa N, Ito M, Kono T, 2001: Cloned micefrom fetal fibroblast cells arrested at metaphase by a serialnuclear transfer. Biol Reprod 64, 44–50.Pavlath GK, Blau HM, 1986: Expression of muscle genes <strong>in</strong>heterokaryons depends on gene dosage. J Cell Biol 102, 124–130.Peura TT, Lewis IM, Trounson AO, 1998: The effect ofrecipient oocyte volume on nuclear transfer <strong>in</strong> cattle. MolReprod Dev 50, 185–191.Pfister-Genskow M, Myers C, Childs LA, Lacson JC,Patterson T, Betthauser JM, Goueleke PJ, Koppang RW,Lange G, Fisher P, Watt SR, Forsberg EJ, Zheng Y, LenoGH, Schultz RM, Liu B, Chetia C, Yang X, Hoeschele I,Eilertsen KJ, 2005: Identification of differentially expressedgenes <strong>in</strong> <strong>in</strong>dividual bov<strong>in</strong>e preimplantation embryos producedby nuclear transfer: improper reprogramm<strong>in</strong>g ofgenes required for development. Biol Reprod 72, 546–555.Prather RS, First NL, 1990: Clon<strong>in</strong>g embryos by nucleartransfer. J Reprod Fertil Suppl 41, 125–134.Prather RS, Sims MM, First NL, 1989: Nuclear transplantation<strong>in</strong> early pig embryos. Biol Reprod 41, 414–418.Raser JM, O’Shea EK, 2005: Noise <strong>in</strong> gene expression: orig<strong>in</strong>s,consequences, and control. Science 309, 2010–2013.Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


416 B ObackRenard JP, Maruotti J, Jouneau A, Vignon X, 2007: Nuclearreprogramm<strong>in</strong>g and pluripotency of embryonic cells: Applicationto the isolation of embryonic stem cells <strong>in</strong> farmanimals. Theriogenology. 68, Suppl 1 S196–S205.Rideout WM, 3rd, Wakayama T, Wutz A, Eggan K, Jackson-Grusby L, Dausman J, Yanagimachi R, Jaenisch R, 2000:Generation of mice from wild-type and targeted ES cells bynuclear clon<strong>in</strong>g. Nat Genet 24, 109–110.Robl JM, Gilligan B, Critser ES, First NL, 1986: Nucleartransplantation <strong>in</strong> mouse embryos: assessment of recipientcell stage. Biol Reprod 34, 733–739.Saito S, Sawai K, Ugai H, Moriyasu S, M<strong>in</strong>amihashi A,Yamamoto Y, Hirayama H, Kageyama S, Pan J, Murata T,Kobayashi Y, Obata Y, Yokoyama KK, 2003: Generationof cloned calves and transgenic chimeric embryos frombov<strong>in</strong>e embryonic stem-like cells. Biochem Biophys ResCommun 309, 104–113.Santos F, Zakhartchenko V, Stojkovic M, Peters A, Jenuwe<strong>in</strong>T, Wolf E, Reik W, Dean W, 2003: Epigenetic mark<strong>in</strong>gcorrelates with developmental potential <strong>in</strong> cloned bov<strong>in</strong>epreimplantation embryos. Curr Biol 13, 1116–1121.Schurmann A, Wells DN, Oback B, 2006: Early zygotes aresuitable recipients for bov<strong>in</strong>e somatic nuclear transfer andresult <strong>in</strong> cloned offspr<strong>in</strong>g. <strong>Reproduction</strong> 132, 839–848.Shi W, Hoeflich A, Flasw<strong>in</strong>kel H, Stojkovic M, Wolf E,Zakhartchenko V, 2003: Induction of a senescent-likephenotype does not confer the ability of bov<strong>in</strong>e immortalcells to support the development of nuclear transferembryos. Biol Reprod 69, 301–309.Simonsson S, Gurdon J, 2004: DNA demethylation isnecessary for the epigenetic reprogramm<strong>in</strong>g of somatic cellnuclei. Nat Cell Biol 6, 984–990.Smith C, Berg D, Beaumont S, Standley NT, Wells DN,Pfeffer PL, 2007: Simultaneous gene quantitation of multiplegenes <strong>in</strong> <strong>in</strong>dividual bov<strong>in</strong>e nuclear transfer blastocysts.<strong>Reproduction</strong> 133, 231–242.Smith SL, Everts RE, Tian XC, Du F, Sung LY, Rodriguez-Zas SL, Jeong BS, Renard JP, Lew<strong>in</strong> HA, Yang X, 2005:Global gene expression profiles reveal significant nuclearreprogramm<strong>in</strong>g by the blastocyst stage after clon<strong>in</strong>g. ProcNatl Acad Sci U S A 102, 17582–17587.Somers J, Smith C, Donnison M, Wells DN, Henderson H,McLeay L, Pfeffer PL, 2006: Gene expression profil<strong>in</strong>g of<strong>in</strong>dividual bov<strong>in</strong>e nuclear transfer blastocysts. <strong>Reproduction</strong>131, 1073–1084.Sullivan EJ, Kas<strong>in</strong>athan S, Kas<strong>in</strong>athan P, Robl JM, Collas P,2004: Cloned calves from chromat<strong>in</strong> remodeled <strong>in</strong> vitro. BiolReprod 70, 146–153.Suzuki T, M<strong>in</strong>ami N, Kono T, Imai H, 2006: Zygoticallyactivated genes are suppressed <strong>in</strong> mouse nuclear transferredembryos. Clon<strong>in</strong>g Stem Cells 8, 295–304.Tada M, Morizane A, Kimura H, Kawasaki H, A<strong>in</strong>scough JF,Sasai Y, Nakatsuji N, Tada T, 2003: Pluripotency ofreprogrammed somatic genomes <strong>in</strong> embryonic stem hybridcells. Dev Dyn 227, 504–510.Takahashi K, Yamanaka S, 2006: Induction of pluripotentstem cells from mouse embryonic and adult fibroblastcultures by def<strong>in</strong>ed factors. Cell 126, 663–676.Tani T, Kato Y, Tsunoda Y, 2001: Direct exposure ofchromosomes to nonactivated ovum cytoplasm is effectivefor bov<strong>in</strong>e somatic cell nucleus reprogramm<strong>in</strong>g. Biol Reprod64, 324–330.Terranova R, Pereira CF, Du Roure C, Merkenschlager M,Fisher AG, 2006: Acquisition and ext<strong>in</strong>ction of geneexpression programs are separable events <strong>in</strong> heterokaryonreprogramm<strong>in</strong>g. J Cell Sci 119, 2065–2072.Tian Q, Stepaniants SB, Mao M, Weng L, Feetham MC, DoyleMJ, Yi EC, Dai H, Thorsson V, Eng J, Goodlett D, BergerJP, Gunter B, L<strong>in</strong>seley PS, Stoughton RB, Aebersold R,Coll<strong>in</strong>s SJ, Hanlon WA, Hood LE, 2004: Integrated genomicand proteomic analyses of gene expression <strong>in</strong> mammaliancells. Mol Cell Proteomics 3, 960–969.Vassena R, Han Z, Gao S, Baldw<strong>in</strong> DA, Schultz RM, LathamKE, 2007: Tough beg<strong>in</strong>n<strong>in</strong>gs: alterations <strong>in</strong> the transcriptomeof cloned embryos dur<strong>in</strong>g the first two cell cycles. DevBiol 304, 75–89.Vassena R, Han Z, Gao S, Latham KE, 2007: Deficiency <strong>in</strong>recapitulation of stage-specific embryonic gene transcription<strong>in</strong> two-cell stage cloned mouse embryos. Mol Reprod Dev74, 1548–1556.Vassena R, Han Z, Gao S, Latham KE, 2008: Erratum:Deficiency <strong>in</strong> recapitulation of stage-specific embryonic genetranscription <strong>in</strong> two-cell stage cloned mouse embryos. MolReprod Dev. 75, 217.Wakayama T, Tateno H, Mombaerts P, Yanagimachi R, 2000:Nuclear transfer <strong>in</strong>to mouse zygotes. Nat Genet 24, 108–109.Wang F, Kou Z, Zhang Y, Gao S, 2007: Dynamic reprogramm<strong>in</strong>gof histone acetylation and methylation <strong>in</strong> the first cellcycle of cloned mouse embryos. Biol Reprod 77, 1007–1016.Wells DN, Laible G, Tucker FC, Miller AL, Oliver JE, XiangT, Forsyth JT, Berg MC, Cockrem K, L’Huillier PJ, TervitHR, Oback B, 2003: Coord<strong>in</strong>ation between donor cell typeand cell cycle stage improves nuclear clon<strong>in</strong>g efficiency <strong>in</strong>cattle. Theriogenology 59, 45–59.Wells DN, Misica PM, Tervit HR, 1999: Production of clonedcalves follow<strong>in</strong>g nuclear transfer with cultured adult muralgranulosa cells. Biol Reprod 60, 996–1005.Wuensch A, Habermann FA, Kurosaka S, Klose R,Zakhartchenko V, Reichenbach HD, S<strong>in</strong>owatz F,McLaughl<strong>in</strong> KJ, Wolf E, 2007: Quantitative monitor<strong>in</strong>g ofpluripotency gene activation after somatic clon<strong>in</strong>g <strong>in</strong> cattle.Biol Reprod 76, 983–991.Yabuuchi A, Kato Y, Tsunoda Y, 2002: Effects of aggregationof nuclear-transferred mouse embryos developed fromenucleated eggs receiv<strong>in</strong>g ES cells on <strong>in</strong> vitro and <strong>in</strong> vivodevelopment. J Reprod Dev 48, 393–397.Yamazaki Y, Mak<strong>in</strong>o H, Hamaguchi-Hamada K, Hamada S,Sug<strong>in</strong>o H, Kawase E, Miyata T, Ogawa M, Yanagimachi R,Yagi T, 2001: Assessment of the developmental totipotencyof neural cells <strong>in</strong> the cerebral cortex of mouse embryo bynuclear transfer. Proc Natl Acad Sci U S A 98, 14022–14026.Yang F, Hao R, Kessler B, Brem G, Wolf E, ZakhartchenkoV, 2007: Rabbit somatic cell clon<strong>in</strong>g: effects of donor celltype, histone acetylation status and chimeric embryocomplementation. <strong>Reproduction</strong> 133, 219–230.Yang J, Yang S, Beaujean N, Niu Y, He X, Xie Y, Tang X,Wang L, Zhou Q, Ji W, 2007: Epigenetic marks <strong>in</strong> clonedrhesus monkey embryos: comparison with counterpartsproduced <strong>in</strong> vitro. Biol Reprod 76, 36–42.Zeng F, Baldw<strong>in</strong> DA, Schultz RM, 2004: Transcript profil<strong>in</strong>gdur<strong>in</strong>g preimplantation mouse development. Dev Biol 272,483–496.Zhang F, Pomerantz JH, Sen G, Palermo AT, Blau HM, 2007:Active tissue-specific DNA demethylation conferred bysomatic cell nuclei <strong>in</strong> stable heterokaryons. Proc Natl AcadSci U S A 104, 4395–4400.Zhou W, Xiang T, Walker S, Farrar V, Hwang E, F<strong>in</strong>deisen B,Sadeghieh S, Arenivas F, Abruzzese RV, Polejaeva I, 2007:Global gene expression analysis of bov<strong>in</strong>e blastocystsproduced by multiple methods. Mol Reprod Dev 75, 744–758.Author’s address (for correspondence): B Oback, AgResearch Ltd.,Ruakura Research Centre, East Street, Private Bag 3123, Hamilton,New Zealand. E-mail: bjorn.oback@agresearch.co.nzConflict of <strong>in</strong>terest: The author declares no conflict of <strong>in</strong>terest.Ó 2008 The Author. Journal compilation Ó 2008 Blackwell Verlag


Reprod Dom Anim 43 (Suppl. 2), 417–422 (2008); doi: 10.1111/j.1439-0531.2008.01193.xISSN 0936-6768Nuclear Transfer of Freeze-Dried Somatic Cells <strong>in</strong>to Enucleated Sheep OocytesP Loi 1 , K Matzukawa 2 , G Ptak 1 , Y Natan 3 , J Fulka Jr 4 and A Arav 31 Department of Comparative Biomedical Sciences, Teramo University, Teramo, Italy; 2 National Institute of Livestock and Grassland Science,Tsukuba, Japan; 3 Institute of Animal Science, Agricultural Research Organization, The Volcani Center, Bet Dagan, Israel; 4 Institute of AnimalProduction, Prague, Czech RepublicContentsLyophilization has been used s<strong>in</strong>ce long time to preserveyeast and bacteria stra<strong>in</strong>s. Subsequently, a great deal ofefforts has been dedicated to the preservation <strong>in</strong> a dry state ofred blood cells and platelets. However, despite more than30 years passed by, no significant progress has been achieved.Recently, it has been reported that freeze-dried mice spermatozoawere able to generate normal offspr<strong>in</strong>g follow<strong>in</strong>g<strong>in</strong>jection <strong>in</strong>to the mature mice oocytes. In this work, weprompted to apply the lyophilization protocol developed formice spermatozoa to sheep somatic cells (lymphocytes andgranulosa cells). More than 350 enucleated sheep oocyteswere <strong>in</strong>jected with granulosa cells, and freeze dried us<strong>in</strong>g theprotocol developed for mice sperm cells. Transplanted nucleiorganized large pronuclei with fragmented DNA, but none ofthem entered the first mitosis. In the second part of theexperiments, trehalose and EGTA were found to reducesignificantly the extent of nuclear damage (65% and 55%<strong>in</strong>tact nuclei <strong>in</strong> lymphocyte and granulosa cells, respectively)follow<strong>in</strong>g freeze dry<strong>in</strong>g. Granulosa cells lyophilized withEGTA ⁄ trehalose and stored at room temperature for 3 yearswere used for nuclear transfer, and the <strong>in</strong>jected oocytes werecultured <strong>in</strong> vitro for 7 days. Approximately 16% of the oocyte<strong>in</strong>jected with freeze-dried cells developed <strong>in</strong>to blastocysts. Toconclude, we demonstrated for the first time that nucleatedcells ma<strong>in</strong>ta<strong>in</strong> genomic <strong>in</strong>tegrity after prolonged storage <strong>in</strong> adry state, and we were able to achieve early embryonicdevelopment follow<strong>in</strong>g <strong>in</strong>jection of these cells <strong>in</strong>to enucleatedsheep oocytes.IntroductionLyophilization has been used for the preservation offowl spermatozoa already <strong>in</strong> the 1950s (Polge et al.1949). The orig<strong>in</strong>al protocol was applied later to otherspecies (Sherman 1954), but the results <strong>in</strong> terms ofoffspr<strong>in</strong>g production were contradictory (Saacke andMalquist 1961). The def<strong>in</strong>itive proof that dry spermatozoareta<strong>in</strong> genetic <strong>in</strong>tegrity was established only whenmicrosurgical procedures were developed for bypass<strong>in</strong>gthe lack of mobility of freeze-dried spermatozoa, andnormal mice were produced by <strong>in</strong>tracytoplasmic sperm<strong>in</strong>jection (ICSI) of freeze-dried sperm (Wakayama andYanagimachi 1998). A follow<strong>in</strong>g paper from the samegroup demonstrated the preservation of genomic <strong>in</strong>tegrity<strong>in</strong> freeze-dried spermatozoa (Kusakabe et al. 2001),and more recently these results have been demonstrated<strong>in</strong> other species (Kesk<strong>in</strong>tepe et al. 2002; Liu et al. 2004).The possibility to store male gametes <strong>in</strong> a dry staterepresents a major breakthrough for stor<strong>in</strong>g and shipp<strong>in</strong>gmale gametes from mutants, transgenic and otherstra<strong>in</strong>s of laboratory mouse. Lyophilization of spermcould also be an attractive way to store spermatozoa <strong>in</strong>farm animal species, and especially <strong>in</strong> human, whereICSI has become a rout<strong>in</strong>e procedure <strong>in</strong> assistedreproduction.The progressive reduction of large animals worldwide(Hilton-Taylor 2000; Margules and Pressey 2000) hassuggested to establish gene banks from species threatenedby ext<strong>in</strong>ction (Myers et al. 2000), with the aim touse the cells for somatic cell nuclear transfer (SCNT,Wilmut et al. 1997).Somatic cell nuclear transfer has <strong>in</strong>deed an obviouspotential for the multiplication of rare genotypes (Corley-Smithand Brandhorst 1999; Loi et al. 2001), but itswide application is prevented by the low efficiency <strong>in</strong>terms of offspr<strong>in</strong>g outcomes. Furthermore, the majorityof the endangered mammals are practically unknownfrom a reproductive po<strong>in</strong>t of view. Therefore, thestorage of somatic cells for future use, once theprocedure of somatic cell clon<strong>in</strong>g will be reliable, iscerta<strong>in</strong>ly a wise step to be undertaken. However, theestablishment of gene banks <strong>in</strong> the form of cell l<strong>in</strong>esencounters several problems, represented by the costs ofliquid nitrogen. Recently, our group demonstrated thatsomatic cells rendered unviable by heat treatmentreta<strong>in</strong>ed their potential to generate a normal lamb afternuclear transplantation (Loi et al. 2002). The ma<strong>in</strong>scope of this work was to demonstrate the feasibility ofus<strong>in</strong>g radical approaches for the nuclear reprogramm<strong>in</strong>gof somatic cells, but, as <strong>in</strong> case of freeze-dried sperm, wealso established that lack of cell viability, as <strong>in</strong>dicated bymassive membrane and cytoplasmic damage, is not anabsolute prerequisite for SCNT.In this work, we developed robust procedures for thepreservation of sheep somatic cells <strong>in</strong> a freeze-driedstate, and we tested their ability to direct early embryonicdevelopment of enucleated oocytes. The ensu<strong>in</strong>gresults demonstrate for the first time the production ofnormal embryos from nuclear transfer of somatic cellsstored freeze dried for more than 3 years at roomtemperature.Materials and MethodsCell collection and freeze dry<strong>in</strong>gIn the first experiment, granulosa cells dissociated from<strong>in</strong> vitro-matured Cumulus Oocyte Complexes (COCs)from Sarda breed ewes were dispersed <strong>in</strong> 1 ml of HepesbufferedDMEM medium supplemented with 10% FCS;then 100 ll aliquots of the cell suspension were loaded<strong>in</strong>to 5 ml ampoules and plunged directly <strong>in</strong>to liquidnitrogen. Frozen cells were then passed on pre-cooledstage of a freeze-dry<strong>in</strong>g apparatus (Freezone 4.5; LabconcoCorporation, Kansas City, MO, USA) andÓ 2008 Teramo University


418 P Loi, K Matzukawa, G Ptak, Y Natan, J Fulka Jr and A Aravlyophilized for 24 h under an <strong>in</strong>let pressure of 1 mtorr at)50°C, follow<strong>in</strong>g the protocol used for sperm celllyophilization (Wakayama and Yanagimachi 1998). Atthe end of the process, each ampoule was flame sealed,placed <strong>in</strong>to cardboard and stored at room temperature(18–23°C) until use.On the light of the massive nuclear damage <strong>in</strong> rehydratedcells lyophilized us<strong>in</strong>g the sperm cell protocol,different solutions were tested for lyophilization. Briefly,peripheral blood lymphocytes were isolated fromperipheral blood (from Assaf ewes) through a Ficoll–Paque density gradient; the purity of the cells wasassessed by an automatic cell counter (Pentra 60; ABX,Montpellier, France), more than 80% of the cells werelymphocytes.Granulosa cells were obta<strong>in</strong>ed by COCs collectedfrom the ovaries of slaughtered Assaf ewes. CumulusOocyte Complexes were matured for 24 h <strong>in</strong> the mediumTCM 199 plus 10% FCS, FSH, LH and oestradiol <strong>in</strong><strong>in</strong>cubator at 38.5°C with 5% CO 2 . At the end ofmaturation, expanded COCs were shortly <strong>in</strong>cubated<strong>in</strong> hyaluronidase (300 USP units ⁄ ml; Sigma-Aldrich,Milano, Italy) and dissociated <strong>in</strong>to s<strong>in</strong>gle-cell populationby repeated pipett<strong>in</strong>g.The freez<strong>in</strong>g solution (2000 ll) was added to the cells(lymphocytes or granulosa) to be lyophilized. Thefreez<strong>in</strong>g solution was composed of 50% FCS and0.1 M trehalose <strong>in</strong> Hepes Talp buffer. Samples at avolume of 2 ml were frozen us<strong>in</strong>g the MTG freez<strong>in</strong>gapparatus (IMT, Nes Ziona, Israel) at a cool<strong>in</strong>g rate of5.1°C ⁄ m<strong>in</strong>. The concentration of the cells ranged from1–6 · 10 6 cells ⁄ ml. After freez<strong>in</strong>g, the samples werestored <strong>in</strong> liquid nitrogen until transfer <strong>in</strong>to the lyophilizer(Freezone Plus 6; Labconco). Samples werelyophilized for 72 h, after which each ampoule wasflame sealed, placed <strong>in</strong>to cardboard and stored at roomtemperature (18–23°C) until use. Aliquots of the sampleswere dispatched by air mail to Teramo, Italy, fornuclear transfer studies.RehydrationImmediately before use for nuclear transfer, the ampouleswere opened and 100 ll or 2000 ll of milliQ water(accord<strong>in</strong>g to the freeze-dry<strong>in</strong>g protocol) were added.Rehydrated cells were washed twice with medium 199plus Hepes, antibiotics and BSA before use for nucleartransfer. For each ampoule, viability was assessed onsmall aliquots cells by propidium iod<strong>in</strong>e sta<strong>in</strong><strong>in</strong>g (200cells counted). DNA fragmentation was assessed withcomet essay accord<strong>in</strong>g the manufacturer <strong>in</strong>struction(R&D Systems, Milano, Italy) <strong>in</strong> every replicate.DNA <strong>in</strong>tegrityDNA <strong>in</strong>tegrity was evaluated us<strong>in</strong>g the s<strong>in</strong>gle cell gelelectrophoresis assay (aka comet assay) (R&D Systems)as described by the manufacturer. Cells were diluted to aconcentration of 105 cells ⁄ ml <strong>in</strong> PBS. The cells werecomb<strong>in</strong>ed with molten LM agarose at a ratio of:1 : 10(v ⁄ v), then 75 ll were placed on comet slides.The slides were put <strong>in</strong> the dark at 4°C for 10 m<strong>in</strong>. Slideswere then immersed <strong>in</strong> a pre-cooled (4°C) lysis solutionfor 1 h at 4°C. Afterward, the slides were immersed <strong>in</strong> afreshly prepared alkali solution, which consisted of 0.6 gNaOH pellets, 250 ll of 250 mM EDTA, pH 10.0 and49.75 ml deionized water, for 60 m<strong>in</strong> <strong>in</strong> the dark atroom temperature. F<strong>in</strong>ally, the slides were washed <strong>in</strong> aTBE buffer (Tris base, Boric acid and EDTA) for 5 m<strong>in</strong>and then the slides were submerged <strong>in</strong> TBE buffer <strong>in</strong> ahorizontal electrophoresis apparatus; and 1 V ⁄ cm wasapplied for 10 m<strong>in</strong>. In addition, each experiment wasconducted with two controls; the first one was of cellsthat were previously treated with 100 lM of hydrogenperoxide for 10 m<strong>in</strong> at 2–8°C as described <strong>in</strong> themanufacturer kit (this served as a positive control forthe assay show<strong>in</strong>g damaged DNA) and the secondcontrol was of fresh cells, <strong>in</strong>dicat<strong>in</strong>g endogenous levelsof damage with<strong>in</strong> the cells. After the samples were dried,they were sta<strong>in</strong>ed with SYBR green. Scor<strong>in</strong>g was carriedout us<strong>in</strong>g a fluorescent microscope (Zeiss, Vertrieb,Germany) connect to a digital camera (Sony, Tokyo,Japan) and analysed us<strong>in</strong>g the Image J free software(NIH, Baltimore, MD, USA).SEMFor evaluation of the morphology of dry samples,Scann<strong>in</strong>g Electron Microscopy (Philips Inc., Milano,Italy) was used. The samples were gold plated beforebe<strong>in</strong>g placed <strong>in</strong> the SEM, and observed with a voltage ofthe electron scatter of 25 kV.Oocyte maturationMethods of <strong>in</strong> vitro embryo production were as previouslydescribed (Ptak et al. 1999a,b). Briefly, follow<strong>in</strong>gcollection of ovaries from adult sheep (Comisana andBergamasca breeds) at slaughter, oocytes were aspiratedand evaluated under a dissect<strong>in</strong>g microscope. Onlyoocytes surrounded by at least two layers of granulosacells and with evenly granulated cytoplasm were selectedfor <strong>in</strong> vitro maturation (IVM). Oocytes were matured<strong>in</strong> vitro <strong>in</strong> bicarbonate-buffered TCM-199 (Gibco) (275mOsm) conta<strong>in</strong><strong>in</strong>g 2 mM glutam<strong>in</strong>e, 100 lM cysteam<strong>in</strong>e,0.3 mM sodium pyruvate, 10% foetal bov<strong>in</strong>e serum(FBS) (Gibco Milan, Italy), 5 lg ⁄ ml FSH (OvagenAuckland, New Zealand), 5 lg ⁄ ml LH, 1 lg ⁄ ml oestradiol<strong>in</strong> a humidified atmosphere of 5% CO 2 <strong>in</strong> air at39°C for 24 h.Oocyte enucleation and nuclear transferMatured oocytes were enucleated generally after 22 h,after the start of the maturation. Cumulus OocyteComplexes were shortly <strong>in</strong>cubated with hyaluronidase,and the granulosa cells removed by repeated pipett<strong>in</strong>g.Oocytes were <strong>in</strong>cubated <strong>in</strong> Hepes-buffered 199 mediumconta<strong>in</strong><strong>in</strong>g 4 mg ⁄ ml of BSA and 7.5 lg ⁄ ml of cytochalas<strong>in</strong>B and Hoechst 33342, 10 mg ⁄ ml for 15 m<strong>in</strong> <strong>in</strong><strong>in</strong>cubator.Enucleation was carried out <strong>in</strong> Hepes-buffered 199medium plus BSA and cytochalas<strong>in</strong> B us<strong>in</strong>g the rout<strong>in</strong>emanipulation procedures.Enucleated oocytes were allowed to recover fromcytochalas<strong>in</strong> B treatment for 10 m<strong>in</strong> <strong>in</strong> the <strong>in</strong>cubator,Ó 2008 Teramo University


Dry Cells for Nuclear Transfer 419then directly <strong>in</strong>jected with freeze-dried cell. Reconstructedoocytes were activated <strong>in</strong> Hepes-bufferedmedium 199 conta<strong>in</strong><strong>in</strong>g 5 lg ⁄ ml Ionomyc<strong>in</strong> for 5 m<strong>in</strong>,then <strong>in</strong>cubated <strong>in</strong> SOF medium plus antibiotics andBSA conta<strong>in</strong><strong>in</strong>g 10 mM dimethylam<strong>in</strong>opur<strong>in</strong>e and7.5 lg ⁄ ml cytochalas<strong>in</strong> B for 3–5 h. Subsamples ofreconstructed embryos were processed for comet assayas described earlier.Embryo cultureReconstructed embryos were transferred <strong>in</strong>to 20-lldrops of SOF enriched with 1% (v : v) basal mediumEagle (BME)-essential am<strong>in</strong>o acids, 1% (v : v) m<strong>in</strong>imumessential medium (MEM)-non-essential am<strong>in</strong>oacids (Gibco), 1 mM glutam<strong>in</strong>e, and 8 mg ⁄ ml fattyacid-free BSA (SOFaaBSA). Zygote cultures werema<strong>in</strong>ta<strong>in</strong>ed <strong>in</strong> humidified atmosphere of 5% CO 2 ,7%O 2 , 88% N 2 at 39°C, and the medium renewed on day 3and day 5 of culture (day 0 = day of activation, 16).Embryonic development was monitored every 24 h, andarrested embryos were fixed <strong>in</strong> acetic acid–methanol3 : 1 for 24 h, then sta<strong>in</strong>ed with 2% aceto-orce<strong>in</strong> forcheck<strong>in</strong>g number and gross morphology of nuclei. Inthree replicates, aliquots of reconstructed embryos werefixed at late nuclear swell<strong>in</strong>g stage (13–16 h postactivation)and processed for Comet analysis to <strong>in</strong>vestigatethe presence and extent of DNA damage.DNA microsatellite analysisGenomic DNA from the pooled blastocysts derivedfrom batches of freeze-dried cells (one ampoule of cellwas used for each replicate), and from the re-hydratedcell used as nuclei donor was prepared follow<strong>in</strong>g thestandard procedures. Microsatellite analyses were performedwith multiplex polymerase cha<strong>in</strong> reaction us<strong>in</strong>gn<strong>in</strong>e microsatellite markers (CP49, FCB11, AE129,FCB304, INRA063, MAF214, PZ963, CSRD247 andHSC <strong>in</strong> table 1). Based on the degree of polymorphismof these microsatellites <strong>in</strong> the sheep population, theprobability that cell l<strong>in</strong>e and the cloned embryos wouldhave the same genotype was determ<strong>in</strong>ed to be smallerthan 2 · 10 )11 . Genotypes were determ<strong>in</strong>ed by polyacrylamidegel electrophoresis us<strong>in</strong>g an automatedDNA sequencer (Perk<strong>in</strong>-Elmer, Bucks, UK) and wereanalysed by Genscan and Genotyper software (GenscanDetection System, Woburn, MA, USA).Stastical analysisThe experimental observations, expressed as percentageof Comet assay positive cells and embryonic developmentto the blastocyst stage, were processed by a onewayanalysis of variance us<strong>in</strong>g the statistical packageSPSS (SPSS, Inc., Chicago, IL, USA) follow<strong>in</strong>g themodel:y ij ¼ l þ a i þ e ij ;where y ij is the experimental observations, l, the generalmean, a i , effect caused by the treatment (i = 1 forcontrol groups (cells and embryos), i = 2 for Cometpositive cells ⁄ freeze-dried cells nuclear transfer embryos,and ij = casual effect of the error (0, r).ResultsThe first embryo reconstructions were made us<strong>in</strong>ggranulosa cells freeze-dried accord<strong>in</strong>g the methods publishedby Wakayama and Yanagimachi 1998 (4). Over350 oocytes were <strong>in</strong>jected <strong>in</strong> these prelim<strong>in</strong>ary experiments.The majority of them (90%) were activated anddeveloped a pronuclear-like structure, but none of themcompleted the first mitosis. Nuclear morphology revealedhighly decondensed pronucleus-like structure,with massive DNA fragmentation (Fig. 2e). Lymphocytesand granulosa cells were unviable follow<strong>in</strong>g rehydration,for all of them sta<strong>in</strong>ed red with propidiumiod<strong>in</strong>e. However, the <strong>in</strong>clusion of trehalose <strong>in</strong> thefreez<strong>in</strong>g medium dramatically reduced DNA damageassessed by Comet, with 65% of lymphocytes and 55%of granulosa negative to the Comet assay (Fig. 2a).Despite the higher proportion of damaged DNA, granulosacells were used for nuclear transfer, for we have anestablished and robust clon<strong>in</strong>g protocol based on the useof granulosa cells as nuclear donor.The positive effect exerted by the <strong>in</strong>clusion of threalose<strong>in</strong> the freez<strong>in</strong>g media was also evident from the SEManalysis of cells (lymphocytes) freeze dried with orwithout threalose. In the presence of threalose, the driedmixture displayed a glassy, smooth appearance (Fig. 2b),on the contrary of the control samples, where the powderhas rather a spl<strong>in</strong>ter-like appearance (Fig. 2c).Embryo development follow<strong>in</strong>g nuclear transfer ofcells frozen with trehalose is <strong>in</strong>dicated <strong>in</strong> Table 2.Cleavage rate was similar to both the control and thefreeze-dried groups, although the latter reached theblastocyst stage (Fig. 2d) at lower extent (15.6% vs21%, p = 0.56). Microsatellite analysis performed withmultiplex polymerase cha<strong>in</strong> reaction us<strong>in</strong>g n<strong>in</strong>e microsatellitemarkers demonstrated early left any doubts thatthe blastocysts obta<strong>in</strong>ed were isogenic with the freezedrieddonor granulosa cells (data not shown).DiscussionWe have conv<strong>in</strong>c<strong>in</strong>gly shown that the procedure whichhas been developed by Wakayama and Yanagimachi1998 for mouse spermatozoa (4) was not suitable tofreeze-dried granulosa cells which needed to serve asTable 1. Microsatellite analysis of cell l<strong>in</strong>es and correspond<strong>in</strong>g clonedblastocysts (three replicates)LocusCell l<strong>in</strong>eClonedblast.Cell l<strong>in</strong>eClonedblast.Cell l<strong>in</strong>eClonedblast.PF PFC PE1 PE1C PE2 PE2COAR CP 49 90 98 90 98 90 98 90 98 90 98 90 98FCB 11 118 122 124 134 124 134 124 134 124 134 118 122OAR AE 129 145 149 145 149 145 149 145 149 145 149 145 149FCB 304 166 170 170 170 170 170 170 170 170 170 166 170INRA 063 175 179 175 179 175 179 175 179 175 179 175 179MAF 214 189 261 189 189 189 189 189 189 189 189 189 261CSRD247 223 227 213 227 213 227 213 227 213 227 223 227HSC 269 269 269 293 – – 269 293 269 293 269 269Ó 2008 Teramo University


420 P Loi, K Matzukawa, G Ptak, Y Natan, J Fulka Jr and A Arav% DNA Intact100806040200aFreshbFreeze-driedLymphocytesFreeze-DriedGranulosa cellsa-b = p < 0.05; b-c = p < 0.05; a,b,c-d = p < 0.001;H 2 O 2 treatedFig. 1. Proportion of nuclei with undamaged DNA (assessed byComet assay; 200 nuclei counted per group, three replicates each).a–b = p < 0.05; b–c = p < 0.05; a,b,c–d = p < 0.001nucleus donor <strong>in</strong> somatic cell clon<strong>in</strong>g. Although apronuclear-like structure was found <strong>in</strong> more than 90%of the reconstructed embryos 7–9 h after activation,cdnone of these embryos entered the first mitosis, andcytological analysis revealed <strong>in</strong> all embryos extensiveDNA damage, like the one observed <strong>in</strong> occurrence ofasynchronous transfer between s-phase nuclei andmitotic cytoplasts (Johnson and Rao 1970).Comet analysis confirmed the presence of DNAfragmentation <strong>in</strong> all the cells after rehydration (Figs 1and 2a). Interest<strong>in</strong>gly, all cloned embryos arrested atbefore the first mitosis, confirm<strong>in</strong>g the presence of a veryrobust DNA damage checkpo<strong>in</strong>t <strong>in</strong> zygotes (Kalogeropouloset al. 2004).These results were <strong>in</strong> part expected. Spermatozoa aresmall cells with a low level of hydration (Ward andZalensky 1996), and with transcriptionally <strong>in</strong>activeDNA tightly packed around the basic protam<strong>in</strong>e core;altogether, this nuclear structure is very suitable forfreeze dry<strong>in</strong>g. On the contrary, somatic cells with largerdiameter, higher water content, and upper most withtheir nuclear organization render them extremely vul-(a)(b)(c)(d)(e)(g)(h)(f)Fig. 2. (a) Comet analysis ofrehydrated lymphocytes lyophilized<strong>in</strong> 0.1 M threalose, 40·. (b)SEM appearance of dry powderlyophilized with 0.1 M threalose,350·. (c) SEM appearance of drypowder lyophilized without threalose,350·. (d) Fragmented DNA<strong>in</strong> a pronuclear-like structureorganized after 7 h post-activation,100·. (e) Normal pronucleus with aregular nuclear membrane <strong>in</strong> controlreconstructed embryos, 100·.(f) Control embryo reconstructedwith fresh granulosa cells; nocomet tail visible. (g) Embryoreconstructed with a freeze-driedcell: note the massive DNA damage<strong>in</strong>dicated by the migration offragmented DNA outside theembryo. (h) Blastocyst producedseven after culture of enucleatedoocytes <strong>in</strong>jected with freeze-driedgranulosa cells, 40·Ó 2008 Teramo University


Dry Cells for Nuclear Transfer 421Table 2. In vitro development of enucleated oocytes <strong>in</strong>jected withfreeze-dried and fresh, control granulosa cellsSource of cells Cultured 2–8 Cells Morula BlastocystGranulosa control 129 43 31 27 (20.9%) aGranulosa freeze-dried 160 52 28 25 (15.6%) bThe cultures were ma<strong>in</strong>ta<strong>in</strong>ed for 7–8 days <strong>in</strong> the medium SOF plus am<strong>in</strong>o-acidsand BSA, with FCS added on day 4. Reconstructed embryos were checked every24 h for development.a–b p = 0.56.nerable to the comb<strong>in</strong>ed osmotic ⁄ dehydration stressimposed the freeze-dry<strong>in</strong>g process.These prelim<strong>in</strong>ary observations, and the follow<strong>in</strong>greports published on the topics (Kaneko et al. 2003),prompted us to formulate alternative test medium forfreeze-dry<strong>in</strong>g somatic cells. Our first target was to avoidthe massive DNA damage evident <strong>in</strong> rehydrated cellsdried with the sperm protocol.Trehalose produced by some plants, yeasts, sporesand a range of unicellular organism confers them theability to survive conditions of almost absence of water(Crowe et al. 1992). The mechanism by which trehaloseconfers desiccation tolerance is not clear. Trehalose perse is thought to replace the shell of water on the surfaceof macromolecules, prote<strong>in</strong>s <strong>in</strong> particular (Prestrelskiet al. 1993) while on lipids it seems that its protectiveeffect is due to the <strong>in</strong>hibition of phase transitiontemperature of membranes (Leslie et al. 1994). Remarkably,the <strong>in</strong>duced threalose expression <strong>in</strong> fibroblastsconferred desiccation tolerance for 5 days (Guo et al.2000). Based on these <strong>in</strong>dications, trehalose was<strong>in</strong>cluded <strong>in</strong> the formulation of the freeze-dry<strong>in</strong>gmedium.As reported <strong>in</strong> results, freeze-dried medium conta<strong>in</strong><strong>in</strong>g0.1 M trehalose was highly effective <strong>in</strong> protect<strong>in</strong>g thenuclear compartment, for about half of the cells had<strong>in</strong>tact DNA after rehydration. The best results observed<strong>in</strong> lymphocytes, compar<strong>in</strong>g to granulosa cells (65% vs55%, respectively), p < 0.05) was probably due to thesmaller size of the former cell.Enucleated oocytes reconstructed with freeze-driedgranulosa cells developed a pronucleus-like structureand started to cleave an equal rates compared withcontrol ones (Table 2), and 16% of them reached theblastocyst stage (Fig. 2). This result was quite surpris<strong>in</strong>g,because if we consider that about half of thegranulosa cells displayed damaged DNA follow<strong>in</strong>grehydration, we would have expected a much lowerproportion of oocytes <strong>in</strong>jected with dried cells develop<strong>in</strong>gto blastocyst stage. Exonuclease and recomb<strong>in</strong>ationactivity both <strong>in</strong>crease dur<strong>in</strong>g oogenesis, and fully growneggs have found to catalyse homologous recomb<strong>in</strong>ation,ligation and illegitimate recomb<strong>in</strong>ation of exogenousDNA (Lehman et al. 1993), suggest<strong>in</strong>g that multiplepathways are available <strong>in</strong> the oocyte’s cytoplasm for therepair of double strand break. Therefore, it is possiblethat DNA damage <strong>in</strong>duced by the dehydration arerepaired by the oocyte and do not affect the development,at least to blastocyst stage.Our results showed for the first time the developmentalpotential of freeze-dried cells after the nucleartransplantation. However, while <strong>in</strong> the orig<strong>in</strong>al reportsperm cells were kept lyophilized for 4 months prior touse for ICSI, the cells used <strong>in</strong> our study were storedfreeze dried for more than 3 years at room temperature.Therefore, we believe that our method is a majorcontribution towards alternative ways or the establishmentand ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g cell l<strong>in</strong>es for nuclear transfer.Clon<strong>in</strong>g by nuclear transfer is a very dynamic field,where jo<strong>in</strong>t efforts of embryologist and molecularbiologist are produc<strong>in</strong>g knowledge of nuclear reprogramm<strong>in</strong>gmechanism at very rapid pace (Gurdon et al.2003; Meissner and Jaenisch 2006), lett<strong>in</strong>g foresee<strong>in</strong>g asuccessful application of clon<strong>in</strong>g procedures <strong>in</strong> themedium term. Unfortunately, the list on endangeredspecies is not limited to mammals, but a wide range ofm<strong>in</strong>or vertebrates are also disappear<strong>in</strong>g quickly. However,clon<strong>in</strong>g procedures have been recently adapted tovery unconventional animal models with two papersreport<strong>in</strong>g about successful clon<strong>in</strong>g of zebra fish (Ju et al.2004) and Drosophila (Haigh et al. 2005) these accomplishmentsbroad the range of species that can potentiallybenefit from clon<strong>in</strong>g technology.To conclude, we demonstrated for the first time thatsomatic cells stored <strong>in</strong> a freeze-dried state are able todirect embryonic development of enucleated oocytes tillblastocyst stage. These results represent a substantialbreakthrough <strong>in</strong>novative for the establishment ofgenetic banks from endangered animals.ReferencesCorley-Smith GE, Brandhorst BP, 1999: Preservation ofendangered species and populations: a role for genomebank<strong>in</strong>g, somatic cell clon<strong>in</strong>g, and androgenesis? MolReprod Dev 53, 363–367.Crowe JH, Hoekstra FA, Crowe LM, 1992: Anhydrobiosis.Annu Rev Physiol 54, 579–599.Guo N, Puhlev I, Brown DR, Mansbridge J, Lev<strong>in</strong>e F, 2000:Trehalose expression confers desiccation tolerance on humancells. Nat Biotechnol 18, 168–171.Gurdon JB, Byrne JA, Simonsson S, 2003: Nuclear reprogramm<strong>in</strong>gand stem cell creation. Proc Natl Acad Sci U S A30, 11819–11822.Haigh AJ, Macdonald WA, Lloyd VK, 2005: The generationof cloned Drosophila melanogaster. Genetics 169, 1165–1167.Hilton-Taylor C, 2000: (compiler): IUCN Red List ofThreatened Species. IUCN ⁄ SSC, Gland, Switzerland andCambridge, UK.Johnson RT, Rao PN, 1970: Mammalian cell fusion: <strong>in</strong>ductionof premature chromosome condensation <strong>in</strong> <strong>in</strong>terphasenuclei. Nature 226, 717–722.Ju B, Huang H, Lee KY, L<strong>in</strong> S, 2004: Clon<strong>in</strong>g zebrafish bynuclear transfer. Methods Cell Biol 77, 403–411.Kalogeropoulos N, Christoforou C, Green AJ, Gill S,Ashcroft NR, 2004: chk-1 is an essential gene and isrequired for an S-M checkpo<strong>in</strong>t dur<strong>in</strong>g early embryogenesis.Cell Cycle 3, 1196–1200.Kaneko T, Whitt<strong>in</strong>gham DG, Yanagimachi R, 2003: Effect ofpH value of freeze-dry<strong>in</strong>g solution on the chromosome<strong>in</strong>tegrity and developmental ability of mouse spermatozoa.Biol Reprod 68, 136–139.Kesk<strong>in</strong>tepe L, Pacholczyk G, Machnicka A, Norris K, CurukMA, Khan I, Brackett BG, 2002: Bov<strong>in</strong>e blastocyst developmentfrom oocytes <strong>in</strong>jected with freeze-dried spermatozoa.Biol Reprod 67, 409–415.Ó 2008 Teramo University


422 P Loi, K Matzukawa, G Ptak, Y Natan, J Fulka Jr and A AravKusakabe H, Szczygiel MA, Whitt<strong>in</strong>gham DG, YanagimachiR, 2001: Ma<strong>in</strong>tenance of genetic <strong>in</strong>tegrity <strong>in</strong> frozen andfreeze-dried mouse spermatozoa. Proc Natl Acad Sci U S A98, 13501–13506.Lehman CW, Clemens M, Worthylake DK, Trautman JK,Carroll D, 1993: Homologous and illegitimate recomb<strong>in</strong>ation<strong>in</strong> develop<strong>in</strong>g Xenopus oocytes and eggs. Mol Cell Biol13, 6897–6906.Leslie SB, Teter SA, Crowe LM, Crowe JH, 1994: Trehaloselowers membrane phase transitions <strong>in</strong> dry yeast cells.Biochim Biophys Acta 1192, 7–13.Liu JL, Kusakabe H, Chang CC, Suzuki H, Schmidt DW,Julian M, Pfeffer R, Bormann CL, Tian XC, YanagimachiR, Yang X, 2004: Freeze-dried sperm fertilization leads tofull-term development <strong>in</strong> rabbits. Biol Reprod 70, 1776–1781.Loi P, Ptak G, Barboni B, Fulka J Jr, Cappai P, Cl<strong>in</strong>ton M,2001: Genetic rescue of an endangered mammal by crossspeciesnuclear transfer us<strong>in</strong>g post-mortem somatic cells.Nat Biotechnol 19, 962–964.Loi P, Cl<strong>in</strong>ton M, Barboni B, Fulka J Jr, Cappai P, Feil R,Moor RM, Ptak G, 2002: Nuclei of nonviable ov<strong>in</strong>e somaticcells develop <strong>in</strong>to lambs after nuclear transplantation. BiolReprod 67, 126–132.Margules CR, Pressey RL, 2000: Systematic conservationplann<strong>in</strong>g. Nature 405, 243–253.Meissner A, Jaenisch R, 2006: Mammalian nuclear transfer.Dev Dyn 235, 2460–2469.Myers N, Mittermeier RA, Mittermeier CG, da Fonseca GAB,Kent J, 2000: Biodiversity hotspots for conservation priorities.Nature 403, 853–858.Polge C, Smith A, Parkes AS, 1949: Revival of Spermatozoaafter vitrification and dehydration at low temperature.Nature 164, 666–667.Prestrelski SJ, Arakawa T, Carpenter JF, 1993: Separation offreez<strong>in</strong>g- and dry<strong>in</strong>g-<strong>in</strong>duced denaturation of lyophilizedprote<strong>in</strong>s us<strong>in</strong>g stress-specific stabilization. II. Structuralstudies us<strong>in</strong>g <strong>in</strong>frared spectroscopy. Arch Biochem Biophys303, 465–473.Ptak G, Dattena M, Loi P, Tischner M, Cappai P, 1999a:Ovum pick-up <strong>in</strong> sheep: efficiency of <strong>in</strong> vitro embryoproduction, vitrification and birth of offspr<strong>in</strong>g. Theriogenology52, 1105–1114.Ptak G, Loi P, Dattena M, Tischner M, Cappai P, 1999b:Offspr<strong>in</strong>g from one-month-old lambs: studies on the developmentalcapability of prepubertal oocytes. Biol Reprod 61,1568–1574.Saacke RG, Malquist JO, 1961: Freeze-dry<strong>in</strong>g of bov<strong>in</strong>espermatozoa. Nature 192, 995–996.Sherman JK, 1954: Freez<strong>in</strong>g and freeze-dry<strong>in</strong>g of humanspermatozoa. Fertil Steril 5, 357–371.Wakayama T, Yanagimachi R., 1998: Development of normalmice from oocytes <strong>in</strong>jected with freeze-dried spermatozoa.Nat Biotechnol 16, 639–644.Ward SW, Zalensky AO, 1996: The unique, complex organizationof the transcriptionally silent sperm chromat<strong>in</strong>. CritRev Eukaryot Gene Expr 6, 139–147.Wilmut I, Schnieke AE, McWhir J, K<strong>in</strong>d AJ, Campbell KHS,1997: Viable offspr<strong>in</strong>g derived from fetal and adult mammaliancells. Nature 385, 810–813.Author’s address (for correspondence): L<strong>in</strong>o Loi, Department ofComparative Biomedical Sciences, Teramo University, Piazza AldoMoro 45, 64100, Teramo, Italy. E-mail: ploi@unite.itConflict of <strong>in</strong>terest: All authors declare no conflict of <strong>in</strong>terests.Ó 2008 Teramo University


The largest on-l<strong>in</strong>e library <strong>in</strong>cludes textbooks, proceed<strong>in</strong>gs, journals,manuals, a calendar, and much more. Free unlimited access forveter<strong>in</strong>arians, veter<strong>in</strong>ary technicians and students.Books <strong>in</strong> Veter<strong>in</strong>ary Medic<strong>in</strong>e:o A Concise Review of Veter<strong>in</strong>ary Virology byCarter, Wise and Floreso Topics <strong>in</strong> Bull Fertility by Chenowetho Recent Advances <strong>in</strong> Small Animal<strong>Reproduction</strong> by Concannon et al.o Recent Advances <strong>in</strong> Equ<strong>in</strong>e <strong>Reproduction</strong> byBallo Recent Advances <strong>in</strong> Yak <strong>Reproduction</strong> by Zhaoand Zhango Recent Advances <strong>in</strong> Anesthetic Managementof Large <strong>Domestic</strong> <strong>Animals</strong> by Steffeyo Recent Advances <strong>in</strong> Goat Diseases byTempestao Recent Advances <strong>in</strong> Laparoscopy andThoracoscopy by Wilsono Recent Advances <strong>in</strong> Veter<strong>in</strong>ary Anesthesiaand Analgesia: Companion <strong>Animals</strong> by Gleedand Ludderso Cl<strong>in</strong>ical Avian Medic<strong>in</strong>e by Harrison andLightfooto Equ<strong>in</strong>e Respiratory Diseases by Lekeuxo Care of Donkeys by Matthews and Tayloro A Guide to Hematology <strong>in</strong> Dogs and Cats byRebar, MacWilliams, Feldman, et al.o Dermatology for the Small Animal Practitionerby Muellero Veter<strong>in</strong>ary Toxicology by Beasleyo Animal Disease Factsheets The Center for FoodSecurity & Public Health - Iowa State UniversityProceed<strong>in</strong>gs <strong>in</strong> Veter<strong>in</strong>ary Medic<strong>in</strong>e:o NAVC - North American Veter<strong>in</strong>ary Conferenceo IPVS - International Pig Veter<strong>in</strong>ary SocietyCongresso WBC - World Buiatrics Congresso AAEP - American Association of Equ<strong>in</strong>ePractitionerso SIVE - Italian Association of Equ<strong>in</strong>eVeter<strong>in</strong>arians Annual Congresso BEPS - Belgian Equ<strong>in</strong>e Practitioners SocietyStudy Dayso Geneva Congress on Equ<strong>in</strong>e Medic<strong>in</strong>e andSurgeryo SCIVAC - Società Culturale Italiana Veter<strong>in</strong>ariper Animali da Compagnia Congresso EENHC - European Equ<strong>in</strong>e Health andNutrition Biannual Congresso AAVPT – American Academy of Veter<strong>in</strong>aryPharmacology and Therapeuticso WSAVA - World Small Animal Veter<strong>in</strong>aryAssociation Annual Congresso Can<strong>in</strong>e Cancer Conference: Genes, Dogs,and Cancero Cross-Species Approach to Pa<strong>in</strong> andAnalgesiao EAVDI - European Association of Veter<strong>in</strong>aryDiagnostic Imag<strong>in</strong>go EAZWV - European Association of Zoo andWildlife Veter<strong>in</strong>arians Biannual Meet<strong>in</strong>gso Lameness <strong>in</strong> Rum<strong>in</strong>ants SymposiumVeter<strong>in</strong>arians, students and Technicians have free access to all books andproceed<strong>in</strong>gs. Visit the IVIS Website at www.ivis.org and register today. For free!!


Table of Contents Volume 43 · Supplement 2 · July 2008 · 1-422CONTENTS cont<strong>in</strong>ued...B. M. A. O. PERERA<strong>Reproduction</strong> <strong>in</strong> <strong>Domestic</strong> Buffalo 200-206P. S. BRAR, A. S. NANDAPostpartum Ovarian Activity <strong>in</strong> South Asian Zebu Cattle 207-212M. J. R. PARANHOS DA COSTA, A. SCHMIDEK, L. M. TOLEDOMother-Offspr<strong>in</strong>g Interactions <strong>in</strong> Zebu Cattle 213-216B. S. PRAKASH, M. SARKAR, M. MONDALAn Update on <strong>Reproduction</strong> <strong>in</strong> Yak and Mithun 217-223F. X. DONADEU, H. G. PEDERSENFollicle Development <strong>in</strong> Mares 224-231M. A. HAYES, B. A. QUINN, N. D. KEIRSTEAD, P. KATAVOLOS, R. O. WAELCHLI, K. J. BETTERIDGEProte<strong>in</strong>s Associated With the Early Intrauter<strong>in</strong>e Equ<strong>in</strong>e Conceptus 232-237Z. ROTHHeat Stress, the Follicle, and Its Enclosed Oocyte: Mechanisms and Potential Strategies to ImproveFertility <strong>in</strong> Dairy Cows 238-244K. - P. BRÜSSOW, J. RÁTKY, H. RODRIGUEZ-MARTINEZFertilization and Early Embryonic Development <strong>in</strong> the Porc<strong>in</strong>e Fallopian Tube 245-251S. PYÖRÄLÄMastitis <strong>in</strong> Post-Partum Dairy Cows 252-259M. G. DISKIN, D. G. MORRISEmbryonic and Early Foetal Losses <strong>in</strong> Cattle and Other Rum<strong>in</strong>ants 260-267N. MANABE, F. MATSUDA-MINEHATA, Y. GOTO, A. MAEDA, Y. CHENG, S. NAKAGAWA, N. INOUE,K. WONGPANIT, H. JIN, H. GONDA, J. LIRole of Cell Death Ligand and Receptor System on Regulation of Follicular Atresia <strong>in</strong> Pig Ovaries 268-272F. F. BARTOL, A. A. WILEY, C. A. BAGNELLEpigenetic Programm<strong>in</strong>g of Porc<strong>in</strong>e Endometrial Function and the Lactocr<strong>in</strong>e Hypothesis 273-279K. C. CAIRES, J. A. SCHMIDT, A. P. OLIVER, J. DE AVILA, D. J. MCLEANEndocr<strong>in</strong>e Regulation of the Establishment of Spermatogenesis <strong>in</strong> Pigs 280-287I. DOBRINSKIMale Germ Cell Transplantation 288-294N. RAWLINGS, A. C. O. EVANS, R. K. CHANDOLIA, E. T. BAGUSexual Maturation <strong>in</strong> the Bull 295-301A. DINNYES, X. C. TIAN, X. YANGEpigenetic Regulation of Foetal Development <strong>in</strong> Nuclear Transfer Animal Models 302-309R. C. BOTT, D. T. CLOPTON, A. S. CUPPA Proposed Role for VEGF Isoforms <strong>in</strong> Sex-Specific Vasculature Development <strong>in</strong> the Gonad 310-316B. K. WHITLOCK, J. A. DANIEL, R. R. WILBORN, T. H. ELSASSER, J. A. CARROLL, J. L. SARTINComparative Aspects of the Endotox<strong>in</strong>- and Cytok<strong>in</strong>e-Induced Endocr<strong>in</strong>e Cascade Influenc<strong>in</strong>gNeuroendocr<strong>in</strong>e Control of Growth and <strong>Reproduction</strong> <strong>in</strong> Farm <strong>Animals</strong> 317-323C. R. BARB, G. J. HAUSMAN, C. A. LENTSEnergy Metabolism and Lept<strong>in</strong>: Effects on Neuroendocr<strong>in</strong>e Regulation of <strong>Reproduction</strong> <strong>in</strong> the Gilt and Sao 324-330C. GALLI, I. LAGUTINA, R. DUCHI, S. COLLEONI, G. LAZZARISomatic Cell Nuclear Transfer <strong>in</strong> Horses 331-337D. RATH, L. A. JOHNSONApplication and Commercialization of Flow Cytometrically Sex-Sorted Semen 338-346J. M. VAZQUEZ, J. ROCA, M. A. GIL, C. CUELLO, I. PARRILLA, I. CABALLERO, J. L. VAZQUEZ, E. A. MARTÍNEZLow-Dose Insem<strong>in</strong>ation <strong>in</strong> Pigs: Problems and Possibilities 347-354C. B. A. WHITELAW, S. G. LILLICO, T. KINGProduction of Transgenic Farm <strong>Animals</strong> by Viral Vector-Mediated Gene Transfer 355-358A. C. O. EVANS, N. FORDE, G. M. O'GORMAN, A. E. ZIELAK, P. LONERGAN, T. FAIRUse of Microarray Technology to Profile Gene Expression Patterns Important for <strong>Reproduction</strong> <strong>in</strong> Cattle 359-367J. P. KASTELIC, J. C. THUNDATHILBreed<strong>in</strong>g Soundness Evaluation and Semen Analysis for Predict<strong>in</strong>g Bull Fertility 368-373G. C. ALTHOUSESanitary Procedures for the Production of Extended Semen 374-378B. LEBOEUF, J. A. DELGADILLO, E. MANFREDI, A. PIACÈRE, V. CLÉMENT, P. MARTIN, M. PELLICER,P. BOUÉ, R. DE CREMOUXManagement of Goat <strong>Reproduction</strong> and Insem<strong>in</strong>ation for Genetic Improvement <strong>in</strong> France 379-385N. KOSTEREVA, M. - C. HOFMANNRegulation of the Spermatogonial Stem Cell Niche 386-392P. MERMILLOD, R. DALBIÈS-TRAN, S. UZBEKOVA, A. THÉLIE, J. - M. TRAVERSO, C. PERREAU,P. PAPILLIER, P. MONGETFactors Affect<strong>in</strong>g Oocyte Quality: Who is Driv<strong>in</strong>g the Follicle? 393-400K. KIKUCHI, N. KASHIWAZAKI, T. NAGAI, M. NAKAI, T. SOMFAI, J. NOGUCHI, H. KANEKOSelected Aspects of Advanced Porc<strong>in</strong>e Reproductive Technology 401-406B. OBACKClimb<strong>in</strong>g Mount Efficiency – Small Steps, Not Giant Leaps Towards Higher Clon<strong>in</strong>g Success <strong>in</strong> Farm <strong>Animals</strong> 407-416P. LOI, K. MATZUKAWA, G. PTAK, Y. NATAN, J. FULKA JR, A. ARAVNuclear Transfer of Freeze-Dried Somatic Cells <strong>in</strong>to Enucleated Sheep Oocytes 417-422

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!