13.07.2015 Views

n~po~ twenty-s i hth rnnurl conference - the Society for Reproductive ...

n~po~ twenty-s i hth rnnurl conference - the Society for Reproductive ...

n~po~ twenty-s i hth rnnurl conference - the Society for Reproductive ...

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.

~ fa [Bu@4M ~ IN ~ POS BAS ~ [8G9~~M~!NJ ~ IPJ(Q)S[P>OST[E~SASfR{lBo9~N~POSTERSASlRi[83°~~M~[NHPOSf[E 0 AS~183°~4M~[N]~POSrE~SA\S~[8°~4M~M~[NJ~rPOSf[E~SASASlRiIBO~4M~lN~PODOSf[ErRUSAS~f8°~0~4M~[N~POS1[E~[E[RSAS~[Bi°~4M~[NHPOSu[E~SASfR{rs09 ~[NJ~[POSuE~SAS 0rs°9)4M~NHPOSuIE 0ASfR{l83°~4M~[N]~POSr[E[RSAS~[8°~4M~ . OSf[El~SASf1l[6°~M1~NHPOSflE~SAS0~4M~!Nl~POS1'E~ r~ SrRIB 0 94M ~lN~ POS1~fR{SASfRH8°Sl4M~lN~ POSllElFRSAS[R1[8Q~4M~fNJ[J°OSulE~SASRrs094M~fNl~POSll~fR{SAS[R{rsO~4M~N~POS"=~"ERSASrRi[80 ~41M] ~ [NJ ~pas1 IE fRiSIAS[Rl [8)°94 ~~ ~ IN ~ PCOSlE I~~§ J~\S!fit fB u@4M~ [NJ ~ Posr IE IR{SAS ~ [8) 094M ~ rNl ~ POS1 rE [RlSIASfRi [8) 094M ~ [1~ II Pcansrrr IE RSAS [R{ 18u~MJ ~ [N] ~ POS1IE RSAS~H8°94M ~ IN ~ rPOSllEfR{SIAS [riH8 U S,4M I] [i\~ ~ POS1E [R1AS[R{f8Q9)4M~lN~ POSulEfRiSAS[Rlf8Q~~rM~ U\JH POSu[E~\SASR[8°~4M~ fNH~(OSu[E ~SAS fR1rs o 94M ~ fNl ~ POSflEfR{SIAS fR1(83°~4M ~ fNj ~ [POS1IE RSAS~[8°~4M~~ PROrEED I ~I~S OF THE "'~~rN~POSf[EIM\SIA~ L I~ u ~SRlBO~~~~N~PO~ TWENTY-S I HTH RNNURLASfR1l8°~CONFERENCErlE lRSASR[8°9)4M ~ IN ~ pCQ)SlrlElKlSAS R[8U~4~tTI ~ [NJ ~ lPo)srr lF~USASt~~ ~~ ~ fpJOS1"IE [R{SIASlRi[8U9d{~M ~ IN~ POSllElKlSIAS~lB5USJc(!lJ,M~ fNl II~\S ·[R1[8°s)4M ~ IN ~ ~051llE RSAS [Rlf8°G)~M~ N ~ ~OSl'lE 11~SASiPOST[E R1S lAS fRU8°9~M ~ N ~poslE~SASRlB O~~M ~ [~~ [POST IE[8 u9J1~rM ~ ~\lH [pOSIT fE RSlAS ~ [8 o~~ 1M ~ [NJ ~ POS1 fE RSAS ~ [8U~~,M~ IN ~ [Posr rE rR1SAS [R{ [8°~4M ~ IN ~ P(() 511 rE fR{SlAS IRi [8 0 ~~, [MH [NJ ~ ~osrr [E lR{§ 10\S L~{ ~ uS)~ ~M~[NHP)OSl~ HILTON HOTEL HR I SHRNE ~~[POSlr[ERSA~fRl~o@4M~ 26-28 SEPTEMBER 1994 :IRl~o~4M~!N]~~h©lrE~~~ J ~~~D~~~~Ie ''J) \\d) lJ::;:;;, ln1~ In ... ..olI ~ U b Ln\ ~ 11\\ ~,2)[8U~~,M~ [N ~ [poSLrlE[R{SASfRl[8U9)~M~[NJ~ rOSlfElRiSASlR\[8)°~&1j}M~[N~ [POSf =[E!RlSAS[R{[B)°9~,M~~~~rPO lJ~rERSASR18U ~rrtA]~N 0(0


The Australian <strong>Society</strong> <strong>for</strong> <strong>Reproductive</strong> Biology Inc.Twenty Sixth Annual ConferenceThe Hilton Hotel, BrisbaneSeptember 26 - 28 1994PROGRAMME AND MINIPOSTERS OF PAPERSCopyright Australian <strong>Society</strong> <strong>for</strong> <strong>Reproductive</strong> Biology, 1994ISSN 0818-4712PersonnelAcknowledgementsHotel mapsProgramme guidesProgrammePlenary LecturersJunior Scientist FinalistsMinipostersAuthor indexCONTENTS11111IVVIIXXIXXX1-1171


THE AUSTRALIAN SOCIETY FOR REPRODUCTIVE BIOLOGY INC.September 1994THE AUSTRALIAN SOCIETY FOR REPRODUCTIVE BIOLOGY INc.wishes to thank <strong>the</strong> following <strong>for</strong> <strong>the</strong>ir supportof <strong>the</strong> 1994 <strong>conference</strong>ChairmanSecretaryTreasurerCommittee MembersOFFICE BEARERSClinical Interests RepresentativePostgraduate RepresentativePROGRAMME COMMITTEEChairmanCommittee MembersProf Alan TrounsonDr Lois SalamonsenDr Leeanda WiltonDr Jim CumminsDr Graham JenkinDr Simon MaddocksDr Kris BattyeDr Conrad SemiaDr Rob NormanMr David MillerDr Simon MaddocksDr Bill BreedDr Rob NormanDr Mark NottleDr Bob SeamarkProfessor Brian SetchellSPONSORSMeat Research Corporation of AustraliaOrganon (Australia) Pty. Ltd.Serono Australia Pty. Ltd.William A. Cook Australia Pty. Ltd.TRADE EXHffiITORSAlphapharm Pty. Ltd.Astra Pharmaceuticals Pty. Ltd.Bayer Australia Ltd.Baxter Diagnostics Pty. Ltd.Bio-mediq DPC Pty. Ltd.Boehringer Mannheim Australia Pty. Ltd.Bristol-Myers Squibb Pharmaceuticals Pty. Ltd.Children's Growth FoundationCiba-Geigy Australia LimitedCryologic Pty. Ltd. - Freeze ControlCSL PharmaceuticalsEli Lilly Australia Pty. Ltd.Fisons Pty. Ltd.General Diabetes ServicesMedtel AustraliaNovo Nordisk Pharmaceuticals Pty. Ltd. (Australia)Organon (Australia) Pty. Ltd. Division of AKZOParke Davis Pty LtdPharmacia (Australia) Pty. Ltd.Roche Products Pty. Ltd.Sandoz Australia Pty. Ltd.Upjohn Pty. Ltd.LOCAL ORGANISING COMMITTEEChairmanCommittee MembersDr Peter KayeDr Len MartinDr Michael Demden (ESA)Mrs Ruth Lillian (ICMS)The contents of <strong>the</strong>se Proceedings have not been edited by <strong>the</strong> <strong>Society</strong> and are reproduced assubmitted. Responsibility <strong>for</strong> <strong>the</strong> accuracy of <strong>the</strong> communications and <strong>for</strong> carrying outexperimental work on animals or humans in accordance with <strong>the</strong> approval of<strong>the</strong> appropriate ethicscommittee, or with <strong>the</strong> appropriate legislation, rests with <strong>the</strong> authors.Material in <strong>the</strong>se Proceedings may not be reproduced without permission of <strong>the</strong> <strong>Society</strong>.Price of <strong>the</strong> Proceedings to Non-members: $25 plus postage.11iii


FLOOR PLAN FOR MEETING VENUE(JTY Jv{APFire Exit Stair 6FACILITIESBarbeques, Picnic Tables @]~Post Offices [§]Taxi Ranks •lllJToiletsTourist In<strong>for</strong>mation [)Car Park LobbyoMargauxsNight CluboNORTHMoreton65sq.my -.-­Xes;;:)TLockyer112 sq.mRedlands105 sq.mYX:50 ::~~.~ :r. >-"'::;~'Ej ~Fire Exil Stair iFire Exil Stair 5Fire Exit Stair ~Ballroom LobbyService Elevators~sunmapSCALEC==lOcO=2:IOO==3IOO==4:I00=::55?~etresivh~e ...nc{f.=i·:;;~r;..Fire Exil Stair 3gillCarparkAccessx X• xo"'0xBallroom C295 SQ.mA. X XIX .0Ir•IJL,Anterooms,~I.-II•Grand Ballroom I' .... .1II-,IIIIIIIII~~JIBallroom B200 sq.mIIIIIIII --,I_IX---~Ballroom A295 sq.mxO I xo XO XOY


Programme GuidesProgramme GuidesMonday September 26 Tuesday September 27ASRB ESA ASRB ESA0800 Snack breakfast in <strong>the</strong> Trade Exhibition area 0800 Snack breakfast in <strong>the</strong> Trade Exhibition area08300930100010301130Concurrent Miniposter sessions:-Hypothalamo-Pituitary-Gondal axis-In-vitro maturation & In-vitrofertilizationTeaConcurrent Miniposter sessions:-Cryopreservation & Transgenesis-Uterine structure & PregnancyPhannacia Lecture: David Hill"Peptide growthfactor interaction duringdevelopment "TeaConcurrent Oral sessions:-Sandoz Junior Scientist Award-Thyroid-clinical-Fetal development-Steroid honnone action-Servier Award0830090009301000Miniposter session:-Growth Factors and <strong>the</strong> UternsTeaMerck Sharp & Dohme Lecture: Ralph Buttyan"Death genes, anti-death genes andprostate cancer "TeaConcurrent Oral sessions:-Endocrine cancers-Effects ofIGFs-Bone/Calcium12001300140015301600Goding Lecture - llpo Huhtaniemi "Molecular aspects of<strong>the</strong> ontogeny of<strong>the</strong> mammalianpituitary-gonadal axis"LunchJunior Scientist OralsTeaThe Great Debate"lcsi or !cnon"LunchConcurrent Poster sessions:-HPA Axis-Stress, metabolism and peptides-Reproduction 1TeaConcurrent Miniposter sessions:-Thyroid-Insulin & IGF's103012001300140014301530Concurrent Miniposter sessions:-Somatic cells and gonadal regulation-Regulation of fertilityHarrison Lecture - Allen Spiegel "Defects in G protein-coupled signal transductionpathways in human disease "LuchConcurrent Miniposter sessions:-Genn cells, spennatogenesis &spenn function-Embryonic DevelopmentTeaLunchConcurrent Poster sessions-Growth Honnone-Clinical endocrinology-Reproduction 2Tea17151815FinishAnnual General MeetingFinish1600Miniposter session:-Growth factors in early pregnancyConcurrent Miniposter sessions-Testis-HPA Axis & 2nd messengers1700 Annual General Meeting Henning-Berlin Lecture: Marc Pannentier"Molecular genetics and transgenic models (~fthyroid proliferative diseases "1800Finish1830 Finishvivii


0800083009001000ASRBProgramme GuidesWednesday September 28ESASnack breakfast in <strong>the</strong> Trade Exhibition areaMRC Lecture: Bill Thatcher"Control and management ofovarianfollicles in cattle to optimize fertility "TeaPincus Taft Lecture: Ron Kahn"Insulin resistance, diabetogenes and <strong>the</strong> causeoftype II diabetes "TeaAUSTRALIAN SOCIETY FOR REPRODUCITVE BIOLOOY INC.PROGRAMME 1994Registration: The registration desk will be located on Level 4 of <strong>the</strong> Hilton Hotel. On ~unday, September25th, it will be open from O9OOh, on Monday - Wednesday between 0830 and 1700h.The Goding and Harrison Lectures, and <strong>the</strong> Junior Scientist finalis,t orals will be presented in Ballroom A,<strong>the</strong> MRC Lecture and <strong>the</strong> Annual General Meeting of <strong>the</strong> <strong>Society</strong> will be held in <strong>the</strong> Redlands Room, witho<strong>the</strong>r session venues as indicated in <strong>the</strong> detailed programme. All venues are on Level 4 of<strong>the</strong> Hilton Hotel.Trade displays will also be found on this level in Ballroom C where morning and afternoon teas will also beserved. Each morning between 0800-0830 a snack breakfast will also be available in <strong>the</strong> Trade Exhibitorarea.Following <strong>the</strong> ASRB Silver Jubilee Symposium on Sunday, September 25th, <strong>the</strong>re will be an in<strong>for</strong>malsocial function at <strong>the</strong> Queensland Art Gallery starting at 1830h.1030Miniposter session:-Artificial insemination & Embryo transferCombined symposium with ADS:Gene TherapyMONDAY SEPTEMBER 261130FinishSession 1 (Concurrent)HYPOTHALAMO·PITUITARY·GONADAL AXIS123013001500LunchLunchCombined symposium with ANZBMS:Corticosteroids and BoneFinishChairman: Dr. lain ClarkeTime: 0830 - 10001Venue: Ballroom BBarker-Gibb,M.L., Boublik,J.H. and Clarke,I.J.NPYeffects on LH secretion in <strong>the</strong> ovariectomised ewe: Y2 receptor involvement, seasonal variationand site ofaction within <strong>the</strong> hypothalamus2 Sawangjaroen,K. and Curlewis,J.D.In vitro effects ofPACAP and VIP on sheep pituitary cells3 O'Brien,G.M., Curlewis,J.D. and Martin, L.Physiological and pharmacological challenges to prolactin secretion in Pteropus species (jlying foxes)4 Drummond,A., Dyson,M., Mercer,J. and Findlay,J.Localisation ofinhibin and relatedpeptides in <strong>the</strong> neonatal rat ovary: potential mediators ofgonadotrophin sensitivity5 O'Shea,T.,Anderson, S.T. and Hillard,M.A.Immunization ofmerino lambs. Effects on inhibin binding and gonadotrophin response toovariectomy6 Klein,R., Clarke,I.J. and Robertson,D.M.Studies in sheep exploring plasma follistatin elevations due to frequent blood sampling or surgery7 Tilbrook,A.J., Clarke,!.J. and de Kretser,D.M.Follistatin suppresses FSH in castrated rams8 Garcia,S., Findlay, J.K. and Mercer,J.E.Regulation ofovarian activin receptor type II expression across <strong>the</strong> rat oestrous cycle9 Rabiee, A.R., Lean, I.J., Gooden, J.M. and Miller, B.G.Glucose and cholesterol uptake by <strong>the</strong> ovary of<strong>the</strong> sheep10 Bergfeld,E., D'Occhio,M.J., Whyte, T.R., Aspden,W.J. and Kinder,J.E.Recovery ofpituitary and testis function in young bulls cifter treatment with <strong>the</strong> LHRH agonistdeslorelinviiiix


MONDAY SEPTEMBER 26 (cont)11 Turner,A., Hemsworth,P.H., Lohuis,H. and Tilbrook,A.J.The role ofstress in boar induced puberty in gilts12 Stobart,S.J. and Fitzpatrick,L.A.Seasonal changes in ovarian activity ofbos indicus crossbred heifers in a tropical environment13 Wright,P.J., Philip,C. and Hariadi,M.Progesterone treatment ofewes reduced corpus luteum weight and oestradiol treatment reducedovulation rate14 Hamemick, D.L., Clay, C.M., VanKirk, E.L. and Moss, G.E.Oestradiol increases amounts ofmRNA<strong>for</strong> gonadotrophin-releasing hormone receptors in sheepSession 2 (Concurrent) : IN-VITRO MATURATION & IN-VITRO FERTILIZATIONChairman: Dr.l..eeanda WiltonTime: 0830 - 1000Venue: Sam<strong>for</strong>d Room15 EarLC.R., Irvine, B.J., Rowe,J.P., Kotaras, P. and Armstrong,D.T.Production ofin vivo matured oocytes from 6 week old calves16 Smith, J.F., Tervit,H.R., McGowan,L.T. and Pugh,P.A.Effect ofaspiration system on <strong>the</strong> recovery and development ofsheepfollicular oocytes17 Udy,G.B., McGowan,L.T., Nemaia,F.A., Ankersmit,A.E.L. and Tervit,H.R.Cattle oocyte recovery is affected more by aspiration pressure than needle size18 Grupen,C.G. Nagashima,H. and Nottle,M.B.Clsteamine enhances synchronous pronuclear<strong>for</strong>mation in porcine oocytes matured andfertilized inVItro19 McMillan,W.H., Pugh,P.A. and Peterson,AJ.Early .ernkryo survival is higherfollowing twin ra<strong>the</strong>r than single transfer offrozen-thawed, but notfresh In VItro produced bovine embryos20 Richings,N.M., Boume,H., Harari,O., Jones,G.M., Ma<strong>the</strong>ws,P.M., McDonald,M.,Baker,H.W.G. and Johnston,WH.Intra-cytoplasmic sperm injection (ICSI) as a treatment<strong>for</strong> severe male infertility in humans and itspotential as a tool in endangered species programmes21 Lacham-Kaplan,O. and Trounson, A.Effect ofsperm motility on <strong>the</strong> fertilization rate by intracytoplasmic sperm microinjection22 Ca~t,J., Morton,M. and Saunders,D.Should all oocytes fenilise after intracytoplasmic sperm injection?23 Smitz.J., J~nssensw~llen,C., Lui,J., Joris,H., Camus,M., Devroey,P. and Van Steirteghem,A.Retrospective analySIS ofsuperovulated cycles <strong>for</strong> ICSI yielding an increased number ofimmatureoocytes24 Fry,. R.C., Simpson,T.L., Squires,T.J. and Findlay,J.K.Follicular growth and oocyte collection in heifers immunised against inhibinMONDAY SEPTEMBER 26 (coot)25 Xia,P., Rutledge,J., Watson,A.J. and Armstrong,D.T.Characterization ofin vitro syn<strong>the</strong>sized proteins secreted by porcine non-attached oviductal epi<strong>the</strong>lialcell spheres treated with estrogenSession 3 (Concurrent)Chairman: Dr. Mark NottleTime: 1030 - 1130CRYOPRESERVATION & TRANSGENESISVenue: Ballroom B26. Brennan,A.P. and Holden,C.A.Cryopreservation ofhuman sperm with pentoxifylline27 Sanchez-Partida, L.G., Zupp,J.L., Maxwell,W.M.C. and Setchell,B.P.Effect oflevels ofglycerol and compatible solutes on <strong>the</strong> post-thaw motility ofram spermatozoa28 Molinia,F.C. and Rodger,J.C.Pellet-jreezing ofmarsupial spennatozoa29 Withdrawn30 Pugh,P.A., Ankersmit,A.E.L., McGowan, L.T. and Tervit,H.R.Effect ofprotein type and concentration in <strong>the</strong> freezing medium on <strong>the</strong> survival ofIVP bovineembryos31 Nagashima,H., Kashiwazaki,N., Ashman, R.J. and Nottle, N.B.In vitro and in vivo survival ofcryopreserved hatched blastocysts in pigs32 Kashiwazaki,N., Nagashima,H., Ashman,R.J. and Nottle,M.B.Cryopreservation ofporcine in vivo and in vitro derived blastocysts with glycerol and ethylene clycol33 Nottle,M.B., Nagashima,H., Kashiwazaki,N., Ashman,R.J., Du,Z., Grupen,C.G.,McIlfatrick,S.M., Harding,M., Cheah,C., Craw<strong>for</strong>d,R.J. and Robins,A.J.Production ofpigs containing a metallothionein porcine growth hormone gene construct34 Sutton,R., Fawcett,A.A., O'Grady,J., Ward,W.G. and Brown,B.W.An evaluation ofa tyrosinase minigene as a marker<strong>for</strong> transgenesis in Quakenbush mice35 Tatham,B.,Giliam,K., Dowsing,A., Mahaworasilpa,T. and Trounson,A.Effects of<strong>the</strong> zona pellucida on electrofusion and development oftwo-cell mouse embryos36 Du,Z.T., Dai,Y.F., Owens,P.C., Armstrong,D.T. and Seamark,R.F.Fertility oftransgenic female rats expressing pig growth hormoneSession 4 (Concurrent)Chainnan: Dr. Len MartinTime: 1030 - 1200UTERINE STRUCTURE & PREGNANCYVenue: Sam<strong>for</strong>d Room37 Riley,S.C., Wong,E., Findlay,J.K. and Salamonsen,L.A.The localization ofneutral endopeptidase in <strong>the</strong> ovine uterus during <strong>the</strong> oestrous cycle and earlypregnancyxxi


MONDAY SEPTEMBER 26 (cont)38 Meier,S., Jenkin,G. Trewhella,M.A. and Fairclough,R.J.Changes in endometrial phospholipids andfatty acids during <strong>the</strong> oestrous cycle and early pregnancyin <strong>the</strong> ewe39 Massa,H. and Martin,L.Porcine-relaxin differentially inhibits uterine longitudinal & circular muscles in vivo in oestrous &early pregnant rats40 Sakoff.J.A. and Murdoch, R.N.The period ofuterine receptivity in <strong>the</strong> pseudopregnanct quackenbush special mouse as defined by <strong>the</strong>deciduogenic lectin, concanavalin A41 Shaw,T.J. and Murphy,C.R.Early deciduogenic response of<strong>the</strong> uterine mucosa to <strong>the</strong> lectine concanavalin A and sesame oil42 Hosie,M.J. and Murphy,C.R.Effects of<strong>the</strong> interaction between clomiphene citrate and ovarian hormones on <strong>the</strong> surfaceultrastructure of<strong>the</strong> uterine luminal epi<strong>the</strong>rlial cells43 Terry,V., Murphy,C.R. and Shorey,C.D.Vaginal epi<strong>the</strong>lium ofovariectomised rats treated with combined clomphene citrate and exogenousoestrogen or progesterone44 Kleemann,D.O., Walker,S.K. and Seamark,R.F.Origin ofenhancedfetal growth in sheep administered progesterone during early pregnancy45 Shaw,G., Gehring,H. and Bell,E.C.Prostaglandin production by placental tissues in late pregnant tammar wallabiesMONDAY SEPTEMBER 26 (cont)49 McDougalLS. and Macmillan,K.L.Anovulatory dairy cows have lower LH pulsefrequency and intrafollicular concentrations ofoestradiol (£2) and testosterone (T) than cyclic cows50 Russell,D.L., Robertson, D.M. and Findlay, J.K.Pla.filma FSH elevation in ewes immunised with inhibin aN subunit is attributed to neutralisation (~fcirculating high molecular weight inhibin51 Robinson,S.J., Squires,E.L., Graham,J.K. and Maxwell,W.M.C.Effects ofcalcium ionophore A23I87 and heparin on stallion sperm52 O'Brien,J.K., Ryan,J.P., Waite,K.M., Maxwell,W.M.C. and Evans,G.Energy metabolism ofin vivo and in vitro matured oocytes from pre-pubertal and adult sheep53 Lane,M. and Gardner,D.K.Eagle's essential amino acids stimulate development ofinner cell mass cells ofcultured mouseembryosSession 7 :Chainnan: Assoc. Professor Jock FindlayTime: 1600 - 1715Si:THE GREAT DEBATE: ICSI OR ICNONRob McLachlanChris O'NeillLeeanda WiltonVenue: Ballroom ANon:Robert SeamarkAlan TrounsonJim Cummins46 Shaw,G., Gehring,H. and Bell,E.C.Prostaglandin production by endomentrium in late pregnant tammar wallabies47 Cox,n.B., Kent,J.C., Owens,R.A. and Hartmann,P.E.Mammary morphological andfunctional changes during pregnancy in womenSession 5Chairman: Prof. Alan TrounsonGODING LECTUREProfessor I. HuhtaniemiMolecular aspects of<strong>the</strong> ontogeny of<strong>the</strong> mammalian pituitary-gonadal axisSession 8 :Chairman: Assoc. Professor Bob SeamarkTime: 0900 - 1000TUESDAY SEPTEMBER 27GROWTH FACTORS & THE UTERUSVenue: Ballroom B54 Wa<strong>the</strong>s,D.C., Perks,C.M. and Stevenson,K.R.Insulin-like growthfactor-l (IGF-l) Syn<strong>the</strong>sis in <strong>the</strong> ovine reproductive tract: possible interactionsbetween uterus and ovaryTime: 1200 - 1300 Venue: Ballroom A 55 Cann,C.H., Fairclough,R.J., Sutton,R. and Gow,C.B.Expression ofinsulin-like growthfactor-l mRNA in endometrium during early pregnancy in <strong>the</strong>sheepSession 6 :Chairman: Assoc. Professor Bob SeamarkTime: 1400 - 1530SERONO-ASRB JUNIOR SCIENTIST AWARDVenue: Ballroom A48 HotzeLM.J., Martin,G.B., Walkden-Brown,S.W. and Fisher,J.S.Nutritional effects on testicular growth and LH and FSH secretion in Suffolk and Merino rams in <strong>the</strong>breeding and non-breeding seasons56 Peterson,A.J., Ledgard,A.M., Hodgkinson,S.C. and Tervit,H.R.IGFBP proteolytic activity in <strong>the</strong> uterineflushings ofpregnant and nonpregnant sheep57 Clements,J., Ehrlich,A., Marsh,M. and Salamonsen,L.Prostate-specific antigen and insulin-like growthfactor binding proteins-3 are co-localized in <strong>the</strong>human endomentrium: patterns ofexpression across <strong>the</strong> menstrual cycle58 Robertson,S.A., Seamark,A.C. and Seamark,R.F.GM-CSF mediates <strong>the</strong> post-mating inflammatory reaction in <strong>the</strong> murine uterusxiixiii


TUESDAY SEPTEMBER 27 (cont)81 Jiang,F.-X. and Short,R.V.Characterisation of<strong>the</strong> primordial germ cells in ratfetuses82 Simorangkir,D.R., de Kretser,D.M. and Wre<strong>for</strong>d,N.G.M.Synergistic effect oftransient neonatal hypothyroidism and neonatal hemicastration on testis size,Sertoli cell and spermatid numbers in adult rats83 Harris, M. and Rodger,J.C.The generaion and partial characterisation ofantibodies against marsupial gametes84 Kim,Y.H., Almahbobi,G., Temple Smith,P.D., de Kretser, D.M. and McFarlane, J.R.Characterisation ofmammalian sperm tail proteins using ratfibrous sheath antibody85 Kim, Y.H., Temple Smith,P.D., de Kretser,D.M. and McFarlane, J.R:Isolation and characterisation ofrat sperm tail outer densefibers proteins86 Loveland,K., Zlatic,K., Hayes,T., de Kretser,D.M. and McFarlane,J.MAP2 expression in <strong>the</strong> adult rat testis87 Setchell.B.P., Zupp, J.L., Ekpe,G., Maddocks,S. and Grigg,G.The effect ofenvironmental temperature on scrotal temperature measured by telemetry in rams atpastureTUESDAY SEPTEMBER 27 (cont)95 Waite,K.M. and Ryan,J.P.Metabolism ofenergy substrates by polyspermic mouse embryos96 Pantaleon,M. and Kaye,P.L.Insulin and IGF-I stimulate glucose transport in mouse blastocysts via <strong>the</strong> IGF-I receptor97 Dunglison,G. and Kaye,P.Endocytosis offluid by mouse blastocysts98 Roberts,C. and Breed,B.Marsupial viviparity - shell membrane, implantation and placentation99 Mate,K.E.,Robinson,E.S. and Pedersen,R.A.A·timetable ofin vivo embryonic development in <strong>the</strong> grey short-tailed opossum (Monodelphisdomestical)100 Whitworth,D.J. and Renfree, M.B.Gonadal sex reversal in female tammar wallabies is induced by MIS101 Buaboocha,W. and Gemmell, R.T.Thyroid gland development in <strong>the</strong> brushtail possum, Trichosurus vulpecula88 Young,S., Bradley, M.P., Gidley-Baird, A. and Hinds,L.A.The effects ofmelatonin implantaton ofspermatogenesis in <strong>the</strong> European redfox (Vulpes vulpes)Session 14 :GROWTH FACTORS IN EARLY PREGNANCY89 Murdoch,R.N. and Jones,R.C.Chairman: Professor Alan TrounsonGlucose and acetate utilization by marsupial epididymal spermatozoa is inhibited by a prostaticsecretion Time: 1600 - 1700 Venue: Ballroom B90 Bailey,J.L. and Storey, B.T.Calcium influx into mouse sperm activated by solubilised mouse zonae pellucidae is inhibited by threeinhibitors of<strong>the</strong> zona-induced acrosome reaction: 3-quinuclidinyl benzilate, tyrphostin A48, andpertussis toxin91 Setiadi,D., Sistina,Y. and Rodger,J.C.Acrosomal integrity oftammar wallaby (Macropus eugenii) spermatozoa after treatment withpenetrating (TCEP) and non-penetrating (aSH) reducing agents92 SetiadLD. and Rodger,lC.The effect of2-mercapto-ethanol (2ME) on <strong>the</strong> motility and acrosome oftammar wallaby spermatozoaSession 13 : (Concurrent) :Chairman: Professor Marilyn RenfreeTime: 1400 - 1530EMBRYONIC DEVELOPMENTVenue: Ballroom B93 Mate,K.E., Villar, A.J. and Pedersen,R.A.Relaxation ofRI9 imprinting in mouse androgenetic embryonic stem cells94 Ryan,J.P. and Waite,K.M.Changes in <strong>the</strong> metabolism ofenergy substrates within <strong>the</strong> first cell-cycle ofmouse embryos102 Cavanagh,A.C., Kaye,P.L. and Morton,H.Early pregnancyfactor (EPF): purification to homogeneity from human platelets and identification aschaperonin 10103 O'Neill,C.Platelet activating factor stimulates cell-cycle progression in 2-cell murine embryos104 Marshall.J.T.A., Brownlee,A.G. and Nancarrow,C.D.In vitro amplification ofan ovine oestrus-associated oviductal glycoprotein cDNA105 Boatman,D.E. and Magnoni,G.E.The role ofan estrogen dependent glycoprotein, oviductin, infertilization in <strong>the</strong> golden hamster106 Harvey,M.B.,Arcellana-Panlilio,M., Zhang,X.Q., Edwards,D.R. and Schultz,G.A.Proteinases important in implantation are down-regulated in par<strong>the</strong>nogenetic mouse blastocysts107 Athanasis-Platsis,S., Kaye,P.L., Cavanagh,A.C. and Morton,H.Early pregnancyfactor is required at implantation to optimize embryonic development108 Somodevilla Torres,M., Murphy,R.M., Hillyard,N.C, Cavanagh,A.C. and Morton,H.Passive immunization ofpregnant mice against chaperonin 10 causes loss ofembryonic viability109 Lash,G. and Legge,M.The identification ofearly pregnancy factor in red deer (Cervus elaphus)xvixvii


TUESDAY SEPTEMBER 27 (cont) PLENARY LECTURERS FOR 1994Session 15 :Chainnan: Professor Alan TrounsenTime: 1700 - 1830Session 16WEDNESDAY SEPTEMBER 30MRC LECTUREProf. Bill ThatcherControl and management ofovarian follicles in cattle to optimize fertilityChainnan: Assoc. Professor Jock FindlayTime: 0900 - 1000ANNUAL GENERAL MEETINGVenue: Redlands RoomVenue: Redlands RoomThe Goding Lecturer: Professor IIpo Huhtaniemillpo Huhtaniemi is Professor and Chainnan of<strong>the</strong> Department ofPhysiology, Medical Faculty at<strong>the</strong> University ofTurku, Finland. He completed an M.D. and <strong>the</strong>n a PhD at <strong>the</strong> University ofHelsinki by1974 and <strong>the</strong>n trained as a specialist in chemical pathology in 1979. He was appointed as a Senior Fellowof <strong>the</strong> Academy ofFinland from 1981-83, and was Associate Professor of Clinical Chemistry at <strong>the</strong>University of Helsinki from 1984-85. He was appointed to <strong>the</strong> Chair of Physiology at <strong>the</strong> University ofTurku in 1986. He has spent time in o<strong>the</strong>r laboratories as a postdoctoral fellow in <strong>the</strong> Department ofObstetrics & GYnaecology at <strong>the</strong> University of Cali<strong>for</strong>nia, San Francisco in 1975-76, as a visiting associateat <strong>the</strong> NICHD with <strong>the</strong> National Institutes ofHealth, USA in 1979-80, and was visiting professor at <strong>the</strong>Center of Molecular Biology, University ofHeidelberg, Germany in 1990-91.Professor Huhtaniemi's research interests include <strong>the</strong> ontogeny and regulation of <strong>the</strong> hypothalamicpituitary-gonadalaxis, testicular endocrinology, <strong>the</strong> molecular biology of gonadotrophins andgonadotrophin receptors, and <strong>the</strong> use of transgenic animals.The Goding Lecture <strong>for</strong> 1994 is entitled "Molecular aspects of<strong>the</strong> ontogeny of<strong>the</strong> mammalianpituitary-gonadal axis".Session 17 :Chainnan: Dr. Chis MaxwellTime: 1030 - 1130ARTIFICIAL INSEMINATION & EMBRYO TRANSFERVenue: Redlands Room110 Smith,J.F., Parr,J., Beaumont,S., Oliver,J. and Upreti,G.C.Comparison ofcervical, transcervical and laparoscopic insemination ofewes with chilled stored ramsemen111 Eppleston, J., Pomares,C.C., Stojanov,T. and Maxwell,W.M.C.In-vitro and in-vivo fertility ofliquid-stored goat spermatozoa112 Vishwanath,R., Piu,C. and Shannon,P.A unique sperm agglutinating factor from egg yolk113 Prendergast,E., Vishwanath,R., Shannon,P. and Molan,P.Charged lipoprotein fractions ofegg yolk in bovine semen diluents114 Chairussyuhur, A., Sanchez-Partida, L.G., Zupp, J.L., Maddocks, S. and Setchell B.P.Motility characteristics ofsemen from rams subjected to scrotal insulation, qfter storage at chilledtemperatures <strong>for</strong> one weekThe MRC Lecturer: Professor William W. ThatcherBill Thatcher is Graduate Research Professor in <strong>the</strong> Department ofDairy and Poultry Sciences at <strong>the</strong>University ofFlorida, Gainesville, USA. He completed a B.Sc. and an M.Sc at <strong>the</strong> University ofMaryland, be<strong>for</strong>e undertaking a PhD at Michigan State University (awarded 1968). He was appointedAssistant Professor in Animal Physiology at <strong>the</strong> University ofFlorida in 1969, and <strong>the</strong>n AssociateProfessor be<strong>for</strong>e becoming Professor ofPhysiology in 1978. He was appointed Graduate ResearchProfessor in 1988. He has been <strong>the</strong> recipient of numerous awards including an NIH predoctoralfellowship, a Fulbright-Hayes award and awards from <strong>the</strong> American <strong>Society</strong> ofAnimal Science and <strong>the</strong>American Dairy Science Association. In 1994 he was awarded <strong>the</strong> <strong>Society</strong> <strong>for</strong> <strong>the</strong> Study of ReproductionResearch Award.Professor Thatcher's research interests are focused on elucidating <strong>the</strong> mechanisms by which <strong>the</strong>oestrous cycle is controlled to optimize reproductive efficiency in cattle. Interactions between <strong>the</strong> embryoand <strong>the</strong> uterine endometrium, <strong>the</strong> hormonal regulation of gene expression ofkey enzymes, honnonereceptors, hormones and paracrine regulators in ovarian follicles are being investigated, as is <strong>the</strong> paracrinedialogue between <strong>the</strong> trophectodenn and uterine endometrial epi<strong>the</strong>lium.The MRC Lecture <strong>for</strong> 1994 is entitled "Control and management of ovarian follicles in cattle tooptimize fertility".115 UI?r~ti,G.C., Jensen,K., Munday,R., Vishwanath,R. and Smith,J.F.Studles on ram spermatozoal aromatic amino acid oxidase116 Stojanov,T., Rhodes,S.L., Maxwell,W.M.C. and Evans,G.The effect ofantioxidants on <strong>the</strong> pregnancy rate after insemination with liquid stored ram spermatozoa117 Lehrer,A.R. and McMillan, W.H.Is peri-oestrous intravaginal impedance an aid in screening <strong>for</strong> prospective pregnant recipient cows?xviiixix


lISIrI!IIIrti ar i alistsMaria HotzelFaculty of Agriculture,UWA, Nedlands, WASimon RobinsonDepartment of AnimalScience, Sydney, NSWIIEScott McDougallDairy Research Corporation,Hamilton, New ZealandJustine O'BrienDepartment of Animal Science,Uni. Sydney, NSWDarryl RussellPrince Henry's Institute ofMedical Research, Melbourne, VICMichelle LaneInstitute of Reproduction andDevelopment, Monash Uni., Clayton, VICxx


NPY EFFECTS ON LH SECRETION IN THE OVARIECTOMISED EWE:Y2 RECEPTOR. INVOLVEMENT, SEASONAL VARIATION AND SITE OF ACTION WITHIN THEHYPOTHALAMUS.Mandi L. Barker-Gibb 1, Jaroslav H. Boublik 2 and lain J. Clarke 11Prince Henry's Institute of Medical Research, P.O. Box 5152, Clayton, Victoria 3168; 2Baker Medical ResearchInstitute, P.O. Box 348 Prahran, Victoria 3181.INTRODUCTIONNeuropeptide Y (NPY) action is mediated throughY1(post-synaptic) and Y2 (pre-synaptic) receptor subtypes.NPY plays a key role in modulating LHsecretion in many species, although <strong>the</strong> extent towhich this involves action through <strong>the</strong> differentreceptor sub-types is not clear. In rats, LH secretionappears to be controlled through activation of <strong>the</strong> Y1receptor, and NPY acts at both <strong>the</strong> hypothalamic andpituitary level (1). In contrast, NPY does not act on<strong>the</strong> pituitary gland in sheep (2). We haveinvestigated possible hypothalamic mechanisms bywhich NPY modulates GnRH/LH release in ewes.Intracerebroventricular (i.c.v.) injections of NPY1-36which acts at <strong>the</strong> Y1 and Y2 receptors and a specificY2 receptor agonist NPY18-36 were given to identifyreceptor specific actions on LH pulse secretion.Microinjections (1 Ill) of NPY were also given into <strong>the</strong>preoptic area (POA) to examine effects at <strong>the</strong> level of<strong>the</strong> GnRH cell bodies. The ewe is also a highlyseasonal species and <strong>the</strong> activity of <strong>the</strong> GnRH 'pulsegenerator' is directly influenced by changes inphotoperiod which occur with season. The effect ofphotoperiod on <strong>the</strong> GnRH system is mediated via anunidentified neuronal circuit. Thus, we alsoinvestigated seasonal variations in <strong>the</strong> effects of NPYon LH secretion by giving NPY1-36 into <strong>the</strong> thirdventricle (3V) during both <strong>the</strong> breeding (BS) and nonbreedingseason (NBS).METHODSOvariectomised (OVX) ewes were fitted with guidetubes into a lateral ventricle (LV), 3V or POA.Jugular venous blood samples were taken every 10min <strong>for</strong> 3h. During <strong>the</strong> SS, sheep were given 10lJgNPY1-36, 100lJg NPY18-36 or saline into <strong>the</strong>LV;101l9,100lJg NPY1-36, and 100119 NPY 18-36 orsaline into <strong>the</strong> 3V, and 1.5lJg NPY into <strong>the</strong> POA.Injections of NPY were also given into <strong>the</strong> 3V (100lJg)and POA (1,5, 10lJg) during <strong>the</strong> NBS to examineseasonal effects Sampling continued <strong>for</strong> 3h postinjection,and samples were assayed <strong>for</strong> LH. Pre- andpost- injection differences were analysed byStudent's paired t-test.RESULTSInjections of 10 (LV) or 100lJg NPY1-36 (3V) had littleeffect on LH secretion in OVX ewes during <strong>the</strong> B.S.except to extend (P5 400::r:C!J 200;--0- PACAP 5 min-+- PACAP 30 min-0- VIP5min-tI- VIP 30 min--0- Control____ PACAP 16 7 M__ VIP16 7 MPACAP 16 6 MVIP 16 6 Mo+---r---,--_r_----.---...,.--r-----.a 234 5 6 7Incubation time (h)80 -0--+-~ --­~ 60§8 40~.s::i 20o0-t---,---r---,----.----,------.----,o ~ ~ 00 ~ 100 1~ 1~Incubation time (min)Fig.1 Time-course relationship <strong>for</strong> PACAP and VIP on intracellular cAMPDose (M)Fig. 2Dose-response curve <strong>for</strong> PACAP and VIPFig. 3Time-course relationship <strong>for</strong> <strong>the</strong> effects of PACAP and VIP on GHsecretion from sheep pituitary cellso+--,---,----,r-~-r-_r_---r-..--~__r_~---.a 2 4 6 8 10 12 14 16 18 20 22 24Incubation time (h)Fig. 4Time-course relationship <strong>for</strong> <strong>the</strong> effects of PACAP and VIPon LHsecretion from sheep pituitary cells1 2


Physiological and pharmacological challenges to prolactin secretion inPteropus species (flying foxes)G.M. O'Brien, J.D. Curlewis & L. MartinDept Physiology & Pharmacology, University of Queensland, Q4072.INTRODUCTIONPlasma prolactin in female flying foxes(Megachiroptera: Pteropus po!iocepha!us & P.scapu!atus) was examined after removal ofsuckling young, and after a dopamine agonist,bromocriptine, or an antagonist, domperidone.METHODS50pL aliquots of plasma were assayed intriplicate, using a heterologous double antibodyRIA (1, 2).RESULTS - SUCKLING".........52-J 4E0)3c............2---l~0-I00-2 -1RESULTS - BROMOCRIPTINE2.5--..~ 2.0E0) 1.5c............-lCt:0..I01.00.51-0.0-1 0• T A Individuals012Time (h)Fig. 1. In 3 lactating P. scapu!atus, little redflying foxes, plasma prolactin was3.01 ±O.74ng mL- 1 (mean±s.e.). Young wereremoved, and after 2h prolactin wassuppressed (1.24± 0.02ng mL- 1 p < 0.05). 5­30min after resumption of suckling, prolactinwas 1.25 ± 0.18ng mL- 1 (p > 0.05) and had notrecovered after 4h.2Time (h)Fig. 2. Early-mid pregnant P. poliocephalus,34354grey-headed flying foxes, had similar initialprolactin levels in test & control groups(1.40 ± 0.33ng mL-' and 1.31 ± O.19ng mL- 1p > 0.05). In vehicle controls prolactin wasunchanged after 2h (p > 0.05) and suppressedin <strong>the</strong> group treated with bromocriptine(0.72 ± 0.19ng mL-' p < 0.05); response wasmaintained at 4h (p > 0.05).RESULTS - DOMPERIDONE: 10I---lE 80)--S 6---l 4~a...b 212 ,..-----r----r--~---r-----r--...,A·•• A • T Domperidoneo ~ 0 \7 Vehicle0'-----'---""-----1.---'----1.__-'-1 o2 3 4 5Time (h)Fig. 3. Prolactin was 2.03 ± 0.22ng mL- 1 &1.35 ± 0.20ng mL-' (p > 0.05) in 2 groups ofpregnant P. poliocepha!us. After Domperidone,prolactin increased to 7.56 ± 1.63ng mL-' by20min (p < 0.05, n = 3: no response in 4thanimal) with no fur<strong>the</strong>r change by 60min(p > 0.05); vehicle controls remained constant(p > 0.05).CONCLUSIONSSuppression of prolactin by removal of <strong>the</strong>suckling stimulus or by bromocriptine, and itsstimulation by domperidone confirm that <strong>the</strong>sensitive heterologous assay is measuringPteropus prolactin. The pharmacologicalresponses indicate that, as in o<strong>the</strong>r species,prolactin secretion is under dopaminergicregulation in flying foxes.REFERENCES1. O'Brien, G.M., Curlewis, J.D. & Martin, L.(1990) Proc Aust. Physio!. Pharmacol. Soc.2..1:65P.2. McNeilly, A.S. & Friesen, H.G. (1978)Endocrinology 102:1539-1547.LOCALISATION OF INHIBIN AND RELATED PEPTIDES IN THE NEONATAL RAT OVARY:POTENTIAL MEDIATORS OF GONADOTROPHIN SENSITIVITY. Ann Drummond, Mitzi Dyson, Julie Mercer and Jock FindlayPrince Henris Institute of Medical Research, Clayton, Victoria 3168, AustraliaIntroductionThe rat ovary at birth is insensitive to gonadotrophins andremains so until approximately day 7 (1), suggesting thatspecific functional receptors are not present during thistime. The acquisition of FSH receptors by preantralfollicles is independent of gonadotrophin stimulation andmay involve <strong>the</strong> inhibin-related peptides. We postulatethat activin is responsible <strong>for</strong> stimulating FSH receptorsin vivo, given that this phenomenon occurs in vitro (2).To investigate this hypo<strong>the</strong>sis, <strong>the</strong> presence of mRNAarid protein <strong>for</strong> <strong>the</strong> a and ~A inhibin/activin subunits in<strong>the</strong> neonatal rat ovary, at times pertinent togonadotrophin sensitivity, were examined. The suitabilityof an in vitro neonatal ovarian cell bioassay <strong>for</strong> testing<strong>the</strong> effects of <strong>the</strong>se peptides on steroidogenesis wasalso assessed.MethodsFrozen sections of ovary from 21 day old PMSG-treatedand neonatal rats were used <strong>for</strong> immunohistochemistryand in situ hybridisation. Digoxygenin-Iabelled riboprobes<strong>for</strong> inhibin-a and inhibin-~A were used to localise mRNAin ovarian tissue. Immunohistochemical localisation ofinhibin/activin subunits, was per<strong>for</strong>med using threepolyclonal antisera raised against syn<strong>the</strong>tic peptidefragments of <strong>the</strong> inhibin/activin a, ~A and ~B subunits(3). For cell bioassays, neonatal rat ovaries werecollagenase/Dnase dispersed and <strong>the</strong> cells cultured in<strong>the</strong> presence or absence of FSH and MIX <strong>for</strong> 48 hours.Conditioned media was assayed <strong>for</strong> androstenedione,progesterone and inhibin by RIA.ResultsThe a, ~A and ~B proteins were present in neonatalovaries (Table 1), but only a could be consistentlydetected in PMSG ovaries, by immunohistochemistry.Granulosa cells, or in less differentiated ovaries, follicularcells, expressed <strong>the</strong>se proteins, whereas ~A was <strong>the</strong>only subunit present in oocytes. Both inhibin-a andinhibin-~A mRNAs were expressed by granulosa cells ofPMSG and neonatal ovaries, as early as day 4. Notevery follicle expressed <strong>the</strong> mRNAs at any time. FSH in<strong>the</strong> presence of MIX, stimulated androstenedione andprogesterone production by neonatal ovarian cells inculture (Fig 1). By day 8, androstenedione production ismore prominent than progesterone. Inhibin productionwas stimulated by FSH (Fig 2), in <strong>the</strong> absence of MIX,but only at day 8.References1. Funkenstein, B., Nimrod, A. and Lindner, H.R. (19aO) Endocrinology106: 9a-106.2. Xiao, S., Robertson, D.M. and Findlay, J.K. (1993) Endocrinology131: 1009-1016.3. Gurusinghe, C.J., Healy, D.L., Jobling, T., Mamers, P. and Burger,H.G. (1994) Gynaecologic Oncology (SUbmitted)Supported by <strong>the</strong> NHMRC of AustraliaFigure 1: Dispersed Neonatal Ovary Culture1816 • progesterone14 & androstenedione12- 10.§g> 8642O~-ccC 10 20 C 1 10 20Day4DaySAll treatments with MIX ± FSH (1-20 ng/ml)a p,0.01 b p


IMMUNIZATION OF MERINO LAMBS. EFFECTS ON INHIBIN BINDING ANDGONADOTROPHIN RESPONSE TO OVARIECTOMYT. O'Shea], S. T. Anderson] and M. A. Hillard 2IDepartment ofPhysiology, University ofNew England, Annidale, NSW 2351, and 2CSIRO, Division ofAnimal Production, Armidale, NSW 2350STUDIES IN SHEEP EXPLORING PLASMA FOLLISTATIN ELEVATIONS DUE TO FREQUENTBLOOD SAMPLING ORSURGERYB Klein 1 , I.J. Clarke and D.M. Robertson1Department of Anatomy and Physiology, RMIT, Bundoora, 3083 and Prince Henry's Institute of Medical Research, P.O. Box 152, Clayton, 3168, AustraliaINTRODUCTIONImmunization of lambs with inhibin preparationsresults in pubertal advancement (1,2), withincreased ovulation rate and testicular growth ratebut no changes in gonadotrophin concentrations(1,3). Fur<strong>the</strong>r studies on Merino sheep immunizedat 3, 7 and 15 weeks of age against a syn<strong>the</strong>ticpeptide fragment (porcine alpha inhibin 1-32,Peninsular Labs., Belmont, Cali<strong>for</strong>nia) conjugatedto albumin (1,3) are reported.MATERIALS AND METHODSThe ewes were ovariectomized at 32 months, onemonth after an additional immunization. Bindingof iodinated native bovine inhibin (supplied by Dr.D. M. Robertson, Monash University, Melbourne)was determined on plasma diluted 1:500. Plasmagonadotrophin concentrations after ovariectomywere analysed by AOV with repeated measures.RESULTSNo binding was seen in <strong>the</strong> control animals, exceptat age 3 weeks by passive transfer from someimmunized mo<strong>the</strong>rs. In one ram passive transferappeared to interfere with active immunization anddata from this ram have been excluded. In peptideimmunized animals plasma binding was seen inboth sexes after <strong>the</strong> first booster immunization,reached a maximum at 24 weeks, and haddecreased at 28 months (Table 1).In ewes itrapidly increased after reimmunization at 31months but <strong>the</strong>n decreased again (29.6 ± 1.11% at32 months; 29.2 ± 0.89% at 33 months; 17.9 ±1.84% at 50 months).Binding of inhibin was less in ram plasma than inewe plasma at all ages (P < 0.01). The rate ofdecline in plasma binding was <strong>the</strong> same inovariectomized ewes (0.65% per month) as inentire ewes (0.62% per month), showing that <strong>the</strong>presence of endogenous ovarian inhibin did notcause a persistent titre.One ewe was undergoing a gonadotropin surge atovariectomy and data from this ewe have beenexcluded. During 8 days after ovariectomy plasmaFSH concentrations were greater (P < 0.006) inewes ovariectomized in <strong>the</strong> luteal phase (4.02 ±0.18 ng Iml) than in <strong>the</strong> follicular phase (3.50 ±0.17), and peptide immunized ewes showed aslower response (P < 0.002) to ovariectomy thancontrol ewes (Fig. 1).Plasma LH concentrations.showed a significant interaction (p < 0.04), suc<strong>hth</strong>at LH concentrations in ewes ovariectomized inluteal phase were greater than those of ewesovariectomized in follicular phase in controlanimals (2.20 ± 0.24 v. 1.66 ± 0.16) but not inpeptide immunized ewes (1.07 ± 0.12 v. 1.16 ±0.13).At 32 days after castration nei<strong>the</strong>r FSH (8.75 ±DAD ng/ml) nor LH (4.51 ± 0.34 ng/ml) showedany residual effect from immunization or phase ofcycle at ovariectomy.DISCUSSION/SUMMARY1) The presence of endogenous ovarian inhibindoes not affect persistence of antibody titres toa syn<strong>the</strong>tic alpha - inhibin peptide.2) The phase of <strong>the</strong> oestrous cycle at ovariectomyaffects <strong>the</strong> subsequent gonadotropin response.3) The delay in <strong>the</strong> post-castration rise in plasmaFSH in peptide immunized ewes is possiblydue to a longer retention of biologically activeinhibin in plasma by antibodies.Table 1: Binding of native bovine inhibin byplasma (diluted 1:500) of Merino sheepimmunized with porcine alpha 1-32 peptide.Results are expressed as percentage cpm bound tototal cpm added. Values are means ± SE <strong>for</strong> 12females and 11 males.AnimalsAge (weeks)8.4 15 16.4 24 32 121Female 6.6% 14.8% 24.0% 24.5% 21.4% 9.2%(0.95) (1.31) (1.57) (1.78) (1.78) (1.17)Males 2.8% 5.5% 10.1% 10.7% 8.2% 2.2%87-;;-E 6rI.l.$ I=l-.5~ 4 -5-:I:f£ 3210(1.06) (1.09) (1.08) (2.32) (1.93) (0.39)**n = 6........•......... ..LT .........••••••••• ..10.•...................48144Fig. 1: FSH after ovariectomy of controls (D) andewes immunized with peptide (. ).96HOURS AFTER OVARIECI'OMYT192REFERENCES1. O'Shea, T. et al. (1989) J. Reprod. Ferti!. Abstr.Series 1-.: Abstr. 6.2. O'Shea, T. et al. (1993) Reprod. Fertil. Dev. ~:173 - 180.3. Anderson, S. T. et aL (1990) Proc. Aust. Soc.Reprod. BioL 22: 133.INTRODUCTIONAprotein hormone termed FSH-suppressing protein (FSP) (Robertson ataI., 1987) or follistatin (Ueno et aJ., 1987) has been isolated from ovarianfollicular fluid which is functionally similar to inhibin in suppressing FSH.In a previous study (Klein et a/., 1993) aimed at exploring <strong>the</strong>physiological role of FSP in plasma, FSP levels were observed to riseacutely during periods of frequent blood collection. This rise in plasmaFSP coincided with <strong>the</strong> time during which animals underwent surgery andperiods in which an increase in bleeding frequency had occurred.The aims of this study were to investigate <strong>the</strong> cause of <strong>the</strong> rapid risein plasma FSP levels by (1) examining <strong>the</strong> effects of <strong>the</strong> anticoagulan<strong>the</strong>parin used during blood sampling; (2) determining whe<strong>the</strong>r frequentbleeding per se acts as a stimulus <strong>for</strong> FSP elevations and (3) and todetermine whe<strong>the</strong>r a rise in plasma FSP seen 10-12 hours after surgerywas due to a concomitant rise in <strong>the</strong> acute phase protein a-2macroglobulin (a-2M) shown to-bind FSP (Vaughan et al., 1993).MATERIALS AND METHODSIn <strong>the</strong> first study eight Corriedale ewes were bled via indwelling venousca<strong>the</strong>ters at hourly intervals <strong>for</strong> 3h, <strong>the</strong>n at ten minute intervals <strong>for</strong> 2h,followed by two samples taken at hourly intervals. The ca<strong>the</strong>ters wereflushed with heparinised (50IU/ml) saline and <strong>the</strong> collection vials wereheparinised. At <strong>the</strong> 4 h point (ha~ way through <strong>the</strong> phase of rapidsampling), 4 sheep received a large intravenous injection of heparin(3200IU/1.5ml) and control sheep received an equal volume of saNne. In<strong>the</strong> second study, 4 ewes were sampled (via jugular ca<strong>the</strong>ters) at hourlyintervals <strong>for</strong> 4h, <strong>the</strong>n at ten minute intervals <strong>for</strong> 2h followed by <strong>the</strong> takingof 3 samples at hourly intervals using <strong>the</strong> anticoagulant citrate glucosedextrose instead of heparin. A fur<strong>the</strong>r 4 ewes acting as controls weresampled at hourly intervals. In <strong>the</strong> third study, samples were taken athourly intervals from 8 ewes <strong>for</strong> 27h, and <strong>the</strong> sheep were <strong>the</strong>nlaparotomised and hOUrly samples were taken 10-12h later. Plasmasamples were collected <strong>for</strong> subsequent determination of a-2M byimmunoprecipitation and FSP by a radioimmunoassay method developed<strong>for</strong> sheep plasma (Klein et aI., 1993).RESULTSPlasma FSP levels were observed to rise acutely (1)


Follistatin Suppresses FSH In Castrated RamsA.J. Ti/brook 1 , !.J. C/arke 2 and D.M. de Kretse,31Department of Physiology and 31nstitute of Reproduction and Development, M.ona~h2prince Henry's Institute of Medical Research, Clayton, 3168, VictoriaUniversity andREGULATION OF OVARIAN ACTIVIN RECEPTOR TYPE II EXPRESSION ACROSS THERAT OESTROUS CYCLESonia Garcia, Jock K. Findlay and Julie E. MercerPrince Henry1s, Institute of Medical Research, PO Box 152, Clayton, Vic. 3168.INTRODUCTIONFollistatin is structurally distinct from inhibin and has beenshown to suppress <strong>the</strong> release of follicle stimulatinghormone (FSH) by pituitary cells in vitro (1). We found thattreatment of castrated rams with a 50pg Lv. injection and800j./g infusion of human recombinant inhibin A (hr-inhibin)over 12h suppressed plasma concentrations of FSH to <strong>the</strong>levels found in intact rams (2). There have been no studiesin sheep to determine if follistatin will also suppressplasma FSH in vivo. There<strong>for</strong>e, we tested <strong>the</strong> hypo<strong>the</strong>sisthat follistatin suppresses <strong>the</strong> plasma concentrations ofFSH in castrated rams. The effect on plasma luteinizinghormone (LH) was also investigated.MATERIALS AND METHODSEight rams, castrated within <strong>the</strong> first 3 weeks of birth,were allocated to 2 groups of 4. One group was given a50j./g Lv. injection followed by an 800pg Lv. infusion ofhuman recombinant follistatin (hr-F) over 12h. The controlgroup was given an Lv. injection and 12h Lv. infusion ofvehicle. Jugular venous samples were collected <strong>for</strong> 3hbe<strong>for</strong>e and <strong>for</strong> up to 132h after treatment. FSH and LHwere measured by radioimmunoassay.RESULTSInfusion of castrated rams with hr-F significantly (P.. I CToSample A2 3 4 5CJ)Sample 82 3 4 5_o:::::l!!!!ilI!I•••S. AetRII eDNAr:::::.::::::::::::::::::::::::::::::::::::::::::J' CTCJ) CJ) (J)::J ::J ::::>::Ja: a: a: c::~~~CJ) CJ) I-CJ) (J)wWW ill0C50a: a:00- 0->- W...Ia:~« ...Iw~AetRII eDNA..(458 bp CT..( 408 bp AetRIIeDNAFig 1 (A) Schematic representation of amplified 408 bp region ofActR11 cDNA and of 458 bp competitive template (CT). Solidregion in CT represents a 50 bp oligonucleotide subcloned into aNarl site. (B) Titration curve of PCR products containing a fixedamount of ActRII cDNA and increasing amounts of CT DNA.Fig.2 Relative levels of ActR11 mRNA in rat ovaries across <strong>the</strong>oestrous cycle. * p< 0.05, different from oestrus.Summary and ConclusionThis increase in ActR11 mRNA levels occurs across<strong>the</strong> period when <strong>the</strong> dominant follicle is developingand persists into, and indeed is maximal at oestrus,when ovulation occurs, consistent with a role <strong>for</strong>activin in development of <strong>the</strong> follicle. These dataprovide fur<strong>the</strong>r evidence <strong>for</strong> <strong>the</strong> important role ofactivin as a local regulator of reproductive function in<strong>the</strong> ovary.7 8


-GLUCOSE AND CHOLESTEROL UPTAKE BY THE OVARY OF THE SHEEPA.R.Rabiee, I.J. Lean, J.M. Gooden and B.G. MillerDepartment of Animal Science, University of Sydney, Camden NSW 2570INTRODUCTIONThe dramatic cyclic changes in growth and steroid and protein output of <strong>the</strong> ovary suggest that <strong>the</strong>ovary is a highly metabolically active organ. Knowledge of ovarian affinity and demand <strong>for</strong> energysubstrates used <strong>for</strong> steroid and protein hormone production should lead to improved understandingof ovarian nutrient requirements to ensure successful reproductive per<strong>for</strong>mance. Also important is <strong>the</strong>study of relationships between uptake of different variables because factors influencing ovarianuptake of nutrients may be identified. We have previously found that <strong>the</strong> small ovarian mass of sheepmay limit our ability to detect small, but significant uptake of metabolites, glucose uptake wassignificant <strong>for</strong> all sheep pooled (P


THE ROLE OF STRESS IN BOAR-INDUCED PUBERTY IN GILTSA.I. Turner 1 • 2 , P.H. Hemsworth 1 , H. Lohuis 1 and A.J. TilbraalCIVIAS, Dept Agriculture and 2Dept Physiology, Monash University, Victoria.IntroductionIt is well accepted that exposure of pre-pubertal gilts toboars stimulates puberty in gilts. This stimulation ofpuberty may be due to stimuli from <strong>the</strong> boar, primarilypheromones (1), and/or an acute stress response (2). Therelative importance of <strong>the</strong>se factors is unknown. In thisexperiment we tested <strong>the</strong> hypo<strong>the</strong>sis that stress plays amajor role in boar-induced puberty in gilts.Materials and MethodsWe used a behavioural approach to reduce <strong>the</strong> stressresponse when gilts are exposed to boars. Frequentexposure of pre-pubertal gilts to boars in an arena mayallow habituation to boars and to <strong>the</strong> arena in which thisexposure occurs. Thus, <strong>the</strong> degree of novelty of thisexposure <strong>for</strong> <strong>the</strong>se gilts should decrease over time andconsequently <strong>the</strong> gilts should experience a lesser stressresponse than gilts inexperienced with this exposure.Evidence of habituation to this exposure should bereflected in a reduced level of exploration (activity) by<strong>the</strong> gilts. We assessed <strong>the</strong> role of an acute stressresponse in boar-induced puberty by comparing <strong>the</strong>effects of boar exposure at 170 days of age on gilts thathad previously been exposed frequently to boars in anarena with gilts which had not received this previousexperience.Twenty four gilts were allocated to <strong>the</strong> following 4treatments (N::;::6). Controls: gilts were introduced from130 days of age <strong>for</strong> 1h daily to an empty pen adjacent to2 pens containing ovariectomised females; DMA 13o : giltswere introduced from 130 days of age <strong>for</strong> 1h daily to adetection mating arena (DMA) that allowed auditory,olfactory and only limited physical contact with boarsthrough <strong>the</strong> arena walls; DMA 17O : gilts were introducedto <strong>the</strong> DMA <strong>for</strong> 1h daily from 170 days of age; INTRO:gilts were introduced to a boar in his own pen <strong>for</strong> 15mindaily from 170 days of age. From 170 days of age <strong>the</strong>standing response to pressure on <strong>the</strong> back of <strong>the</strong> gilt wasused daily to determine <strong>the</strong> occurrence of puberty.Ovaries were examined at slaughter to confirmattainment of puberty. With <strong>the</strong> exception of <strong>the</strong> INTROgroup, <strong>the</strong> level of activity (locomotion) of <strong>the</strong> gilts wasmeasured during treatment by recording movementacross a grid marked on <strong>the</strong> floor of <strong>the</strong> arena.ResultsThe mean (±SEM) age of puberty (Fig 1) did not differbetween <strong>the</strong> control and DMA I30 treatments (P>0.05). Incontrast, <strong>the</strong> DMA 170 (P


PROGESTERONE TREATMENT OF EWES REDUCED CORPUS LUTEUM WEIGHTAND OFSfRADIOL TREATMENT REDUCED OVUI.ATION RAlEPatrick J. Wright. C. Philip and M. HariadiDepartment of Veterinary Sciences, University of Melbourne, Werribee 3030OESTRADIOL INCREASES AMOUNTS OF mRNA FORGONADOTROPHIN-RELEASING HORMONE RECEPTORS IN SHEEPD.L. HamernilC, C.M. Clay2, E.L. VanKirk!, G.E. MostIDept. Veterinary & Biomedical Science, University of Nebraska, Lincoln, NE; USA.2Dept. Physiology, Colorado State University, Ft. Collins, CO; USA.3Dept. Animal Science, University of Wyoming, Laramie, WY; USA.INIRODUCTIONWe have assessed i) <strong>the</strong> effect of exogenousprogesterone on corpus luteum (CL) weight (todetermine whe<strong>the</strong>r <strong>the</strong> reduction of CL weightsin ewes of reduced nutrient stat~ (1) could bedue to increasing plasma' progesteroneconcentration reflecting reduced metabolicclearance rate (2», and ii) <strong>the</strong> effect ofexogenous oestradiol on ovulation rate (todetermine whe<strong>the</strong>r ovulation rate could bemodulated by an inhibitoryeffect of oestradiol).MATERIAL AND METIiODSIn Study 1 mature Merino (M) ewes (n=20,body condition score 1.5-2) receivedprostaglandin (PG; Cloprostenol, 125 ug im)each 14 days on 4 occasions to synchroniseoestrous cycles. Ten of <strong>the</strong>se ewes were treatedwith progesterone <strong>for</strong>. 4 cycles from Day 6-14(Day 0 = day of PG treatment) (group Mp).Pessaries (CIDR-G) were inserted on Day .6,replaced on Day 10 and removed on Day 14/0.The remaining 10 PG-treated ewes wereuntreated controls (Group Me). On .Day 14 of<strong>the</strong> 4th progesterone-treatment cycle <strong>the</strong> eweswere ovariectomized and CLs counted andweighed.In Study 2 mature Border Leicester (BL) ewes(n=22, body condition score 3-4) were treatedwith an oestradiol implant (3.5 em sc; groupBLo, n=11), or were untreated controls (groupBLc, n=11). The occurrence of oestrus (crayonmarks from vasectomised rams) was observedover 5 weeks. The ewes were <strong>the</strong>n treated twicewith PG 14 days apart to synchronise oestrouscycles, ovariectomized 14 days later and <strong>the</strong>corpora lutea counted and weighed.RESULTSIn Study 1, in <strong>the</strong> single ovulators (9 ewes ineach treatment group), progesterone-treatedewes had lighter CL's (367 (38), mean (sem»than did control ewes (489.3 (24.3» (P=0.02,Fig. 1).In StUdy 2, oestrogen treatment did not affectoestrous cyclicity, but did result in a lowerovulation rate (1.8 (0.2» than in control ewes(2.45 (0.16» (P=O.04, Fig. 2).DISCUSSION/SUMMARY/CONCLUSIONSThe results indicate that plasma concentrationsof progesterone and oestradiol may be factorsinfluencing corpus luteum weight and ovulationrate in ewes undergoing normal oestrous cycles.Ṣ5500:i:'ij '" ...5031E:JII-4OD2" :Je- 3500u.s~c:~ c""5>o550 P = 0.023002.700ControlContlolTreatmentlrealll1entProgesteroneFig. 1: Corpus luteum weights (mean, sem) inprogesterone-treated (Mp, n=9) and in control(Me, n=9) ewes.OestradiolP = 0.041.700L__~lIgll_~mm~ -Fig. 2: Ovulation rate in oestradiol-treated(BLo, n=11) and in control (BLc, n=ll) ewes.REFERENCES1. Wright, PJ, Pantke, P and Clarke IJ (1991)Proc. AS.R.B. abstr 82.2 Parr, RA (1992) Reprod. Fertil. Dev. 4, 297­300.INTRODUCTIONGonadotrophin-releasing hormone (GnRH)stimulates syn<strong>the</strong>sis and secretion ofgonadotrophins in mammals by binding tomembrane-bound receptors on gonadotrophs.Recently, cDNA encoding <strong>the</strong> GnRH receptor(GnRH-R) were cloned from several species,including sheep (1). In sheep, amounts of mRNA<strong>for</strong> GnRH-R increased prior to <strong>the</strong> preovulatorysurge of LH (1). Oestradiol (E), alone or incombination with inhibin also increased amountsof GnRH-R mRNA in sheep pituitaries in vitro(2). This study was conducted to test <strong>the</strong>hypo<strong>the</strong>sis that E increases amounts of mRNA <strong>for</strong>GnRH-R during <strong>the</strong> periovulatory period in ewes.MATERIALS AND METHODSPituitarieswere collected from five ewes on d14 of<strong>the</strong> oestrous cycle (dO=oestrus). Ano<strong>the</strong>r 20 eweswere ovariectomised (OVX) on d14 of <strong>the</strong>oestrous cycle, and E implants were inserted in 5OVX ewes (OVXE) to mimic low concentrationsof E during <strong>the</strong> early follicular phase of <strong>the</strong>oestrous cycle. Sixteenhours after OVX, pituitarieswere collected from 5 OVX and 5 OVXE ewes,and <strong>the</strong> remaining 10 OVX ewes received anintramuscular injection of E (25JLg) to induce apreovulatory-like surge of LH (OVX+E).Pituitaries were collected from OVX+ E ewes 18or 54 hr after injection of E (n=5/group). Bloodsamples were collected at hourly intervals followinginjection of E <strong>for</strong> measurement of LH. Number ofGnRH-R were quantitated in half of eachpituitary. Polyadenylated RNA was isolated fromhalf of each pituitary, applied to Nor<strong>the</strong>rn and slotblots, and hybridised with a radioactive cDNAprobe encoding <strong>the</strong> ovine GnRH-R. Hybridisationsignals were quantitated in arbitrary densitometricunits.RESULTSInjection of E induced a preovulatory-like surge ofLH within approximately 18 hr in OVX ewes.Nor<strong>the</strong>rn blot analysis revealed GnRH-Rtranscripts of - 5.6, 3.8, 2.1, 1.3, and 0.8 kbp. Eimplants increased amounts of mRNA <strong>for</strong> GnRH-R at 16 hr in OVXE ewes compared to <strong>the</strong>amount of GnRH-R mRNA at d14 of <strong>the</strong> oestrouscycle or at'16 hr in OVX ewes (Table 1). Amountsof GnRH-R mRNA in OVX ewes were alsoelevated (p < .05) 18 hr after E injection comparedto 0 or 54 hr after E (Table 1). Number of GnRH­R were elevated (p


PRODUCTION OF IN VNO MATURED OOCYTES FROM 6 WEEK OID CALVESC.R. Earll, B.J. Irvinel, J.P. Rowel, P.Kotaras 2 , and D.T. Armstrong 21 Strnan Research Centre, S.A Research & Development Institute, Naracoorte, Australia 2Department of Obstetrics and Gynaecology, University of Adelaide, Australia1N1RODUCTIONIt has recently been demonstrated that viableembryos can be produced from young lambsand calves using in vitro techniques onoocytes collected from follicles stimulated bya combination of FSH and PMSG followedby an injection of GnRH (1, 2). Preliminarydata obtained from 6 week old ewe lambs (1)suggests that <strong>the</strong> development rate offoetuses from oocytes is greater from in vivothan in vitro matured oocytes (3). Oocytes atcollection were classified into two groups:mature, those with expanded cumulus cells;immature, those with tight, intact cumulusinvestments. For <strong>the</strong>se experiments with <strong>the</strong>young lambs, mature oocytes at collectionwere submitted directly to IVF whileimmature oocytes underwent IVM cultureprior to IVF. The young lamb differs from<strong>the</strong> calf in that it appears much easier toobtain in vivo matured oocytes from <strong>the</strong> lambthan <strong>the</strong> calf.The current study <strong>the</strong>re<strong>for</strong>e investigatedthree different in vivo maturation inducingtreatments in an attempt to increase <strong>the</strong>percentage of matllre oocytes obtained fromcalves at aspiration.MATERIAlS AND METHODSThe stimulation treatment employed was acombination of 200mg of FSH s.c.(Folltropin, Vetrepharm, Canada) and 400J.U. PMSG Lm. (Folligon, Intervet, Holland).Progesterone sponges were inserted 5 daysprior to <strong>the</strong> stimulatory injections andremoved 48 hours later. The 3 maturationinducing treatments were; Treatment 1, 500J.Lg GnRH Lm.; Treatment 2, 40 mg FSH Lv.with 10 mg LH Lv. (Lutropin, Vetrepharm,.Canada); Treatment 3, 1500 J.U. HCG i.v.and 1500 J.U. Lm. (Chorulon, Intervet,Holland). The maturation treatments weregiven 24 hours post sponge pull. Oocyteswere aspirated 24 hours post maturationtreatment and scored <strong>for</strong> maturation status.RESULTSTreatmentFolliclesAspiratedOocytes 6%Recovered Mature1 (n=8) 35.9 ± 23.1 17.1 ± 10.9 43%2 (n=7) 56.5 ± 34.8 31.9 ± 23.5 69%3 (n=8) 23.9 ± 21 10.9 ± 8.9 82%The between calf variation in follicle numbersand oocytes collected was large so that wit<strong>hth</strong>e small number of calves in each treatment(n=8) <strong>the</strong> differences in follicle numbersbetween treatments were not significant.Treatment 3 produced a greater percentageof oocytes that were mature but a lowernumber of matured oocytes than treatment 2(p < 0.05). It is of interest that such a largedifference in <strong>the</strong> number of oocytes collectedcan be caused by <strong>the</strong> different maturationinducing treatments.DISCUSSIONExperiments to determine <strong>the</strong> relativedevelopment rates of in vivo and in vitro calfoocytes are in progress. If in vivo maturedcalf oocytes develop to embryos at ratesbetter than in vitro matured oocytes <strong>the</strong>ntreatment 2 would be a better maturationtreatment than those previously employed.REFERENCES(1) Armstrong, D.T., Irvine, B.J., Earl, C.R.(1994) S.S.R in press.(2) Earl, C.R., Irvine, B.J., Armstrong D.T.(1994) Proc. Aust. Soc. Animal Prod. (inpress).(3) Armstrong, D.T., Holm, P., Irvine, B.,Petersen, B.A., Stubbings, R.B.,McLean, D., Stevens, G., Seamark R.F.(1992) Therio 38:667-678.Supported by Meat Research Corporation.EFFECT OF ASPIRATION SYSTEl\1 ON THE RECOVERY AND DEVELOPMENT OF SHEEPFOLLICULAR OOCYTES.IF. Smith, H.R. Tervit, L.T. McGowan and P.A. PughAgResearch, Ruakura Agricultural Research Centre, PB 3123, Hamilton, N.Z.INTRODVCTIONRepeated collection of ooctyes from living animals followedby IVMlIVF/IVC has <strong>the</strong> potential to produce moreoffspring than traditional MOET procedures. Varioussystems <strong>for</strong> follicular aspiration in <strong>the</strong> sheep have beenreported (1, 2!) and we compare here two systems involvingcombinations of needle types and vacuums.l\IETHODSSixty four. Coopworth ewes were synchronised withintravaginal sponges (Repromap Upjohn! NZ) <strong>for</strong> 14 daysand given 0.96 units FSH (Ovagen; Iep! NZ) in 6 equaldoses on days 11! 12 and 13 of sponge treatment. Follicleaspiration was per<strong>for</strong>med via laparoscopy on day 14. AStortz (STZ; 20 g double lumen; 45 cm long) needle at 3vacuums (15, 25! 50 mm Hg) was compared with a systemsimilar to that reported by o<strong>the</strong>rs (2, A). It involved needlesof two gauges (20 v 18)! lengths (Long, 45cm; Short, 22cm)at 15 mm Hg. Two operators per<strong>for</strong>med <strong>the</strong> aspirations over4 days and <strong>the</strong> STZ 50mm Hg treatment was used as <strong>the</strong>control treatment on each day. Recovered oocytes wereIVM, IVF and rvc in our standard system (3) anddevelopment to blastocysts recorded. Oocytes recoveredfrom slaughtered untreated ewes were run as controls.RESULTSThe mean number of follicles aspirated in <strong>the</strong> treated eweswas 14.7 and mean number of oocytes recovered was 8.0(54.1 %). Recovery rate was influenced by operator (62.9 v44.7%; P


CATTLE OOCYTE RECOVERY IS AFFECTED MORE BY ASPIRATION PRESSURE THANNEEDLE SIZEG. B. Udy, L T. McGowan, F. A. Nemaia, A. E. L. Ankersmit and H. R. Teryit.AgResearch, Ruakura Agricultural Research Centre, PB 3123 Hamilton, New Zealand.CYSTEAMINE ENHANCES SYNCHRONOUS PRONUCLEAR FORMATION INPORCINE OOCYTES MATURED AND FERTILIZED IN VITROC. G. Grupen,:\H. Nagashima and M. B. Nottie ,Bresatec Limited, Department of Obstetrics and Oynaecology, University of Adelaide, Adelaide, SA,5005, AustraliaINTRODUCTIONA range of needles (16G-20G; single and doublelumen) and vacuum pressures (50-200mmHg)have been used <strong>for</strong> oocyte recovery in humansand domestic animals (1). Oocyte recovery andquality vary with some combinations used (2).To enable <strong>the</strong> appropriate needle and vacuumchoice to be made <strong>for</strong> cattle transvaginal oocyterecovery (TVR), we investigated <strong>the</strong> effects ofneedle size and vacuum pressure.MATERIALS AND METHODSFollicles (2-7mm) were aspirated from ovariescollected from slaughtered cattle using a 50cmlong, single lumen needle. Oocytes were graded<strong>for</strong><strong>the</strong> presence of intact cumulus (G1), partiallydamaged cumulus (G2) and partially orcompletely denuded (G3). Between 210 and 220follicles were aspirated <strong>for</strong> each of three needlesizes (16, 17 or 18G) at four vacuum pressures(25, 50, 75 & 100mmHg).In addition to <strong>the</strong> in vitro ovarian aspirations,TVR was per<strong>for</strong>med on 25 fifteen-month oldHere<strong>for</strong>d nulliparus cattle using a 50cm, 17Gneedle with 75mmHg vacuum pressure. AnAloka (SSD-500, Japan) 7.5MHz linearultrasound probe was used to visualise <strong>the</strong>ovaries.RESULTSThe mean number offollicles aspirated per ovarywas 13.1 (2575/197) and overall oocyte recoveryrate was 53.2%. Recovery rate was affected byrecovery day (P


EARLY EMBRYO SURVIVAL IS HIGHER FOLLOWING TWIN RATHER THAN SINGLETRANSFER OF FROZEN-THAWED, BUT NOT FRESHIN VITRO PRODUCED BOVINE EMBRYOSW.ij. McMillan, P.A. Pugh and AJ. PetersonAgResearch Rualrura, PB 3123, Hamilton, New ZealandINTRODUCTIONThere appears to be little published data comparingsingle and twin post-transfer embryo survival· (ES)of in vitro-produced (IVP) bovine embryos indesigned experiments. In one report where ei<strong>the</strong>rsingle or twin IVP embryos were transferred fres<strong>hth</strong>ere was a tendency <strong>for</strong> higher survival in singleembryos (1). The survival of each of twintransferredIVP embryos appears to be a dependentevent when transferred fresh, but an independentevent when transferred frozen-thawed (2). Thepotential <strong>the</strong>re<strong>for</strong>e exists <strong>for</strong> an interaction between<strong>the</strong> number of embryos transferred and freezethawing.The aim of <strong>the</strong> current study was to test<strong>the</strong> hypo<strong>the</strong>sis that <strong>the</strong> difference in ES betweensingle and twin transferred embryos was independentoftransfer as fresh or frozen-thawed embryos.MATERIALS AND METHODSNinety (Trial 1) and 109 (Trial 2) I8-month oldHere<strong>for</strong>d x Friesian heifer recipients were randomlyallocated to one of 4 treatment groups in a 2 x 2factorial design (single or twin transfer; fresh orfrozen-thawed transfer). ES (n=300 embryos) wasassessed by slaughtering a random sample of heifersat d 18 (n=85 embryos assessed); slaughtering nonreturningheifers at d 25 (n=49 embryos); recordingreturn oestrus to d 50 (n=118 embryos) andslaughtering <strong>the</strong> remaining non-returning heifers at d50 (n=48 embryos). ES was defmed as <strong>the</strong> number ofembryos surviving to <strong>the</strong> time of assessment/totalembryos transferred, summed over all assessmenttimes.RESULTSAn interaction was apparent in Trial 1. There wasno difference in ES between embryos transferredfresh as singles (n=23 embryos) or as twins (n=46)(39 vs. 28%, Table 1). However, ES was higher <strong>for</strong>twin (n=46) compared with single (n=21) transfers ifembryos had been frozen-thawed (26 vs. 0%, P =0.025, Table I). Single fresh embryos were morelikely to survive than single frozen-thawed embryos(39 vs. 0%, P


EFFECT OF SPERM MOTILITY ON THE FERTILISATION ~TE BYINTRACYTOPLASMIC SPERM MICROINJECTIONOrly Lacham-Kaplan and Alan trounsonInstitute of Reproduction, Centre <strong>for</strong> Early Human Development,Monash Mniversity, 246 Clayton Rd, Clayton, Melbourne, Ausralia.INTRODUCTIONThe use of intracytoplasmic spermmicroinjection(lCS1) as a treatment <strong>for</strong> severe male infertilityhas enabled most of <strong>the</strong>se pateints to fertilize<strong>the</strong>ir wives oocytes(l). The insertion of asingle sperm into <strong>the</strong> ooplasm results in anincreased fertilization rate when compared to invitro insemination or sub zonal sperm injection.High recovery rates of motile sperm can beobtained from most samples of ejaculated andepididymal sperm (70%-100%). However, in somemale factor patients and frozen thawedepididymal sperm <strong>the</strong> recovery of motile sperm isbelow 10%. The present study examines <strong>the</strong> effectof sperm motility on <strong>the</strong> fertilization rateusing intracytoplasmic sperm microinjection ofejaculated and epididymal sperm.MATERIALS AND METHODSEjaculated sperm were washed by high speedcentrifugation(1800g)through a Percoll gradient(90%,45%) <strong>for</strong> 20 min. The 90% percoll layer wasretained and washed with HTF+30mg/ml BSA (1800gx 5min), followed by swim up of <strong>the</strong> pelletedsperm into fresh HTF+30mg/ml BSA until <strong>the</strong>y wereneeded <strong>for</strong> microinjection. The top flraction of<strong>the</strong> sperm solution which contained <strong>the</strong> motilesperm was separated, and washed (1800g x 5min)in PBS+3mg/ml BSA. The pelleted sperm werediluted to lx10" sperm/ml in PBS+3mg/ml BSA.Fresh epididymal sperm which were obtainedsurgically from <strong>the</strong> epididymis (2) and frozen/thawed epididymal sperm were treated <strong>the</strong> same asejaculated sperm. A 100pl drop of <strong>the</strong> spermsolution was placed on <strong>the</strong> microinjectionchamber (3). Cumulus oocyte complexes obtainedafter follicular aspiration were transferredinto HTF+10% pateint serum. The cumulus cellswere removed. by a brief exposure to 0.1%hyaluronidase. Up to 5 mature oocytes wereplaced in <strong>the</strong> sperm drop located on <strong>the</strong>microinjecton chamber. A single sperm wasselected, immobilized and ins~rted into <strong>the</strong>oocytes as described previously (3). Inaddition, fully motile and mechanicallyimmobilized sperm from a motile popUlation wereinjected into oocytes. The ocytes were examined<strong>for</strong> fertilization 17-19h after microinjection.RESULTSThere was a significant reduction, in <strong>the</strong>fertilization rate by non-motile ejaculatedsperm and epididymal sperm when compared tomotile sperm (Table 1 and Table 2) .Only 38% of<strong>the</strong> oocytes were fertilized by immotileejaculated sperm compared to 59% of oocytesinj ected with sperm from a motile population(Table 1) (p


RETROSPECTIVE ANALYSIS OF SUPEROVULATED CYCLES FOR ICSI YIELDINGAN INCREASED NUMBER OF IMMATURE OOCYTESJ.Smitz C. Janssenswillen J. Liu, H. Joris, M. Camus, P. Devrgey and A. Van Steirtegh~mCentre <strong>for</strong> <strong>Reproductive</strong> Medicine, University Hospital, Dutch-speaking Brussels Free University, BelgiumINTRODUCTIONWomen undergoing in-vitro fertilization by intracytoplasmicsperm injection are stimulated routinely by a combinedgonadotropin releasing-hormone agonist (GnRHa) andhuman menopausal gonadotropin (HMG) desensitisationprotocol. The final oocyte maturation is induced by humanchorionic gonadotropin (HCG). The nuclear maturationcan be precisely evaluated by removal of <strong>the</strong> surroundingcumulus-corona granulosa cell layer (CCG) and observation<strong>for</strong> <strong>the</strong> presence of <strong>the</strong> first polar body (PB). The cohort offollicles stimulated by HMG is usually asynchronous andfollicles of different diameter are punctured 36 hours afterHCG injection.In a first series of of 227 consecutive GnRHa and HMGstimulations 80% of <strong>the</strong> oocytes were at <strong>the</strong> metaphase IIstage (MIl) at <strong>the</strong> moment of retrieval and 20% of <strong>the</strong>oocytes were ei<strong>the</strong>r at <strong>the</strong> metaphase I stage (MI) orgerminal vesicle (GV) stage.In <strong>the</strong> present study we wanted to evaluate <strong>the</strong> stimulationprofiles and <strong>the</strong> outcome of cycles which yielded more than40% immature oocytes. We questioned whe<strong>the</strong>r <strong>the</strong>presence of an elevated oocyte immaturity rate wouldinfluence fertilisability, developmental and implantationpotential of <strong>the</strong> mature oocytes from <strong>the</strong> same cycles.PATIENTS AND METHODSThe origin of infertility in <strong>the</strong>se couples was severeandrological impairment. The majority of women in thisgroup were regularly menstruating. Out of 1471consecutive IC51-treatments (over a 2 year-period) <strong>the</strong>cycles with ~ 40% immature oocytes were listed. For thisanalysis we considered exclusively <strong>the</strong> cycles with a normalovarian response i.e. when ~ 5 oocytes were recovered.These cycles were compared to matched control cyclesper<strong>for</strong>med in <strong>the</strong> same period but with a percentage ofimmature oocytes < than 40%.Ovarian stimulation treatmentAll cycles were stimulated with a combined Buserelin andHMG long protocol as described previously (1). Thedecision <strong>for</strong> HCG injection was decided on <strong>the</strong> presence offollicular diameters of <strong>the</strong> largest group of folliclesexceeding 17 mm and/or concentrations of serum17 ~-estradiol > 300 ng/L per large follicle. Lutealsupplementation with 600 mg micronised naturalprogesterone intravaginally was systematically given (2).Oocyte retrieval fertilization and embryo cultureOocyte retrieval was done 36 hours after <strong>the</strong> ovulatorystimulus. The CCG were kept <strong>for</strong> less than 1 minute inmedium with <strong>the</strong> hyaluronidase and <strong>the</strong> removal of <strong>the</strong>CGG was enhanced by aspirating <strong>the</strong> oocytes in and outhand-drawn pipettes. The oocytes were put under aninverted microscope to assess <strong>the</strong> maturation stage by <strong>the</strong>observation of a germinal vesicle (GV) and <strong>the</strong> presence(Mil) or absence (MI) of <strong>the</strong> first polar body (PB). Alloocytes that had extruded <strong>the</strong> first polar body were injectedwith a single sperm (3). Fertilization was observed byvisualisation of 2 pronuclei 16 hours after single sperminjection and embryo quality was based on <strong>the</strong> equality ofsize of <strong>the</strong> blastomeres and <strong>the</strong> proportion of anucleatedfragments (4). Transfer of embryos was limited to 3embryos. Supernumerary embryos with < 20% fragmentswere cryopreserved (5). .RESULTSOut of 1471 stimulation cycles <strong>the</strong> absence of a secondpolar body was revealed in more than 40% of <strong>the</strong> retrievedoocytes in 124 cycles (110 women). In 13 women <strong>the</strong>appearance of a high proportion of GV oocytes happenedrepeatedly. We compaired <strong>the</strong> frequency distribution of <strong>the</strong>number ofoocytes per cycle in <strong>the</strong> GV ~ 40 % group wit<strong>hth</strong>e matched control group. The two distributions had asimilar profile. We analysed <strong>the</strong> outcome of <strong>the</strong>se cycles inaccordance with <strong>the</strong> number of oocytes per retrieval. Theresults are summarised in table I.We aimed to find differences in <strong>the</strong> ovarian stimulationprofiles that might explain <strong>the</strong> abnormally high rate ofimmature oocytes per retrieval. These figures are listed inTable III.OocyteslcyclePreovulatory E2 conc (mglL)(P fJglL)N° of days of exponentialE2 riseTotal number of HMGamps usedN° of follicles with meandiameter ~ 18 mmon day of HCG injectionN° offollicles ~ 18 mm perN° of retrieved oocytes (%)Study group Control groupGV ~ 40% GV < 40%Oocytes/cycle < 15 ~ 15 < 15 ~15N° cycles 75 49 84 40Fertilization (%) of Mil oocytes 65±28 56±23 64±20 59±28Cleavage rate (%) to embryosof good and/or fair quality 74±27 69±24 67±28 66±29N° of embryos transferred 1.93 2.51) 2.50 2.63per cycle (± 1.03) (±0.65) (±0.68) (±0.54)Implantation rate (%)per embryo 12.5 20.2 22.5 25.0N° of embryos frozen <strong>for</strong>N° of patients 16 <strong>for</strong> 8 69 <strong>for</strong> 23 96 <strong>for</strong> 32 146 <strong>for</strong> 23The detailed results of <strong>the</strong> embryo transfers shown intable II.Study group (GV >40%)Study group< 15 ~ 151831 2527(±646) (±530)0.84 0.86(± 0.58) (± 0.60)7.6 6.2(±1.93)a (±0.93)a39 25.5(±19)b (±9.1)b4.1 5.1(±1.95)c (±2.52)da, b, c, e, f: P < 0.001; d : P < 0.01; g, h : P < 0.05Control group< 15 ~ 151899 2621(±537) (±649)0.83 0.87(±0.92) (±0.71)7.0 6.7(±1.43) (±1.1)35 27.2(±15) (±10.5)5.4 6.6(±1.84)c (±2.69)d48.6 26.2 56.9 33.7(±26.1)e,g (± 15.5)e,h (±25.7) f,g (± 16.1) f,hCONCLUSIONSIn 8.4% of consecutive GnRHa/HMG cycles an oocyteimmaturity rate of more than 40% was observed. Themetaphase II oocytes from <strong>the</strong>se cycles had a similarfertilization and cleavage potential than those from <strong>the</strong>matched control group. The lower implantation and takehome-babyrates in cycles with a high proportion ofimmature oocytes are due to <strong>the</strong> lower availability of goodquality embryos <strong>for</strong> transfer.A lower proportion of preovulatory follicles with a meanfollicular diameter of ~18 mm in cycles was found incycles with a high oocyte immaturity rate. In <strong>the</strong> cyclesproducing more than 15 oocytes <strong>the</strong> ovulatory stimulus wasgiven when only 25 to 35% of <strong>the</strong> follicles were larger than18 mm diameter.REFERENCES1) Smitz et aL, Hum. Reprod. 3, 585-590,19882) Smitz et aL, Hum. Reprod. 7, 168-175, 19923) Van Steirteghem et aL, Hum. Reprod. 8, 1055-1060, 19934) Staessen et aL, J. Vitro Fert. Embryo Transf. 6, 22-29, 19895) Van Steirteghem et aL, Hum. Reprod. 2, 593-598, 1987ControlsN° of cycles without a transfer 11 9Embryo transfer (% of total)with 1 embryo 21 12 embryos 35 383 embryos 44 61Transfer of 3 embryosNumber 51 65Embryo quality distribution% of E's with exellent 13 16good quality 67 74fair 20 10FOLLICULAR GROWTH AND OOCYTE COLLECTION IN HEIFERS IMMUNISEDAGAINST INHIBIN.R. C. Fry, T.L. Simpson, T.J. Squires and 1.K Findlay *.VIAS, Werribee, Vic 3030 and *PHIMR, Clayton, Vic 3168IntroductionThe repeated aspiration of follicles from elite heifers canproduce numerous oocytes <strong>for</strong> embryo production (1).We can aspirate all follicles >2 mm diameter on <strong>the</strong>ovaries using a 17 gauge needle attached to a 6.5 MHzultrasound probe (Toshiba Capasee, 2). This studyinvestigated <strong>the</strong> influence of immunisation againstinhibin' or repeated FSH on follicular growth, oocyterecovery and oocyte quality in heifers.Materials and MethodsTwenty five 15 month old Friesian heifers were allocatedto 5 groups (n=5). Four groups (PLC, INHI5, INH30,INH250) were immunised against inhibin (Biotech Aust)at dose rates of 0, 15, 30 and 250 Ilg given 4 weeks priorto and at <strong>the</strong> start of <strong>the</strong> treatment period. The 5th group(FSH) received 16mg FSH (Follitropin) + 200 ill PMSGat each aspiration. All heifers received progestagen earimplants (Crestar) throughout <strong>the</strong> experiment to preventovulation and serum samples were taken <strong>for</strong> antibodyresponse.All heifers were aspirated twice weekly <strong>for</strong> 1 week priorto and during <strong>the</strong> 4 week treatment period. At eachaspiration, follicle number and size were recorded, allcandidate follicles aspirated, and <strong>the</strong> oocytes collectedand graded according to <strong>the</strong> cumulus vestment. Results<strong>for</strong> <strong>the</strong> 4 week treatment period were analysed byANOVA (Genstat).ResultsThere was a mean of 16 follicles 2mm in diameter orgreater observed on <strong>the</strong> ovaries at each aspiration (9x 2­4mm, 4 x5-9mm, 2 xl0+mm). A mean of 9.0 follicleswas aspirated and 4.6 oocytes recovered (51 %). Of <strong>the</strong>se,3.6 (78%) were classified as viable <strong>for</strong> IVMIIVF(1.5A,O.8B,1.4C) while 0.5 were Denuded and 0.6Expanded cumulus). Antibody levels against inhibin weredetected at <strong>the</strong> time of booster and reached a plateau 2weeks later (Fig. 1). Immunisation against inhibin ortreatment with FSHlPMSG did not alter <strong>the</strong> total numberof follicles on <strong>the</strong> ovary, but did change <strong>the</strong> follicle sizedistribution, with less 2-4 mm follicles and more 10+mm follicles (P.".I:>0:;; 20c:«


CHARACTERIZATION OF IN VITRO SYNTHESIZED PROTEINS SECRETED BYPORCINE NON-ATTACHED OVIDUCTAL EPITHELIAL CELLSPHERES TREATED WITH ESTROGEN.Ping Xia, Jean Rutledge, Andrew J. Watson and David T. AnnstrongDept. of Obstetrics and GynaecoIogy, The University of Western Ontario, London, ON,Canada N6A 5A5INTRODUCTIONRecent studies by Buhi et a1. (1992) have demonstrated thatestrogen is responsible <strong>for</strong> induction ofde novo syn<strong>the</strong>sis andsecretion of certain oviductal secretory proteins (OSP) andinhibition of o<strong>the</strong>r OSP in porcine oviductal explant cultures. In<strong>the</strong> baboon (Boice 1990), sheep (Gandolfi 1991), hamster (Kan1988) and mouse (Kapur 1988), E:2-dependent oviductalglycoproteins contribute to <strong>the</strong> extracellular matrix of<strong>the</strong> embryoby associating with <strong>the</strong> zona pellucida and to <strong>the</strong> exclusivemicroenvironment of <strong>the</strong> embryo by entering <strong>the</strong> perivitellinespace. Studies in our laboratory have shown that E:2-treatedoviductal epi<strong>the</strong>lial cells facilitate early porcine embryonicdevelopment in vitro (Xia and Armstrong, 1994). The presentwork was undertaken to detect in vitro syn<strong>the</strong>sis of secretoryproteins by ~-treated porcine oviductal epi<strong>the</strong>lial cells used <strong>for</strong>coculture with early embryos.MATERIALS AND METHODSEpi<strong>the</strong>lial cell sheets were squeezed from ampullae of oviductsand cultured in TCM 199 with 10% FCS with or without E 2 (1~g/ml) <strong>for</strong> 48 hr, after which <strong>the</strong> oviductal cell aggregates<strong>for</strong>med epi<strong>the</strong>lial cell spheres (ECS). The ECSs (40 ECSs) werecultured in 200 ~I MEM without methionine containing 5 mg/mlglucose, 500 ~Ci/ml L)SS-Methionine and with or without E:2 (1~g/ml) under paraffin oil <strong>for</strong> 6 hr. The culture medium wasdialysed against 10 mM Tris-HCI buffer, pH 8.2, and deionizedwater. Dialysed culture medium (10,000 or 50,000 cpm) wasanalyzed by both ID- and 2D-SDS-PAGE.RESULTSThe results from ID-SDS-PAGE showed that <strong>the</strong> in vitrosyn<strong>the</strong>tic pattern of secretory proteins by ECS was altered by E 2treatment. These alterations included <strong>the</strong> appearance of onepolypeptide (33,000 Mr) and <strong>the</strong> disappearance of a secondpolypeptide (82,000 Mr) in <strong>the</strong> E:2-treated group, compared to <strong>the</strong>200-200-116-97-116-97- 66-82~tV 45-ctVC ~~45-31-33~31- 21-I I I2 3 4 7.6 7.0 6.0Figure 1. ID-SDS-PAGE.patterns observed <strong>for</strong> <strong>the</strong> controls. Additional proteins ofapproximate Mr 45,000 and 97,000 were increased in abundanceby E:2 treatment (Figure 1). Upon analyses by 2D-SDS-PAGE ofECS culture medium, <strong>the</strong> Mr 97,000 protein band was resolvedinto two acidic proteins of PI 4.5 and PI 5.1. The PI 5.1 proteinwas clearly suppressed in <strong>the</strong> ~-treated group (arrow 7) whereas<strong>the</strong> PI 4.5 protein was increased by E:2 treatment (arrow 8). TheMr 45,000 protein complex in <strong>the</strong> ~-treated group was resolvedinto three acidic proteins, Mr 45,000 and 43,000, both with PI 5.5(arrow 4,5) and a Mr 36,000-45,000 protein complex with PI 4.8(arrow 6). Two basic proteins with PI 8.0 (Mr 36,000) (arrow 1)and PI 6.8 (Mr 25,000) (arrow 3) were both inhibited by E:2treatment. Ano<strong>the</strong>r protein (Mr 25,000, PI 7.6) was increased in<strong>the</strong> E:2-treated group (arrow 2).CONCLUSIONIn vitro syn<strong>the</strong>tic patterns of secretory proteins by porcineoviductal epi<strong>the</strong>lial cells used <strong>for</strong> embryonic coculture areinfluenced by E:2 treatment.REFERENCESBuhi WC et aI., 1992 J Exp ZooI262:426-435.Boice ML et aI., 1990 Bioi Reprod 43:340-346.Gandolfi C et aI., 1991 Eur J Basic Appl Histochem 35:383-392.Kan FWK, (1988) J Histochem Cytochem 36:1441-1447.Kapur RP and Johnson LV (1988) Anat Rec 221:720-729.Xia P and Armstrong DT (1994) Therigenology 41 :340.Figure 1: 1. ECS conditioned medium, control.2. E:2-treated ECS conditioned medium.3. ECS, control.4. E:2-treated ECS.Figure 2: A: ECS conditioned medium, control.B: E:2-treated ECS conditioned medium.Figure 2. 2D-SDS-PAGE11'1 A2 3I I I L5.1 4.5 7.6 7.0pHI6.0IBI5.1 4.5-CRYOPRESERVATION OF HUMAN SPERM WITH PENTOXIFYLLINEMonash IVF, 185 RoddIe Street, Richmond, Victoria 3121INTRODUCTIONAddition of 3mM pentoxifylline (PF), a cyclicadenosine 3'-5'-monophosphate (cAl\.1P) phosphodiesteraseinhibitor, to cryoprotectant has been shownto increase recovery of motile sperm post-thaw (1).This improvement may be due to (i) increasedintracellular cAl\.1P levels and direct stimulation ofmotility or (ii) PF acting as an antioxidant <strong>the</strong>rebyreducing membrane damage associated with <strong>the</strong>fr~eze-thaw process. We have previously shown thatmotility and normal acrosome morphology (AM) aresignificantly reduced by cryopreservation and thatdecreases in normal AM may be due to cell death or tosubletllal damage including membrane disruption (2).The aim of this study was to investigate <strong>the</strong> effect ofPF on sperm during cryopreservation and to assess itspotential usefulness in improving <strong>the</strong> recovery offunctional sperm post-thaw.MATERIALS AND METHODSSemen samples from donors (n=15) with normalsemen characteristics (WHO, 1992) were assessed <strong>for</strong>progressive and total motility and AM. Motility wasassessed on a warm-stage-equipped phase contrastmicroscope. AM was assessed by labelling ethanolfixed sperm with fluorescein-conjugated concanavalinA lectin (2). Samples were diluted 1: 1 with HumanSperm Preservation Medium (3) and divided into 4aliquots to which PF was added to <strong>the</strong> following finalconcentrations; 0 (control), 1, 3 and 10mM. Thesealiquots were mixed, aspirated into O.5m1 straws,frozen in liquid N 2 vapour and stored in liquid N 2 •Samples were thawed at 25°C <strong>for</strong> 10 minutes <strong>for</strong> reassessmentofmotility and AM which were reported asa percentage of<strong>the</strong>ir initial values (mean±sem).RESULTSPost-thaw progressive motility was found to beincreased in <strong>the</strong> presence of all concentrations of PFcompared to control (Fig. I). However, this increasewas only significant (p


-EFFECT OF LEVELS OF GLYCEROL AND COMPATIBLE SOLUTES ON THEPOST-THAW MOTILITY OF RAM SPERMATOZOAL.G. sanchez-Partida l , J.L. Zuppl, W.M.C. Maxwelz2 and B.P. Setchelzi.lDepartment ofAnimal Sciences at 1Waite Agricultural Research Institute, University ofAdelaide, Glen Osmond,SA 5064 and 2University ofSydney, NSW 2006.INTRODUCTIONSuccessful cryopreservation ofram spermatozoadepends on <strong>the</strong> presence of glycerol in <strong>the</strong>freezing medium. However it has been suggestedthat glycerol may have a negative effect onfertility and lambing rates (1). The addition ofcompatible solutes (CS) has been reported toenhance post-thaw motility (PTM) of ramspermatozoa in <strong>the</strong> presence of 5% glycerol (2).In order to investigate <strong>the</strong> effect of differentlevels of glycerol and CS <strong>the</strong> following studieswere conducted.MATERIAL AND METHODSTwo factorial experiments were conducted,using tris-citrate diluents containing six levelsof glycerol (0,1,2,3,4,5 % v/v) and three levelsof CS (0, 54 and 81 mM). The CS were prolinein experiment 1 and glycine betaine i"nexperiment 2. All <strong>the</strong> diluents contained 15%egg yolk (v/v) at pH 7.0. Semen collection,dilution and freezing was done as previouslydescribed (2). Three pellets per diluent per ramwere individually thawed and assessed using aHamilton-Thorn motility analyser afterincubation in a water bath (37°C) <strong>for</strong> 0,4 or 8 h.REFERENCESRESULTSResults are presented in Figures 1 and 2. Theaddition of54 mM ofCS to <strong>the</strong> diluents improvedPTM in <strong>the</strong> presence of glycerol (P


EFFECT OF PROTEIN TYPE AND CONCENTRATION IN THE FREEZING MEDIUMON THE SURVIVAL OF IVP BOVINE EMBRYOS.P.A. Pugh. A.E.L., Ankersmit, L.T. McGowan and H.R. TervitAgResearch, Ruakura Agricultural Research Centre, PB 3123, Hamilton, N.Z.·- Paper withdrawn -INTRODUCTIONThe survival of in vitro produced (IVP) bovineembryos following freezing can be low (1) and isinfluenced by <strong>the</strong> type of culture system used (2).There. are reports of successful freezing of NPembryos in media with raised protein levels (3). Sinceproteins can have cryoprotective effects (4,5), thisstudy examined <strong>the</strong> effect of freezing medium BSAor FCS concentrations on <strong>the</strong> survival of IVP bovineembryos.MATERIALS AND l\1ETHODSEmbryos were produced by IVMlIVF and <strong>the</strong>n IVC inSOF/amino acidslBSA as described previously (1).Good quality late morulae/early blastocysts; (LMlEB;n=187); expanded blastocysts (ExB, n=332) andhatching/hatched blastocysts (HB, n=142) wereselected <strong>for</strong> freezing. The basal freezing medium(FBI, 4 mg/ml BSA) was supplemented with 10%,20% or 50% v/v FeS or 5, 10 or 25 mg/ml BSA and1.5M ethylene glycol. Embryos were equilibrated atroom temperature in <strong>the</strong> freeze solution, loaded intostraws and frozen at -0.6 0 C/min to -35 0 C be<strong>for</strong>eplunging into liquid nitrogen. Straws were thawed(30 0 C) and recovered embryos held in PB1 + 0.5Msucrose at room temperature <strong>for</strong> 5 min. Embryoswere <strong>the</strong>n cultured in SOF + 5% FCS <strong>for</strong> 48 h. Thosethat had re-expanded or hatched and maintainedintegrity were judged to have survived freezing.RESULTSSurvival rates are presented in Figure 1. There was asignificant negative effect of FCS concentration onsurvival (control, 64%; BSA, 64%; FCS, 53%;P


IN VITRO AND IN VIVO SURVIVAL OF CRYOPRESERVEDHATCHED BLASTOCYSTS IN PIGSH. Nagashima, N. Kashiwazaki, R.I. Ashman, M.B. NattIeBresatec Limited, Department of Obstetrics and Gynaecology, The University of Adelaide, Adelaide, S.A. 5005CRYOPRESERVATION OF PORCINE IN VIVO AND IN VITRO DERIVED BLASTOCYSTSWITH GLYCEROL AND ETHYLENE GLYCOLN. Kashiwazaki, H. Nagashima, R.I. Ashman, M.B. NattIeBresatec Limitid, Department of Obstetrics & Oynaecology, University of Adelaide, SA 5005INTRODUCTIONThe production of live piglets from cryopreserved embryos isstill in its infancy and is restricted to embryos from <strong>the</strong> perihatchingstage (1). Survival of cryopreserved embryosfollowing transfer is extremely limited (1) and may bebecause <strong>the</strong> cryoprotective effect of 1.5M glycerol used inprevious studies is insufficient. The addition of 0.25Msucrose to freezing media containing glycerol has also beenreported to be beneficial <strong>for</strong> post-thaw survival of cattleembryos (2). The present study was undertaken to determinewhe<strong>the</strong>r increasing <strong>the</strong> concentration ofglycerol or addition ofsucrose to <strong>the</strong> freezing media could improve <strong>the</strong> efficiencywith which pig embryos can be successfully frozen.MA1ERIALS AND METHODSEarly hatched blastocysts were collected from superovulatedcrossbred (Large White/ Landrace) gilts six days after matingwith Large White boars. Embryos were cryopreserved in <strong>the</strong>presence of 1.5M glycerol + 0.25 M sucrose (GS), 1.5Mglycerol (1.5G) or 1.8M glycerol (1.8G) using a conventionalslow cooling method. Embryos were cooled directly fromroom temperature to -6.5°C, seeded, and <strong>the</strong>n cooled at a rateof 0.3°C/min to -30 (GS) or -35°C using a programnlablefreezer (Freeze Control, CryoLogic) be<strong>for</strong>e being plunged intoliquid nitrogen. Mter thawing in a 30°C water bath followedby stepwise dilution of <strong>the</strong> cryoprotectant, <strong>the</strong> post-thawsurvival of embryos was assessed by in vitro culture <strong>for</strong> 24 hin Whitten's medium supplemented with 15mg/ml BSA andtransfer. Frozen-thawed embryos were classified according to<strong>the</strong>ir morphology into 3 categories (A: prefreeze morphology,B: damaged, C: extensively damaged) after culture.RESULTSAs shown in Table 1, post-thaw survival rates of embryoswere significantly improved by ei<strong>the</strong>r increasing glycerolconcentration (1.8G) or adding sucrose to <strong>the</strong> freezingmedium (GS). More A-grade embryos were obtained in GSgroup compared with 1.5G and 1.8G groups. Transfer offrozen-thawed embryos from <strong>the</strong> GS, 1.5G and 1.8G groupswhich had been graded after culture <strong>for</strong> 24 h to 4 recipients didnot result in pregnancy (Table 2). Transfer of embryos fromGS group to a recipient 3 h after thawing without beinggraded resulted in <strong>the</strong> birth of4 piglets (Table 2; Fig.!).DISCUSSION/SUMMARY/CONCLUSIONSThe results demonstrate that addition of 0.25M sucrose to <strong>the</strong>freezing media improves post-thaw survival compared withfreezing with glycerol alone. A marked feature of thisimprovement was <strong>the</strong> increase in <strong>the</strong> percentage ofblastocysts with little or no visible sign of damage (A-grade).Frozen-thawed embryos were evaluated morphologically afterin vitro culture <strong>for</strong> 24 h in an attempt to ensure that onlyviable embryos were transferred to recipient animals. Nei<strong>the</strong>rtransfer, however, resulted in <strong>the</strong> establishment ofpregnancy.In contrast, a litter of 4 live piglets was obtained following<strong>the</strong> transfer of 66 embryos, 3 h after thawing to one recipient.Although this result needs to be confirmed using fur<strong>the</strong>rnumbers of transfers, it is possible that in vitro culture afterthawing may have had a detrimental effect on embryoviability. In conclusion, <strong>the</strong> present study has demonstratedthat <strong>the</strong> in vitro survival of hatched blastocyst followingfreeze-thawing using 1.5M glycerol can be increased by <strong>the</strong>addition of 0.25M sucrose and that this treatment can be usedto produce live piglets from frozen embryos.Table 1. Survival of cryopreserved porcine hatchedblastocyst in vitro.Cryopro- Embryo DeyelQpment after culture fQr 24 htectants survival A-grade B-grade C-grade(%) (%) (%)1.5G 40148 4 8 28(85.1)a (10.0)C (20.0) (70.0)1.8G 55/61 16 26 13(91.6)b (29.1)d (47.3) (23.6)GS 136 63 36 20(95.2)b (52.9)e (30.1) (16.8)a vs b, c vs d P


PRODUCTION OF PIGS CONTAINING A METALLOTHIONEIN PORCINE GROWTHHORMONE GENE CONSTRUCTM.B. Nottle, H. Nagashima, N. Kashiwazaki, R.I. Ashman, Z. Du, C.G. Grupen, S.M. McIlfatrick,M. Harding, C. Cheah, R.I. Craw<strong>for</strong>d, A.I. RobinsBresatec Limited. PO Box 11, Rundle Mall, Adelaide, South Australia, 5005, Australia-An evaluation of a tyrosinase minigene as a marker <strong>for</strong> transgenesis inQuackenbush mice.R. Sutton, ~.A. Fawcett, J. '0 Grady, W.G. Ward, and B.W. BrownCSIRO Division of Animal Production, Locked Bag 1, Delivery Centre, Blacktown, NSW,2148INTRODUCTIONIntegration rates <strong>for</strong> pigs containing variousgrowth hormone gene constructs vary from 0.04to 1.73 % of embryos injected and from 5.6 to35.3 % of piglets born (1). In this abstract wereport integration rates <strong>for</strong> pigs containing ametallothionein porcine growth hormone geneconstruct. Data are from two trials conducted inApril (Trial 1) and November (Trial 2) 1993.MATERIALS AND METHODSLarge White x Landrace gilts were mated at <strong>the</strong>irsecond oestrus and <strong>the</strong>n aborted by intramuscularinjection (Lm.) of 1 mg of <strong>the</strong> prostaglandin F2uanalogue, cloprostenol (Estrumate; Pitman-MoorePty. Ltd., New South Wales, Aust.) between 25 and40 days after mating, followed by a secondinjection of 0.5 mg cloprostenol 24 h later.Animals were superovulated with 1000 Lu. eCGLm. (Pregnecol, Heriot AgVet, Vic., Aust.) given at<strong>the</strong> same time as <strong>the</strong> second cloprostenol injection.Ovulation was induced by Lm. injection of 500Lu. hCG (Chorulon, Intervet Pty. Ltd., New SouthWales, Aust.) 72 hours later. Animals werenaturally mated 28-30 hours after hCG injectionand in Trial 2 were also inseminated with freshsemen (lxl0 10 sperm) 2 hours after naturalmating. Ova were recovered by retrogradeflushing of <strong>the</strong> oviducts with PBS plus 1% FCS,50-56 hours after hCG injection. Zygotes werecentrifuged at 12,000 x g <strong>for</strong> 8 minutes to enable<strong>the</strong> pronuclei to be visualised. Zygotes were heldin Dulbecco's Minimal Essential Media with 25mM Hepes and 5mg/ml BSA. One pronucleus wasinjected with 4~10 picolitres of 10 ng/Ill DNA inPBS. 25-35 microinjected embryos weretransferred to each recipient. Integration of <strong>the</strong>transgene was determined by PCR using DNAfrom a sample of tail tissue taken at birth.RESULTSEmbryo and animal data are shown in Tables 1and 2.Table 1. Mean (± sem) ovulation, recovery andfertilisation rates and <strong>the</strong> numbers of zygotes injected <strong>for</strong><strong>the</strong> two trials. Values are expressed on a per donor sowbasis.Ovulation No. ova No. ova No.rate recovered fertilised zygotesinjectedTrial 1(n=229) 16.6± 0.3 13.3±0.3 6.8 ±0.4 5.7±0.4Trial 2(n=151) 18.9+0.3 14.9+0.4 10.8+ 0.5 10.1+0.5Table 2. Number of recipients, farrowing rate, number ofpiglets born live and <strong>the</strong> number of <strong>the</strong>se that weretransgenic <strong>for</strong> <strong>the</strong> two trials.Trial 1(n=229)Trial 2(n=151)No. of No. No. No.recipients recipients piglets transgenicfarrowing born live48472524DISCUSSIONACKNOWLEDGMENTSThe authors gratefully acknowledge <strong>the</strong> assistanceof Bunge Meat Industries Ltd., Corowa, New SouthWales, Australia.REFERENCES148141Data from <strong>the</strong> two trials indicate that between 5-10zygotes suitable <strong>for</strong> microinjection can beobtained per donor using <strong>the</strong> current protocol andthat 3-6 donors are required to provide 25-35embryos <strong>for</strong> one transfer. Overall integration rates<strong>for</strong> <strong>the</strong> transgene were 2.9 % of embryos injectedor 30.4% of piglets born live.(1) Brem, G. (1992). In 'Embryonic Developmentand Manipulation in Animal Production' EdA. Laurie and F. Gandolfi . p 147. PortlandPress. London.(2) French, AJ. et al., (1991) Theriogenology 35:202.4345INTRODUCTIONThe aim of <strong>the</strong> present study was to test <strong>the</strong> utility ofa tyrosinase minigene (ptrTYR5) as a marker <strong>for</strong>transgenesis as described by Beermann et al.(1). Thisgene can genetically rescue albino mice, restoringpigment to hair and eyes.These authors suggested that ptrTYR5 could be coinjectedas a marker with o<strong>the</strong>r transgenes (3 out of 5of <strong>the</strong>ir transgenics carried both genes). The coinheritanceof pigmented hair with a transgene ofinterest would greatly reduce <strong>the</strong> amount of screeningrequired to maintain <strong>the</strong> transgenic line.MATERIALS AND METHODSWe generated transgenic mice by microinjection ofptrTYR5 into pronuclei of single cell newly fertilisedeggs. This 11.1 kb construct includes a short 270bppromoter, <strong>the</strong> first exon and first intron from <strong>the</strong>mouse gene; and remaining 4 exons from mouseeDNA; and 850bp SV40 polyadenylation signal. Wetested tested <strong>for</strong> transgenesis by phenotype, PCR andSou<strong>the</strong>rn blotting. We compared <strong>the</strong> microinjection ofptrTYR5 into Quackenbush mice at 2 concentrations,with that of 3 o<strong>the</strong>r genes (all 5ng/ul) injected into F1mice. Gene 1 was injected be<strong>for</strong>e <strong>the</strong> 5ng/ul series ofptrTYR5; Gene 3 and <strong>the</strong>n Gene 5 were microinjectedin <strong>the</strong> period as <strong>the</strong> 2.5ng/ul ptrTYR5 often on analternate days in <strong>the</strong> same week.TABLE 1. TRANSGENESIS SUCCESS RATESRESULTSWe had a lower success rate with this transgene inQuackenbush mice than o<strong>the</strong>r genes microinjectedinto <strong>the</strong> CBAxC57BU6(F1) mice normally used in thislaboratory. In Quackenbush mice, <strong>the</strong> pronuclei werevisible in fewer eggs (57% cf 72%) so that fewerembryos could be injected, embryo survival of <strong>the</strong>microinjection was similar, so that <strong>the</strong> numbers ofembryos transferred are comparable; and <strong>the</strong> level oftransgenesis is lower than usual. The 5ng/ulconcentration appeared to be toxic.DISCUSSIONWe have successfully restored <strong>the</strong> pigmentation toalbino Quackenbush mice by transgenesis with atyrosinase mini-gene, ptrTYR5.However, we would not at this stage recommend <strong>the</strong>co-microinjection of ptrTYR5 as a marker with gene ofinterest using albino Quackenbush mice as analternative to use of C57Black/F1 mice. O<strong>the</strong>r optionswhich could be explored would be to use an F1 crossof 2 albino lines, to engineer a shorter transgene andto fur<strong>the</strong>r reduce <strong>the</strong> concentration of <strong>the</strong> tyrosinaseminigene used.REFERENCEBeermann, F., Ruppert, S., Hummler, E. andSchutz,G.(1991) Nucl. Acids Res. 19:958Total Embryos Embryos Pups TransgenicEmbryos Injected Transferred Born MiceptrTYR5 314 183 140 9 0(5ng/ul) 58% 77% 6% 0%ptrTYR5 462 260 194 10 1(2.5ng/ul) 56% 75% 5% 10%Gene 1 748 68% 69% 11% 5% (n=2)Gene 3 505 76% 78% 13% 13% (n=5)Gene 5 748 73% 75% 13% 28% (n=15)33 34


Effects of zona pellucida on electrofusion and development of two .cell mouse embl)'osBrendan Tatham, Kim Giliam, Aneta Dowsing, Tohsak Mahaworasilpa 1 , and Alan TrounsonCEHD Monash University Melbourne and IDepartment Physi~s University New South Wales SydneyAustralia.INTRODUCTIONElectrofusion is a process where electric fields induce cell to cellfusion and is a valuable technique used to fuse embryonic cells <strong>for</strong>nuclear transfer. The modification of <strong>the</strong> nuclear transfer procedurewhich alleviated <strong>the</strong> requirement <strong>for</strong> micromanipulation so that oocyteswere enucleated by centrifugation. The new technique involved <strong>the</strong>removal of <strong>the</strong> zona pellucida, <strong>the</strong> effect of this on <strong>the</strong> electrofusionproperties ofcells is unknown. This study in vestigated <strong>the</strong> electrofusionofbiastomeres of 2 cell mouse embryos with <strong>the</strong> zona intact or absent'and <strong>the</strong> development of <strong>the</strong>se tetraploid embryos when cultured invitro.MATERIALS AND :METHODSFemale C57/BIJxCBAlWehi mice were superovulated <strong>the</strong>n matedbe<strong>for</strong>e embryos at <strong>the</strong> 2 cell stage were collected. The zona pellucidawas removed with Pronase. Embryos were equilibrated <strong>for</strong> 10 minutesin fusion media (0.25 M sucrose, pH 7.4) \vith 4 washes. Blastomereswere <strong>the</strong>n aligned <strong>for</strong> 5 to 10 seconds with an AC pulse of 8 voltsamplitude in an electrofusion chamber which consisted ofparallel wireswith a diameter of 0.5 mm and a separation of 0.5 mm. Electrofusionwas <strong>the</strong>n achieved with a single DC pulse of 10, 50 or 100 ~s duration,with <strong>the</strong> amplitudes from 0.4 to 4.8 kV/cm . Electrofusion was scored1 hour after electrical treatment when groups of control and fusedembryos were cultured in M16 until <strong>the</strong> blastocyst stage.RESULTSThe results of approximately 1500 electrofusion attempts showthat when DC pulses of less than 2.0 kVIcm amplitude were used <strong>the</strong>electrofusion was not significantly different <strong>for</strong> embryos with <strong>the</strong> zonaintact (52.0%) or absent (61.5%). At amplitudes greater than 2.0 kV/cmDC <strong>the</strong> electrofusion of embryos with <strong>the</strong> zona intact (61.3%) wassignificantly greater than when absent (33.8%; P


· THE LOCALIZATION OF NEUTRAL ENDOPEPTIDASE IN THE OVINE UTERUS DURINGTHE OESTROUS CYCLE AND EARLY PREGNANCYSimon C Riley, Evelyn Wong, Jock K Findlay &Lois A SalamoQsenPrince Henry's Institute of Medical Research, PO Box 5152, Clayton, Victoria, 3168Changes in Uterine Endometrial Phospholipids and Fatty Acids during <strong>the</strong>Oestrous Cycle and Early Pregnancy in <strong>the</strong> Ewe,8 Meier, MA Trewhella, RJ Fairclough, G JenkinDepartment of Physiology Monash University Clayton Vic. 3168 AustraliaINTRODUCTIONNeutral endopeptidase (NEP; EC 3.4.24.11;'enkephalinase') is a membrane-bound cell surfaceectoenzyme with an extracellular active site thatcleaves and inactivates several bioaetive peptides. Itis expressed in <strong>the</strong> uterus in women and rats (1,2)and its substrates, which include oxytocin andendo<strong>the</strong>lins are important in <strong>the</strong> control of uterinefunction. In women, NEP has been implicated in <strong>the</strong>control of menstration, because NEP activitydecreases during <strong>the</strong> pre-menstrual phase allowing anincrease in <strong>the</strong> availability of endo<strong>the</strong>lin to inducevasoconstriction of <strong>the</strong> spiral arterioles. The role ofNEP in <strong>the</strong> ovine uterus during <strong>the</strong> oestrous cycle andat <strong>the</strong> time of implantation, in which peptidesubstrates of NEP have been implicated (3) has notbeen determined. This study examines <strong>for</strong> <strong>the</strong>presence and localization of NEP in <strong>the</strong> sheep uterusduring this period and investigates its regulation. byovarian steroids in vivo.MATERIALS AND METHODSThe uteri of parous Corriedale ewes (n=64; 4/group)throughout <strong>the</strong> oestrous cycle and <strong>the</strong> first 20 days ofpregnancy, and of ovariectomised ewes receiving notreatment (ovx), or progesterone (P), or oestradiol (E),or E+P, were flushed and collected by hysterectomy.Pregnancy was confirmed by identification of anembryo. Tissues were fixed in <strong>for</strong>malin <strong>for</strong>immunohistochemistry. Specific NEP immunostainingwas identified using a monoclonal antibody tocommon acute lymphoblastic leukemia antigen (CloneJ5; Coulter Corp.), and visualised using <strong>the</strong> VectorABC kit. Sections were scored on a scale of 0-3 (0 =no staining; 3 = strong staining throughout a cell type)in comparison with both positive and negative controlsections.ReSULTSSpecific_ NEP immunostaining was present in <strong>the</strong>uterus throught <strong>the</strong> oestrous cycle and early pregnancy.In <strong>the</strong> endometrium,' NEP was' localised instromal cells except those immediately underlying <strong>the</strong>epi<strong>the</strong>lia, . in increasing intensity to I day 14 of <strong>the</strong>oestrous cycle, which was maintained from Days 15-.20 of pregnancy (Fig.1). Weak staining was presentin luminal and glandular epi<strong>the</strong>lium. Strong stainingwas observed in myometrium, in particular in a bandadjacent to <strong>the</strong> endometrium, and in endo<strong>the</strong>lial andsmooth muscle cells of <strong>the</strong> vasculature (Fig.1). In <strong>the</strong>Day 20 conceptus, staining was localized inuninucleate, but not in binucleate cells, and in heartand brain of <strong>the</strong> embryo. In ovx ewes untreated, ortreated with E or P, immunostaining in all tissuecompartments was strong, whereas E+P treatmentresulted in a weaker pattern of staining (Fig.1).DISCUSSIONNEP immunoreactivity is present in <strong>the</strong> ovine uterusand is regulated at least in part by ovarian steroids.NEP is distributed in specific regions that may conveya partitioning effect within <strong>the</strong> tissue on its bioactivepeptide substrates. The band of NEP in <strong>the</strong>myometrium may prevent <strong>the</strong> passage of oxytocin andendo<strong>the</strong>lin, potent myometrial constrictors frompassing into <strong>the</strong> myometrium. The presence of NEPwithin <strong>the</strong> vasculature may regulate <strong>the</strong> paracrineeffect of endo<strong>the</strong>lin vascular tone, and <strong>the</strong> passage ofpeptides between <strong>the</strong> uterus and systemic circulation.The absence of NEP in luminal and glandular epi<strong>the</strong>liawould maintain <strong>the</strong> high concentrations of suchpeptides that are required <strong>for</strong> preparation of <strong>the</strong> uterusand <strong>the</strong> implantation processes in <strong>the</strong> uterine lumen.The presence of NEP in <strong>the</strong> trophoblast and embryoindicates an ability to regulate bioaetive peptides atthis early stage of development.FIGURE 1: Specific NEP immunostaining in caruncular andintercaruncular stroma (a,c) and myometrium, vascularsmooth muscle and endo<strong>the</strong>lium (b,d) during earlypregnancy (a,b) and in steroid-treated ovariectomised ewes(c,d).3 (a)2.521.5E' 1'c 'ro§ 0.52_ 0..ut+-'-..lCf-I--t4-l-l"l""'-"4-JL....t'4-L.t.::j--1-l:'t-'-E.4 8 10 12 14 16 17 20'0,?;~ (e)~3j 2.521.510.5O~~t:a4-~l-l-Ovx P E E+PEarty pregnancy3 (b)2.521.510.5oOvariectorrised steroid-trsated4 8 10 12 14 16 17 20Ovx P E E+PREFERENCESCasey, et al. (1991) J BioI. Chem 266, 23041-7.Ottlecz, et al. (1991) J Cell Biochem 45,401-11.Riley, et al. (1994) J Reprod Fert 100, 4551-9.Supported by <strong>the</strong> NH&MRC and a PHIMR VacationScholarship to EW.IntroductionThis study was conducted to determinechanges in endometrial phospholipid andfatty acid content in association withprostaglandin syn<strong>the</strong>sis and release duringluteolysis and early pregnancy.MethodsUterine endometrium was collected ondays 3, 12 and 15 of <strong>the</strong> oestrous cycleand on corresponding days of pregnancy.Lipids were extracted using chloro<strong>for</strong>m andmethanol (2: 1 v/v), containing <strong>the</strong> antioxidant, BHT. Phospholipids wereseparated from total lipids using thin layerchromatography (TLC), with a solventsystem of chloro<strong>for</strong>m:mathanol:aceticacid:water (65:60:6:4 v/v). Areascorresponding to standard phospholipids(PC, PS, PI, PE) were removed separately.The fatty acids associated with eachphospholipid were measured as <strong>the</strong> fattyacid methyl esters (FAME), using gaschromatography, and a BPX-70 columnpacked with 70% cynopropyl siloxane,(0.25 ~m; Ld. 0.22 mm;25 m length). EachFAME was quantitated against an internalstandard C17:0. Different stages of <strong>the</strong>cycle and pregnancy were comparedusing ANOVA and expressed as <strong>the</strong> mean± standard error of <strong>the</strong> mean (sem).ResultsTotal lipids extracted from <strong>the</strong> uterineendometrium during <strong>the</strong> oestrous cycle andearly pregnancy are summarised inTable 1.TABLE 1. Total Lipids (mg/g tissue)Stage * Mean semDay 3 c. 17.93a,o 3.79Day 12 c. 20.53°'c 1.72Day 15 c. 27.62 c 3.64Day 3 p. 15.77 a 1.01Day 12 p. 25.72°'c 1.68Day 15 p. 17.03 a 1.15* c. oestrous cycle, p. pregnancya,b,c statistical significance (p


PORCINE-RElAXIN DIFFERENTIALLY INHIBITS UTERINE LONGITUDINAL & CIRCULAR MUSCLESIN VIVO IN OESTROUS & EARLY PREGNANT RATS.Helen Massa and Len MartinDepartment of Physiology and Pharmacology, University of Queensland, Q4072, AustraliaThe period of uterine receptivity in <strong>the</strong> pseudopregnant Quackenbush Special mouse asdefined by <strong>the</strong> deciduogenic lectin, Concanavalin AJennette A. Sakofjand R. N MurdochDepartment ofBiological Sciences, The University ofNewcastle, N.S.W., 2308INTRODUCTION Myometrial contractions increase incomplexity and frequency in early pregnant rats (1,2). Thecontractions are essential <strong>for</strong> normal embryo spacing andorientation since <strong>the</strong>se are disturbed when myometrialactivity is inhibited with porcine relaxin (p-RXN; 3,4).MATERIALS AND METHODS Oestrous (Oe) & day 5pregnant rats (d5P; d1=day of sperm) underwent videolaparoscopy(VL; 1,2). VL started at 09:00h, 1h afteranaes<strong>the</strong>sia (Ketamine:Xylazine;60:8 mg kg- 1 ) and insertionof a jugular cannula, and lasted <strong>for</strong> 3.5h. In each rat, vehicle(V, 0.9% NaCI), oxytocin (Ot, 20mU) & p-RXN (0.2, 1, 5~g,one dose/rat, 5 rats/dose) were given 20 min apart asboluses (0.5mL IV) in <strong>the</strong> sequence V, Ot, p-RXN, Ot, Ot(Fig 1). Myometrial activity, analysed from video-tape replay(1,2) was recorded as <strong>the</strong> number 10min- 1 of longitudinal &circular contractions propagating towards <strong>the</strong> cervix or ovary(respectively Lc, Cc La, Co) and examined by ANOVA.RESULTS At Oe, most contractions were Lc: on d5P, <strong>the</strong>irfrequency fell whilst that of Lo, Cc & Co rose, as be<strong>for</strong>e(1,2). A bolus of Ot increased Lo & Co in both Oe & d5Prats [P


EARLY DECIDUOGENIC RESPONSE OF THE UTERINE MUCOSA TO THELECTIN CONCANAVALIN A AND SESAME OILINTRODUCTIONThe Decidual Cell Reaction (DCR) is vital <strong>for</strong> <strong>the</strong>establishment of pregnancy and involves <strong>the</strong>specialised differentiation of <strong>the</strong> uterine stromasurrounding an implanting blastocyst. It has longbeen recognised that agents o<strong>the</strong>r than a blastocystare capable of initiating a Decidual like response.Few studies have investigated <strong>the</strong> interactionbetween deciduogenic agents and <strong>the</strong> uterineepi<strong>the</strong>lium -<strong>the</strong> first point of contact between <strong>the</strong>blastocyst and maternal tissue. Given <strong>the</strong> widelydiffering composition of deciduogenic agentsavailable, it would be of interest to compare <strong>the</strong>manner in which <strong>the</strong>se agents interact with <strong>the</strong>uterine epi<strong>the</strong>lium. This investigation compares <strong>the</strong>initial stages of deciduomata fonnation using oil, acommonly used deciduogenic agent and <strong>the</strong> lectinConcanavalin A (ConA).MATERIALS AND METHODSPseudopregnancy (PSP) was induced on <strong>the</strong>morning of oestrus in adult virgin rats. On <strong>the</strong>morning of day 5 PSP, Animals were anaes<strong>the</strong>tisedand given an intraluminal injection of ei<strong>the</strong>r 1 mgof ConA in 50 pl of saline, 50 JIl of sesame oil or50 JII of saline alone. Animals were sacrificed 6, 12and 24 hours after injection. Fifteen minutes prior tosacrifice animals were injected with 0.3 ml of 0.1 %Pontamine Sky Blue to visualise stromal oedema.Tissue was processed <strong>for</strong> routine light microscopyand Electron Microscopy. Observations of tissuemorphology were made from wax serial sectionsand 1 JIm thick semi-thin sections.RESULTSTwelve and 24 hours following injection of ei<strong>the</strong>rConA or oil, blue bands were clearly visible within<strong>the</strong> uteri. Tissue sections revealed marked stromaloedema. O<strong>the</strong>r indicators of a decidual-like reaction,including differentiation of stromal cells and <strong>the</strong>fonnation of <strong>the</strong> Primary Decidual Zone (PDZ)were visible in <strong>the</strong> uterine hom of both ConA andoil treated groups at <strong>the</strong>se times. The reaction of <strong>the</strong>epi<strong>the</strong>lium of <strong>the</strong> two groups however variedsignificantly. The f epi<strong>the</strong>lium of animals injectedwith oil was greatly attenuated within <strong>the</strong> bluebands (fig 1) and in places was entirely missing,often appearing to be contained within luminal oildroplets. The epi<strong>the</strong>lium of ConA injected rats didnot display 'such damage. However, within blueTJ. Shaw and C.R. MurphyDept. Anatomy and Histology, University of Sydney.bands from ConA injected animals, white bloodcells were often aggregated under <strong>the</strong> epi<strong>the</strong>lium,and preliminary results suggest that within <strong>the</strong> blueband <strong>the</strong> white cells appeared to breach <strong>the</strong> uterinelumen (fig 2).DISCUSSIONWe conclude that both ConA and Oil Inducedeciduomata <strong>for</strong>mation on Day 5 of PSP. Themanner in which <strong>the</strong>.se two agents interact with <strong>the</strong>uterine mucosa, however, differs significantly. Oilappears to cause marked uterine trauma supporting<strong>the</strong> <strong>the</strong>ory of Lundkvist 1982, that oil is in fact atraumatic deciduogenic agent. The exact manner inwhich ConA induces deciduomata is unclear. It ishowever apparent th&t ConA alone does not cause<strong>the</strong> degree of trauma associated with oil, and it ispossible <strong>the</strong> white blood cells present followinginjection are <strong>the</strong>mselves involved in <strong>the</strong> initiation ofdeciduomata.Figure 1. Uterine mucosa following injection of oil,attenuated epi<strong>the</strong>lial cells (arrow) are visible. x830.Oil droplet is out of field of view.Figure 2. Uterine mucosa following injection ofConA, White blood cells are visible in lumen (A),and arrow indicates point of epi<strong>the</strong>lial crossing.x830.Lundkvist, O. & Nilsson, B.O. 1982, Cell Tiss Res.225,355-364.EFFECTS OF THE INTERACTION BETWEEN CLOMIPHENE CITRATE AND THE OVARIAN HORMONESON THE SURFACE ULTRASTRUCTURE OF THE UTERINE LUMINAL EPITHELIAL CELLSMJ.Hosie and C.R.Murphy, Dept Anatomy and Histology, The University of Sydney, NSW, 2006.INTRODUCTION: The widely used super-ovulatory drug,Clomiphene citrate (CC) is a syn<strong>the</strong>tic non-steroidal impededoestrogenlantioestrogen thought to act via <strong>the</strong> hypothalamicpituitaryaxis resulting in <strong>the</strong> maturation of multiple follicles (1).Although CC is a valuable and efficient super-ovulator (70-80%of women) <strong>the</strong> ensuing pregnancy rates are relatively low (2). Ithas. been suggested that this may be due to altered endometrialdevelopment (3), which is thought to be critical <strong>for</strong> implantation(4). Currently <strong>the</strong>re is little research on <strong>the</strong> ultrastructural effectsof CC on <strong>the</strong> uterus. This study <strong>the</strong>re<strong>for</strong>e focuses on <strong>the</strong> surfaceultrastructural effects of CC at <strong>the</strong> time of blastocyst implantation(around day 6 of pregnancy in <strong>the</strong> rat). Scanning electronmicroscopy (SEM) was used to evaluate uterine surfaceultrastructure from ovariectomized rats treated with CC andoestradiol-17p (E2) or progesterone (P 4 ) or <strong>the</strong>se two hormonesgiven in combination, as well as giving CC prior to <strong>the</strong> P4-P4.P4E2sequence of hormones, given over 3 days, which has been shownto induce both implantation and an ultrastructural picture similarto that seen at implantation in ovariectomized rats.MATERIALS AND METHODS: This study used 60, 12-14week old sexually mature rats, bilaterally ovariectomized andallowed to recover <strong>for</strong> 4 weeks be<strong>for</strong>e treatment. The animalswere treated with: O.lml injections of: 0.51lg E 2 ; 5.0mg P 4 ; and/or0.25mg CC or 1.25mgCC. The experimental groups consisted of:1) P4+E2; 2)E2+O.25mgCC; 3)P4+O.25mgCC; in a single dose; 4)P­P-PE; 5)0.25mgCC-P-P-PE; 6)1.25mgCC-P-P-PE; 7)P-P-P; <strong>the</strong>sewere given over 4 days. Group 8 consisted of day 6 pregnantcontrols, and group 9 consisted of <strong>the</strong> appropriate placebo groupswhere <strong>the</strong> results were combined as <strong>the</strong> tissue appeared similar.The rats were killed 24 hours after <strong>the</strong> last treatment and <strong>the</strong>tissue routinely processed <strong>for</strong> SEM.RESULTS: It was found that when CClE2 (Fig 3) wereadministered toge<strong>the</strong>r, <strong>the</strong> surface ultrastructure was similar inmany respects to that seen in <strong>the</strong> control (Fig 1) or E/P4 (Fig 2)or P4 alone were administered, suggesting that in this situation CCis behaving as an anti-oestrogen or' a progesterone. Typicalfeatures include short relatively sparse microvilli and raised cellborders. At low power <strong>the</strong>se groups all resembled each o<strong>the</strong>r inthat <strong>the</strong> tissue had an overall smooth appearance with few smallgland openings. When CCIP 4(Fig 4) was given <strong>the</strong> result wasoestrogenic with oval to round domed cells with recessed borderscovered with dense blunt sometimes long MV. At low power <strong>the</strong>epi<strong>the</strong>lium had a roughened appearance with some foldingevident, suggesting that CC is acting as an oestrogen in thissituation and/or in some way preventing progesterone action in <strong>the</strong>expression of <strong>the</strong> progestogenic features usually qbserved. WhenCC is given prior to <strong>the</strong> PPPE sequence (Fig 5), <strong>the</strong> surfaceultrastructure of <strong>the</strong> tissue most closely resembled that observedwith CC alone showing very long microvilli, many droplets andareas of doughnut shaped cells (Fig 6). CC appears to override <strong>the</strong>usual surface ultrastructure seen with PPPE (Fig 7). Features seenwith both PPPE and day 6 pregnant tissue (Fig 8) include a lossof mv, and <strong>the</strong> appearance of many irregular protrusions anddroplets.CONCLUSION: These results indicate that when CC is given inconjunction with E2 or P4 it is capable of ei<strong>the</strong>r behaving in asimilar fashion to P4 or E2. Clomiphene administration prior to <strong>the</strong>PPPE sequence of hormones appears to change <strong>the</strong> tissuetopography to a surface similar to that seen when CC is used onits own (5). Thus CC changes <strong>the</strong> ultrastructural appearance of <strong>the</strong>uterine epi<strong>the</strong>lium and so may interfere with implantation by thismechanism.l.Clark lA, B.M. Markaverich (1982) Pharmac.Ther. 15,467-519.2.Navot D., N. Lufer (1989) IReproduct.Med. 34,3-9.3.Cummings A.M. et al (1991) Fundam.Appl.Toxicol. 16,506-16.4.Martel D.C. et al (1981) (Ide Brux, R.Martel, IP.Gautry,eds). Plenum Publishing Corporation.5.Hosie MJ., C.R.Murphy (1992) Acta Anat. 145,175-178.All Micrographs x6000.Fig 1, Placebo control.Fig 3, CCIE 2 .Fig 5, CCIPPPE Droplet (D).Fig 7, PPPE Protrusion (P).Fig 2, E21P4.Fig 4, CCIP 40Fig 6, CCIPPPE (X) Pit.Fig 8, Day 6 pregnancy_41 42


-VAGINAL EPITHELIUM OF OVARIECTOMISED RATS TREATED WITH COMBINEDCLOMIPHENE CITRATE AND EXOGENOUS OESTROGEN OR PROGESTERONEV. Terry, C.R. Murphy, C.D. ShoreyDept. Anatomy and Histology, University of Sydney, NSW, 2006INTRODUCTIONClomiphene citrate (CC), a non-ste.roidaloestrogen derivative, has been used In <strong>the</strong>induction of ovulation in anovulatory women <strong>for</strong>many years (1) .Emphasis has been placed onresearching <strong>the</strong> effect of this compou~d on. <strong>the</strong>reproductive tract of neonates WIth httleattention to <strong>the</strong> adult reproductive tract alreadyexposed to steroidal hormones. Our previousstudies with CC on <strong>the</strong> adult rat vagIna haverevealed an oestrogenic component of activityand an ability to modify cell appearance (2,3).Since CC rarely acts in isolation, or in a systemdevoid of hormonal priming, we decided tostudy <strong>the</strong> interactive effects of CC and oestrogenor progesterone on <strong>the</strong> vagina of ovariectomisedadult rats.MATERIALS AND METHODSThirty-five Female virgin Wistar rats (12-14weeks of age) were bilaterally ovariectomisedunder halothane anaes<strong>the</strong>sia and maintail1ed <strong>for</strong>4 weeks prior to experimentation. Groups 1,2,3and 4 were designated control groups and givenno treatment or a single injection of carrier A orB or A+B only. O<strong>the</strong>r groups were treated asfollows: Group 5 - 0.25mg CC + 5mg P4 (PC);Group 6 - 0.25mg CC + O.51lg E2 (EC); Group 7- 5mg P4 + O.5Jlg E2(PE). Animals were sacrificed 24 hrs afterinjection and tissue processed <strong>for</strong> light andscanning electron microscopy.RESULTSResults indicate that a 24 hr exposure to <strong>the</strong>combined treatments can stimulate a dormantvaginal epi<strong>the</strong>lium by initiating cell division,cause cell proliferation and modification.Analysis showed that <strong>the</strong>re was no statistical orstructural differences between CC incombination with exogenous oestrogen orprogesterone (Fig 2). Both treatments resulted inincreased epi<strong>the</strong>lial area, height, number oflayers and mitotic activity compared to controltissues (Fig 1). Statistical comparison with ourprevious study indicated that <strong>the</strong> two combinedtreatments using CC were similar to E2 alone.Scanning Electr~n microscopy revealed achanged surface morphology in all threetreatment groups. Control groups exhibitedlongitudinal vaginal wall folding, <strong>the</strong> covering ofwhich comprised polygonal cells clearlydemarcated by ridges (Fig 3a + 3b). Treatedgroups exhibited increased folding; surface cellswere irregularly arranged with slightly raised43apicies and separated by recessed borders.Numerous cells exhibited individually elongatedmicrovilli and/or rosette <strong>for</strong>mations (Fig 4a +4b).CONCLUSIONIn conclusion it can be suggested that when CCis given in conjunction with P4 <strong>the</strong>re is anenhancement of CC's oestrogenicity plus anincreased ability. to modify cell surfaceappearance. When CC is administered with E2<strong>the</strong>re is no apparent cumulative effect from thatof E2 alone, but <strong>the</strong>re is a modification of <strong>the</strong>surface appearance.Fig 1. Control (x 500)Fig 3a. Control (x 920)Fig 2. PC, EC, PE (x 500)Fig 3b. Control (x 4600)Fig 4a. PC, EC, PE (x 920) . Fig 4b. PC, EC, PE (x 4600)1. Greenblatt R.B.,Barfield W.E., Junck E.C., Ray A.W.(1961) 1. Am. Med. Assoc. 178,101-1042. Terry V., Murphy C.R., Shorey C.D. (1992) Acta Anat.145,212-2153. Terry V., Murphy C.R., Shorey C.D. (1993) Acta Anat.148, 14-21..vOrigin of enhanced fetal growth in sheep administeredprogesterone during early pregnancyD. O. Kleemann J , S.K. Walker and R.F. Seamark1South Australian Research and Development Institute, TurretfieldResearch Centre, Rosedale, SA 5350 and 2 Department of Obstetrics andGynaecology, University of Adelaide, SA 5000INTRODUCTIONModification of <strong>the</strong> embryo's early environment can actto influence subsequent development. For example, insheep <strong>the</strong> transient exposure of day 3 embryos <strong>for</strong> threedays to an advanced uterine environment resulted inlarger fetuses compared with control fetuses whenmeasured on day 37 of pregnancy (1). Similarly,subjecting sheep zygotes to in vitro culture <strong>for</strong> 3-5 days(2) or transferring bovine nuclei to host oocytes (3) canresult in progeny of excessive birthweight. In <strong>the</strong> cow(4) and sheep (5) progesterone administered early inpregnancy enhanced fetal development compared wit<strong>hth</strong>at observed in control animals . In all <strong>the</strong>se studies,changes to <strong>the</strong> embryo or to <strong>the</strong> embryo's environmentoccurred within <strong>the</strong> first few days of development thushighlighting <strong>the</strong> significance of this period on subsequentfetal per<strong>for</strong>mance. In <strong>the</strong> present experiment weexamined if <strong>the</strong> effect of progesterone on fetal growth ismediated during <strong>the</strong> days of treatment or is longer termin nature and whe<strong>the</strong>r any effect of progesterone isdependent on <strong>the</strong> exposure of embryos to <strong>the</strong> uterineenvironment during supplementation.MATERIALS AND METHODSZygotes (n = 949) were collected and transferred to <strong>the</strong>oviducts of initial recipient Merino ewes (n = 36; 6ewes per replicate) that were randomly allocated toei<strong>the</strong>r a control group or a group treated withprogesterone pessaries (CIDR G) <strong>for</strong> three dayscommencing 13-18 h after <strong>the</strong> expected time ofovulation. In <strong>the</strong> latter group embryos were ligated in<strong>the</strong> oviducts of 12 of 24 ewes. Embryos from eachtreatment group were recovered on day 3 andtransferred to uterine horns of final recipient ewes. Halfof <strong>the</strong>se ewes were similarly treated with progesteronepessaries, giving a 3 x 2 factorial design. Bloodsamples were taken from initial recipients on day 3 andassayed <strong>for</strong> plasma progesterone concentrations. Eweswere slaughtered on day 76 and <strong>the</strong> number, weight, sexand crown-rump length of fetuses was recorded.RESULTSPeripheral progesterone concentrations were significantly(P


Prostaglandin production by placental tissues in late pregnant tammarwallabiesG. Shaw, H. Gehring and E.C. Bell.Department of Zoology, The University of Melbourne, Victoria Australia 3052.Prostaglandin production by endometrium in late pregnant tammarwallabiesG. Shaw, H. Gehring, and E.C. Bell.Department of Zoology, The University of Melbourne, Victoria Australia 3052.INTRODUCTIONProstaglandins (PGs) are released at parturitionin tammar wallabies, leading to a brief peak ofPGF2 metabolite (PGFM) in <strong>the</strong> peripheralplasma (1). There is increased PO production by<strong>the</strong> uterine endometrium at term (2). This studyinvestigates <strong>the</strong> contribution of <strong>the</strong> placenta to PGprcxluction in late pregnancy.MATERIALS AND METHODSTammars have a thin, membranous,choriovitelline, or yolk sac, placenta. It ispossible to separate <strong>the</strong> placenta andendometrium cleanly, by gently pulling <strong>the</strong>membranes free, even at'full term. Females atday 18 of pregnancy (primitive streak: stage) tod26 of pregnancy (full term) were killed byanaes<strong>the</strong>tic overdose, <strong>the</strong> gravid uterus wasopened, and <strong>the</strong> fetus and placenta were gentlyremoved. Placenta was chopped into pieces 0.5 ­1 mm across. Portions of chopped tissue (10-20mg) were cultured on polycarbonate membranesat <strong>the</strong> liquid-air interface in 5 ml Ram l s-F10medium with antibiotics. Two replicates fromeach animal were set up. After a 4 hour preincubation<strong>the</strong> medium was replaced. At 24h <strong>the</strong>medium was removed and frozen at -80°C inindomethacin coated tubes until assayed byspecific RIAs <strong>for</strong> PGF2a, PGE2 and PGFM.Tissue from each well was collected and assayed<strong>for</strong> protein content against BSA standards.Results are expressed as mean ± s.e.m. of PGproduced (ng per day per ~g protein). Data aregrouped by stage: d18-22 (early organogenesis);d23-24; and d25-26 (term: viable neonates can beobtained at day 25, though <strong>the</strong>y are usually bornon day 26).RESULTSPlacenta produced both PGF2a and PGE2, butlittle or no PGFM Figure 1. Production ofPGF2a was about 10 times that of PGE2.Production rates of by placenta increasedsignificantly during late pregnancy200180160e: 140ot5::J ..-.. 120-0 .f:o Q)0.. e 100e: Q.::g 0) 80~.€0)0)jg ..s 60enea.. 4020oI ~ .PGE2PGF2a~PGFMd18-22d23-24Days of gestationd25-26Figure 1. Prostaglandin production by explal1tsof yolk sac membrane from late pregnant tammarwallabies.DISCUSSIONPG production by <strong>the</strong> placenta increasesdramatically at term. What regulates this increaseis unknown. This increased placental PGproduction may contribute to <strong>the</strong> fetal (or fetoplacental)signal that triggers parturition.REFERENCES(1) Lewis PR, Fletcher TP & Renfree MB(1986) J. Endocrinol. 111:103-109.(2) Shaw G, Gehring, H & Bell, EC (1994)These Proceedings.INTRODUCTIONProstaglandins (ros) playa key role at parturitionin mammals, inducing uterine contractions andsoftening <strong>the</strong> cervix. In tammars <strong>the</strong>re is a briefpulse ofPO F2a metabolite (PGFM) in peripheralplasma in <strong>the</strong> minutes around birth (1) probablyassociated with <strong>the</strong> uterine contractions that expel<strong>the</strong> fetus. PGs may also synchronise <strong>the</strong>stereotyped parturient behaviour with birth (2).The source of <strong>the</strong> PGs has been assumed to be <strong>the</strong>uterus. This study examined <strong>the</strong> profile of POproduction by uterine tissue through latepregnancy.MATERIALS AND METHODSFemales at day 18 of pregnancy (primitive streak:stage) to d26 of pregnancy (full term) were killedby anaes<strong>the</strong>tic overdose and <strong>the</strong> uteri dissected out.Tammars have two separate uteri with separatecervices, and only carry a single fetus, so it waspossible to compare gravid and non-gravid uteri ateach stage. The uteri were opened, <strong>the</strong> fetus andplacenta were gently removed, and <strong>the</strong>endometrium removed <strong>for</strong>m <strong>the</strong> myometrium using<strong>for</strong>ceps. Endometrial tissues were chopped intopieces 0.5 - 1 mm across. Portions of choppedtissue (10-20 mg) were cultured on polycarbonatemembranes at <strong>the</strong> liquid-air interface in 5 mlRam l s-FI0 medium with antibiotics. Tworeplicates <strong>for</strong> each tissue type from each animalwere set up. After a 4 hour pre-incubation freshmedium was supplied. At 24h <strong>the</strong> medium wasremoved and frozen at -80°C in indomethacincoated tubes until assayed by specific RIAs <strong>for</strong>PGF2cx., PGE2 and PGFM. The tissue from eachwell was collected and assayed <strong>for</strong> protein contentagainst BSA standards. Results are expressed asmean ± s.e.m. of PG produced (ng per day per Jlgprotein). Data are grouped by stage: d18-22 (earlyorganogenesis); d23-24; and d25-26 (term: viableneonates can be obtained at day 25, though <strong>the</strong>yare usually born on day 26).RESULTSProduction rates of PGF2a and PGE2 in <strong>the</strong>gravid endometria increased dramatically towardsterm, and substantially more PGF2a wasproduced than ei<strong>the</strong>r PGE2 or PGFM. Nongravidendometrium produced significantly lessPGs than gravid endometrium, and production didnot peak: at term.DISCUSSIONThus, <strong>the</strong> uteri may be <strong>the</strong> source of <strong>the</strong> peak: ofPO seen in peripheral plasma at parturition. Thea) Gravid Endometrium1001 o IIlII PGF2aPGE280 0 PGFMe:ot5::J..-.. 60-oSo Q)0.0l-e: Q.:0 0) 40~.€0l0)cu e:w-e 20a..e:ot5od18-22 d23-24 d25-26Days of gestationb) Non-gravid Endometrium::J C 60 -0 ._o Q)0..0l-e: Q.:0 0) 40.ffi.€0)0)cu e:w-o 200::100rb PGF2a1_ PGE280 Iq PGFMd18-22 d23-24 d25-26Days of gestationgravid uterus is more likely than <strong>the</strong> non-graviduterus to be <strong>the</strong> major source because thiscontains 2-3 times more tissue (3), and is moreactive in PG syn<strong>the</strong>sis at term. The mechanismsregulating <strong>the</strong> increasing PG syn<strong>the</strong>tic activitytowards term are still unknown.REFERENCES(1) Lewis PR, Fletcher TP & Renfree MB(1986) J. Endocrinol. 111:103-109.(2) Shaw G (1990) J. Reprod. Fert. 88: 335­342.(3) Renfree MB & Tyndale-Biscoe CH (1973)Dev. BioI. 32: 28-40.45 46


MAMMARY MORPHOLOGICAL AND FUNCTIONAL CHANGES DURING PREGNANCY IN WOMEN:METHODSIn this ongoing study of five women, we have measuredbreast volume during pregnancy, using <strong>the</strong> ComputerizedBreast Measurement (CBM) System (1) and <strong>the</strong> 24-houtput of lactose in urine, using a spectrophotometricassay (2).RESULTS AND DISCUSSIONThe timing and magnitude of <strong>the</strong> increase in <strong>the</strong> output oflactose in urine during gestation was similar involunteers 1-3, with volunteers 4 and 5 showing lowlactose excretion during <strong>the</strong> early stages of pregnancy(Figure 1).Changes in breast volume are shown in Figure 2 with acomputer generated breast reconstruction <strong>for</strong> volunteer 3shown in Figure 3. Although <strong>the</strong>re was close agreementin <strong>the</strong> changes in breast volume between right and leftbreasts of <strong>the</strong> individual volunteers. considerablevariation was observed between <strong>the</strong> volunteersthroughout pregnancy (Figure 2). In addition, <strong>the</strong>relative volumes of <strong>the</strong> right and left breasts of volunteerI were considerably different. The preconception breastvolume of volunteer 5 (indicated by asterisk), is likely tobe elevated as a consequence of her breastfeeding aprevious infant at <strong>the</strong> time of measurement, who wasweaned 4 weeks prior to conception (Figure 2).Assuming that <strong>the</strong> 24-h urinary lactose is a qualitativemeasure of <strong>the</strong> amount of lactose syn<strong>the</strong>sized, <strong>the</strong>seobservations suggest that lactogenesis 1 begins duringmid-gestation in women. However, <strong>the</strong> growth of <strong>the</strong>breast during pregnancy, measured by <strong>the</strong> increase inbreast volume, showed considerable differences between<strong>the</strong> women. These differences may be explained byvariation in <strong>the</strong> extent to which breast adipose tissue wasreplaced with lobular-alveolar tissue. We conclude from<strong>the</strong>se preliminary studies that <strong>the</strong> measurement ofchanges in <strong>the</strong> output of lactose in urine and in <strong>the</strong>volume of <strong>the</strong> breasts during pregnancy may offer a noninvasivemeans of relating <strong>the</strong> endocrinology ofpregnancy to mammogenesis and lactogenesis 1 inwomen.DB. Co."), i.e. KetU l , R.A. Owens 2 and PE. Hartmantz llThe Department of Biochemistry, 2The Department of Computer ScienceThe University of Western Australia, Nedlands 6009INTRODUCTIONDuring pregnancy <strong>the</strong> mammary gland undergoesextensive lobular-alveolar growth (mammogenesis). Atsome time during this growth, <strong>the</strong> epi<strong>the</strong>lial cellsdifferentiate and initiate <strong>the</strong> secretion of milkconstituents albeit at a low rate (lactogenesis I). Lactosesyn<strong>the</strong>sized by <strong>the</strong>se newly-differentiated alveolarepi<strong>the</strong>lial cells is reabsorbed into <strong>the</strong> blood and excretedin <strong>the</strong> urine. We have measured changes in breastvolume and urinary lactose during pregnancy, todetermine <strong>the</strong> relationship between mammogenesis andlactogenesis LREFERENCES1) Daly, S.EJ. et al. (1992) Exp. Physiol. 77:79-872) Arthur, P.G. et al. (1989) Anal. Biochem. 176:449-456ACKNOWLEDGEMENTS -- NH&MRC432o1000900- 5-Q)800E::::l0 700>.....C/)~~ 600.0Q).2:~Q)a:500400300o 10 20 30 40Time post-conception (weeks)Figure 1. Excretion of lactose in <strong>the</strong> urine duringpregnancy. Volunteer 10 ,2" ,3. ,4+,5....200*~~~-10 0 10 20 30 40Time post-conception (weeks)Figure 2. Breast volume changes during pregnancy, Opensymbols, Right breast, closed symbols, Left breast.Volunteer l' ,2" ,3. ,4+,5....Figure 3. CBM generated reconstructions of left breastshape during trimesters 1,2 and 3, <strong>for</strong> volunteer 3...pNutritional effects on testicular growth and LH and FSH secretion in Suffolkand Merino rams during <strong>the</strong> breeding and <strong>the</strong> non-breeding seasonsMaterials and MethodsTwo experiments were carried out, one in <strong>the</strong> non-breeding season (July/August; n=6/group) and one in <strong>the</strong> breeding season(FebruarylMarch; n=5/group). Merino and Suffolk rams were fed to maintain liveweight (0.9 kg chaff + lOOg lupins;"Maintenance") or <strong>the</strong> Maintenance diet plus 1.5 kg lupins ("Lupins"). On Days -1 and 12 after <strong>the</strong> change in diet, blood wassampled every 20 minutes <strong>for</strong> 24 hours and assayed <strong>for</strong> LH and FSH. Liveweight and scrotal circumference were measuredweekly. Data were analysed by repeated measures analysis of variance. In <strong>the</strong> case of LH and FSH, where an interaction wasfound to be significant, analysis of <strong>the</strong> change between Days -1 and 12 was carried out.BREEDING 30% change in LW % change in SC FSH (ng/ml) LH pulses/24h2015 3 1210102 8SEASON(February~arch) 10 5 6NON-BREEDING 30M.J. HotzeIl, G.B. Martin 1 ,2, S.W. Walkden-Brown1 and J.S. Fisher11Faculty of Agriculture, The University ofWestern Australia, Nedlands, WA 6009, Australia2CSIRO Division of Animal Production, Private Bag, PO Wembley, WA 6014, AustraliaIntroductionTesticular growth parallels changes in live weight in Merino rams, but in Suffolk rams testicular growth appears to be driven byphotoperiod, independently of changes in live weight (1). We tested <strong>the</strong> hypo<strong>the</strong>sis that <strong>the</strong> reproductive responses to changes indiet differs during <strong>the</strong> breeding and <strong>the</strong> non-breeding seasons in Suffolk, but not Merino rams.ResultsLupin supplementation increased live weight in both breeds in both seasons (p


ANOVULATORY DAIRY COWS HAVE LOWER LH PULSE FREQUENCY ANDINTRAFOLLICULAR CONCENTRATIONS OF OESTRADIOL (E 2 ) AND TESTOSTERONE (T)THAN CYCLIC COWS.S. McDougall and K.L. MacmillanDairying Research Corporation, Private Bag 3123, Hamilton, NZINTRODUCTIONSome pasture-fed dairy cows in New Zealandhave extended (>50 day) periods of postpartumanovulation (ppa) despite <strong>the</strong> presence of largefollicles (>10 mm) undergoing regular turnover.Low LH pulse frequency (1) and lowintrafollicular E 2 concentration (2) are reported insuckled beef cows during ppa. Failure ofovulation may occur due to a low LH pulsefrequency producing insufficient T and hence E 2to induce <strong>the</strong> pre-ovulatory gonadotrophin surge.As intrafollicular E 2 concentrations vary with <strong>the</strong>stage of follicle development (3), this hypo<strong>the</strong>siswas tested at two stages of follicle developmentin <strong>the</strong> ovaries of ppa dairy cows.MATERIALS AND METHODSUsing a 2 x 2 design, 28 anovulatory (Anov) orcycling (Cyc) cows at ei<strong>the</strong>r growing (Grow) orplateau (Plat) stage of development of <strong>the</strong>largest follicle (as determined by daily transrectalultrasound) were ovariectomised 24 h afterinduction of luteolysis (25 mg of Dinoprost). LHconcentration was determined in jugular samplescollected at 15 minute intervals <strong>for</strong> <strong>the</strong> 8 h be<strong>for</strong>eovariectomy and <strong>the</strong> number of pulses definedusing an alogorithm (4). The largest follicle wasdissected out, its diameter (Diam) measured and<strong>the</strong> E 2 , T and P4 concentrations of its follicularfluid determined. Data were analysed by GLMwith cow status, stage of follicular developmentand <strong>the</strong> interaction as <strong>the</strong> main effects. Thesteroid concentrations were log trans<strong>for</strong>medbe<strong>for</strong>e analysis. The relationships among LogE 2 , Log T and LH pulse frequency wereexamined by regression analysis.RESULTSAnov cows had a lower LH pulse frequencyGrowPlatAnoveycAnovCyeLH tpUlses/8h3.1 bc5.4 ab2.4 c6.0 aDiam tmm10.0 c11.4 bc16.2 a15.0 ab58.6 a274.2 b37.6 a504.9 band lower E 2 , T and P4 concentrations than eyccows. Plat follicles were larger, had lower Tconcentrations and had higher E 2 :T ratios and P4concentrations than Grow follicles (Table 1). Thestatus by stage interactions were not significant.The Log E 2 and Log T concentrations werepositively related to <strong>the</strong> number of LH pUlses. 2(Log E 2 = 0.12xLH freq.+1.56, R =26.3, p


EFFECTS OF CALCIUM IONOPHORE A23187 AND HEPARIN ON STAWON SPERM1l0r--r------------,-------------rllO"'0 70 . ~..~.~~ ,. 1.?9..~1.l}................................. 70Q)'0::J"if..fDJUalS.J. Robinson\ E.L. Squires 2 , J.K. Graham 2 and W.M.C. Maxwell 110epartment of Animal Science, University of Sydney, NSW 20062Animal Reproduction and Biotechnology Laboratory, Colorado State University, Fort Collins, CO 80523 USAIntroductionIn vitro capacitation of sperm is an essential pre-requisite <strong>for</strong> in vitrofertilisation (IVF). Alterations to <strong>the</strong> sperm head membranes duringcapacitation enable <strong>the</strong> sperm to undergo <strong>the</strong> acrosome reaction(1), and thus <strong>the</strong> proportion of sperm which have reacted may beused to assess sperm capacitation. Incubation with calciumionophore A23187 (2) and heparin (3) increase <strong>the</strong> occurrence of<strong>the</strong> acrosome reaction in stallion sperm. Experiments wereconducted to study <strong>the</strong> effects of A23187 and heparin on <strong>the</strong>motility and acrosome reaction of stallion sperm in vitro.Materials and MethodsSemen was collected from four stallions on separate days.Immediately after collection, <strong>the</strong> gel-free fraction was extended to50 x 10 6 sperm/ml with a modified, glucose-free TALP medium(mTALP-g; 2mM Ca 2 +) at 3rC. The extended semen was <strong>the</strong>nwashed < once by centrifugation (500g, 15 min) to remove <strong>the</strong>seminal plasma. The sperm were <strong>the</strong>n resuspended to 50 x 10 6sperm/ml in mTALP-g and <strong>the</strong> sperm suspension divided into 6parts (A,B,C,D,E,F), equilibrated at 37.5°C. A stock solution ofA23187 in DMSO was added to each part so that <strong>the</strong> finalconcentration was 0 (A,B), 2.5~M (C,D) or 5.0~M (E,F). The spermsuspensions were <strong>the</strong>n incubated at 37.5°C <strong>for</strong> 5 min be<strong>for</strong>ewashing by doubling <strong>the</strong> volume with mTALP-g and centrifugation(300g, S min). The sperm were resuspended to 25 x 10 6 sperm/mlin mTALP·g, and a stock solution of heparin (in mTALP·g) wasadded to parts B,D & F so that <strong>the</strong> final concentration was1OO~g/ml. The sperm suspensions were <strong>the</strong>n incubated <strong>for</strong> 2 hoursat 37.5°C in 5% CO 2 in air. Immediately (0) and at 30, SO and 120min, aliquots were removed <strong>for</strong> assessment of % motile sperm(objective assessment; Hamilton-Thorn motility analyser), %acrosome reacted sperm (spontaneous), and <strong>the</strong> % of sperm whichwere able to undergo induced acrosome reactions. The % of liveacrosome-intact and acrosome-reacted sperm were determined byviewing a drop of <strong>the</strong> sperm suspension under fluorescentmicroscopy after staining with propidium iodide and fluoresceinconjugated~sativum agglutinin«4). To assess <strong>the</strong> ability ofsperm to undergo an induced acrosome reaction, <strong>the</strong> sperm wereincubated in mTALP~g containing 1OO~g/ml Iysophosphatidylcholine (LPG) <strong>for</strong> a fur<strong>the</strong>r 15 minutes (5) and <strong>the</strong> acrosomereaction assessed as above. Mean % of motile and acrosomeFigure 1: Effects of A23187 and heparin on <strong>the</strong> motility andacrosome reaction of stallion sperm. Means with different lettersdiffer (P.EJ A-IVVu 00 800[g A-IVT;:e;(l)~.~ 600~~CI:l 400,.0:::lCI:l"0~ 200 03 H-Glu 14 C-Glu 14 C-Glut 14 C-PyrEnergy substrateFigure 1. Metabolism of energy substrates by in vivo(IVV) and in vitro (IVT) matured pre-pubertal (P) and adult(A) sheep oocytes.* P-IVV oocytes not allocated to <strong>the</strong>se treatments.# 14C_Pyr (fmol x 10-1).1000~ >..u 8000.Qn"'dQ) S.~ c3 600=3 eno +1Q)I::::Cd C'::l.b (l) 400CI:l S,.0 "-"0CI)"0 200~oPre-pubertalAdultOocyte classificationFigure 2. Metabolism of glutamine by mature pre-pubertaland adult sheep oocytes (in vivo and in vitro data pooled).REFERENCES1. Palma, G. A., Clement-Sengewald, A. & Krefft, H. (1993).Therio. 39, 278 abstr.2. Pinkert, C. A., Kooyman, D. L., Baumgartner, A. &Keisler, D. H. (1989). .T. Reprod. Fert. 87, 63-66.3. Ryan, 1. P., Hunton, 1. R. & Maxwell, W. M. C. (1992).Reprod. Fert. Dev. 4, 91-97.4. O'Fallon, .T. V. & Wright, R. W., .Tf. (1986). BioI. Reprod.34, 58-64.51 52


Eagle's Essential Amino Acids Stimulate Development of Inner Cell Mass Cellsof Cultured Mouse EmbryosMichelle Lane and David K. GardnerEmbryo Physiology Laboratory, Institute of Reproduction and Development, Monash University, Clayton, Victoria.IntroductionDevelopment and cleavage of mouse zygotes to <strong>the</strong> blastocyststage in culture are significantly increased by Eagle's non-essentialamino acids and glutamine (NEGLN), whilst essential amino acidswithout glutamine (ESS) have no apparent beneficial effects ondevelopment in vitro (1) However subsequent studies have shownthat NEGLN do not effect fetal development after blastocysttransfer. In contrast, fetal development is significantly increasedwhen blastocysts are developed in <strong>the</strong> presence of ESS (2). It ispossible that <strong>the</strong> two groups of amino acids have different effectson <strong>the</strong> cell lineages in <strong>the</strong> blastocyst. The aim of this study was<strong>the</strong>re<strong>for</strong>e to assess <strong>the</strong> role of amino acids on <strong>the</strong> development ofinner cell mass (lCM) and trophectoderm (IE) cells in culture andsubsequent blastocyst viability.Materials and MethodsZygotes were collected from Fl hybrid (C57BU6 x CBA/Ca)female mice and cultured in amino acid free medium mMTF (1) ormMfF supplemented with NEGLN, ESS or Eagle's 20 aminoacids (20AA). Embryos were cultured in 20111 drops ofmedium ingroups of lOin 5% CO 2in air at 37'C <strong>for</strong> an initial 48h in one of<strong>the</strong> above groups and <strong>the</strong>n allocated <strong>for</strong> a second 48h ofculture inone of <strong>the</strong> three amino acid treatments. Control embryos werecultured in mMfF throughout giving a total of 10 cultureconditions. In vivo derived blastocysts were recovered 88hpost-hCG. lCM and IE cells in blastocysts were assessed usingimmunosurgery and differential nuclear staining (3). Viability ofblastocysts was detennined by transfer to pseudopregnant femalesand fetal development assessed on day 15 ofpregnancy.ResultsTotal blastocyst cell munber and number of IE cells weresignificantly increased by culture with NEGLN <strong>for</strong> <strong>the</strong> first 48h,irrespective of <strong>the</strong> medium in <strong>the</strong> second 48h (Fig. 1). Culturewith ei<strong>the</strong>r ESS or 20M <strong>for</strong> <strong>the</strong> second 48h period significantlyincreased <strong>the</strong> number of rCM cells compared to NEGLN or noamino acids and were equivalent to in vivo derived controls.Similarly, fetal development was significantly increased by ESS or20AA <strong>for</strong> <strong>the</strong> second 48h culture period compared to NEGLN(Fig.2). Fetal development in all treatments was significantlyreduced compared to in vivo derived embryos. There was apositive correlation between <strong>the</strong> mean number of inner cell masscells in blastocysts in each treatment and subsequent fetaldevelopment (Fig.3).DiscussionThis study demonstrates that <strong>the</strong> preimplantation mouse embryoundergoes a switch in nitrogen requirements as developmentproceeds from <strong>the</strong> zygote to <strong>the</strong> blastocyst stage. NEGLN stimulatecleavage when present during culture of. <strong>the</strong> pre-compactedembryo whilst ESS stimulate <strong>the</strong> development of <strong>the</strong> rCM ofcultured embryos post-compaction. This has significantimplications <strong>for</strong> <strong>the</strong> <strong>for</strong>mulation of media <strong>for</strong> <strong>the</strong> culture of <strong>the</strong>preimplantation embryo as currently only one medium is used <strong>for</strong>all stages of development. This study demonstrates a positivecorrelation between allocation of cells to rCM and subsequentfetal development. Detennination of rCM number in blastocysts is<strong>the</strong>re<strong>for</strong>e a useful in vitro viability index.80"070~c:'*60~ 5002:-.cE40l.1JQ) 30j807060a; 50 .cE:J40zQj()3020100c: 20~OJ0..102520a;.cE:J15c:::EQ1050010aNo M N/N EIN 2O/N NIE EIE 2O{E N/20 E/20 20/20 In V'NoFig.! Effect of amino acids on <strong>the</strong> development of rCM (solidbars) and IE (open bars) in cultured mouse blastocysts. a-f: likepairs significantly different <strong>for</strong> rCM; P


EXPRESSION OF INSULIN-UKE GROWTH FACTOR-! mRNA IN ENDOMETRIUM DURING EARLYPREGNANCY IN THE SHEEPC.R. Cann, RJ. Fairclough 1 ,R. Sutton 2 and C.B. GowSchool of Agriculture~La Trobe University~ Bun.doora, Vic.~ 3083.1 Dept Food Technology~ Victoria University ofTechnology~ Werribee, Vic.• 3030.2 Division of Animal Production~ CSIRO~ Blacktown~ NSW, 2148.IGFBP PROTEOLYTIC ACTIVITY IN THE UTERINE FLUSHINGS OF PREGNANTAND NONPREGNANT SHEEPA.J. Peterson, A.M. Ledgard, S.C. Hodgkinson and H.R. TervitAgResearch, Ruakura Agricultural Research Centre, Hamilton, New ZealandINTRODUCTIONInsulin-like growth factor-I (IGF-l) of maternal origin hasbeen implicated in <strong>the</strong> cellular proliferation anddifferentiation of <strong>the</strong> preimplantation mammalian embryo.Although mouse embryos do not produce mRNA encodingIGF-I until after implantation (1), mRNA encoding <strong>the</strong> IGF­I receptor is present in mouse embryos from <strong>the</strong> 8-cell stage(2). Studies of uterine flushings from ewes at days 10-16 ofpregnancy have shown <strong>the</strong> presence of immunoreactive IGF­I (3). indicating that <strong>the</strong> ovine embryo is in <strong>the</strong> presence ofIGF-I prior to implantation. This study aimed toinvestigate <strong>the</strong> expression patterns of mRNA encoding IGF­I in <strong>the</strong> endometrium of early pregnant ewes.MATERIALS AND METHODSEndometrium and uterine flushings were collected fromMerino ewes at oestrus (day 0) and on days 3, 6~ 8, 15 and20 of pregnancy. There were 5 ewes per group except onday 20 where n=4. Uterine flushings were examined <strong>for</strong> <strong>the</strong>presence of an embryo to confrrm pregnancy. Ewes wereincluded in this study if an embryo was found in <strong>the</strong> uterineflushings.Endometrium was immediately snap-frozen and stored at-70°C. Nor<strong>the</strong>rn blotting was per<strong>for</strong>med to screen <strong>for</strong>mRNA transcripts <strong>for</strong> IGF-I using sheep liver as a positivecontrol <strong>for</strong> IGF-I mRNA expression. Total RNA wasextracted using <strong>the</strong> Guanidine lsothiocyanatelCsCI cushionmethod. Total RNA (15 mg) was electrophoresed on 1%agarose/<strong>for</strong>maldehyde gels be<strong>for</strong>e transfer to nylonmembranes (Hybond-N+; Amersham. Australia).Membranes were hybridised with a cDNA probe encodingpart of <strong>the</strong> protein coding region <strong>for</strong> ovine IGF-I. The probewas labelled with 32p-dCTP using random primingprocedures. Prehybridisation was <strong>for</strong> 30 min at 65°C inRapid Hybridisation Buffer (Amersham, Australia) andhybridisation was <strong>for</strong> 3 hours 65°C in Rapid HybridisationBuffer (Amersham, Australia) with <strong>the</strong> addition of <strong>the</strong> 32p_labelled probe. Blots were washed to a stringency of 0.1 xSSC/0.1 % SDS at 65°C prior to autoradiography <strong>for</strong> -6days.Fold differences in IGF-I mRNA signals were quantitatedusing scanning densitometry. Signals <strong>for</strong> IGF-I mRNAwere adjusted <strong>for</strong> RNA sample loading differences determinedfrom densitometric analysis of photographs of <strong>the</strong> 28Sribosomal RNA bands on gels stained with Ethidiumbromide prior to transfer. Signals <strong>for</strong> IGF-I mRNA werealso adjusted <strong>for</strong> differences between autoradiographs. Datafrom one ewe on both days 3 and 6 of pregnancy wereexcluded from analysis due to poor preparation of samples.Data were analysed by Analysis of Variance to evaluate <strong>the</strong>effect of day ofpregnancy on expression of IGF-I mRNA.RESULTSA major transcript of -8.3 kb in size encoding IGF-I wasdetected in <strong>the</strong> endometrium of early pregnant ewes. Therewas an effect (p-0.002) of day of pregnancy on <strong>the</strong>expression of IGF-I mRNA. The highest level of expressionof IGF-I mRNA occurred on <strong>the</strong> day of oestrus whereas <strong>the</strong>lowest level occurred on day 15 of pregnancy. The temporalvariation in expression of IGF-I mRNA~ relative to <strong>the</strong>mean value recorded on day 15, is shown in Figure 1.Relatively high IGF-I mRNA expression at oestruscompared to various stages of <strong>the</strong> cycle and pregnancy havebeen previously reported in endometrium of <strong>the</strong> cow (4).Our study extends <strong>the</strong>se findings to early pregnancy in <strong>the</strong>sheep~ suggesting a role ofovarian steroids in <strong>the</strong> regulationof IGF-I mRNA. Previous studies have also shown thatuterine flushings from ewes on day 16 of pregnancy havelower concentrations of immunoreactive IGF-I compared toewes on day 10 ofpregnancy (3). We have also shown verylow expression ofIGF-I mRNA in <strong>the</strong> endometrium of eweson day 15 of pregnancy. Fur<strong>the</strong>r studies are underway todetermine if <strong>the</strong> present observations are pregnancy specific~but it appears that <strong>the</strong> syn<strong>the</strong>tic capacity of <strong>the</strong> uterus toproduce IGF-I may playa role in <strong>the</strong> development of <strong>the</strong>ovine embryo.REFERENCESo 3 6 8 15 20Day of PregnancyFigure 1: Changes in endometria! IGF-I mRNAe~ cillirge8rty pregl8I1Cy In <strong>the</strong> ewe. Values are expr8ssed re&aliw 10 <strong>the</strong>mean varue <strong>for</strong>-IGF-I mRNA expression on day 15 of pregl8I1Cy.Values presented are means; wrticaJ lines are $EM.DISCUSSION(1) Heyner. S., Rao~ V., Jarrett~ L.• & Smith, R.M. (1989)Developmental Biology 134:48-58.(2) Heyner, S., Smith~ R.M.~ & Schultz, G.A. (1989)Bioessays 11:171-176.(3) Ko~ Y.~ Lee, C.Y.~ Ott, T.L., Davis, M.A.• Simmen.R.C.M., Bazer, F.W., and Simmen, F.A. (1991) Biology ofReproduction 45: 135-142.(4) Geisert. R.D., Lee. C-Y., Simmen. F.A.~ zavy. M.T.,Fliss, A.E.~ Bazer, F.W. & Simmen. R.C.M. (1991)Biology ofReproduction 45:975-983.INTRODUCTIONInsulin-like growth factors (IGFs) -I and -II belong toa family of peptide hormones that includes relaxinand insulin and share a high degree of structuralsimilarity with proinsulin(1). They are importantmitogenic peptides and are significant autocrineparacrinefactors in cell proliferation(2). They arecomplexed to a family of at least six binding proteins(IGFBPs) which modulate <strong>the</strong>ir mitogenic activity invivd 3 ).Recently it has become apparent that <strong>the</strong>bioavailability of IGF is influenced not only by <strong>the</strong>IGFBPs <strong>the</strong>mselves but also by specific IGFBPproteases(4). IGF-I and -II are present in <strong>the</strong> uterinelumenal fluid of sheep and IGFBPs of 16-18 kDa and22-24 kDa, specific <strong>for</strong> IGF-II, are seen in bothpregnant and non-pregnant sheep only on days 10­15 after oestrus. The aim of <strong>the</strong> present study wasto determine whe<strong>the</strong>r <strong>the</strong>se low molecular weightIGFBPs were proteolytic fragments of an unidentifiedparent IGFBP.MATERIALS AND METHODSTimed uterine flushings were obtained from pregnantand non-pregnant sheep. After centrifugation, <strong>the</strong>ywere lyophilized, reconstituted in sterile water andprotein concentration determined by <strong>the</strong> Brad<strong>for</strong>dmethod. Recombinant human (rh) non-glycosolatedIGFBP3 and rh IGFBP2 were iodinated usingiodogen. Labelled rh IGFBP3 and IGFBP2 wereincubated overnight at 37°C with uterine flushingsequivalent to 30 ~g protein followed by SDS-PAGEunder non-reducing conditions. The gels were driedand autoradiographed. In a separate experiment <strong>the</strong>proteinase inhibitors EDTA (10 mM) and PMSF(10 mM) were added.The flushings were also subjected to Weaternimmunoblotting using anti-b IGFBP2 and anti-hIGFBP4.RESULTSDay 15 uterine flushings from both pregnant and nonpregnantsheep reduced both rh IGFBP3 and IGBP2to lower molecular weight fragments; <strong>the</strong> <strong>for</strong>mer wascleaved to four fragments of 5.5, 14, 16 and 20­22 kDa, <strong>the</strong> latter to two fragments of 14 kDa and18 kDa. This proteolytic activity was inhibited byabout 70% by EDTA but was unaffected by PMSF.The activity was absent on days 3 to 10 and presenton days 12 to 15 after oestrus in both pregnant andnon-pregnant sheep. It disappeared in both casesfrom days 16 to 17 after oestrus.Although similar Western immunoblotting revealedIGFBP2 and IGFBP4 in ovine and bovine follicularfluid(s) negative results were obtained from <strong>the</strong> ovineuterine flushings.DISCUSSIONThe profile of proteolytic activity suggests that in bothpregnant and non-pregnant sheep it is underprogesterone control. Thus in <strong>the</strong> latter it appearsduring <strong>the</strong> luteal phase and disappears duringluteolysis. In <strong>the</strong> <strong>for</strong>mer it also disappears despiteelevated plasma progesterone levels, suggesting that<strong>the</strong> conceptus is secreting a factor that inhibits IGFBPproteolysis. The physiological significance of <strong>the</strong>IGFBP2 and 3 proteases in uterine fluid is unknowna~hough <strong>the</strong>y appear to be divalent cation dependent.The parent binding proteins <strong>the</strong>mselves have notbeen detected in sheep and <strong>the</strong> source and regulationof <strong>the</strong> IGFBP proteases must await purification andsubsequent identification.REFERENCES1. Daughadey, W., Rotwein, P. (1989) Endocr. Rev.10: 68-91.2. Rechler, M.M., Nissley, S.P., Roth, J. (1987) N.Engl. J. Med. 316: 941-3.3. Rechler, M.M., Brown, A.L. (1992) Growth Reg.2: 55-68.4. Binoux, M. (1994) Growth Reg. 4 (SuppI.1): 53.5. Peterson, A.J., Ledgard, A.M., Hodgkinson, S.C.(1994) Growth Reg. 4 (SuppI.1): 113.55 56


PROSTATE-SPECIFIC ANTIGEN Ar;.JD INSULIN-LIKE GROWTH FACTOR BINDING PROTEIN-3 ARECO-LOCALIZED IN THE HUMAN ENDOMETRIUM: PATTERNS OF EXPRESSION ACROSS THEMENSTRUAL CYCLE.Judith Clements. Allison Ehrlich, Marion Marsh and Lois Salamonsen.Prince Henry's Institute of Medical Research, PO Box 152, Clayton, Vic. 3168.IntroductionProstate-specific antigen (PSA) is a member of <strong>the</strong>human kallikrein (KLK) gene family of serine proteasesand is encoded by <strong>the</strong> gene KLK3. We have recentlydemonstrated that KLK3 and two o<strong>the</strong>r KLK genes(KLK1 and 2) are expressed in <strong>the</strong> human endometrium(1). Seminal PSA has been shown to degrade <strong>the</strong>insulin-like growth factor (IGF) binding protein, IGFBP-3,so that it is no longer capable of binding to IGF-I or IGF­II (2). These growth factors have been implicated inei<strong>the</strong>r <strong>the</strong> proliferative response mid-cycle (IGF-I) ordecidual cell differentiation (IGF-II) in <strong>the</strong> humanendometrium (3). PSA may thus act to regulate <strong>the</strong>bioavailability of <strong>the</strong> IGFs in <strong>the</strong> uterus as has beensuggested <strong>for</strong> <strong>the</strong> prostate. In this study, we wished todefine to which cell-type(s) PSA localized in <strong>the</strong> humanendometrium, its pattern of expression across <strong>the</strong>menstrual cycle, and whe<strong>the</strong>r it was co-localized withIGFBP-3.Materials and methodsEndometrial tissue (n=31) was collected from essentiallynormal cycling women (cycle days 2-28) undergoingdilatation and curettage during routine investigation ofinfertility and was histologically dated. Tissue blocksused <strong>for</strong> PSA immunocytochemistry were divided into 5arbitrary phases of <strong>the</strong> menstrual cycle: menstrual (day2-4, n=4), proliferative (day 6-12, n=11), and early (day17, n=4), mid (day 21-24, n=8) and late (day 25-28, n=4)secretory phases. Ten of <strong>the</strong> 31 tissue blocks werechosen <strong>for</strong> subsequent immunocytochemistry <strong>for</strong>IGFBP-3. Immunocyto-chemistry was per<strong>for</strong>med withcommercially available antibodies to PSA (Dako) andIGFBP-3 (UBI) using <strong>the</strong> avidin-biotinylated horseradishperoxidase complex method (Vectastain ABC kit).Intensity of staining was graded, by 3 independentobservers, using a scale of 0-4 to indicate absence ofstaining (0) to intense staining (4).ResultsPSA was localized primarily to <strong>the</strong> glandular and luminalepi<strong>the</strong>lium and was also present in secretions fromindividual glands. There was less staining in <strong>the</strong> stroma.There was no apparent difference in staining intensity orpattern of localization across <strong>the</strong> menstrual cycle (Fig.1). The endo<strong>the</strong>lium and/or <strong>the</strong> smooth muscle cells ofendometrial blood vessels were also positive <strong>for</strong> PSA tovarying degrees, but with no consistent pattern,throughout <strong>the</strong> cycle. IGFBP-3 was localized to <strong>the</strong>glandular and luminal epi<strong>the</strong>lial cells, stromal cells andendometrial blood vessels in a similar fashion to that ofPSA (Fig. 2).References1. Clements J & Mukhtar A. 1994. J Clin Endocrinol Metab 78:1536-1539.2. Cohen P, Graves HCB, Peehl DM, Kamarei M, Guidice LC,Rosenfeld RG. 1992. J Clin Endocrinol Metab 75: 1046-1053.3. Seppala M 1994. In Molecular biology of <strong>the</strong> femalereproductive system. Academic Press p379Fig. 132.5o~ 2en£1.5encQ)E 10.5Fig. 2PSA3-2.5-~ 2-;:;:Mens.(4)Prolif.(11 )o glandularepi<strong>the</strong>lium~ ::::>'1 5 .....~ . -:::: ITC .;.; II~ 0.: ~.::!·.!:i=.:::~.i,.:.:. •.:::.::.• ::.:i••iUla-'-'-",""",,"L-.,&;:;';": :.c.::::;w:.I}:--.I.':"':'=Mens. Prolif. Sec.(1) (5) (4)Early(4)Mid.(8)IGFBP-3Late(4)SecretoryI28l luninal ED stromaepi<strong>the</strong>liumII!IMens. Prolif. Sec.(1) (5) (4)Staining intensity scores <strong>for</strong> PSA alone (Fig. 1)and PSA/IGFBP-3 co-localisation (Fig. 2) in <strong>the</strong>menstrual', proliferative and secretory phases of<strong>the</strong> menstrual cycle. n/group is indicated inparen<strong>the</strong>ses below each figure.Summary and ConclusionPSA and IGFBP-3 are co-localized to <strong>the</strong> same celltypesin <strong>the</strong> human endometrium. These data suggestthat PSA, as in <strong>the</strong> prostate, may function as aprocessing enzyme <strong>for</strong> IGFBP-3 in <strong>the</strong> uterus to regulate<strong>the</strong> bioavailability of IGF-I and IGF-II. The lack of anyapparent change in intensity of staining <strong>for</strong> PSA or itssubstrate, IGFBP-3, across <strong>the</strong> menstrual cyclepresumably reflects <strong>the</strong> need <strong>for</strong> bioactive IGF-I and/orIGF-II in <strong>the</strong> human endometrium at all phases of <strong>the</strong>menstrual cycle.GM-CSF mediates <strong>the</strong> post-mating inflammatory reaction in <strong>the</strong> murine uterusSarah A Robertson, Anna C Seamark and Robert F SeamarkDepartment of Obstetrics and Gynaecology, The University of Adelaide, Adelaide, SA 5005INTRODUCTIONGranulocyte-macrophage colony-stimulating factor (GM­CSF) is a lymphohaemopoietic cytokine syn<strong>the</strong>sised byoestrogen-primed epi<strong>the</strong>lial cells in <strong>the</strong> murine uterus. Adramatic but transient peak in its release is evoked by seminalplasma-mediated stimulation of epi<strong>the</strong>lial cells at mating1.In recent studies we have investigated <strong>the</strong> role of GM­CSF in <strong>the</strong> pre-implantation uterus. Transcripts <strong>for</strong> both <strong>the</strong> ex.and ~ components of <strong>the</strong> GM-CSF receptor have beendetected by RT-PCR in uterine tissue at oestrus and during<strong>the</strong> pre-implantation period, with expression being severalfold higher following mating than at oestrus (S. Robertson,unpublished). In inflammatory sites, GM-CSF regulates <strong>the</strong>recruitment and activation of myeloid leukocytes, and cellsexpressing GM-CSF receptor in <strong>the</strong> day 1 pregnant uterushave been identified as leukocytes on <strong>the</strong> basis of <strong>the</strong>irpredominantly sub-epi<strong>the</strong>lial distribution in <strong>the</strong> endometrium,using autoradiography following incubation of ethanol-fixedfrozen tissue sections in 125I-GM-CSF2. The aim of <strong>the</strong>current study was to determine whe<strong>the</strong>r epi<strong>the</strong>lial GM-CSFhas a role in <strong>the</strong> post-mating inflammatory response which ischaracterised by infiltration of large populations ofmacrophages, eosinophils and neutrophils into <strong>the</strong>subepi<strong>the</strong>lial endometrial stroma and luminal cavity.EXPERIMENTALTo investigate <strong>the</strong> effect of GM-CSF on <strong>the</strong> number anddistribution of uterine leukocytes, groups of fourovariectomised mice were anaes<strong>the</strong>tised and given anunilateral intraluminal injection of ei<strong>the</strong>r 40, 200, or 1000 Uof recombinant GM-CSF in 50 jll of 1% BSA, or carrieralone, through a dorsal incision. Sixteen hours later, <strong>the</strong> micewere sacrificed and <strong>the</strong> numbers and distribution ofleukocytes in right and left uterine horns were assessedimmunohistochemically using lineage-specific mAbs.RESULTSIn each of two experiments, <strong>the</strong> numbers of leukocytes inendometrial tissues were altered dramatically by intraluminalGM-CSF (Fig. 1). The most notable of <strong>the</strong> cellular changeswas a dose dependant effect of GM-CSF on <strong>the</strong> numbers ofeosinophils (endogenous peroxidase+) within <strong>the</strong> endometrialstroma and <strong>the</strong> myometrium (p = 0.005), with <strong>the</strong> numbers ofmacrophages and neutrophils (F4/80+ and Mac-l+ cells), andto a lesser degree activated macrophages/dendritic cells (Ia+)also increased in response to GM-CSF. F4/80+ and Ia+ cellswere distributed throughout <strong>the</strong> stroma but were mostconcentrated in areas underlying <strong>the</strong> epi<strong>the</strong>lium. Neutrophils(Mac-l+ F4/80- polymorphonuclear cells) were absent incarrier-treated mice but present in large numbers in GM-CSFtreated mice where <strong>the</strong>y were located predominantly betweenepi<strong>the</strong>lial cells in a pattern suggestive of trafficking into <strong>the</strong>luminal cavity, similar to that observed during <strong>the</strong> postmatinginflammatory response.Interestingly, GM-CSF also had a dose-dependant effecton both <strong>the</strong> number of endometrial glands and <strong>the</strong> diameter of<strong>the</strong> uterus, with a four-fold increase in <strong>the</strong> mean number ofglands (p = 0.0001), and a 70% increase in <strong>the</strong> mean uterinediameter (p =0.0001) after intraluminal injection of 200 U ofGM-CSF. Both <strong>the</strong> number of glands (r = 0.845, p = 0.0001)and uterine diameter (r = 0.859, p = 0.0001) correlated wit<strong>hth</strong>e number of endometrial eosinophils.No differences in leukocyte numbers or endometrialglands were found between injected and ipsilateral horns,most probably reflecting <strong>the</strong> contiguity of <strong>the</strong> lumen ra<strong>the</strong>rthan any systemic actions of th~ injected cytokine.Figure 1: Immunohistochemical localisation of leukocytes(LCA+ cells: A,B), eosinophils (endogenous peroxidase+cells: C,D) and macrophages / neutrophils (Mac-l+ cells: E,F)in <strong>the</strong> endometrium of ovariectomised mice treated ei<strong>the</strong>rwith 200U of rGM-CSF (B,D and F) or carrier (A,C and E)SUMMARYThese data identify specific populations of GM-CSFresponsivemyeloid leukocytes within <strong>the</strong> uterus, and support<strong>the</strong> view that GM-CSF produced by uterine epi<strong>the</strong>lial cellshas a physiological role in regulating <strong>the</strong> trafficking andbehaviour of <strong>the</strong>se cells to participate in <strong>the</strong> dramaticremodelling processes characteristic of this tissue during <strong>the</strong>oestrous cycle and during pregnancy.1. Robertson SA, Mayrhofer G and Seamark RF (1992),BioI. Reprod. 46, 1069-1079.2. Robertson SA, Mayrhofer G and Seamark RF (1993), inSerono Symposia 97 '<strong>Reproductive</strong> Immunology' (Dondero Fand Johnson P, eds), pp 117-122, Raven Press, New York.57 58


ENDOTHEllN CONTENT AND CHARACTERIZATION IN THE SHEEP UTERUS DURINGTHE OESTROUS CYCLE AND AT IMPLANTATIONSimon C Riley, Lois A Salamonsen & Jock K FindlavPrince Henry's Institute of Medical Research, PO Box 5152, Clayton, Victoria, 3168--Endo<strong>the</strong>lin in <strong>the</strong> Endometrium of Women with MenorrhagiaM.M. Marsh1, D.L. Healy2, J.K. FindlaY' & L.A. Salamonsen 1 • .1Prince Henry's Institute of Medical Research and 2Monash University Department ofObstetrics and Gynaecology Clayton Vic 3168.INTRODUCTIONEndo<strong>the</strong>lins (ET) are a family of three 21 amino acidpeptides with potent vasoconstrictors and mitogeniceffects. ETs have been demonstrated in <strong>the</strong> ovineuterus in luminal and glandular epi<strong>the</strong>lial cells usingimmunohistochemical techniques (1) with <strong>the</strong> greateststaining intensity at <strong>the</strong> time of implantation.However, <strong>the</strong>ir role in <strong>the</strong> control of ovine uterinefunction has not been defined. These studies havecharacterised and measured <strong>the</strong> content of ET within<strong>the</strong> ovine uterus during <strong>the</strong> oestrous cycle and earlypregnancy around <strong>the</strong> time of implantation to try andelucidate <strong>the</strong> role of ET in <strong>the</strong>se processes.MATERIALS AND METHODSUteri of Corriedale ewes (n=75) were flushed withsaline and removed by hysterectomy. Pregnancy wasconfirmed by <strong>the</strong> presence of an embryo.Endometrium and myometrium were dissected andfrozen. Tissues were homogenised in 1M acetic acidand <strong>the</strong> homogenates were boiled, centrifuged and<strong>the</strong> supernatant collected and Iyophylized. ET contentof <strong>the</strong> extracts and flushings were measured by aradioimmunoassay that recognises ET-1, -2 and -3equally, but not <strong>the</strong> precursor big ET. The iso<strong>for</strong>m(s)of ET present in <strong>the</strong> uterus were characterised byreverse-phase HPLC.RESULTSET-1 was <strong>the</strong> principal ET iso<strong>for</strong>m present in bothtissue extracts and uterine flushings. In tissueextracts <strong>the</strong>re was no significant change in ET contentduring <strong>the</strong> oestrous cycle, but during pregnancy, ETcontent significantly increased on and after Day 10-12(Table 1).TABLE 1 Amount of IR-ET (±SEM; pg g-1n=4/group) in endometrial extracts.Period Day IR-ETOestrous 4 327 ±9S acycle12 476±97 a16 629 + 129 aPregnancy 4 279 +40 a10-12 453 +39 015-17 477+25°20 616 +43 cwt wt;Within anyone group, values denoted with <strong>the</strong> samesuperscript are not significantly (P>0.05) different.In uterine flushings, <strong>the</strong> amount of ET increased significantlyon Day 14 of <strong>the</strong> oestrous cycle <strong>the</strong>ndecreased by Day 16 (Fig. 1). During early pregnancy,ET content in flushings increased on Day 14and increased fur<strong>the</strong>r on Days 15 and 16 (Fig. 1).CONCLUSIONSET-1 was present in <strong>the</strong> ovine uterus in tissue extractsand in uterine flushings in increasing amounts around<strong>the</strong> time of implantation (Day 16).FIGURE 1. Immunoreactive ET content (±SEM) inuterine flushings of sheep during <strong>the</strong> oestrous cycleand early pregnancy.300022500~.... 2000gg. 1500~ 1000~a,b4 10 12 14 16 4 7 8 9 10 12 14 15 16oestrous cycleearlyDaypregnancyWithin anyone group, values denoted with <strong>the</strong> samesuperscript are not significantly (P>0.05) different.REFERENCES(1) Riley et aJ (1994). J. Reprod. Fert. 100: 451­459.(2) Marsh et al (1994). J. Clin. Endo. Metab. (inpress).Supported by <strong>the</strong> NH&MRC.This is <strong>the</strong> sameiso<strong>for</strong>m as produced by <strong>the</strong> human endometrium invitro (2), and is present in <strong>the</strong> uterus in similarconcentrations to that found in o<strong>the</strong>r tissues where afunction <strong>for</strong> ET has been demonstrated. ET mayhave a role in <strong>the</strong> control of uterine blood flow duringimplantation. ET may elicit a mitogenic effect on <strong>the</strong>blastocyst during <strong>the</strong> phenomenal expansion thatoccurs between Days 13-16, and on <strong>the</strong> endometriumin association with <strong>the</strong> remodelling and angiogenicresponses related to implantation and placentaldevelopment.x,IntroductionEndo<strong>the</strong>lin (ET) a growth factor and potentvasoconstrictor (1) is present in human endometriumthroughout <strong>the</strong> normal menstrual cycle. Expression ofET protein and mRNA shows cyclical variation,maximum levels being observed in <strong>the</strong> premenstrualphase (2). The aim of this .study was t? ex~mi~e <strong>the</strong>cellular localization of ET In endometnal bIopSies ofwomen with documented menorrhagia and to comparethis to that observed in <strong>the</strong> normal menstrual cycle.Materials and MethodsEndometrial biopsies (n=17) were obtained within<strong>for</strong>med consent from women who presented withmenorrhagia confirmed by a monthly menstrual bloodloss greater than 80 mls. These samples were classifiedhistologically according to Noyes criteria (3) intoproliferative (n=6), early secretory (n=3), mid-secretory(n=4) and late secretory phase (n=4). Normalendometrium (n=55; proliferative n=21 , early secretoryn=11, mid secretory n=10, late secretory n=5 andmenstrual n=8) was obtained from women of provenfertility undergoing laparoscopic sterilisation or womenundergoing assessment of tubal patency. Tissue wasfixed in 10% buffered <strong>for</strong>malin, paraffin embedded,sectioned and subjected to immunohistochemistry usinga polyclonal antibody to ET (cross-reactive with ET-2 &ET-3) and <strong>the</strong> components of <strong>the</strong> StrepAviGensupersensitive kit (2) and counterstained with Mayer'shaematoxylin (Sigma). ET immunoreactivity was scoredby two independent observers in relation to knownpositive and negative controls included in each staining.Individual cellular compartments within each tissuesection were scored from 0 (negative) to 4 (maximalstaining intensity) and trans<strong>for</strong>med to a percentageimmunostaining intensity where negative = 0 and 4 =100%. Mean immunostaining intensity ± SEM wascalculated <strong>for</strong> each cellular compartment within eachphase of <strong>the</strong> menstrual cycle.ResultsET immunoreactivity in <strong>the</strong> endometrium from thosewomen with menorrhagia (Fig 1) is substantially differentfrom that of <strong>the</strong> normal menstrual cycle (Fig 2).Compared to normal endometrium in menorrhagia <strong>the</strong>reis little variation in <strong>the</strong> immunostaining pattern across<strong>the</strong> menstrual cycle. ET immunoreactivity is less intensein glandular and luminal epi<strong>the</strong>lium than that observedat <strong>the</strong> same stage of <strong>the</strong> normal menstrual cycle, andalthough <strong>the</strong> stromal ET immunoreactivity is of similarintensity to <strong>the</strong> normal menstrual cycle, at all stages itremains more intense than that of luminal and glandularepi<strong>the</strong>lium.Acknowledgments: Supported by <strong>the</strong> NH & MRCFigure 1:e 'E 100o()~0) 75c::'c 'cu(j) 50o c:::::JE.§ 25I­ w§O-+J~"""""(l)~Figure 2:--- e 'E 100o()~(6, 2, 4, 4) (6, 3, 4, 4)Luminal Epi<strong>the</strong>lium Glandse..... 750)c::'c 'cu(j) 50o c:::::JE.§ 25Iii~(l)0 -+-J:......&.t-(21,11,10,6,8) (9, 5, 4, 5, 8) (20,10,10,5,8)~ Stroma Luminal Epi<strong>the</strong>lium GlandsFigure LegendsCellular localization of ET immunoreactivity (Mean±SEM)across <strong>the</strong> menstrual cycle - D Proliferative, [21 EarlySecretory, II Mid Secretory, E8I Late Secretory, and •Menstrual phase. Numbers below each bar denote <strong>the</strong> numberof samples in each cellular compartment. Endometrial biopsiesfrom subjects with menorrhagia (fig1 ) and normal endometrium(fig 2).Summary & ConclusionsThe immunostaining pattern of ET in <strong>the</strong> endometriumin menorrhagia exhibits a different pattern to thatobserved during <strong>the</strong> normal menstrual cycle. Inmenorrhagia lower levels of immunoreactivity areobserved at all stages of <strong>the</strong> menstrual cycle in luminaland glandUlar epi<strong>the</strong>lium compared to <strong>the</strong> normalmenstrual cycle where maximal staining intensity isobserved in <strong>the</strong> premenstrual phase. This difference isconsistent with a role <strong>for</strong> ET in <strong>the</strong> control of menstrualbleeding. Fur<strong>the</strong>r studies are being per<strong>for</strong>med toelucidate <strong>the</strong> function of ET in normal and abnormalmenstrual bleeding.References1. Masaki, T. (1993) Endocrine Reviews 14:256~268.2. Salamonsen L.A., et aI., (1992) Am J Obstet Gynecol.,167: 163-167.3. Noyes, R.W., et aI., (1950) Fertility & Sterility 1:3~25.59 60


· LEUKAEMIA INHIBITORY FACTOR (LIF) IN OVINE ENDOMETRIUM DURING THEOESTROUS CYCLE AND EARLY PREGNANCYDaphne Vogiagis1,2, Richard C. Fry2 & Lois A. Salamonsen1.1 Prince Henry's Institute of Medical Research, PO Box 152, Clayton, Vic, 3168.2 Victorian Institute of Animal Science, Werribee, Vic, 3030.THE USE OF THE POINT SM1PLED INTERCEPT F.M1ILY OF N1ETIIODS FOR TIIEESTIMATION OF SERTOLI CELL NUCLEAR VOLUME.Nigel G Wre<strong>for</strong>d l • 2 andAnne O'Connor l ,Institute ofReproduction and Development! and Dept. ofAnatomy 2, Monash University,Wellington Rd. Gayton, Victoria 3168 , AustraliaINTRODUCTIONLeukaemia inhibitory factor (L1F) is a cytokine with abroad range of actions on various in vitro culturesystems. In <strong>the</strong> mouse its uterine expressioncoincides with <strong>the</strong> onset of blastocyst implantationand females lacking a L1F gene are fertile but <strong>the</strong>irblastocysts fail to implant (1,2). This study wasdesigned to investigate <strong>the</strong> expression and cellularlocalisation of L1F in <strong>the</strong> ovine endometrium, during<strong>the</strong> oestrus cycle and early pregnancy. Specifically itwas of interest to establish whe<strong>the</strong>r L1F is temporallyassociated with ei<strong>the</strong>r embryo hatching, developmentor blastocyst implantation. Previously we havelocalised L1F immunohistochemically in humanendometrium throughout <strong>the</strong> menstrual cycle wheremaximal staining was detected in <strong>the</strong> mid-latesecretory phase particularly in <strong>the</strong> glands prior tomenstruation (3).MATERIALS AND METHODSParous Corriedale ewes (n=56) were mated wi<strong>the</strong>i<strong>the</strong>r castrate or fertile rams (day 0 = oestrous) andhysterectomised on given days of <strong>the</strong> oestrous cycle(days 4-16) or early pregnancy (days 4-20) (n=3-4per day). Endometrial tissues were snap frozen <strong>for</strong>RNA extraction and fixed in Carnoy's fixative <strong>for</strong>immunohistochemistry. Ovine endometria (total RNA,20l-lg per lane) were tested <strong>for</strong> L1F expression byNor<strong>the</strong>rn blot analysis using an ovine L1F eDNA probe(0.6-kilobase) a generous gift from WEHI. Thecorresponding protein was localised in <strong>the</strong>se tissuesby immunohistochemistry with a commerciallyavailable rabbit anti-human L1F antibody whichrecognises both native and recombinant human L1F(AMRAD, Pharmacia Biotech, Victoria). TheVectastain ABC immunostaining kit was used <strong>for</strong>detection. Intensity of immunostaining was graded bytwo observers, to indicate absence of staining (0) tointense staining (4).RESULTSImmunoreactive L1F was present in all endometrialsamples and was markedly diminished followingpreadsorption of <strong>the</strong> antibody with humanglycosylated L1F. Positive immunoreactivity waspresent in all cellular compartments of <strong>the</strong>endometrium (intercaruncular luminal and glandularepi<strong>the</strong>lium and stroma, caruncularluminal epi<strong>the</strong>liumand stroma, blood vessels and myometrium).However, staining intensity was low in <strong>the</strong> bloodvessels and myometrium. In <strong>the</strong> majority of <strong>the</strong>cellular compartr'f1ents <strong>the</strong>re was a maximal increaseof L1F immunoreactivity at day 12 of <strong>the</strong> oestrouscycle and early pregnancy (Fig. 1 & 2).Nor<strong>the</strong>rn analysis also demonstrated <strong>the</strong> expressionof <strong>the</strong> 4.2-kilobase L1F messenger RNA transcript inall samples of ovine endometrium, confirming <strong>the</strong>immunohIStochemical data.Figuresl.A 2.A4 43 3>.~c2 2~ 1 1go 0'c'(ij1.8 2.8"@ 4 4c~ 3 3§221 10 0I I I4 10 12 14 16 4 7 8 9 10121415161720DayDayDistribution and relative intensity of positiveimmunoreactivity <strong>for</strong> L1F in ovine endometrium during <strong>the</strong>oestrous cycle (Fig. 1) and early pregnancy (Fig. 2). A;caruncular, luminal epi<strong>the</strong>lium, B; intercaruncular stroma.CONCLUSIONSIn <strong>the</strong> ovine endometrium, expression of L1F is notconfined to <strong>the</strong> time of blastocyst implantation as it isin <strong>the</strong> mouse. We postulate that in <strong>the</strong> sheep, L1Fmay have a paracrine role in endometrial function andblastocyst development since it is expressed andlocalised throughout early pregnancy and <strong>the</strong>oestrous cycle. Whe<strong>the</strong>r L1F is essential <strong>for</strong>implantation in <strong>the</strong> sheep, as has been demonstratedin <strong>the</strong> mouse remains to be established.REFERENCES1. Bhatt, H. et al. (1991) Proc. Natl. Acad, Sci. USA.88:11408-12.2. Stewart, C.L. et al. (1992) Nature. 359:76-79.3. Vogiagis, D. et al (1994) Proc. Endo. Soc. Aust.(abst).DV is supported by <strong>the</strong> Victorian EducationFoundation.LAS is supported by NH&MRC of Australia.INTRODUCTIONThe volume of a nucleus or cell is animportant but little used parameter indicatingits functional activity. In combination withvohune density data, it can also be used toobtain numerical density. Traditionally,nuclear volume has been detennined using ageometric model based approach. This workswell in <strong>the</strong> case ofspherical nuclei but is notsuited to irregular or oval nuclei. Thealternative method of serial reconstructionwith sunnnation of areas has been usedextensively in <strong>the</strong> testis but is extremely timeconsuming and dependent on an accuratedetennination of section thickness.The recent development of<strong>the</strong> point sampledintercept (psi) family ofmethods (1,2&3) hasprovided <strong>the</strong> basis <strong>for</strong> <strong>the</strong> rapid and unbiasedestimation of nuclear and cell vohune. Themethods can be applied in 2 contexts. In <strong>the</strong>:first, nuclei are assumed to be isotropicallyoriented (ie. as a group have similar geometricproperties in all directions) and in <strong>the</strong> second(vertical section method), no assumptions aremade with respect to spatial orientation andnuclei may be anisotropic (ie have differentgeometric properties in different directions).With both approaches, nuclei can be sampledin ei<strong>the</strong>r <strong>the</strong> vohune (psi) or nmnberdistributions (nucleator and rotator) to giveestimates of volume (vv) or nmnber (v N )weighted mean volume.In this report, <strong>the</strong>se methods are appliedindependently to <strong>the</strong> estimation ofSertoli cellnuclei.MATERIALS AND 11ETHODSSections (1~) were obtained from Epon­Araldite embedded glutaraldehyde fixed rat(120 days) testis (n=5). Two sectionorientations were used. In <strong>the</strong> first, <strong>the</strong> usualtransverse sections were prepared whilst in <strong>the</strong>second sections were cut parallel to <strong>the</strong> longaxis (vertical sections). Vertical sections weresampled to allow different rotations in <strong>the</strong>horizontal plane (2). Sections were sampledusing a motorised stage to give systematicuni<strong>for</strong>m random fields. Nuclei were sampledwith a point grid (i.e. with a probabilityproportional to <strong>the</strong>ir volmne) <strong>for</strong> <strong>the</strong>detennination of Vv and unifonnly using <strong>the</strong>nucleolus <strong>for</strong> determination of v N .Measurements were per<strong>for</strong>med on a videoimage at a magnification of 2708x using aninteractive software package (GRID,Medicosoft, Denmark). This package willgenerate a variety of grids as well assystematic sets of lines of appropriate (sineweighted) directions <strong>for</strong> application to verticalsections.RESULTSl\1ean ± sd Setfuli cell nuclear vol~ (,mf)~~uredltiing different ~dtods.Assumption Isotropy NoneSectionOrientationVv (Psi)V N (nucleator)V N (rotator)TransverseVertical666 ± 76 m,± 105553 ± 42 612± 55672±48DISCUSSIONThe assumption of isotropy carries aconsistent negative bias in both <strong>the</strong> psi andnucleator estimates. There is reasonableagreement between <strong>the</strong> nucleator and rotatorestimates applied to vertical sections. Thesquare root of <strong>the</strong> ratio (Vv/v N ) minus 1 (i.e.0.4-0.5) is equal to <strong>the</strong> coefficient ofvariationofnuclear volmne in <strong>the</strong> number distributionsuggesting a substantial intra animal variationin Sertoli cell volume at this stage. Thequestion of whe<strong>the</strong>r this variation is stagedependent remains to be investigated.(1) Gundersen, R1.G. (1993) J. Microsc. 170: 3-45(2) Baddeley, AJ. (1986) 1. Microsc. 142: 259-276.(3) Vedel Jensen, E.B. & Gundersen RJ.G (1993) 1.Mcrosc.170: 35-44.61 62


SITES OF SYNlHFSIS AND ACIION" OF PDGF IN 1HE RAT TESTISKate Loveland, Kristina Zlatic, Gail Risbridger, Alicia Stein-Oakley* and David deKretserInstitute of Reproduction and Development and *Dept ofMedicineMonash University, Clayton, VictoriaIntroduction: The importance of platelet-derived growth Figure: Nor<strong>the</strong>rn blots hybridized with cDNAsfactor (pOOF) in growth and differentiation has been encoding POOF ligand and receptor subunits. Eachdocumented in a wide variety of normal and pathological lane contains 10J.lg of total RNA from (a) Leydig cells,systems, though only recently has expression of receptors (b) Sertoli cells, (c) cytoplasts, (d) primary,<strong>for</strong> this growth factor been described in <strong>the</strong> adult testis. spennatocytes, (e) round spermatids. Positions ofBinding sites <strong>for</strong> POOF have been described on rRNA bands are indicated.preparations ofrat Leydig (I) and peritubular myoid cells(2), and mRNA encoding <strong>the</strong> a-subunit of <strong>the</strong> POOF abc d ereceptor (3) has been detected in isolated rat Leydig andSertoli cells. POOF has been implicated in regulation oftestosterone production by Leydig cells (3,4) and in PDGF-Ramodulation of peritubular myoid cell function in vitro(2,5). To more fully explore <strong>the</strong> role of POOF in <strong>the</strong>testis, we sought to determine <strong>the</strong> sites of its syn<strong>the</strong>sisand action. ....288EXTRACELLULAR MATRIX PRODUCTION BY BOVINE GRANULOSA CELLSH.F. Rodgers*, T.C. Lavranos, C.A. Vella, K. Scott and R.I. RodgersDepartments ofMedicine, and *Anatomy and Histology,Flinders University ofSouth Australia, Bed<strong>for</strong>d Park, S.A., 5042, Australia.Bovine granulosa cells were cultured underanchorage-independent conditions (1), and <strong>the</strong>ECM produced by <strong>the</strong>se cells examined bymicroscopy and Western immunoblotting.The colonies of cells (Fig 1, x190) producedan ECM (*) identified as basal lamina collagentype IV (~) by immunohistochemistry (Fig 2,x190).Materials and Met/rods: Nor<strong>the</strong>rn blot analysis wasper<strong>for</strong>med using 32p random· primed cDNA probesspecific <strong>for</strong> each of <strong>the</strong> POOF ligand (A and B) andreceptor (pOOF-Ra and POOF-Rf3) subunits. Leydigcells, pachytene spermatocytes, round spermatids, andcytoplasts were isolated from adult rats as previouslydescribed (3); Sertoli cells from 20 day old rats (3) werecultured in serum-free medium <strong>for</strong> 24 hours prior tocollection. RNA was prepared as described (3), and 10~g of total RNA was loaded in each lane of afonnaldehyde agarose gel. RNA was transferred toHybond N membrane (Amersham) and hybridizationper<strong>for</strong>med using Rapid Hyb (Amersham). Membraneswere washed to O.2X SSC, 0.1% SDS at 65°C.PDGF-~PDGFB---288Fibronectin was detected by immunostaining andWestern immunoblotting (Fig 5).Results: The POOF-Ra cDNA hybridized to mRNAs inLeydig and Sertoli cells,confrrming our previous work(3). No signal was evident in <strong>the</strong> germ cell preparationswith this probe. In addition, <strong>the</strong> Leydig cells containmRNA encoding both POOF-Rf3 and POOF B, althoughreactivity with <strong>the</strong> POOF A cDNA was barely detected.The Sertoli cell preparations contained mRNA whichhybridized with cDNAs encoding <strong>the</strong> POOF-Rf3, POOFB and POOF A cDNAs. No cross-reactivity of any of<strong>the</strong>se probes with germ cell mRNAs was detected. Allbands observed were of expected sizes.Discussion: Our results demonstrate that bothSertoli cells and Leydig cells have <strong>the</strong> capacity tosyn<strong>the</strong>size POOF and to respond to this growth factor.Thus <strong>the</strong> role of POOF in <strong>the</strong> testis is likely to be muchmore extensive than has previously been appreciated, asboth paracrine and autocrine interactions are likely to beinvolved. POOF thus has <strong>the</strong> potential to mediatecommunication between <strong>the</strong> interstitial and seminiferousepi<strong>the</strong>ilium compartments.PDGFAReferences:(1) Gnessi L, Emidi A, Farini D, Scarpa S, Modesti A,Ciampani T, Silvestroni L, Spera G (1992)Endocrinology 130:2219-24.2) Gnessi L, Emidi A, Scarpa S, Palleschi S, Ragano­Caracciolo M, Silvestroni L, Modesti A, Spera G(1993) Endocrinology 133: 1880-90.(3) Loveland KL, Hedger MP, Risbridger GP, HerszfeldD, deKretser DM (1993) Mol Reprod Dvlt 36: 440-7.(4) Risbridger GP (1993) Mol Cell Endo 97:125-8.(5) Tung PS, Fritz IB (1991) J Cell Physiol 146:386-3.Ru<strong>the</strong>nium red stalrung <strong>for</strong> proteoglycansrevealed spaces between cells (Fig 4a, x27500,b, x65000) that were rich in fibers decoratedwith ru<strong>the</strong>nium red (--»). Follicular fluid alsohas ru<strong>the</strong>nium red postive proteoglycangranules.ABC D EF. Lanes: A - preincubation, B - control, C - bFGF,D dibutyryl cArv1P, E - Matrigel, F - plasmafibronectinIn <strong>the</strong>se studies control colonies produced ECMand components, bFGF (50ng/ml) promoted<strong>the</strong>ir production and <strong>the</strong> cells remainedundifferentiated. Dibutyryl cAMP (1mM)inhibited ECM production and <strong>the</strong> cellsdifferentiated as determined by <strong>the</strong>irultrastructure, progesterone production andlevels ofsteroidogenic enzymes.Granulosa cells produce basal lamina whoseproduction can be regulated.1 Lavranos TC, Rodgers HF, Bertoncello I,Rodgers RJ (1994) Expt. Cell Res. 211, 245­251.63 64


Steroidogenesis Is Mediated By Heparan Sulphate ProteoglycansIn The Adult Rat Leydig CelLJim Mc Farlane, Andrew Laslett, Milton Hearn*, David de Kretser, & Gail Risbridger.Institute ofReproduction and Development and *Department ofBiochemistry, Monash University,Clayton, Victoria 3168.Introduction: LH stimulated androgen production by adult and immatureLeydig cells has been shown to be modulated by paracrine stimulatory andinhibitory factors. The aim of this study was to investigate <strong>the</strong> role cellsurface heparan sulphate proteoglycans might play in <strong>the</strong> regulation ofsteroidogenesis in <strong>the</strong>se cells.Materials & Methods: The Leydig cell bioassay used in this study has beenpreviously described l , briefly; cells are purified by centrifugal elutriation andPercoll density gradients, yielding Leydig cells at greater than 95% purity.Endogenous and exogenous heparin binding proteins were removed by <strong>the</strong>addition of heparin (275 J.lg/ml) to <strong>the</strong> media during purification The cellswere plated at 5 x 10 4 cells/well (48 well plates), preincubated <strong>for</strong> 1h in0.25ml ofDMEMJF12 containing 0.1 % BSA at 32°C, 5% CO 2 . This mediawas <strong>the</strong>n removed and replaced with fresh media containing 0.1mg/ml bovine.lipoproteins and test substances in <strong>the</strong> presence or absence of LH (16 ng/ml).The cells were incubated <strong>for</strong> a fur<strong>the</strong>r 20h and assayed <strong>for</strong> testosterone. Usingthis experimental protocol varying doses of heparin (10, 100, 200 ug/m1),protamine sulphate (0.1,1,10 ug/ml) an inhibitor of heparin binding growthfactor activity, and sodium chlorate (3.13, 6.25, 12.5, 25mM) which blocks<strong>the</strong> sulphation of<strong>the</strong> heparin sulphate proteog1ycans were examined <strong>for</strong> <strong>the</strong>ireffect on testosterone production..Results: Heparin increased <strong>the</strong> testosterone production in unstimulated cellsin a dose dependant manner, in some experilnents <strong>the</strong> stimulation was to alevel achieved by a maximum dose LH (Figure 1). The effect ofheparin onLH stimulated cells was variable, but was predominantly inhibitory at dosesof 100-200 ug/ml (50%). Protamine sulphate had no effect on unstimulatedLeydig cell testosterone at <strong>the</strong> highest dose used (10ug/ml), however in LHstimulated cells <strong>the</strong> testosterone production was inhibited by 55% at <strong>the</strong> samedose. The addition of sodium chlorate had no effect on testosteroneproduction by unstimulated cells but was able to inhibit testosteroneproduction in a dose dependant manner up to 87% at <strong>the</strong> maximum dose usedas shown in Figure 2.Discussion: These results demonstrate that cell surface heparan sulphateproteog1ycans toge<strong>the</strong>r with an autocrine factor or factors are an obligatorypart of <strong>the</strong> LH stimulated testosterone production pathway in <strong>the</strong> adult ratLeydig cell. Fur<strong>the</strong>nnore <strong>the</strong>se data rein<strong>for</strong>ce <strong>the</strong> emerging view that cellsurface and extracellular matrix heparin sulphate proteoglycans toge<strong>the</strong>r withautocrine and paracrine factors play an important, if not fundamental role in<strong>the</strong> regulation ofcell function.1. Sun, X-R. et al (1993) Endocrinology, 132: 186-192.50o 10 100Heparin (ugml)Figure 1: The effect of heparin ontestosterone production byunstimulated adult rat Leydig cells.~ a; 75(0ooo 3 6 13 25&xflll1 Qiorate (ntv'!)Figure 2: Effect of sodium chlorateon testosterone production by basaland LH stimulated Leydig cells.REDUCTION OF OVULATION RATE IN THE RAT BY ADMINISTRATION OF ANEUTROPHIL.DEPLETING MONOCLONAL ANTIBODYMats Brannstrom, Nigel Bonello, Robert J. Norman, Sarah Robertson.Department of Obstetrics and Gynaecology, The University of Adelaide, Queen Elizabeth Hospital andMedical School Campuses, Adelaide, South Australia.INTRODUCTIONNeutrophilic granulocytes have been implicated as importantcells in ovarian physiology, particularly in <strong>the</strong> processes ofovulation and luteolysis. There is an 8-fold increase in <strong>the</strong>density of neutrophils in <strong>the</strong> <strong>the</strong>ca layer during <strong>the</strong> ovulatoryprocess in <strong>the</strong> rat (1) and peripheral leukocytes amplify <strong>the</strong> LHinducedovulation response in vitro (2). In <strong>the</strong> present study wehave examined <strong>the</strong> importance of infiltrating neutrophils inovul·ation by investigating whe<strong>the</strong>r selective depletion ofneutrophils from <strong>the</strong> peripheral blood prior to ovulation affects<strong>the</strong> ovulation rate.METHODSImmature 25 day-old rats were primed with 10IU eCG, prior tobeing injected intraperitoneally with 2.5m1 ascites fluidcontaining RP-3, a monoclonal antibody (mAb) known toselectively deplete rat neutrophils (3), on <strong>the</strong> day be<strong>for</strong>e and on<strong>the</strong> day of an ovulation-inducing lOIU hCG injection. At <strong>the</strong>same times, rats in <strong>the</strong> control group were administered withascites fluid containing an irrelevant mouse anti-rat mAb.Neutrophil counts in <strong>the</strong> blood were detennined at Oh and 20hrelative to hCG administration and ovulation was examined bycounting <strong>the</strong> released ova found in <strong>the</strong> ampullary region of <strong>the</strong>oviduct 20h after hCG injection. The number of neutrophils in<strong>the</strong> walls of ruptured follicles were measuredimmunohistochemically using MCAI49, a mAb directedagainst a cytoplasmic detenninant in neutrophils. Circulatingprogesterone levels at 20h post-hCG were assayed byradioimmunoassay. All data was statistically analysed usingtwo-tailed unpaired student-t tests, except where o<strong>the</strong>rwiseindicated.RESULTSThe total white blood cell count in peripheral blood was notaffected by administration of RP-3 (RP-3-treated group:5.8±O.4x10 6 cells/ml, Control group: 6.6±Q.3x10 6 cells/ml).The proportion of neutrophils in differential counts ofperipheral blood samples froJ!1 ~-3~freated rats wassignificantly decreased ~pared to ·>proportions at hCGinjection and 20h later- in <strong>the</strong> control group (Fig 1). Theovulation rate in <strong>the</strong> RP-3-treated group (32.4±3.6 ovulationsper treated animal) was significantly lower than in <strong>the</strong> controlgroup (44.1±3.1) (Fig 2), indicating that depleting peripheralblood neutrophils has partial inhibitory effects on ovulation ratein <strong>the</strong> rat. Immunohistochemical staining with MCA149revealed that at 20h post-hCG <strong>the</strong> density of neutrophils in <strong>the</strong>ruptured follicles of RP-3-treated animals was significantly(p ='3530~ 25:a§"20tla3 15Z~ 105.~~ 200 10**ControlTreatmentD Control ~ RP30l..L-_--.J~~~.L-______I.____l_J~~~Oh20hBlood Sample Time Post-hCGFigure 1. Depletion of peripheral neutrophils as % of totalwbc (mean ± SEM) measured Oh and 20h post-hCG injectionfollowing <strong>the</strong> administration of <strong>the</strong> RP-3 antibody. **significantly different (p


-IntroductionThe penneability of testicular blood vessels is known to increase after administration of human ChorionicGonado~phin(hCG) ~d is associated with <strong>the</strong> adherence ofpolymorphonuclear leukocytes to <strong>the</strong> vascularendo<strong>the</strong>lIum (1). T~stIcular .macrophages are found in <strong>the</strong> testicular interstitium in close proximity to both <strong>the</strong>~stoster?neprodUCIng LeydIg cells and ~e blood. v~ssels ?f <strong>the</strong> testi~ ~d v:hile <strong>the</strong> hCG response does not appear tod~ectly Involve androgens or prostaglandins (2), It IS abolIshed by elImInatIon of <strong>the</strong> Leydig cells with ethanedImethane s~lphonate (EDS) (1). The removal of <strong>the</strong> TMs does not affect this response (3). Nitric oxide (NO) is amolecule whIch has a broad s~trum ofpotent effects including <strong>the</strong> regulation of vascular penneability, immuneresponses and leukocyte adhesIOn (4).Release ?~ N? by <strong>the</strong> ~ and/or Leydig cell may contribute to <strong>the</strong> regulation of blood flow and <strong>the</strong> vascularpermeabIlIty In <strong>the</strong> test!s. !n th~ present study, we have assessed production ofNO by isolated testicularmacrophages cultured In vltro In <strong>the</strong> presence of <strong>the</strong> non specific stimulant, lipopolysaccharide (LPS), indomethacin(IDM) an~ hCG. In order to ~elate <strong>the</strong>se observations to an in vivo situation, we have also observed NO productionby <strong>the</strong> testIcular macrophage In <strong>the</strong> presence of Leydig cells (LC),MethodTMs and LC. w~re isolated b~ a three step procedure involving mechanical dispersion of <strong>the</strong> seminiferous tubules,cellular elutnatIon and a denSIty-dependant separation with Percoll which allowed highly purified cultures of both celltypes to be obtained (5). The TM were <strong>the</strong>n cultured alone (5xl05 cells/ml, lml) or in co-cultures consisting ofequal !1umbers ofLC and TM (5xloS cells/ml total) in RPMI1640/Hams F12 with 10%FCS and 2mM glutamine at34°C In 5% C0 2 in air. Combinations of LPS (10Jlg/ml), IDM (10Jlg/ml) and hCG (lOng/ml) were <strong>the</strong>n added toTM, LC and T.M/LC co.-e~lt~es. After 24 hours <strong>the</strong> TM conditioned medium (TMCM) was collected, centrifuged:m d frozen untIl.use. NItrIte ~n culture supernatants was me~~red by a microplate assay method described in Ding etal, .1988 (4). Bnefly, 50JlI ~IqU?ts were removed from condItIOned medium and incubated in equal volume withGness reagent (1 % s~phanI1amIde, 0.1 % napthylethylene diamine dihydrochloride and 2.5%H3P04) at roomtemperature ~or 10 mI~utes. 1J1e ab~~bance at 550nm was detennined by a microplate reader and NO concentrationswere dete~med by ~smg sodium rntnte as a standard. The cell free medium alone was measured <strong>for</strong> NOconcentratIon and thIS value was subtracted from <strong>the</strong> value obtained with <strong>the</strong> cells.40[NO] 30JlM 2010oResultsNITRIC OXIDE SECRETION BY MACROPHAGES AND LEYDIG CELLS ISOLATEDFROM THE RAT TESTISS. Kern and S. MaddocksDepartment of Animal Science, The University of Adelaide Waite Campus, Glen Osmond 5064.* *li4 ~ mIDItlPS ~G i~ h~~ LCffM L~&MValues are <strong>the</strong> mean±SEM <strong>for</strong> n=6 experiments; *=P


Immunosuppressive activity in <strong>the</strong> rat testis that is not temperature or pH labileINTRODUCTIONThe mammalian testis is regarded as animmunologically privileged site, and testisextract is capable of suppressing lymphocyteproliferation in-vitro (1). In <strong>the</strong> present study wehave investigated some characteristics of <strong>the</strong>immunosuppressive components of a rat testisextract (TE).METHODSTesticular extract (TE) was prepared byhomogenising decapsulated testes in phosphatebufferedsaline (PBS) and centrifuging this at250g <strong>for</strong> 15 min and <strong>the</strong>n 10,OOOg <strong>for</strong> 30 mins.Aliquots of this extract were heated <strong>for</strong> 10 minsat 25 0 , 56 0 , 80 0 or 100 0 C, <strong>the</strong>n cooled at 40 Cand recentrifuged. Additional aliquots had <strong>the</strong>irpH lowered to 1.9 using 1M HCl, raised to 11.2using IN NaOH, or kept at pH 7.2 wi<strong>the</strong>quivalent amounts of water added <strong>for</strong> controls.These aliquots were kept at room temperature <strong>for</strong>60 mins, <strong>the</strong>n neutralised to pH 7.2, cooled to4 0 C and centrifuged. Fur<strong>the</strong>r aliquots of <strong>the</strong>initial extract were separated into fractions ofMW >5000, 1000-5000 and 5000 but no activity in <strong>the</strong> fractions ofMW 3500 Da)and was inhibited by a human recombinant IL-I receptorantagonist (IL-lRA) (Fig. 1A). The inhibitory activity wasretained following dialysis or ultrafiltration (100 kDa cutoff),and was not affected by <strong>the</strong> amine oxidase inhibitor,hydroxylamine (0.1 mM). Lymphocyte viability, asindicated by exclusion of propidium iodide, was notaffected by culture with IF. Purified testicularmacrophages secreted a similar non-dialysable (> 3500 Da)lympho-inhibitory activity in vitro (Fig. IB). Samples ofmedium collected from purified Leydig cell andseminiferous tubule cultures were inactive or stimulatoryin <strong>the</strong> assay, respectively. Inhibitory activity in testicularIF was reduced to 38 % of control by treatment with EDS(2 weeks) and to 18% of control by cryptorchidism (4weeks) (Fig. IC).DiscussionThe data indicate that IL-I, presumably from <strong>the</strong> Sertolicells, is secreted into <strong>the</strong> interstitial tissue, but its activityis prevented by a potent large molecular weight inhibitorof lymphocyte activation. This inhibitory activity is notattributable to testosterone, prostaglandins or oxidizedpolyamine products. The inhibitory factor may beproduced by <strong>the</strong> macrophages under Leydig cell control.References(1) Wang J, et al. (1994) BioI Reprod 51: in press.(2) Gustaffson K, et aI. (1988) J Reprod Irnmunol 14: 139-150.(3) P611linen P, et aI. (1992) J Reprod Irnmunol. 21:257-274.Figure 1: Mouse lymphocyte bioassay.A: Activity ofhuman IL-l (IV) and testicular IF (1.0 p.L), with andwithout addition of IL-IRA (10 p.g/mL). B: Activity oftesticular macrophage conditioned medium (MCM),with and without LPS-stimulation. Values are mean ±SEM (n = 4); * p < 0.05. C: Relative bioactivity (±95% CL) of <strong>the</strong> IF inhibitor from untreated control(CONT), DMSO-treated control (DMSO), EDS-treated(EDS) and 4-week cryptorchid (Co) rats, assayed atmultiple dilutions.C?0 no IL-1RAT""x:::JE0a.50 ~. +IL-1RA I -LPS rf..£.L()Q?.z010~~+~~0 0Q.10:~0 ffi0 w~ >wza* *~w0:~>-:cl- a a aI h1L-1 (1 U) IF (1.0 uL) a 50 100 CONT EDS Co~MCM (uL)DMSO60JA DB 2 C69 70


DEPENDENCE OF FLUID REABSORPTION IN THE EFFERENT DUCTS(ED) OF THE RAT TESTIS ON LUMINAL NA AND CLLA. Hansen, J. Clulow & R.C. JonesDepartment of Biological Sciences, University of Newcastle, NSW, 2308INTRODUCTIONPrevious studies from our laboratory (1,2) havedemonstrated that, in <strong>the</strong> rat, <strong>the</strong> efferent ducts,ra<strong>the</strong>r than <strong>the</strong> ductus epididymidis, are <strong>the</strong>principal site of net reabsorption of testicular fluid.By <strong>the</strong> time sperm have traversed <strong>the</strong> efferentducts, around 95% (2) of <strong>the</strong> rete testis fluid (RTF)accompanying <strong>the</strong> sperm has been reabsorbed.This fluid reabsorption occurs with no significantchange in <strong>the</strong> luminal osmotic pressure, and Na(RTF=136.9±2.7 mM, distal ED=136.8±2.4 roM) isreabsorbed at <strong>the</strong> same rate as water (isotonicreabsorption). These data ,suggest that fluidreabsorption in <strong>the</strong> efferent ducts is related to, andpossibly dependent upon, <strong>the</strong> transport ofNa across<strong>the</strong> epi<strong>the</strong>lium, as has been demonstrated <strong>for</strong> <strong>the</strong>homologous proximal kidney tubule (3). Thecurrent study was undertaken to test thishypo<strong>the</strong>sis.MATERIALS AND METHODSIndividual efferent ducts of anaes<strong>the</strong>tized Wistarrats were perfused as reported previously (4) wit<strong>hth</strong>e following isotonic solutions: (a) control with161 mM NaCl (b) varying concentrations of NaClmade isotonic with mannitol (c) Na only solutionsmade with sodium isethionate and mannitol and (d)Cl only solutions made with choline cWoride andmannitol.RESULTS AND DISCUSSIONThe results of perfusions at 0.1 pI min-I are shownin Fig. 1. The results show that perfusion with 161mM NaCl results in a high rate of fluidreabsorption similar to rates reported <strong>for</strong> KrebsBicarbonate Ringer or native RTF (4). As NaCl isreplaced with mannitol, reabsorption is abolished at[NaCl] = 81 roM. Above this concentration, fluidreabsorption occurs, below this concentration,secretion occurs. Removal of ei<strong>the</strong>r Na or CI from<strong>the</strong> perfusion fluid is sufficient to abolishreabsorption. We conclude from <strong>the</strong>se results thatfluid reabsorption in <strong>the</strong> efferent ducts of <strong>the</strong> rat isdependent on <strong>the</strong> presence of both Na and Cl in <strong>the</strong>71luminal fluid at <strong>the</strong> high concentrationsdemonstrated to occur physiologically (2).z0j::::c.4030a:0enaJ~w 20a:- cE 10- EE0,...- c- 0z0i=w-10a::0w en-20-0- ChCI--.- Nal___ NaCILUMINAL CONCENTRATION (mMJI)200Fig.1 Fluid transport in <strong>the</strong> BD after perfusion withvarying concentrations of NaCI, choline chloride (ChCl)and sodium isethionate (NaI).REFERENCES(1) Jones, R.C. & Jurd, K.M. (1987) Aust. J. BioI. Sci.40: 79-90.(2) Clulow, J., Jones, R.C. & Hansen, L.A. Bxperim.Physiol. (in press).(3) Giebisch, G., Klose, R.M., Malnic, G., Sullivan,W.J. & Windhager, E.B. (1964) J. Gen. PhysioI. 47:1175-1194.(4) Clulow, J., Hansen, L.A. & Jones, R.C. (1992) Proc.Aust. Soc. Reprod. BioI. 24: 10.IMPROVEMENT IN THE DEGREE OF SYNCHRONY OF THE PROSTAGLANDIN-INDUCEDOESTRUS IN COWSMas'ud Hariadi, D.Broomfield and P.J.WrightDepartment of Veterinary Clinical Sciences, University of MelbourneWerribee 3030, VictoriaINTRODUCTIONTreatment of cows with prostaglandin (PG) on day 12(D12) of <strong>the</strong> cycle (oestrus = DO) results in greaterspread in <strong>the</strong> occurrence of oestrus than treatment ato<strong>the</strong>r stages (1). This spread of oestrus reflects <strong>the</strong>stage on follicular maturity at luteolysis (2). We havestudied <strong>the</strong> effect of treatments on D5 of <strong>the</strong> cycle,aimed at synchronising follicular waves, on <strong>the</strong> degreeof synchrony of prostaglandin-induced oestrus in cows.MATERIALS AND METHODSThe study, comprising two experiments, used nonlactatingdairy cows of different breeds (Friesian, Jerseyand Crossbred) which exhibited normal oestrous cycles.In Experiment 1 cows (n = 56) were devided into threegroups. Cows were treated on D4 and D5 of <strong>the</strong> cyclewith saline (control), or charcoal extracted bovinefollicular fluid (CFF, 16 ml, iv, 12h apart), or on D5treated with oestradiol benzoate (ODB, 5 mg', im) andprogesterone (pessary/CIDR-B inserted <strong>for</strong> 7 days). InExperiment 2 cows (n = 92) were divided into four equalgroups, on 05 of <strong>the</strong> cycle Group 1 received 1 ml saline(control), Group 2 had CIDR-B inserted <strong>for</strong> 7 days andODS (5 mg, im). Groups 3 and 4 had CIDR-B and ODBrespectively. All cows in both experiments received500 /1g c1oprostenol (CS) on D12. Blood samples <strong>for</strong>progesterone determination were taken on D12 (Exp.1)and on D5, 09, D12, D14 and at oestrus (Exp.2). Cowswere fitted with heat mount detector and observed twicedaily <strong>for</strong> sign of oestrus.RESULTSTreatments with CIDR-B/ODB tightened (P=0.002) andwith CFF worsened (P=O.01) <strong>the</strong> synchrony of PGinducedoestrus in cows (Fig.1, Exp.1). In Experiment 2,CIDR-B/ODB had better synchrony than saline treatedcows, and ODB treated group was <strong>the</strong> tightest(P


OESTROUS BEHAVIOl)R IN THE ABSENCE OF A DOMINANT FOLLICLEOR OVULATION AFTER NORGESTOMET TREATMENT IN HEIFERSA. Niasari-Naslaji"S, M.J. D'Occhio', T. Whyte' and D. Jillella!fCSIRO Division of Tropical Animal Production, Tropical Beef· Centre, Rockhampton, Queensland;!Department of Farm Animal Medicine and Production, University of Queensland, Brisbane, QueenslandIntroductionCrestar® is an oestrous synchronisation treatmentthat is used extensively in artificial inseminationand embryo transfer programs in beef and dairycattle. It involves <strong>the</strong> i.m. injection of oestradiol(OE 2 ) valerate (5 mg) and <strong>the</strong> syn<strong>the</strong>ticprogestagen norgestomet (17a-acetoxy-11 p­methyl-19-norpreg-4-en3,20-dione, 3 mg),toge<strong>the</strong>r with a s.c. implant of norgestomet (3 mg)that is removed after 8 to 10 days. Oestroususually occurs within 48 hours of implant removal.Crestar® has been used to induce oestrus inprepubertal and peripubertal heifers, butconception rates to artificial insemination (A.I.)have generally been relatively low (1, 2). In astudy in ovariectomised cows and heifers, astandard Crestar® treatment induced oestrousbehaviour in 55% and 57% of females,respectively (3). This finding suggested that inentire females Crestar® could induce oestrus in <strong>the</strong>absence of ovulation, which has importantpractical implications. There<strong>for</strong>e, in <strong>the</strong> presentstudy, we examined whe<strong>the</strong>r Crestar® could induceoestrous without ovulation in peripubertal heifers.Materials and MethodsOvarian status (follicle diameter, presence of acorpus luteum (Cl)) was determined in 23, 18­month-old Brahman (80S indicus) heifers (341 ± 6kg) by (1) trans-rectal ultrasonography using anAloka 210 real-time linear scanner and 7.5 MHztransducer and (2) assay of plasma progesterone(P4) concentrations by RIA. Heifers <strong>the</strong>n received astandard Crestar® treatment described above (Day0). On Day 7, heifers were injected withcloprostenol (Estrumate®) and ovarian statusrecorded. Norgestomet implants were removed onDay 8 and oestrous behaviour was monitored over<strong>the</strong> next 3 days with teaser bulls and twice dailyobservation of group behaviour. Fur<strong>the</strong>rassessment of ovarian status was carried out 6days after oestrus. Follicles were classified asmorphologically 'dominant' if <strong>the</strong>y were ~ 10 mmin diameter. Plasma P4 concentrations ~ 2 ng/mlwere taken as indicative of a functional CL.Results are presented as means ± SEM.ResultsOn Day 0, 3 heifers had a functional Cl suggestingthat <strong>the</strong> majority were acyclic. At <strong>the</strong> same time,13 heifers had a dominant follicle of diameter 10.9± 0.4 mm. On Day 7, only one heifer had adominant follicle and <strong>the</strong> diameter of <strong>the</strong> largestfollicle in o<strong>the</strong>r heifers was 5.7 ± 0.9 mm (n =22). After norgestomet implant removal on Day 8,16 (70%) heifers showed oestrous behaviour wit<strong>hth</strong>e distribution shown in Table 1. On Day 6 afteroestrus no heifer had a functional CL.Table 1 Distribution of oestrous behaviour afterremoval of norgestomet implants.Within 24 h9/16Within 48 h4/16Within 72 h3/16DiscussionThe relatively small follicles on Day 7 of treatmentwould not have been expected to secretesignificant OE 2 over <strong>the</strong> next three days. This,toge<strong>the</strong>r with <strong>the</strong> absence of elevated P4 on Day 6after oestrus, strongly suggested that oestrousbehaviour occurred in <strong>the</strong> absence of folliclegrowth or ovulation. Escobedo et 8/ (4) reportedthat in 80S indicus cross heifers ranging from 8 to16 months of age norgestomet treatment <strong>for</strong> 9days induced oestrus in 60% of heifers, but noanimals showed a typical pre-ovulatory LH surge.A possible explanation <strong>for</strong> <strong>the</strong> above findings isthat circulating OE 2 concentrations were reportedto remain elevated at least to Day 8 after injectionof 5 mg oestradiol valerate (5). The elevated OE 2could presumably induce oestrous behaviour uponwithdrawal of norgestomet.The present data, toge<strong>the</strong>r with previousobservations (4), have important implications. -Forexample, <strong>the</strong> results may explain low conceptionrates to A.1. after synchronisation of peripubertalheifers. Also, in superovulation and embryotransfer programs, heifers should be assessed <strong>for</strong>ovarian status (ovarian palpation, ultrasound, P4assay) to confirm <strong>the</strong> presence of a CL aftersynchronisation, and be<strong>for</strong>e FSH <strong>the</strong>rapy is started.References(1) Short RE et 8/ (1976) J Anim Sci 43: 1254(2) Spitzer JC (1982) Theriogenology 17: 373(3) McGuire WJ et 8/ (1990) Theriogenology 34:33(4) Escobedo F et 8/ (1989) J Anim Sci 67 (Suppl1): 410(5) Kojima N et 8/ (1992) Bioi Reprod 47: 1009-Ovulation and Oestrus among Dairy Cows with Anovulatory AnoestrusFollowing Progesterone Pre-TreatmentK.L. Macmillan, S. McDougall. V.K Taufa andA.M DayDairying Research Corporation, Private Bag 3123, Hamilton, New Zealand.INTRODUCTIONAnovulatory anoestrus (A-A) is <strong>the</strong> mostcommon <strong>for</strong>m ofinfertility among New Zealand'spasture-fed dairy cows. Even when <strong>the</strong>y dorecommence ovulating, it will frequently occurwithout oestrus. Studies in lactating dairy cowson responses to injected oestradiol benzoate(ODB) during <strong>the</strong> postpartum period showed that5 days of pre-treatment with progesterone (P4)increased <strong>the</strong> incidence of oestrus from less than40% without P4 priming to over 70% (1, 2).Ovulation usually accompanied <strong>the</strong> oestrusfollowing P4 priming. These interactions werestudied <strong>for</strong> <strong>the</strong>ir possible relevance as treatmentregimens <strong>for</strong> A-A.MATERIALS AND METHODSTwo trials were conducted with cows in 8 dairyherds in 1992 or 1993. Animals diagnosed withanovulatory anoestrus (A-A) were randomisedwithin herd taking account of age, breed andcalving date into 3 (Trial 1) or 5 sub-groups(Trial 2). A CIDR device (InterAg, Hamilton)was inserted into <strong>the</strong> vagina of each animal toprovide a source ofP4. This device was removed5 or 7 days later at which time some animals wereinjected with 400 ill eCG (NZ Pastoral REFERENCESainseminated through to 14 days after deviceremoval when <strong>the</strong> uninseminated animals werepresented <strong>for</strong> a second veterinary examinationand classified as unresponsive and still A-A, orovulating without being detected in oestrus (Ov­Oe). In Trial 1, a milk sample was obtained fromevery cow 14 days after device removal. Thissample was assayed <strong>for</strong> P4, with concentrationsof>1 ng/ml being accepted as indicative of lutealactivity.RESULTSThe ODB injection increased <strong>the</strong> percentage ofanimals inseminated within 2 to 4 days of deviceremoval (p


-PREPARTUM SlJPPLEMENTATION OF BOS INDICUS COWSENHANCES POSTPARTUM OVARIAN FUNCTIONLee A. Fitzoatrick!, Geoff Fordyce and Keith Entwistle.31 Department of Biomedical & Tropical Veterinary Sciences, James Cook University, Townsville, 48112 Queensland Department of Primary Industries, Swan's Lagoon Beef Cattle Research Station, Millaroo, 48073 Present address: Faculty of <strong>the</strong> Sciences, University of New England, ArmidaJe, 2351Immunisation Against Growth Hormone-Releasing Factor (GRF) Does Not affectThe Testicular Response To Nutrition In RamsS,W Walkden-Brown 1 , M,J. H6tzel 1 , R,D.G. RigbyZ and G.B. Martin 1 ,31Faculty of Agriculture, University of Western Australia, Nedlands, WA 6009. 2CSIRO Division of Animal Production,Blacktown, NSW 2148. 3CSIRO Division of Animal Production, Wembley, WA 6014.INTRODUCTION Introduction ResultsThe growth and function of ovarian follicles in <strong>the</strong> postpartumcow are considered to reflect <strong>the</strong> net effect of endocrineactivity at any particular time postpartum. However, previousstudies have shown that prepartum supplementation canenhance postpartum ovarian follicle populations, in <strong>the</strong>absence of bodyweight (BW)/body condition (BC) effects (1).The aim of this study was <strong>the</strong>re<strong>for</strong>e to test <strong>the</strong> hypo<strong>the</strong>sis thatshort-term prepartum supplementation of Bos indicus crosscows can improve postpartum ovarian function by enhancingfolliculogenesis.MATERIAL AND METHODSFifty pregnant grade Brahman cows (~-% Bos indicus) wereallocated to ei<strong>the</strong>r receive 1 kg of cotton seed meal/head/day(44% crude protein) fed twice weekly <strong>for</strong> six weeks prior to<strong>the</strong> expected date of commencement of calving (CM), orremain unsupplemented controls (CaNT). Cows wereallocated to be ei<strong>the</strong>r ovariectomised at 80 days postpartum orto remain intact. Blood samples were taken every 10 days <strong>for</strong>plasmaprogesterone (P,) estimation. Postpartum ovulation wasregarded as having occurred when plasma P, was >1 ng/m!.Ovarian follicles (~mm) were dissected, measured andcounted, and follicular fluid collected <strong>for</strong> oestradiol (Ez) andprogesterone (P,) estimation. Aromatase activity studies werecarried out on granulosa cells from <strong>the</strong> ovaries of 12 cowsfrom each treatment group by measuring E, production by4xlO' viable granulosa cells in tissue culture medium withadded testosterone (T,), luteinising hormone (LH) and(or)follicle stimulating hormone (FSH), after 3 hours incubation(2). To simplify statistical analyses, follicle data were pooledinto five diameter classes (2-3,4-5, 6-7, 8-10 & ~ll mm).RESULTSThere were no differences in BW or BC between <strong>the</strong> groupsat <strong>the</strong> end of supplementation or during <strong>the</strong> postpartumperiod. Supplemented cows ovulated 30 days earlier followingcalving (101 compared to 131 days <strong>for</strong> CaNT cows; P=0.08).At any given time 80 to 180 days after calving, <strong>the</strong> proportionof cows with cyclic ovarian activity tended to be higher <strong>for</strong>CM cows, although this was only significant at 100 dayspostpartum (64% and 10%; P5 mm) from CM cows produced more E, thansimilar follicles from CaNT cows (p=O.053), while smallfollicles (2-5 mm) from CM cows produced less P, thansimilar follicles from CaNT cows (p=0.003) (Figure 1).Supplementation had no effect on numbers of granulosa cellsin diameter-classes. However, follicles from CM cowscontained more granulosa cells overall than follicles fromCONT cows (80.3±12.6 x 10' vs 31.5±7.3 x 10'; P=0.OO3).Fur<strong>the</strong>rmore, granulosa cells from CM cows had greateraromatase activity compared to CONT cows (po etSmall Large Small LargeFollicles Follicles Follicles FolliclesCO NTCMFigure 1 Least squares means (±SEM) <strong>for</strong> a and P, in follicular fluidof small (2-5 rnrn) and large (>5 rom) follicles from CONT and CMcows0.40~E 0.35~ 0.30--S 0.25o 0.20""'C 0.15aJ:: 0.10(/)(J) 0.05o0.00CONCLUSIONS. T40 T4+FSH T4+LH T4o+FSH+LHCulture Medium CompositionFigure 2 Least squares means (±SEM) <strong>for</strong> aromatase a of granulosacells from CM and CONT cows, in <strong>the</strong> presence ofT., T,+FSH, T.+LHor T,+FSH+LHWe conclude that prepartum supplementation can enhancepostpartum ovarian function ofBos indicus cows. The effectsof this nutritional input on reproductive function may simplybe due to changes in gonadotrophin secretion, or local effectson <strong>the</strong> ovary mediated by metabolic hormones and (or)growth factors. However, <strong>the</strong> exact mechanisms involvedremain to be determined.1. Fitzpatrick LA (1994) PhD Thesis, James Cook University. Townsville2. McNatty et aI. (1984) Journal ofEndocrinology 102: 189-198This work was partially supported by <strong>the</strong> Meat Research CorporationNutrition-induced changes in sperm production inrams are only partially explained by changes ingonadotrophin secretion (Martin et al., 1994). We<strong>the</strong>re<strong>for</strong>e investigated <strong>the</strong> role of <strong>the</strong> hormones of<strong>the</strong> somatotrophic axis by immunising rams againstGRF and observing <strong>the</strong>ir nutritional responses.Immunisation • 2 year old rams were immunisedagainst hGRF [1-29] conjugated to ovalbumin (GRFi),or ovalbumin alone (Control), on day -56, Boosterswere given on days 0 and 28 (n=10/group).Diet • Days -56 to -18. At pasture• Days -17to -1. Maintenance (M) in animal house• Days 0 to 35. 2.5 x maintenance (2.5M)• Days 36 to 77, 5 rams in each group returned to MMeasurements • Weekly Liveweight, scrotalcircumference and jugular plasma sample.• Days -1, 34 and 69 Plasma sampled at 20 minintervals <strong>for</strong> 12h <strong>for</strong> pulsatile-hormone assay.• Day 77 Slaughter and carcass analysis. Spermproduction estimated from testicular homogenates.~ 40ca) 70.5±1.


Use of a GnRH vaccine <strong>for</strong> immunocastration of coltsK.F. Dowsett*, U. Tshewang*, L.M. Knott*, A.E. Jackson*, DAV Bodero* and T.E. Trigg#*Departments of Farm Animal Medicine and Production, The University of Queensland, st. Lucia, Queensland;Sufiivan &Nicolaides Pathologists, PO Box 344, Indooroopi/ly, Queensland; #peptide Technology Ltd, PO Box 444, Dee Why, NSW.-Timing of progesterone supplement and embryonic survival inoverfed giltsIntroduction: Following an experiment with an oil based ovalbumenconjugated GnRH vaccine (Dowsett et al., 1991), a water soluble<strong>for</strong>mulation was developed <strong>for</strong> immunocastration of horses. While<strong>the</strong> oil based vaccine produced effective antibodies to GnRH andsuppressed testicular function it caused severe tissue reactions. Tominimise this effect but maintain antigenicity and efficacy of <strong>the</strong>vaccine, a watersoluble ovalbumen conjugate was tested in colts.Methods: Once testosterone concentrations and semen quality hadattained pUbertal levels, ten 2 year old colts were allocated to twogroups: 4 x 200 mg and 6 x 400 mg. One 2 year old monorchid wasalso treated with 200 mg. They were vaccinated on 6/3/92 and3/4/92. Additional vaccinations were given individually, as determinedby antibody titres and testosterone concentrations, to maintainimmunosuppression during <strong>the</strong> 1992/1993 breeding season.Colts were weighed and scrotal diameters measured weekly. Bloodsamples were collected twice weekly <strong>for</strong> testosterone assays,androstenedione assays and GnRH antibody titres until <strong>the</strong>conclusion of <strong>the</strong> experiment. Two semen samples were examinedprior to vaccination and after testosterone concentrations andantibody titres returned to normal.Following castration, <strong>the</strong> left testis of each colt was perfused fixed <strong>for</strong>histology and electron microscopy. The right testis was used toestimate daily sperm production. The volumes of <strong>the</strong> seminiferoustubules, intertubular areas and Leydig cells were estimated by <strong>the</strong>point counting method. The testis of <strong>the</strong> monorchid was used <strong>for</strong> allexaminations.Results: Nine of <strong>the</strong> colts required third vaccinations. These wereadministered to two colts (200 and 400 mg) and <strong>the</strong> monorchid on26/5/92,three colts on 9/7/92 (2 x 200 mg and 1 x 400 mg) and <strong>the</strong>remainder (1 x200 mg and 2 x 400 mg) on 6/8/92. Third vaccinationswere not reqUired by <strong>the</strong> o<strong>the</strong>r two (400 mg). GnRH titres rangedfrom 0 to 1:4200 some 4 weeks after primary vaccination. Peak titresin <strong>the</strong> 200 mg and 400 mg groups ranged from 1:3300 to 1:9000and 1:2500 to 1:42000 respectively 2 to 4 weeks after <strong>the</strong> booster(Fig 1). Effective titres (1:1000) were maintained <strong>for</strong> 26 and 27 weeksin two 400 mg colts. The o<strong>the</strong>r 400mg colts had effective titres <strong>for</strong> 12to 20 weeks. Third vaccinations maintained effective titres <strong>for</strong> afur<strong>the</strong>r 8 to 24 weeks in <strong>the</strong>se colts. The 200 mg colts andmonorchid developed effective titres <strong>for</strong> 12 to 20 weeks. Thirdvaccinations maintained effective titres <strong>for</strong> a fur<strong>the</strong>r 12 to 22 weeks.Prior to vaccination, testosterone concentrations varied between 0.31and 1.33 ng/ml (mean 0.65 ng/ml). They decreased to 0.08 ng/ml 6weeks after <strong>the</strong> booster and returned to normal concentrations <strong>for</strong>colts (>0.30 ng/ml) 31 to 43 weeks after primary vaccination. (Fig 2.)Mean androstenedione concentrations followed a similar pattern totestosterone. They decreased from 0.51 ng/ml to 0.08 ng/ml 4 weeksafter <strong>the</strong> primary vaccination. Normal concentrations (>0.25 ng/ml)were reached 31 to 43 weeks after primary vaccination (Fig 3.)Mean scrotal width and length be<strong>for</strong>e vaccination was 9.1 em and 9.7em respectively. These decreased to 6.5 em and 7.6 em followingvaccination. Mean scrotal width and length at castration was 9.4 and11.0 em.Mean semen characteristics be<strong>for</strong>e vaccination were gel free volume16.5 ml, sperm concentration 295.5 x 10 6 /ml, total sperm perejaCUlate 4041 x 10 6 and 32% live normal spermatozoa. Be<strong>for</strong>ecastration <strong>the</strong>se parameters were gel free volume 13.0 ml, spermconcentration 319.4 x 10 6 /ml, total sperm per ejaCUlate 4729 x 10 6and 38.7% live normal spermatozoa <strong>for</strong> <strong>the</strong> 200 mg dose group.Be<strong>for</strong>e castration <strong>the</strong> 400 mg live normal spermatozoa.The testes of all animals showed active spermatogenesis with Leydigcells present. The mean testicular parenchyma weights <strong>for</strong> <strong>the</strong> 200and 400 mg group were 129.0 g and 109.8 g. Daily sperm productionper testis and per gram of testis <strong>for</strong> <strong>the</strong> 200 mg group was 3.7 x 10 6and 2.8 x 10 6 , while those of <strong>the</strong> 400mg group were 2.3 x 10 8 and2.0 x 10 6 respectively.180001600014000.. 12000,Ii!~ 10000.Jl 8000~ 60004000200010·10 10 20 30 40 50 60Weeks from primary vaccination1.20.80.60.40.2Fig.1. Antibody titres follOWing vaccinationPrima!y vaccinationPrimary vaccination10Dose ofvaccination-0-- 200mg-400mg10 20 30 40Weeks from primary vaccinationFig 2. Androstenedione levels following vaccinationDose ofvaccination2030Weeks from primary vaccinationFig 3. Testosterone levels following vaccination4050Surgical castrationo +-~--;"'-~~~-,---~--r-~-~~--,--..-'--,·101.2i 0.8..~ 0.6~.!!j 0.40.25060Surgical castranoDiscussion: The results indicate that <strong>the</strong> water soluble vaccinesuppressed testicular function,and both testosterone andandrostenedione production. Androstenedione appears to provide amore accurate measure of <strong>the</strong> effect of <strong>the</strong> vaccine on testicularfunction than testosterone does. Semen data, testicular histology anddaily sperm production data suggest that <strong>the</strong> effects of <strong>the</strong> vaccineare reversible. Fur<strong>the</strong>r work is required to determine <strong>the</strong> reason <strong>for</strong>individual variation in antibody production.References:Dowsett, K.F., Pattie, W.A., Knott, L.M., Jackson, A.E., Hoskinson,R.M., Rigby, R.P.G. and Moss, B.A. (1991) A preliminary stUdy ofimmunological castration in colts. J. Reprod. Fert., Suppl.44, 183­190.60. R.A.Par;, M.A.Miles 1 , M.P. Cash 2 , and J.M. Waters 1IVictorian Institute of Animal Science, Werribee, Vic.3030, 2University of Sydney, NSW2006,INTRODUCTIONGilts which are overfed during early pregnancy may havean increased incidence of embryonic mortaltiy. This hasbeen related to reduced levels of peripheral progesterone(prog) and may be corrected with exogenous progtreatment (1). We have previously shown that in sheep,<strong>the</strong>re is a discrete period (days 11-12 after mating) ofenhanced sensitivity to prog reductions (2).Our aim in this study was to determine if a similarlydiscrete period of response to prog supplement could bedemonstrated in overfed gilts.MATERIALS AND METHODSLarge White x Landrace gilts (n=61) were housed ingroup pens until first pubertal oestrus when <strong>the</strong>y weretransferred to individual pens and fed 2 kg/day offinisher diet. Gilts were double mated at <strong>the</strong>ir nextoestrus (Day 0) and from day 2, <strong>the</strong>y were fed 3.5 kgdaily, split into two feeds. Treatments commenced onday 4 according to <strong>the</strong> following groups. Group 1­Control, arachis oil days 4 to 22; Group 2-Prog days 4 to22; Group 3-Prog days 4 to 10; Group 4-Prog days 10 to16; Group 5- Prog days 16 to 22. Prog (50mg/ml) wasdissolved in arachis oil and Iml was injected (im) eachday into alternate sides of <strong>the</strong> shoulder during <strong>the</strong>morning feed. One ml of oil was injected daily whenprog was not given. Gilts were slaughtered on day 23 ofpregnancy and reproductive tracts were collected.RESULTS and DISCUSSIONThere was a main effect of prog treatment in whichembryo survival was increased (Fig 1). For individualgroups, this difference was only significant when progwas given between days 16-22 but did not occur in giltstreated between days 4-22. In groups 2 and 5, CLweight was reduced, indicating a feedback effect whengiven prog between days 16-22 (Fig 2). The greaterembryo weight from gilts treated with prog from days 4­10 was not confirmed when gilts were treated with progfrom days 4-22 (Fig.3). The precise role of progesteronein <strong>the</strong> overfed pig requires fur<strong>the</strong>r study.This work was funded by <strong>the</strong>Pig Research andDevelopment Corporation.....u~ 0.4­:c.~ 0.3~0.0 L.--l-co-n-tr.J.ol-llpllrJ..JogLlJ.--I._ pr- oḡ• .I..-I'::pCL.roL£g."'--n;p~ro~g""'.'------0.35~ 0.20~·ir 0.10""10090BO70--0 60ḳ E:50.,40 ;;s~ 30lJIt200.70.6100.50.20.100.300.250.100.05Control Prog.(011) DaY'"Day. 4-224-22Fig.1 Percentage of viable embryos at Day 23 aftermating(Oil) Days Days Doys DoysDays 4-22 4-10 10-16 16-224-22Fig.2 Mean (±se) weight of corpora lutea from gilts onday 23 after mating0.00 L....l__-I--.LlUl-l..LU.JIL--=:==---'""",Control Pra9. Pro;(all) Oay. Dal'"Coy. 04--22 4-10+-22ProgCo)'.16-22Fig. 3 Mean (±s.e) embryo weights <strong>for</strong> gilts on Day 23after mating(1) Parr,R.A., Hughes,P.E., Davis,LF., Miles,M.A. andFerlazzo,lC. (1993) Proc. 4th Int. Conf. on Pig Reprod.93.(2) Parr,R.A. (1992) Reprod. Fertil. Dev., 4, 297-300.77 78


PROGESTERONE WITHDRAWAL, PLASMA LACTOSECONCENTRATIONS AND PIGLET PERFORMANCE: IS THERE ARELATIONSHIP?Brett D. Glencross, Janine K. Toussaint, Robert C. Tuckey and Peter E. HartmannDepmtment of Biochemistry, The University of Western Australia, Nedlands W.A. 6009IntroductionThe initiation of copious milk production(lactogenesis II) in <strong>the</strong> sow is triggered by <strong>the</strong>withdrawal ofprogesterone from <strong>the</strong> circulatorysystem (1). Poor piglet perfonnance has beenassociated with high post partum plasmaconcentrations of progesterone (2). It washypo<strong>the</strong>sised that low milk production was <strong>the</strong>primary cause of this poor perfoffilance. Theconcentration of lactose in sow plasma aroundparturition provides a temporal measure oflactogenesis II (3). We have examined <strong>the</strong>relationship between progesterone withdrawaland <strong>the</strong> increase. in plasma lactose concentrationsin 5 sows and compared <strong>the</strong>se changes to <strong>the</strong>growth of<strong>the</strong>ir litters.MethodsBlood samples were collected from sows from70 hours pre- until 44 hours post-partum at 4­hourly intervals (samples were only collectedfrom sow 2 up to 24 hours post-partum). Thesamples were analysed <strong>for</strong> progesterone andplasma lactose by radio-immunoassay andluminometric techniques respectively (Fig 1aand b). In addition <strong>the</strong> birth weights andgrowth of<strong>the</strong> piglets were recorded at 12-hourlyintervals <strong>for</strong> <strong>the</strong> first 3 days after birth and <strong>the</strong>weight gain was standardized by expressing <strong>the</strong>piglet's growth as a ratio of its birth weight.Litter perfoffilance was detennined by <strong>the</strong> meangrowth rate of all <strong>the</strong> piglets within <strong>the</strong> litter.Results and ConclusionsThis preliminary study supports <strong>the</strong> observationmade by de Passille et at. (2), that incompletepr?gesterone withdrawal from sow plasma(~IgS la, lb and 2) is associated with poorpIglet perfoffilance. In addition, <strong>the</strong> positiverelationship between lactose in sow plasma (Fig3) and litter growth rate supports <strong>the</strong> hypo<strong>the</strong>sisthat poor postnatal growth is caused by asuppression oflactogenesis II (2).References(1) Martin, C.E., Hartmann, P.E. & Gooneratne, A.(1978) Aust. J. BioI. Sci., 11: 517-525.(2) de Passille, A.M.B., Rushen, J.• Foxcroft,G.R., Aherne, EX. & Schaefer, A. (1993) J.Anim.Sci.1l.: 179-184.(3) Hartmann, P.E., Whitely, J.L.& Willcox, D.L.(1984) J. PhysioI. 347: 453-463.AcknowledgementsPig Research and Development Corporation255? 205Q)15t::0I-


»THE GENERATION AND PARTIAL CHARACTERISATION OF ANTIBODIES AGAINST MARSUPIAL GAMETESM.S.Harris and I.C.RodgerDepartment of Biological Sciences, University of Newcastle, NSW 2308INTRODUCTIONThe elucidation of <strong>the</strong> mechanisms involved in gametematuration and <strong>the</strong>ir interactions requires an understanding of<strong>the</strong> molecular nature of <strong>the</strong>se cells. Monoclonal antibodieshave proven powerful tools in dissecting <strong>the</strong> molecularcharacter of eu<strong>the</strong>rian gametes, <strong>the</strong>ir maturation andsubsequently <strong>the</strong> fertilisation process. Such characterisationof marsupial spermatozoa has been initiated with <strong>the</strong>production of monoclonal antibodies against tammar wallaby(Macropus eugenii) spermatozoa.CHARACTERISATION OF MAMMALIAN SPERM TAIL PROTEINS USING RAT FIBROUS SHEATHANTIBODYY.H. Kim~ G. Almahbobi, P.D. Temple Smith*, D.M. de Kretser & J.R. McFarlane, Institute of Reproduction andDevelopment, Department ofAnatomy*, Monash University, Clayton, Victoria 3168, AustraliaINTRODUCTIONSperm movement in <strong>the</strong> mammalian species depends on <strong>the</strong>ir flagellum which consists of several cytoskeletalcomponents, including <strong>the</strong> axoneme, outer dense fibres and fibrous sheath (FS). FS is a unique principal piececomponent ofsperm tail, which is composed oflongitudinal columns and numerous transverse ribs (1). The aim of<strong>the</strong> study was to characterise and compare sperm tail fibrous sheath proteins in different species using a polyclonalantibody to rat fibrous sheath by Western blotting and immunofluorescence microscopy.MATERIALS AND METHODSMale Balb/c mice (6-8wks) received immunisations (i.p) ofei<strong>the</strong>r ejaculated or cauda epididymal spermatozoa at 3-4 weekintervals in Freunds adjuvant prior to a final i.v boost. Themouse spleen cells were <strong>the</strong>n fused with NS-1 myeloma cellsand resulting hybridomas screened <strong>for</strong> antibody production byELISA and indirect immunofluorescence (IIF) usingpara<strong>for</strong>maldehyde fixed methanol permeabilised spermatozoa.Positive clones were subsequently single cell cloned. Cellsurface distribution was assayed by IIF on live wallabyspermatozoa at 40C. Species cross-reactivity was evaluatedusing eu<strong>the</strong>rian sperm fixed as above. Electrophoreticseparation of wallaby sperm proteins was per<strong>for</strong>medfollowing a sequential protein extraction. Proteins were <strong>the</strong>ntransferred to nitrocellulose and immunodetection ofmonoclonal antibody binding per<strong>for</strong>med using an anti-IgGalkaline phosphatase conjugated secondary antibody.RESULTSTwo promising monoclonal cultures were produced, wallabysperm 1 and 2 (WS-1 & WS-2). In immunofluorescence studiesWS-1 strongly labelled <strong>the</strong> tail, midpiece and ei<strong>the</strong>r <strong>the</strong> wholehead or <strong>the</strong> acrosomal region of fixed wallaby spermatozoa(fig 1). The antigen recognised by <strong>the</strong> WS-1 antibody hasbeen shown to arise in <strong>the</strong> testis and its cellular distributionremained unaltered up to and after ejaculation. WS-1 did notlabel live spermatozoa implying· an intracellular as opposedto cell surface distribution. The WS-1 antibody alsodemonstrated species cross-reactivity with <strong>the</strong> fixedspermatozoa of ano<strong>the</strong>r marsupial, <strong>the</strong> common brushtailpossum (Trichosurus vupecula). The midpiece of <strong>the</strong> possumspermatozoa labelled most strongly however <strong>the</strong> head and tailalso labelled distinctly. The WS-1 determinant was alsopresent on mouse, rat and rabbit spermatozoa. Again <strong>the</strong>midpiece and tail were most strongly labelled, weakerreactivity being associated with <strong>the</strong> acrosome.Ano<strong>the</strong>r antibody, WS-2, recognised an antigen localised to<strong>the</strong> tail, midpiece and peri-acrosomal ring of fixed wallabyspermatozoa (fig 2). Only <strong>the</strong> midpiece of live spermatozoawas labelled by this antibody suggesting both a surface andintracellular distribution of this antigen. The WS-2 antigenwas shown to be epididymal in origin and was not present onpossum, rat, mouse or rabbit spermatozoa. Immunoblottingrevealed that <strong>the</strong> WS-2 antibody recognised epitopes on threeproteins of approximate relative molecular weights 30, 35and 60kDa.Fig.I. Immunofluorescent localisation of WS-1 to <strong>the</strong>acrosome (a) , midpiece (m) and tail (t) of a tammar wallabyspermatozoan.Fig.2. Immunofluorescent localisation of <strong>the</strong> WS-2 antibodyto <strong>the</strong> peri-acrosomal ring (pa), midpiece. (m) and tail (t) of <strong>the</strong>tammar wallaby spermatozoan.DISCUSSIONSix fusions following whole sperm immunisations haveproduced clones exhibiting similar immunolocalisationpatterns. Work is <strong>the</strong>re<strong>for</strong>e continuing with immunisationsusing an acrosomal extract produced after <strong>the</strong> spermatozoahave been induced to undergo an in vitro acrosome reactionusing <strong>the</strong> diacylglycerol DiCg (1). Sperm were induced toundergo <strong>the</strong> acrosome reaction and <strong>the</strong>n removed bycentrifugation. The supernatant containing <strong>the</strong> plasmalemmaoverlying <strong>the</strong> acrosome, <strong>the</strong> outer acrosomal membrane and<strong>the</strong> acrosomal contents were <strong>the</strong>n concentrated <strong>for</strong> use asantigen.REFERENCES(1) Sistina, Y, Lin,M, Mate, K.E and Rodger,J.C. (1993)J.Reprod. Fert. .22.:335-341.MATERIALS & METHODSThe rat spenn tail fibrous sheath was isolated as previouslydescribed (2), and was solubilised in 1% SDS, 2mM DIT, and 25mM Tris-HCl. The proteins were emulsified in a Freund's completeadjuvant and injected into New Zealand white rabbits (75 ug perrabbit). The rabbits were boosted with antigen at 2 week intervalsand bled 1-2 weeks after each boost. Rat cauda epididymal spermand ejaculated rabbit and human sperm were prepared <strong>for</strong> indirectimmunofluorescence as previously described (3) or solubilised inurea, SDS, and DIT by shaking at room temperature and <strong>the</strong>nsperm tail proteins were isolated from <strong>the</strong> gel-like DNA materialby centrifugation.RESULTSSDS-PAGE of <strong>the</strong>se sperm tail proteins showed CoomassieBrilliant Blue bands of similar intensity, corresponding to 116kDa, 80-89 kDa, 57 kDa, 31 kDa, 28 kDa, and 15.5 kDa, althoug<strong>hth</strong>ere were some species specific bands of different molecular sizespresent (Figure 1). Western blots of sperm tail proteins with anti­FS reacted strongly with 116 kDa and 80 kDa polypeptides of<strong>the</strong>rat, rabbit and human sperm tail proteins. Rat and rabbit sperm tailproteins have similar cross reactivities in 80-89 kDa, 57 kDa, 31kDa and 28.5 kDa, but <strong>the</strong>re was no cross reaction or weak crossreaction with human sperm tail proteins (Figure 1). Usingimmunofluorescence, isolated intact sperm denatured by 1% TritonX-I00 in PBS, reacted with rat fibrous sheath antibody mainly in<strong>the</strong> principal piece of <strong>the</strong> sperm tail, but in <strong>the</strong> rabbit also reactedwith <strong>the</strong> equatorial segment of<strong>the</strong> head (Figure 2).STO 1 2 3116 ....66 ....45 ....31 ....21.5 ....14.4'"4Figure 1: SDS-PAGE of rat (1), rabbit (2), andhuman (3) sperm tail proteins, and rat FS (4).Western blot using anti-rat FS antiserum ofhuman (A), rabbit (B) rat (C) and rat FS (D).Figure 2: Immunofluoresence micrograph ofrabbit sperm using anti-rat FS antiserum.Immmunoactivity was seen on <strong>the</strong> equatorialsegment (E) of <strong>the</strong> head, and principal piece(P) but not <strong>the</strong> middle piece (M) of <strong>the</strong> spermSUMMARYSperm tail proteins are immunologically similar in <strong>the</strong> 3 species tail.although <strong>the</strong>re were some species differences. The cross reactivity of<strong>the</strong> antiserum with <strong>the</strong> equatorial segment inrabbit sperm heads suggest that this protein has antigenic similarity to that ofrat FS proteins, interestingly no crossreactivity was seen in human or rat sperm.REFERENCES(1) Fawcett, D.W. Dev. BioI 1975;44:394-436(2) Kim, Y.H. et al., Proceeding of<strong>the</strong> XIIth North American Testis Workshop. 1993; p 51(3) Almahbobi et al., Histochemical J 1993; 25:14-183 84


•· ISOLATION AND CHARACTERISATION OF RAT SPERM TAIL OUTER DENSE FIBRE PROTEINS MAP2 EXPRESSION IN THE ADULT RAT TESTISY.H. Kim, P.D. Temple Smith*, D.M. de Kretser & lR. McFarlane, Institute of Reproduction and Development,Department ofAnatomy*, Monash University, Clayton, Victoria 3168, AustraliaINTRODUCTIONThe outer dense fibres which surround <strong>the</strong>microtubules of <strong>the</strong> axoneme are located along <strong>the</strong>entire length of <strong>the</strong> sperm tail, and <strong>the</strong>y may beimportant in maintaining <strong>the</strong> passive elasticstructures and elastic recoil of <strong>the</strong> sperm tail (1).The assembly ofrat outer dense fibres is initiated at<strong>the</strong> proximal end of <strong>the</strong> flagellum and progressesdistally during step 16 of spermiogenesis (2). Theisolation of rat sperm outer dense fibres has beenpreviously reported (3,4). The aim ofthis study wasto extend this work and fur<strong>the</strong>r examine <strong>the</strong> proteincomposition of outer dense fibres, by isolating andcharacterising <strong>the</strong> individual protein components of<strong>the</strong> outer dense fibres.DISCUSSIONA1nino acid analysis of<strong>the</strong> structure shows some similarity to <strong>the</strong> fibrous sheath, both being rich in aspartic acid,glutamic acid and serine (5,6), however it contains a higher content of proline and arginine. Although <strong>the</strong> 28-34kDa protein of<strong>the</strong> rat outer dense fibres has been identified as being alternatively spliced <strong>for</strong>ms of<strong>the</strong> rt7 (7) gene,<strong>the</strong> derivation of<strong>the</strong> o<strong>the</strong>r proteins has yet to be determined.REFERENCES(1) Phillips, D.M. (1972) J. Cell BioI. 53:561-573.(2) Irons, MJ, Clermont, Y. (1982) Am J Anat 165:121-130.(3) Vera et al., ( 1984) J BioI Chern 259:5970-5977.(4) Oko, R. (1988). BioI. Reprod., 39:169-182.(5) Brito et al (1989) Gamete Res. 22:205-217.(6) Kim Y.H. et al (1994) Proc 8th European Testis Workshop p126.(7) Van Der Hoorn et al (1990) Dev. BioI. 142: 147-154.14.4.......METHODSFigure 1: Electron micrograph and SDS-PAGE of isolated ratouter dense fibres.Sperm were collected from <strong>the</strong> cauda epididymidesof 12 adult Sprague-Dawley rats. The sperm were filtered through a Nitex screen, and decapitated by sonicationand layered through a sucrose step gradient. Isolated sperm tails were assessed by phase contrast microscopy andextracted in 1% SDS, 2 mM DTI and 25 mM Tris-HCI pH 8 <strong>for</strong> 90 min with shaking at room temperature.Finally, <strong>the</strong> solubilised sperm tail suspensions were centrifuged through a sucrose step gradient and <strong>the</strong> purificationofouter dense fibres was verified by electron microscopy. Electroelution following SDS-PAGE was used to isolateabundant proteins.RESULTSAfter exposure to SDS-DTI, most sperm tail components were solubilised including axoneme, fibrous sheath andmitochondrial sheath while <strong>the</strong> more resistant outer dense fibres remained (Figure 1). SDS-PAGE ·of <strong>the</strong> outerdense fibres revealed at least 13 bands of varying abundance (Figure 1). The range and molecular size pattern of<strong>the</strong>se proteins was similar to that reported by Vera (3) and Oko (4). The most abundant proteins were a large bandbetween 28 kDa-34 kDa, a band at 82 kDa, followed by one 21.5 kDa and 15.5 kDa. We have isolated 8 of <strong>the</strong>most abundant proteins. A1nino acid analysis of whole ODF shows this structure is rich in aspartIc (11 %) andglutamic acid (9%), serine (13%), proline (10%), arginine (10%) and leucine (10%).STOKate Love1Cl;nd, Kristina Zlatic, Terri Hayes, David de Kretser, and Jim McFarlaneInstitute ofReproduction and Development, Monash University, Clayton, Victoria, Australia 3168IntroductionThe testis is an abundant source ofmicrotubule networks which include mitotic and meioticspindles, <strong>the</strong> spermatid manchette and axoneme, and Sertoli cell cytoskeleton. Microtubulesare comprised of CJ. and ~ tubulin subunits, which are polymerised and stabilised by a varietyof microtubule associated proteins (MAPs). Previous studies have shown that <strong>the</strong> testiscontains a variety ofMAPs, and a putative MAP2 protein has been reported inbovine spenn(1). M.AP2 has been shown to bind to and induce polymerisation of microtubules, and isinvolved in cytoskeletal cross-linking and binding to <strong>the</strong> regulatory subunit of cAMPdependentkinase IT (2,3). MAP2 mRNA is processed into at least three alternatively splicedvariants, designated MAP2~ MAP2b and MAP2c. Of<strong>the</strong> 5.7 kb ofcoding sequence presentin <strong>the</strong> 9 kb mRNA which encodes MAP2a and MAP2b, approximately 4 kb of internalsequence is deleted to produce mRNA encoding MAP2c, which consists of only <strong>the</strong> aminoand carboxy terminal regions ofMAP2 (3).o 519 1500 2000-4603t='D-----I~--------1JJ___J'-.-----MAP2R.(--MAP2R2--MAP2R1- -MAP2R3-Figure 1: MAP2 coding sequence and cDNAprobes used in this study.Methods and ResultsMicrotubule preparations were isolated from adult rat testis and brain using taxol-mediatedpolymerisation (4). The polymerised testis samples were analysed by ELISA and Westernblot using an anti-MAPs polyclonal antibody (M7273) which is reported to primarilyrecognise MAP2 and Tau, anti MAP2 (IIM:-2) monoclonal antibody which recognises all<strong>for</strong>ms ofMAP2, and anti-MAP2a+b (AP-20) monoclonal antibody which recognises MAP2aand MAP2b only.Immunoactivity was detected by ELISA using all three antibodies. Western Blots using <strong>the</strong>anti-MAPs antibody (Figure 2, lane A) showed numerous bands ranging from greater than200 kDa to less than 14 kDa with a major band ofimmunoactivity at 70 kDa, ra<strong>the</strong>r than <strong>the</strong>expected 280-300 kDa region <strong>for</strong> MAP2. The anti MAP2 antibody (Figure 2, lane C)revealed a major band at 70 kDa, while <strong>the</strong> MAP2a+b antibody failed to recognise anyproteins (Figure 2, lane B), indicating that MAP2c is present in <strong>the</strong> adult rat testis. Theseresults were confinned using Nor<strong>the</strong>rn blot analysis of total RNA from adult rat brain andtestis, and cDNAprobes which distinguish between MAP2a and MAP2b and MAP2c (Figure3). Brain contains a predominant mRNA of9 kb corresponding to MAP2a and MAP2b and aminor species at 6 kb corresponding to MAP2c. In contrast, <strong>the</strong> testis contains only onepredominant mRNA of6 kb which corresponds to MAP2c.References1. Carr DW and Acott TS (1990): BioI Reprod 43:795-805.2. Garner et al (1988) JBC 106:779-783.3. Kindler et al (1990) JBC 265: 19679-84.4. Valle RB (1982): J Cell BioI 92:435-442.5700200-Discussion 28 S ...These data demonstrate by ELISA, Western and Nor<strong>the</strong>rn Blot analysis that a MAP2c-likeprotein is present in<strong>the</strong> testis. Nor<strong>the</strong>rn blots also indicate that multiple MAP2 mRNA arepresent in <strong>the</strong> testis including species of appoximately 9 kb which could encode MAP2a orMAP2b proteins. These will most like be in association with discrete cell types at specificstages ofdifferentiation and it will be important to establish <strong>the</strong> nature and site ofproductionof<strong>the</strong> various MAP2 mRNAs.66-116 ­97 -55-ARl R2T B T BBR3TBcFigure 2: Western blots ofpurifiedmicrotubule preparations from adultrat testis probed with (A) anti MAPs,(B) anti MAP2a+b, and (C) antiMAP2 antibodies.R4TBFigure 2: Nor<strong>the</strong>rn blot ofadultrat testis (T) and brain (B) totalmRNA probed with (1) Rl, (2)R2, (3) R3, and (4) R4 cDNAprobes.85 86


TIlE EFFECf OF ENVIRONMENTAL TEMPERATURE ON SCROTAL TEMPERATUREMEASURED BY 1ELEMETRY IN RAMS AT PASTURE.Brian P.Setchell 1 , James L.Zupp l, Grace Ekpel, Simon Maddocks 1 and Gordon Grigg 2 .1: Department ofAnimal Science, University ofAdelaide (Waite), Glen Osmond, SA 5064.2:Departrnent ofZoology, University of Queensland, Brisbane, Q 4072.It has been known <strong>for</strong> many years (see (1)) that underlaboratory conditions, <strong>the</strong> temperature of <strong>the</strong> scrotum andhence <strong>the</strong> testes is maintained at about 33°C, considerablybelow body temperature. However, <strong>the</strong>re is littlein<strong>for</strong>mation available on <strong>the</strong> temperature of <strong>the</strong> scrotum ofanimals out in <strong>the</strong> field. It is important to know <strong>the</strong> rangeoftemperatures to which <strong>the</strong> testes oframs are exposedunder field conditions to put into context our studies (2, 3)on <strong>the</strong> effects on semen quality, fertility and embryonicmortality of intermittent scrotal insulation, which raisesscrotal temperature by between 1.5 and 2.5°C.Two adult Merino rams weighing about 55 kgwere anaes<strong>the</strong>tized with pentobarbitone sodium andfluothane, and a small incision made in <strong>the</strong> skin over <strong>the</strong>centre of <strong>the</strong> posterior smface of <strong>the</strong> scrotum, level with <strong>the</strong>middle of <strong>the</strong> testes. A pocket was made in <strong>the</strong>subcutaneous tissue between <strong>the</strong> testes and a temperaturesensitive transmitter (Sirtrack NZ) inserted. The signalfrom this device was a pulse, <strong>the</strong> frequency ofwhich wasrelated linearly to temperature. The skin was sutured withsilk, and after recovery, <strong>the</strong> rams were released into apaddock, from which <strong>the</strong> signal from <strong>the</strong> transmitters couldbe recorded with a Telonics TR-2E 150/154 receiver in <strong>the</strong>nearby laboratory. The transmitters were implanted onJanuary 1 1994, and recordings continued several timesdaily until <strong>the</strong> end of February. At <strong>the</strong> time ofeachrecording, air temperature was measured with a wet and drybulb <strong>the</strong>rmometer fixed about 1 m from <strong>the</strong> ground on a4038~ (1);:,iiiQj 36Q.EQI....ea 34e()tIJ• • y = 31.611 + 0.12361x R"2 = 0.51132 ~Y = 31.459 + 0.11357x R"2 = 0.618 Ram (1)I]f] Y = 31.767 + 0.13356x RI\2 = 0.562 Ram (2)3010 20 30 40 50Air Temperature (degrees Celsius)post on <strong>the</strong> edge of <strong>the</strong> paddock.The summer of 1994 proved to be a particularlymild one, with only one day on which <strong>the</strong> temperature inAdelaide exceeded 400c. Never<strong>the</strong>less, sufficient recordingswere obtained to show that scrotal temperature was linearlyrelated to both dry and wet bulb temperatures. The linearcorrelations derived from 256 observations with airtemperatures ranging from 14 to 42°C, were highlysignificant (P < 0.(01) <strong>for</strong> both rams and <strong>for</strong> <strong>the</strong> pooleddata. Similar relationships were obtained with wet bulbtemperatures. At any given ambient temperature, smalldifferences in scrotal temperatures between <strong>the</strong> two ramsprobably were related to <strong>the</strong> fact that <strong>the</strong> transmitter in ram2 remained between <strong>the</strong> two testes, whereas that in ram 1was located at <strong>the</strong> bottom of <strong>the</strong> scrotum, wheretemperatures are usually about 0.5 0 C cooler (4).It is apparent from this study that at dry bulb airtemperatures above 400C, scrotal temperatures of greaterthan 360C can be anticipated. There<strong>for</strong>e our studies withintermittent scrotal insulation, which raise scrotaltemperature to between 34.5 and 35.5°C <strong>for</strong> as little as8h/day, and produce abnormalities in sperm morphology,fertilising ability and embryo survival (2,3) are entirelyrelevant to <strong>the</strong> field situation, and <strong>the</strong>re<strong>for</strong>e heat stress to<strong>the</strong> males must be considered as a possible factor in <strong>the</strong>high level ofembryonic mortality observed in <strong>the</strong>Australian sheep population.(1). Setchell, B.P. (1978) The Mammalian Testis. Elek Books, London. (2). Mieusset, R., Quintana Casares, P.,Sanchez Partida, L.G., Sowerbutts, S.F., Zupp, J.L. & Setchell, B.P. (1992) J. Reprod.Fertil. 94, 337-343.(3). Ekpe, G., Zupp, J.L., Seamark, R.F. & Setehell, B.P. (1993) Pmc ASRB, 25, 88. (4). Fowler, D.G. (1968)Aust.J.exp.Agric'. 8, 125-132.THE EFFECTS OF MELATONIN IMPLANTATION ON SPERMATOGENESIS IN THEEUROPEAN RED FOX (VULPES VULPES)S. Young AB , M.P. BradleyA, A. Gidley-Baird B and L.A. Hinds A .CRC <strong>for</strong> Biological Control ofVertebrate Pest Populations, CSIRO Division of Wildlife and Ecology, PO Box 84, Lyneham,ACT 2602, Australia A . Department ofAnimal Science, University of Sydney, NSW 2001, Australia B .INTRODUCTIONThe European red fox (Vulpes vulpes) is aseasonal breeder. The mating season occurs during Julyand August in <strong>the</strong> Sou<strong>the</strong>rn hemisphere, and <strong>the</strong> gonadsof each sex exhibit an annual cycle ofregression andredevelopment. Both artificial photoperiod and melatoninimplantation have been used to advance <strong>the</strong> breedingseason in <strong>the</strong> silver fox, a colour mutant of<strong>the</strong> red fox.The aim ofthis study was to advance spermatogenesis in<strong>the</strong> male red fox by administration ofexogenousmelatonin commencing 5 weeks after <strong>the</strong> summer solstice.METHODSOn January 25, 1993, 6 male red foxes (GroupMLT) were implanted with melatonin (Regulin®, 18 mg),and 6 were used as controls (Group C). Fortnightly bloodsamples were collected <strong>for</strong> measurement ofplasmamelatonin, and testis volume was determined weekly. Toassess <strong>the</strong> sensitivity of<strong>the</strong> pituitary-gonadal axis,challenge with a standard dose ofgonadotrophin-releasinghormone (GnRH, 40 ~g) was per<strong>for</strong>med at monthlyintervals (February - July). On March 31 and May 26,blood samples were collected throughout a 24-hour periodto determine <strong>the</strong> pattern ofendogenous melatoninsecretion in V. vulpes. Four foxes (2 from each group)were autopsied in May, June and July, <strong>for</strong> determinationof histological differences in <strong>the</strong> testes and epididymides,and expression ofa fox-specific testis gene (FSA-l).RESULTSGroup lYfLT showed advances in spermatogenicactivity, expression ofFSA-l, and epididymalspermatozoal content. Testis volume was significantlygreater in Group lYfLT from April 11 - June 12 (Fig. 1).No change in GnRH-induced luteinising hormone (LH)release occurred as a function oftreatment or time. Incontrast, Group lYfLT showed a significantly lower releaseof testosterone in response to GnRH challenge duringFebruary - April~ <strong>the</strong>reafter, testosterone release increasedto equal that of Group C in June. A diurnal pattern ofmelatonin secretion was absent in Group C foxes on bothMarch 31 and May 26, and in Group lYfLT foxes on May26. In contrast, all members of Group lYfLT showed aconsistent nocturnal increase in plasma melatonin onMarch 31 (Fig. 2).CONCLUSIONThis study indicates that <strong>the</strong> continuous presenceof elevated plasma melatonin commencing 5 weeks after<strong>the</strong> summer solstice is effective in advancing <strong>the</strong> onset ofspermatogenesis in <strong>the</strong> male red fox by approximately 4weeks. These results extend findings of previous studies(1,2) in <strong>the</strong> silver fox in which melatonin implants placed3 or 8 weeks earlier were also effective in advancingtesticular redevelopment by 4 weeks.'i~ 140 El'l E 120.£.:E:OJ100'Qj~ 80>-"0.0 60iiiE:J40(5> 20III~


»Glucose and acetate utilization. by marsupial epididymal spermatozoa is inhibited by aprostatic secretionR. N. Murdoch and R. C. JonesINTRODUCTIONA knowledge of <strong>the</strong> energy metabolism of epididymalsperm is basic <strong>for</strong> understanding sperm storage in <strong>the</strong>epididymis and <strong>the</strong> effects of <strong>the</strong>ir dilution in seminalplasma during ejaculation. However, <strong>the</strong>re are noreports on <strong>the</strong> metabolism of epididymal sperm from amarsupial and only one report of a metabolic study ofmarsupial spenn (1). The study (1) used ejaculates from<strong>the</strong> brush tailed possum and concluded that <strong>the</strong>endogenous metabolism of <strong>the</strong> sperm was greater thanreported <strong>for</strong> eu<strong>the</strong>rian sperm. Also, <strong>the</strong> rate was notaffected by addition of glucose, or N-acety 0­glucosamine (NAG) which is <strong>the</strong> naturally occurringsugar in accessory gland secretions of marsupials.However, <strong>the</strong> study confounded <strong>the</strong> metabolism ofsperm with that of prostatic bodies which were alsopresent in <strong>the</strong> semen (2).This report describes <strong>the</strong> metabolism of epididymalsperm from <strong>the</strong> mo~t studied marsupial (<strong>the</strong> tammarwallaby, Macropus eugenil).MATERIALS AND METHODSSamples of luminal fluid (1 00-200 ~I) were flushed withparaffin oil from <strong>the</strong> distal cauda epididymidis of <strong>the</strong>tammar, washed once in 0.5 ml Krebs-Ringerphosphate (pH 7.4) and incubated (1 ml; 0.7-1.6 x 10 8sperm! flask) at 37°C in Warburg flasks containing 0.1ml 20% (w/v) KOH in <strong>the</strong> centre well. Depending upon<strong>the</strong> design of <strong>the</strong> experiment compounds were added to<strong>the</strong> incubation media: D-[U- 14 C]glucose (5 ~moles; 500nCi), C 4 C]acetate (5 ~moles; 500 nCi) and NAG (5~moles). Oxygen consumption was measured by usingair as <strong>the</strong> gas phase. Glucose and acetate oxidationwere detennined by measuring 14C02 trapped in <strong>the</strong>KOH while glucose and lactate concentrations weremeasured by enzymatic methods. The data shown inTable 1 are <strong>for</strong> <strong>the</strong> first 2 h of incubation.RESULTSA preliminary study showed that in <strong>the</strong> absence ofexogenous substrate sperm can oxidise endogenoussubstrate <strong>for</strong> up to 8 hours. Table 1 shows that additionof exogenous substrate stimulated oxygenconsumption, with acetate having a greater stimulatoryeffect than glucose or NAG. Experiment 1 shows thatglycolysis produced more lactate from NAG thanglucose, and that glucose utilization was completelyinhibited by <strong>the</strong> presence of NAG. Experiment 2 showedthat NAG also reduced <strong>the</strong> utilization/oxidation ofacetate, but only by about 50%.Department of Biological Sciences,University ofNewcastle, NSW, Australia 2308DISCUSSIONIt is concluded that tammar epididymal sperm have ametabolic rate about <strong>the</strong> same as bull and ram sperm,but probably have a greater store of endogenoussubstrate <strong>for</strong> energy metabolism than sperm fromeu<strong>the</strong>rian mammals. Oxygen consumption by tammarsperm can be stimulated by <strong>the</strong> addition of exogenoussubstrate with glucose and NAG having about <strong>the</strong> sameeffect, but a smaller effect than acetate. However,glycolysis is greater with NAG than glucose assubstrate, and <strong>the</strong> presence of NAG reduces <strong>the</strong>utilization of glucose and acetate by tammar epididymalsperm. Since glucose utilization is completely inhibitedby <strong>the</strong> presence of NAG it is concluded that NAG actsto inhibit <strong>the</strong> transport of glucose into <strong>the</strong> cell.Table 1. Effects of exogenous substrate on <strong>the</strong>metabolism ofsperm from <strong>the</strong> tammar wallaby. Means± sem from 5 tammars expressed as rate/ 1(f sperm!2hours.SubstrateOxygenuptake(~I)Glucoseutilized(~mole)Lactateproduced(~mole)Glucoseoracetateoxidised(~mole)Experiment 1:None 62 ± 4.8 0.0 ± 0.00Glucose 79 ± 7.3 1.8±0.47 2.2 ± 0.40 0.5 ± 0.1NAG 78 ± 8.2 4.2 ± 0.42Glucose+ NAG 79 ± 7.9 0.0 ± 0.00 3.7 ± 0.39 0.01 ± 0.0Experiment 2:None 47 ± 5.7 0.0 ± 0.00Acetate 75 ± 6.4 1.3 ± 0.00 1.3 ± 0.1NAG 54 ± 3.9 2.3±0.12Acetate+ NAG 58 ± 4.9 2.0 ± 0.23 0.7 ± 0.01REFERENCES1. Rodger, J.C. & Suter, D.A.1. (1978) GameteResearch 1, 111-116.2. Rodger, J.C. & White, I.G. (1976) J. Reprod. Fert.46, 467-469.CALCIUM INFLUX INTO MOUSE SPERM ACTIVATED BY SOLUBILIZED MOUSE ZONAEPELLUCIDAE IS INHIBITED BY THREE INHIBITORS OF THE ZONA-INDUCED ACROSOMEREACTION: 3-QUINUCLIDINYL BENZILATE, TYRPHOSTIN A-48, AND PERTUSSIS TOXIN.Janice L. Bailey and Bayard T. StoreyDepartment of ObstetriCs & Gynecology, University of Pennsylvania Medical Center, Philadelphia, PA 19104, USAINTRODUCTION. The acrosome reaction in mouse sperm bound to <strong>the</strong> zona pellucida of <strong>the</strong> mouse egg is induced by <strong>the</strong>zona glycoprotein component ZP3 [1]; this induction is blocked by <strong>the</strong> muscarinic antagonist, 3-quinuclidinyl. bcnzilate(QNB) [2]. Exogenous calcium has long been known to be obligate <strong>for</strong> occurrence of <strong>the</strong> acrosome reactIOn. Wehypo<strong>the</strong>sised that a zona-induced rise in intracellular calcium [Ca2+]i would be an early precursor reaction to acrosomalexocytosis and that acrosome reaction inhibitors should block <strong>the</strong> zona-induced [Ca 2 +]i increase. We report here a test ofthis hypo<strong>the</strong>sis, using <strong>the</strong> fluorescent Ca 2 + indicator fluo-3 to monitor mouse sperm [Ca 2 +li.MATERIALS AND METHODS. Male mice were CD1 retired breeders. Sperm were obtained from excised caudaepididymides by swim-out into bovine serum albumin (BSA)-free medium MJB [3] and incubated with 5 pM fluo-3 tetraacetoxymethyl(AM) ester <strong>for</strong> 20 min at 25° to load <strong>the</strong> cell-penneant ester, which <strong>the</strong>n hydrolyses to g~ve <strong>the</strong> ce~limpenneanttetracarboxylate indicator. Exogenous dye was removed by centrifugation, ~d <strong>the</strong> cells w~re .<strong>the</strong>~ lllcu?a~ed IIIMJB containing 20 mg/mL BSA <strong>for</strong> capacitation. Microscope examination showed um<strong>for</strong>m fIuo-3 dlstnbutlOn wlthlll <strong>the</strong>cells; chlortetracycline (Cfe) assay [4] showed 75-80% capacitated spenn. Spenn [Ca 2 +]i was monitored at 37° by fIuo-3fluorescence at 530 nm, using 498-436 nm dual wavelength excitation. Intracellular fluo-3 fluorescence was stable over <strong>the</strong>experiments' duration in control tests, showing acceptable dye retention. Acrosome reactions were scored using <strong>the</strong> CfCassay [4]. Solubilised mouse zona pellucida protein (SMZP) was prepared as described by Ward et al. [5].RESULTS. The parallel effects of added SMZP in <strong>the</strong> absence and presence of 50 pM QNB; 12.5 pM tyrphostin A48, atyrosine kinase inhibitor; and 10 ng/mL pertuss"is toxin (PTX) , an inactivator of Gi proteins, on [Ca 2 +]i and inducedacrosome reactions(% AR) incapacitated mouse spenn are 150....,----------------------,-50shown in Figure 1.40Fig. 1 Increase in [Ca 2 +]i~ 100c:over baseline of 231 ±58 nM ,..:r(filled bars) and in % AR of ~intact cells (stippled bars» ~ 50over 20±5 % spontaneous ARinduced by SMZP (X ± SE, n:±ZP, 9; +QNB, 7; +A48,5; o+PTX,4).noZP+ZP_..0.&.-+-0+ ZP + QNB + ZP + A48 + ZP + PTXACKNOWLEDGMENT. This work was supported by U.S. National Institutes of Health grant HD-06274.REFERENCES.1. Bleil, J.D., and Wassarman, P.M. (1983) Dev. BioI. 95, 317-324.2. Horman, H.M., and Storey, B.T. (1982) Dev. BioI. 91, 121-130.3. Storey, B.T., Hourani, c.L., and Kim, J.B. (1992) Mol. Reprod. Dev. 32,41-50.4. Ward> C.R., and Storey, B.T. (1984) Dev. BioI. 104,287-296.5. Ward, C.R., Storey, B.T., and Kopf, G.S. (1992) 1. BioI. Chern. 269, 13254-13258.30 a:(20 #The increase in [Ca 2 +]i induced by addition of SMZP (+ ZP) was significant (P < 0.05) compared to control (no ZP);increases in [Ca 2 +]i induced by SMZP in <strong>the</strong> presence of inhibitors were not significant compared to control (P =0.65).Addition of <strong>the</strong> ionophore Br-A23187 after establishment of <strong>the</strong> SMZP-induced stea~y state [Ca2+li resulted in <strong>the</strong> followingfur<strong>the</strong>r [Ca 2 +]i increases (nM; X ± SE, n as in Fig.I): no ZP, I04±29; + ZP, 110±17; + ZP + QNB, 157±42; + ZP +A48, 114±42; + ZP + PTX, 124±49. These fur<strong>the</strong>r increases in [Ca2+:k were not significantly different (P = 0.65). Theincrease in percent acrosome reacted sperm was significant only <strong>for</strong> + ZP, P < 0.001; <strong>for</strong> <strong>the</strong> o<strong>the</strong>r conditions, P = 0.2.DISCUSSION. The increase in [Ca 2 +]i induced by SMZP is tightly correlated with induction of <strong>the</strong> acrosme reaction, inagreement with results reported earlier [2]. Of particular interest is <strong>the</strong> observation that <strong>the</strong> three inhibitors, QNB, A48, andPIX, each of which acts on a different intracellular signal transduction reaction, all block <strong>the</strong> SMZP-induced [Ca2+]iincrease and <strong>the</strong> SMZP-induced acrosome reaction. We conclude from this observation that <strong>the</strong> sperm [Ca 2 +Ji increaseinduced by <strong>the</strong> zona pellucida is an early reaction in <strong>the</strong> sequence leading to zona-induced exocytosis. Still to be resolved is<strong>the</strong> question of whe<strong>the</strong>r <strong>the</strong> Br-A23187-induced increase in [Ca 2 +]i, which is not affected by <strong>the</strong> inhibitors and appears to beaddtive to <strong>the</strong> SMZP-induced increase, represents a pathway <strong>for</strong> Ca 2 + influx independent of that <strong>for</strong> <strong>the</strong> zona-induced ea 2 +influx, and whe<strong>the</strong>r Ca 2 + influx per se is sufficient to induce acrosomal exocytosis.1089 90


Acrosomal Integrity of Tammar Wallaby (Macropus eugenii)Spermatozoa After Treatment with Penetrating (TCEP) and Non-Penetrating (GSH)Reducing AgentsDadi Setiadi, Yulia Sistina and John C. RodgerDepartment of Biological Sciences, University of Newcastle, NSW 2308INTRODUCTIONStudies of <strong>the</strong> acrosome of marsupial spermatozoa have shownthat it is relatively stable compared with that of placentalmammals and this appears to be at least partially due to <strong>the</strong>properties of <strong>the</strong> acrosomal membranes (1-2). The marsupialacrosome contains significant concentrations of disulphides andthiols, which are likely to contribute to its stability (3). This studyfur<strong>the</strong>r examined <strong>the</strong> basis of marsupial acrosome stability byexamining <strong>the</strong> effects of S-S reducing agents on acrosomalintegrity and sperm motility. The two agents used were nonmembranepermeable agent glutathione (GSH) and permeable Tris(2-carboxyethyl)-phosphine Hydrochloride (TCEP).MATERIAL AND METHODSSpermatozoa were obtained by electroejaculation, washed by"swim up" in Phosphate Buffer Saline (PBS) and adjusted to afinal concentration of 60x10 6 sperm/ml. Glutathione (GSH,Sigma, St. Louis, USA) and Tris(2-carboxyethyl)-Phosphinehydrochloride (TCEP, Pierce, illinois, USA) were diluted in PBSand added to spermatozoa to achieve final concentrations of: 0.0,0.5, 1.0,2.0,4.0, 8.0, and 16.0 mM; <strong>the</strong>n incubated <strong>for</strong> 180 minat RT. At <strong>the</strong> end of incubation. motility was assessed andspermatozoa fixed with 2% para<strong>for</strong>maldehyde in PBS; stained byBryan's stain and acrosomes classified as normal, altered orabsent (2). In addition spermatozoa were processed <strong>for</strong> mBBrlabelling of SH groups as described previously (3). Two hundredspermatozoa per treatment were counted <strong>for</strong> each replicate.Statistical analysis was by Student t test.RESULTSBryan's staining revealed that <strong>the</strong> number of altered acrosomeswas maximum at 8.0 mM GSH (7.2%10.60) and 4.0 mM TCEP(15.9%±2.68). The percentage of spermatozoa without anacrosome was maximal with: 16.0 mM GSH (98.4%±0.44) and8.0 mM TCEP (99.5%±0.14). Moreover <strong>the</strong> percentage ofspermatozoa without an acrosome was significantly greater <strong>for</strong>TCEP treatment compared with GSH treatment at 2.0, 4.0 and 8.0mM (P81 % and 3.5) or 1.0 mM TCEP (>79% and 3.0).However, all spermatozoa were immotile with; 8.0 mM GSH at60 min and 4.0 mM TCEP at 30 min incubation. After 3 hoursincubation in 4.0 mM GSH or TCEP, 75% and 97% ofspermatozoa respectively had fluorescent acrosomes. In additionmBBr labelling also allowed visualisation of swollen alteredacrosomes (Fig. 1).DISCUSSIONThe motility data suggested that both TCEP and GSHcompromised <strong>the</strong> survival of tammar wallaby spermatozoa andTCEP was deleterious at a lower concentration. Similarlyacrosomalloss and alteration was seen at lower concentrations ofTCEP than GSH. The greater potency of TCEP was probablydue to <strong>the</strong> fact that it penetrated <strong>the</strong> plasma membrane. Theeffects of GSH, which does not penetrate <strong>the</strong> plasma membrane,Fig. 1. Acrosome (arrow) of tammar wallaby spermatozoa treatedwith <strong>the</strong> fluorescent SH label mBBr. (a) Untreated - Acrosome isnot visible (b) TCEP treated - A veil of moderately fluorescentacrosomal material lies above <strong>the</strong> brightly f1uorescent condensedacrosome seen in lateral view.must have been mediated through <strong>the</strong> surface of <strong>the</strong> spermatozoa.Wallaby spermatozoa appear very sensitive ~o GSH. Guinea pigspermatozoa retained 85% motility after incubation in 40 mM <strong>for</strong>70 min (4). GSH at concentrations >4 mM and TCEP > 2 mMlead to rapid loss of motility by wallaby spermatozoa.CONCLUSIONSTreatment with ei<strong>the</strong>r reducing agent resulted in disruption of <strong>the</strong>acrosomal contents (altered) and acrosomal loss with <strong>the</strong>penetrating agent being effective at lower concentrations.However, in both cases major acrosomalloss was not seen untildeleterious concentrations of reducing agent were present. SinceGSH treatment had little effect on <strong>the</strong> acrosomes of livingspermatozoa S-S stabilisation of<strong>the</strong> plasma membrane is unlikelyto playa part in <strong>the</strong> stability of <strong>the</strong> marsupial acrosome.REFERENCES1) Mate,KE. and Rodger,J.C.(l991) 1.Reprod.Ferti1.91,41-48.2) Sistina, Y., et al. (1993) Reprod. Ferti!. Dev. 5,1-14.3) Mate, KE., et al. (1994) Molec. Reprod. Dev. 37, 318-325.4) Fleming, A.D., et al.,(l986) Gamete Res. 13, 93-102.The Effect of 2-Mercapto-ethanol (2-ME) on <strong>the</strong> Motilityand Acrosome of Tammar Wallaby SpermatozoaDadi Setiadi and John C. RodgerDepartment of Biological Sciences, University of Newcastle, NSW 2308INTRODUCTIONFertilisation in vitro is now a routine procedure in many speciesof placental mammals, but has not been achieved <strong>for</strong> anyAustralian marsupial. The remarkable stability of <strong>the</strong> marsupialacrosome (1) may contribute to <strong>the</strong> problem of obtaining-spermeggbinding in vitro. The stability of <strong>the</strong> acrosomal membranesis due at least in part to disulphide (S-S) bonds (2) and reductionof S-S may have a role in capacitation. The present studyexamined <strong>the</strong> potential of <strong>the</strong> reducing agent 2-ME to convert S-Sbonds to SH (thiol) groups at concentrations which maintainedmotility.MATERIAL AND METHODSSpermatozoa were collected by electroejaculation, washed by"swim up" into Phosphate Buffer Saline (PBS) and <strong>the</strong>irconcentration adjusted to approximately 6OxlQ6 sperm/ml. One mlof spermatozoa was added to 1 ml 2-ME (Sigma, St. Louis, USA)<strong>for</strong> final concentrations of: 0.0, 0.05, 0.5, 5.0, 25.0, 50.0 and100.0 mM and <strong>the</strong>n incubated <strong>for</strong> 3 hours at RT. The motility(%) and progressive motility (1-5) were assessed at 0,30,60, 120and 180 minutes. Fluorescent labelling of thiol groups withmonobromobimane (mBBr) (Calbiochem, La Jolla, CA) wascarried out as previously described (1). Acrosomes were classifiedas Non-Fluorescent, Fluorescent around <strong>the</strong> Perimeter or EntirelyFluorescent. Two hundred spermatozoa were assessed pertreatment and <strong>the</strong> data were analysed using Student's t test aftersquare root trans<strong>for</strong>mation.RESULTSPrior to culture in vitro less than 10% of untreated wallabyspermatozoa had an entirely fluorescent acrosome. The numberof spermatozoa with fluorescent acrosomes (Fig. 1) was higher inall treated groups than in <strong>the</strong> control. After treatment with 5, 25,50 and 100 mM 2-ME <strong>the</strong> percentage of spermatozoa wi<strong>the</strong>ntirely fluorescent acrosomes was >95%, and with 0.05, 0.5 mM2-ME, it was between 85-90%. However after 180 min, 50% ofcontrol spermatozoa also had fluorescent acrosomes. Spermmotility remained high during <strong>the</strong> incubation in 0.05,0.5 and 5.0mM 2-ME, but motility dropped to: 35% after 120 min in 25 mM2-ME; 28% after 60 min in 50 mM 2-ME and 13% after 30 minin 100 mM 2-ME. Progressive motility remained high (>3.5)during 3 hours incubation with concentrations of 2-ME under 25roM, but it dropped to: 1.5 after 60 min with 25 mM 2-ME , 1.33after 30 min with 50 mM 2-ME and 1.00 after 30 min with 100mM 2-ME (Fig. 2).DISCUSSIONIncubation of wallaby spermatozoa with or without addedreducing agent resulted in reduction of acrosomal disulphidebonds as indicated by an increase in <strong>the</strong> number of spermatozoawith entirely fluorescent acrosomes. However, essentially allspermatozoa had fluorescent acrosomes in 2-ME treated groups.In addition spermatozoa treated with 2-ME, below 25 mM werehighly motile <strong>for</strong> up to 60 min. Spermatozoa retained highmotility (>80%) and a high percentage of entirely fluorescentacrosomes (>90%) after 180 min incubation in 5 mM 2-ME. Theresponse of spermatozoa to 2-ME was more encouraging thanwith o<strong>the</strong>r reducing agents (GSH and TCEP) which resulted inFig. 1 The entirely fluorescent acrosome (arrow) of tammarwallaby spermatozoa after treatment with 2-ME.-.LOI~'-"4>.....-+oJ0-1.r-!-+oJ~ 3CIJ~ 2MQCJ5__---I~~"I_--...f'AoM0-.1o 60 120 180Incubation Time (min)o.0mM\l .05 mM .5 rnM.6.=5mM• 25 mM.1 ". 50 mM• 100 mMFig. 2 The effect of 2-ME on progressive motility of tammarwallaby spermatozoa.loss of <strong>the</strong> acrosome and poor motility at low concentrations (3).We are now examining 2-ME treatment as a capacitating strategyon <strong>the</strong> assumption that after S-S cleavage <strong>the</strong> acrosomalmembrane may become less stable, able to bind <strong>the</strong> zonapellucida and undergo an acrosome reaction.REFERENCES1) Mate, KE. and Rodger, J.C. (1991) 1. Reprod. Fertil.91,41-48.2) Mate, KE., et al' (1994) Molec. Reprod. Dev. 37,318-325.3) Setiadi, D., et al. (1994) Proceedings ASRB.91 92


------------------------------------------------------1IIIIIIIIIIIIl.-----------------------------~RELAXATION OF H19 IMPRINTING IN MOUSE ANDROGENETIC EMBRYONIC STEM CELLSKaren E. Mate, Angela J. Villar & Roger A. PedersenLaboratory of Radiobiology & Environmental Health, University of Cali<strong>for</strong>nia, San Francisco, USAINTRODUCTIONMammalian development is unique in itsrequirement <strong>for</strong> both <strong>the</strong> maternal and paternal genomes,owing at least in part to genomic imprinting, which resultsin inequality in <strong>the</strong> activity of paternally and maternallyderived genes (1). The abnormal development ofpar<strong>the</strong>nogenetic and androgenetic embryos has <strong>the</strong>re<strong>for</strong>ebeen attributed to inappropriate expression patterns of<strong>the</strong>se imprinted genes. R19, one of <strong>the</strong> five imprintedgenes identified to date, is expressed exclusively from <strong>the</strong>maternal allele in <strong>the</strong> postimplantation mouse embryo, andis lethal when over expressed (2). We have <strong>the</strong>re<strong>for</strong>eexamined <strong>the</strong> expression ofR19 in wild type, androgeneticand par<strong>the</strong>nogenetic embryonic stem (ES) cells todetermine whe<strong>the</strong>r parental-specific gene expressioncontributes to <strong>the</strong> abnormal development of isoparentalembryos.MATERIALS AND METHODSES cell lines derived from wild type, androgeneticand par<strong>the</strong>nogenetic embryos were used to overcome someof<strong>the</strong> constraints ofworking with isoparental embryos. EScells are <strong>the</strong> in vitro counterpart of blastocyst inner cellmass cells and <strong>the</strong>ir differentiation into embryoid bodiesmimics events that occur in <strong>the</strong> peri-implantation embryo.Androgenetic ES cell lines were derived from androgeneticembryos produced by pronuclear transfer using hybridzygotes from <strong>the</strong> mating of CD-l females (GPIa) andB6CBA Fl (GPIb) males. Par<strong>the</strong>nogenetic ES cells werederived from par<strong>the</strong>nogenetic embryos produced byactivation with 7% ethanol <strong>for</strong> 4 min at ambienttemperature. ES cells were plated initially with STO cellfeeders in medium containing cytokine leukemia inhibitoryfactor (LIF). Be<strong>for</strong>e cells were harvested <strong>for</strong> RNApreparation, undifferentiated ES cells were grown in feederfree gelatine coated dishes <strong>for</strong> at least three passages inmedium containing LIF. Differentiated embryoid bodieswere produced by culturing feeder free ES cells inuncoated bacteriological plates. Total RNA was preparedby <strong>the</strong> GuSCN-CsCI gradient ultracentrifuge technique and15 Ilg per lane was loaded onto a <strong>for</strong>maldehyde gel <strong>for</strong>nor<strong>the</strong>rn blotting.RESULTS & DISCUSSIONThe morphology of wild type, androgenetic andpar<strong>the</strong>nogenetic ES cells appeared similar during cultureand after differentiation into embryoid bodies, indicatingthat <strong>the</strong>y followed a similar developmental timetable. Anysignificant contribution to <strong>the</strong> levels of Ri9 mRNAdetected in ES cells by contaminating STO feeder cells wasdiscounted, as less than 151lg of STO cell RNA did notcontain detectable levels of H19 RNA.RI9 RNA was detected at varying levels in threelines of undifferentiated androgenetic ES cells (Fig la).Overall <strong>the</strong> level of expression was similar to that detectedin wild typeES cells. The levels ofRi9 RNA increased inall three lines of androgenetic cells after differentiation intoembryoid bodies. Ri9 RNA was detected in par<strong>the</strong>nogeneticES cells at high levels (Fig 1b), consistent withover-expression due to <strong>the</strong> presence of two activelytranscribed alleles. The levels ofR19 RNA increased afterdifferentiation of par<strong>the</strong>nogenetic ES cells into embryoidbodies.Detection ofR19 expression in androgenetic EScells indicates an alteration of <strong>the</strong> imprint status normallyassociated with this gene. Alternatively <strong>the</strong>se levels ofR19may reflect those present in normal early embryonic cells,implying <strong>the</strong> existence of a multistep imprinting process asrecently suggested (3). The over expression of R19 inpar<strong>the</strong>nogenetic ES cells raises <strong>the</strong> possibility thatexcessive R19 mRNA may be a factor in <strong>the</strong> abnormaldevelopment of par<strong>the</strong>nogenetic embryos, which is fur<strong>the</strong>rsupported by <strong>the</strong> in utero lethality ofR19 transgenic mice(2). The occurrence of relaxed imprinting also providesopportunities <strong>for</strong> examining <strong>the</strong> concordance between lossor maintenance of parental allele-specific patterns oftranscription and methylation of specific sites in imprintedgenes.(a)Figure 1: Expression of H19 by (a) three lines ofandrogenetic ES cells (ES) and embryoid bodies (EB); (b)four lines ofpar<strong>the</strong>nogenetic ES cells (BS) and two lines ofembryoid bodies (EB).REFERENCES(1) Surani, M.A.H., Barton, S.C. & Norris, M.L. (1984)Nature 308, 548-550.(2) Brunkow, ME. & Tighlman, S.M. (1991) Genes Dev.5, 1092-1101.(3) Latham, K.E., Doherty, A.S., Scott, C.D. & Schultz,R.M. (1994) Genes Dev. 8, 290-299.CHANGES IN THE METABOLISM OF ENERGY SUBSTRATES WITHIN THEFIRST CELL-CYCLE OF MOUSE EMBRYOSINTRODUCTIONMATERIALS AND METHODSRESULTSExperiment 1: Data <strong>for</strong> <strong>the</strong> metabolism ofenergy substrates byQs embryos is presented in Table 1. The metabolism of allsubstrates examined varied with time, though <strong>the</strong> pattern ofchange varied between substrates (Figure 1). Utilization ofglucose via glycoll.sis eH-Glu) and <strong>the</strong> amount of glucoseoxidized to CO eC-Glu) decreased with time, <strong>the</strong> greatestdecrease being observed between time periods commencing at 24and 28 hours post hCG. Pyr metabolism was characterised by anincrease between 24 and 28 hours with a significant decreaseobserved between 28 and 32 hours post hCG. Lact metabolismremained relatively constant <strong>for</strong> <strong>the</strong> first two time periods butdecreased significantly during <strong>the</strong> time period commencing 32hours post hCG. The metabolism of GIn <strong>for</strong> embryos developingin vivo was characterised by a substantial increase between 24 and28 hours post hCG followed by a decrease at 32 hours.Experiment 2: Whereas time-course changes in Glu, Pyr andLact metabolism <strong>for</strong> in vitro grown embryos did not differsignificantly from <strong>the</strong>ir in vitro cultured counterparts (Experiment1), <strong>the</strong> metabolism of GIn was markedly different (Figure 1). Thatis, in contrast to <strong>the</strong> increase observed at 28 hours post hCG <strong>for</strong>freshly collected embryos, no significant change with time wasobserved <strong>for</strong> embryos cultured in vitro. Significant differencesdue to strain of male used <strong>for</strong> matings were also observed <strong>for</strong> 14C_Glu (P=0.025) and GIn (P=0.034) parameters (Figure 2). Forboth substrates, values observed <strong>for</strong> embryos derived from Qsmales were higher than <strong>for</strong> C57BL males at all observation times,<strong>the</strong> pattern of change with time also differing <strong>for</strong> GIn metabolism.DISCUSSIONChanges in metabolism between embryos of different cell-stageshave been well documented in <strong>the</strong> mouse (3,4,5). The presentstudy demonstrates significant variations in <strong>the</strong> metabolism ofenergy substrates within <strong>the</strong> first cell-cycle of <strong>the</strong> mouse embryoand <strong>for</strong> glutamine metabolism, a marked interaction with <strong>the</strong>extent of culture in vitro prior to <strong>the</strong> period of assay. Thesignificant effect of paternal genotype on metabolism wouldappear <strong>the</strong>re<strong>for</strong>e to reflect embryos at varying stages of<strong>the</strong> firstJohn P. Ryan and Kathleen M. WaiteHuman Reproduction Unit, Royal North Shore Hospital, St.Leonards, NSW, 2065.A previous study (1) demonstrated differences in <strong>the</strong> metabolismof energy substrates of pronucleate and 2-cell mouse embryosderived from Quackenbush strain (Qs) females mated to males ofdifferent strains. In <strong>the</strong> mouse, paternal influences have also beenobserved <strong>for</strong> <strong>the</strong> duration of S phase of <strong>the</strong> first cell-cycle andtime to first cleavage (2). The present study investigated whe<strong>the</strong>r<strong>the</strong> metabolism of energy substrates differed within <strong>the</strong> first cellcycleof mouse embryos.Experiment 1: Pronucleate embryos were collected at 24,28 and32 h post hCG from superovulated Qs females that had beenmated to Qs malrs. The ab~lity of embryos to conv~rt 1.0 mMD[5- 3H]glucose ( H-Glu) to H 0 and 1.0 mM D[U- C]glucose,0.4 mM [2-14C]p:xruvate (Pyr); 4.0 mM DL[1- 14 C]lactate (Lact)and 0.5 mM L[U-14C]glutamine (GIn) to 14CO were immediatelyassessed during 2 hour incubation period~ using a methodpreviously described by O'Fallon and Wright (1986) (3).Experiment 2: Pronucleate embryos were collected at 24 posthCG from Qs females mated to Qs or C57BL males and ei<strong>the</strong>rprocessed immediately to assess <strong>the</strong>ir abilities to utilize <strong>the</strong> abovementioned substrates or cultured in Human Tubal Fluid (HTF)media + 3 mg BSA!ml until 28 or 32 h post hCG at which time<strong>the</strong>y were assessed. Statistical analysis: Data were analysed byanalysis of variance with means compared by Newman-Keulstests.cell-cycle as a result of possible differences in <strong>the</strong> timing offertilisation, rate of early embryonic development or <strong>the</strong>irsusceptibility to in vitro culture conditions. Fur<strong>the</strong>r studies appearwarranted to assess <strong>the</strong> association between metabolism andspecific phases of<strong>the</strong> cell-cycle and to investigate <strong>the</strong> relationshipbetween changes in metabolism and subsequent embryonicdevelopment.Table 1. Metabolism of energy substrates (fmoles!embryo.h- 1 ,mean ± s.e.m.) by Qs derived embryos 24, 28 and 32 hours posthCG.Expt Time 3 t~belledEnergy Substrate(h) H-Glu C-Glu Pyr Lact:( c:h


METABOLISM OF ENERGY SUBSTRATES BY POLYSPERMIC MOUSE EMBRYOS INSULIN AND IGF-I STIMULATE GLUCOSE TRANSPORT IN MOUSE BLASTOCYSTS VIA THEIGF-I RECEPTORKathleen M. Waite and John E RyanMarie Pantaleon and Peter KayeDepartment of Physiology and Pharmacology, The University ofHuman Reproduction Unit, Royal North Shore Hospital, St.Leonards, NSW, 2065.Queensland, Brisbane, 4072, AustraliaINTRODUCTIONIn <strong>the</strong> mouse, significant increases in <strong>the</strong> metabolism ofglucoseand pyruvate have been observed as a result offertilization (1).No such effect was observed <strong>for</strong> par<strong>the</strong>nogenetically activatedoocytes, suggesting that <strong>the</strong> fertilizing sperm or a sperm relatedfactor plays an important role in initiating and/or maintainingappropriate metabolic processes within <strong>the</strong> developing embryo.This study investigates <strong>the</strong> metabolism of energy substrates bypolyspermic mouse embryos.MATERIALS AND METHODSExperimental animals. Oocytes were collected fromsuperovulated Quackenbush strain (Qs) females at 8 weeks of age,15 hours post hCG. Cumulus cells were removed in Hepes­Human Tubal Fluid (HTF) media supplemented with 150i.u.hyaluronidase/ml. The zonae pellucidae were removed from <strong>the</strong>majority of oocytesby brief exposure to an acidified Tyrode'ssolution (pH 2.5). Zona-free oocytes \\jere rcubated in RTF + 3mg BSA/ml in <strong>the</strong> presence of 10 -10 motile sperm /ml,collected from Qs males. Experimental design. A randomselection of <strong>the</strong>se oocytes were assessed individually during a 2hour incubation period ~t 6 h post igsemination <strong>for</strong> <strong>the</strong>ir abilitiesto convert 1.0 mM D[5- H]glucose ( H-Glu) to 3H 0 (an estimateoft~i total amount of ~lucose utilised via glycolysis) and 1.0 mM[U- C] glucose to 1 CO (an estimate of <strong>the</strong> total amount ofglucose oxidised), simultineously (2). Th amounts of 0.4 mM[2_r 14 C]pyruvate (Pyr) or 0.5 mM L[U- 4C]glutamine (GIn)oxidised to 14CO during 2 hour incubation periods were alsoassessed. Zona-intact (ZI) and zona-free (ZF) oocytes not exposedto sperm were used as controls. Following <strong>the</strong> assay period, alloocytes were fixed, stained with orcein and examined <strong>for</strong> <strong>the</strong>presence of sperm tails and pronuclei (PN, includingdecondensing sperm heads). Statistical analysis. Data wereanalysed by analysis of variance with means compared byNewman-Keuls tests.RESULTSThere was no significant difference in metabolism of glucose,pyruvate or glutamine observed between zona intact and zonafree oocytes. Glucose metabolism, both <strong>the</strong> total amountutilised via glycolysis and <strong>the</strong> amount oxidised to COincreased significantly with fertilization as evidenced by th~presence of 2 PN (Figure 1). A linear increase in <strong>the</strong> metabolismof glucose was also observed as <strong>the</strong> number of PN identified inpolyspermic embryos increased. In contrast to glucose, nosignificant changes in <strong>the</strong> metabolism of pyruvate or glutaminewere observed irrespective of <strong>the</strong> number of PN (Figures 2 and3).DISCUSSIONThese data support a role <strong>for</strong> sperm or a sperm related factor inregulating <strong>the</strong> metabolic competency of <strong>the</strong> early mouse embryo.The significance of<strong>the</strong> increased metabolism ofglucose with anincrease in <strong>the</strong> number ofpronuclei is unknown. It may, however,represent a mechanism whereby development of polyspermicembryos is different to normally fertilised oocytes. The two mainhypo<strong>the</strong>ses proposed as to how sperm cause oocyte activation and<strong>the</strong> generation of calcium oscillations are by <strong>the</strong> binding of spermto cell-surface receptors and <strong>the</strong> introduction into <strong>the</strong> oocyte of acytosolic sperm fraction (3). The present study has not addressedwhe<strong>the</strong>r it is <strong>the</strong> fusion of multiple sperm to <strong>the</strong> plasma membraneor an increased dose of a sperm specific factor into <strong>the</strong> oocyte thatis responsible <strong>for</strong> <strong>the</strong> observed effect on glucose metabolism.How <strong>the</strong> metabolism of energy substrates might be associated to<strong>the</strong> complex series of events associated with oocyte activationawaits fur<strong>the</strong>r investigation.fmoles/embr."o/h (mean +/- s.e.m (n»2500,...------"------------------..,_ Total Utilisation l22:I Oxidisedd2000 .1500 ': .1000 .30500 .200ZI ooc."te ZF ooc."teZI ooc."te ZF ooc."te 2 PH 3 PHTrea1;Ment GroupZI ooc."te ZF ooc."te 2 PH 3 PHTrea1;Men1; Group4 PH4 PH4 PH5 PHFigure 1. Metabolism of 1.0 mM glucose eH-Glu, 14C_Glu) bymouse oocytes and embryos having 2, 3, 4 or 5 pronuclei. Values<strong>for</strong> each metabolic parameter with different superscripts differsignificantly (P


•ENDOCYTOSIS OF FLUID BY MOUSE BLASTOCYSTSGreta Dunglison 112 and Peter Kaye 11Department of. Phtsiol~gy and Pharmacology, The University of Queensland, Brisbane,4072, Australla; Nuffleld Department of Obstetrics and GynCEcology, John RadcliffeHospital, Ox<strong>for</strong>d, OX3 9DU, UK .INTRODUCTIONEndocyto~s and subsequent lysosomal hydrolysis provide b1astocysts with accessto considerable pools of external maternal protein in <strong>the</strong> extra cellular fluid (ECF).This protein may be transported into <strong>the</strong> cells in <strong>the</strong> bulk fluid phase as well asadsorbed to invaginated plasma membrane. Using iodinated bovine serum albumin(BSA), we estimated that <strong>the</strong> blastocyst internalises fluid at a rate about equivalentto <strong>the</strong> whole blastocyst volume per hour (1). However, as albumin may adhere to<strong>the</strong> cell membrane (2), it may be endocytosed in <strong>the</strong> two phases mentioned aboveand so <strong>the</strong> measured rate of uptake may overestimate fluid transport. Toaccurately assess fluid phase endocytosis we used a non-adsorbing solute, 3Hdextran.MATERIALS AND METHODSBlastocysts were collected from superovulated mice by standard procedures andincubated under varying conditions with 3H-dextran (17.4 ~M, 18,5 GBq/LI 73 kD)in BMOC2 medium containing 1 g BSA/L. They were washed free of external 3Hdextranand radioactivity analysed.RESULTSAt 4°C in BMOC2 b1astocysts took up dextran at a rate equivalent to 0.9 ± 0,2pL/rnin/b1as~ when incubated at 3rC, <strong>the</strong> initial rate of uptake rose to 4.3 ±0,6pL/min/blast Insulin stimulated uptake at 3rC by 30% to 5.7 ±0.7 pL/min/b1ast lP


A TIMETABLE OF IN VIVO EMBRYO~ICDEVELOPMENT IN THE GREY SHORT-TAILED OPOSSUM(MONODELPHIS DOMESTICA)Karen E. Mate!, Edward, S.Robinson 2 & Roger A. Pedersen!•Gonadal sex reversal in female tammar wallabies is induced by MISD.l. Whitworth and M.B. RenfreeDepartment of Zoology, University of Melbourne, Parkville, Victoria 3052, Australia!Laboratory of Radiobiology & Environmental Health,University of Cali<strong>for</strong>nia San Francisco &2Department of Genetics, Southwest Foundation <strong>for</strong> Biomedical Research,San Antonio, TexasINTRODUCTIONThe grey short-tailed opossum (Monodelphisdomestica), has emerged in recent years as one of <strong>the</strong> majormarsupial models, and offers great potential <strong>for</strong> cellular andmolecular analyses ofmarsupial embryo development. Thetiming of early cleavage and some later stages ofMonodelphis domestica embryo development have beenexamined (1,2), however a comprehensive timetable ofembryonic development throughout gestation has not yetbeen established. The aim of this study was to establish <strong>the</strong>sebasic embryological parameters <strong>for</strong> Monodelphis.MATERIALS & METHODSThe timing of development was examined in a total of501 embryos from female Monodelphis domestica, collectedat known time intervals after video recorded matings. Whena pair mated more than once, <strong>the</strong> first mating was taken as<strong>the</strong> initiating event. A total of 60 pregnant females weretransported by air from San Antonio to San Francisco,usually<strong>the</strong> day after mating. An additional 23 animals werekilled and examined in San Antonio to collect early stages ofgestation and to ensure that transportation had not caused adelay in development. Embryos were recovered andexamined fresh, or in <strong>the</strong> case of blastocysts also afterfixation and sectioning.RESULTS & DISCUSSIONA total of 403 embryos (mean 8.8/female, range 1-17)as well as 47 degenerate embryos (mean 1.0/female, range 1­12) were recovered from 46 of<strong>the</strong> animals shipped to UCSF.The remaining animals contained only unfertilized eggs ordegenerate embryos. Preimplantation development inMonodelphis occupied nine of <strong>the</strong> 13.5 days of gestation.Most of <strong>the</strong> time during <strong>the</strong> preimplantation period wasoccupied by cleavage and <strong>the</strong> bilaminar blastocyst stage(Table 1). The interval from copulation to birth inMonodelphis was approximately 14.5 days.The developmental sequence and timetable ofMonodelphis is similar to that of <strong>the</strong> o<strong>the</strong>r well characterizeddidelphid, Didelphis virginiana (3), but with several notabledifferences: (i) transit of fertilized eggs through <strong>the</strong> oviductwas rapid (as quick as 6-12 h), which may contributing to <strong>the</strong>variable asymmetric mucoid covering of Monodelphisembryos; (ii) early cleavage stages were slower than inDidelphis, in particular <strong>the</strong> four-cell stage was variable andlasted up to 24 h; (iii) primary endoderm cells proliferatedfrom an already distinct embryonic area of <strong>the</strong> non-expandedunilaminar blastocyst.Table 1: A summary of development in <strong>the</strong> grey short-tailedopossum, Monodelphis domestica.Day AfterMatingDay 12Day 13Day 14No. oraEmbryosDay 1 96 (7)Day 3 46 (5)Day 4 32 (3)Day 5 30 (3)Day 6 30 (4)Day 7 9 (2)Day 8 14 (2)Day 9 36 (4)Day 10 40 (4)Day 11 34 (4)72 (7)14 (2)10 (2)a No of animals in paren<strong>the</strong>sesMcCrady Stage (m bold)1: Ovulation, fertilization, PN <strong>for</strong>mation2-5: 2-6 cell (mostly 4-cell)4,6: 4-8 cell (mostly 4-cell)8-9: 16-32 cell9-10: Complete unilaminar blastocyst11-14: Primary endoderm <strong>for</strong>mation;complete bilaminar blastocyst (335~)14: Bilaminar blastocyst (607llm)14: Bilaminar blastocyst (Imm) withobvious embryoblast18-22: Primitive streak & first somite14-26: Heart & blood vessel <strong>for</strong>mation,fusion of neural folds, cephalic flexure,amnion <strong>for</strong>mation, implantation30: Cervical flexure, paddle-like<strong>for</strong>elimbs, lateral amniotic folds <strong>for</strong>m32: Digital buds on <strong>for</strong>elimb; club-likehind limb, well developed allantois33-34: Claws on <strong>for</strong>elimb digits, digitalridges on hind limb, mouth/nostrils openDay 15 29 (3) 35: Elongated body, digital buds on hindlimb, birthREFERENCES(1) Baggott, L.M. & Moore, H.D.M. (1990)-J. Zool. Lond.222,623-639.(2) Harder, J.D., Stonerook, MJ. & Pondy, J. (1993) J. Exp.Zool. 266, 463-479.(3) McCrady, E. (1938) Am. Anat. Mem. 16, 14-226.IN1RODUCTIONJost's observations of <strong>the</strong> freemartin condition in cattleshowed that fetal ovaries could be trans<strong>for</strong>med into testislikestructures by hormones derived from <strong>the</strong> male co-twin(1). MIS could be <strong>the</strong> factor responsible, since testicularstructures develop in fetal rat ovaries exposed to MIS invitro (2). Similarly, fetal mouse ovaries transplanted under<strong>the</strong> skin of adult male hosts also develop seminiferoustubule-like structures (3)t and female mice transgenic <strong>for</strong>MIS show <strong>the</strong> transitory appearance of seminiferous tubules(4).This study was designed to determine if an XXundifferentiated gonad could be transfonned into a testis-likestructure in <strong>the</strong> tammar wallaby (Macropus eugenii) bytransplantation into older male pouch young. We alsocultured undifferentiated tammar ovaries with MIS todetennine ifthis could cause testicular development.MATERIALS AND :METHODSTransplantsOn <strong>the</strong> day of birth, gonads with or without mesonephrosattached were removed from male and female pouch young,and transplanted under <strong>the</strong> abdominal skin of 10 day oldmale and female recipients. Donor gonads from each sexwere transplanted into recipients of <strong>the</strong> opposite sex and <strong>the</strong>same sex. After surgery, recipients were reattached to <strong>the</strong>irmo<strong>the</strong>rs teat and returned to <strong>the</strong> pouch. After 25 days,recipients were killed and <strong>the</strong> transplants removed.Transplants were fixed in Bouin's fixative, paraffinembedded,sectioned at (7Jlm)t and stained withhaematoxylin and eosin be<strong>for</strong>e analysis with lightmicroscopy.Organ cultureGonads with or without mesonephros attached wereremoved from female pouch young on <strong>the</strong> day of birth andplaced onto individual polycarbonate membranes.Membranes were floated onto D:MEM/Ham's FI2 culturemedium containing L-glutamine, to which had been addedpenicillin/streptomycin (1 OOU/mVIOOJlg/ml), transferrin(5llg/ml), insulin (20Jlg/ml) and retinoic acid (50ng/ml).Recombinant human MIS (9Jlg/ml)1 was added to onegonad from each pair. The corresponding gonad from eachpair was cultured without MIS as a control. Gonads werecultured <strong>for</strong> 4 or 8 days be<strong>for</strong>e processing <strong>for</strong> routinehistology.RESULTSThe majority (11/15) of male gonads transplanted into malerecipients were ei<strong>the</strong>r resorbed by <strong>the</strong> hostt or uponhistological analysis were found to be dysgenic. However,of those which did survive, all continued to developmorphologically as normal testes, with Sertoli cells alignedinto seminiferous tubules surrounding <strong>the</strong> primordial germcells. A tunica albuginea and Leydig and peritubular myoidcells were also visible.Of <strong>the</strong> 2/8 female gonads transplanted into female recipientswhich survived, both developed as normal ovaries, with1We thank Dr. P. Donahoe and R. Ragin, PediatricSurgery, Massachusetts General Hospitalt <strong>for</strong> providing <strong>the</strong>rhMIS.mesenchymal cells delineating nests of mitotically-activegerm cells.Of 18 female gonads transplanted into male recipients, 4survived and all developed seminiferous tubule-likestructures which were devoid ofgenn cells (Fig. la).Figure 1: Transdifferentiated ovaries after transplantationinto male recipients (a)t or after culture with added MIS (b)showing <strong>the</strong> <strong>for</strong>mation of seminiferous tubule-likestructures. Note <strong>the</strong> lack of germ cells in (a).We did not find that <strong>the</strong> mesonephros had any effect ontransplant survival, or to <strong>the</strong> degree of development as atestis or an ovary.Of <strong>the</strong> 4/8 female gonads which developed seminiferoustubules in vitro, 3 were cultured with <strong>the</strong>ir mesonephrosattached. These gonads showed seminiferous tubules<strong>for</strong>ming around clusters of germ cells, in a patterncharacteristic of male control gonads (Fig. Ib). The singlegonad cultured without attached mesonephros was muchmore masculinised and <strong>the</strong> well-developed tubules weredevoid ofgerm cells. One ovary remained undifferentiatetd..All three gonads which were cultured <strong>for</strong> 8d becamedysgenic, suggesting that <strong>the</strong> medium cannot support <strong>the</strong>tissue <strong>for</strong> this length of time.CONCLUSIONSMIS can induce <strong>the</strong> masculinisation of neonatal tammarovaries in vitro, and could be <strong>the</strong> factor responsible <strong>for</strong> <strong>the</strong>transdifferentiation of ovaries transplanted into male pouchyoung. Thus, MIS may playa role in normal testiculardifferentiation.REFERENCES(1) Jost, A., Vigier, B. and Prepin, J. (1972) J. Reprod.Fert. 29, 349-379.(2) Vigier, B., Watrin, F., Magre, S., Tran, D. and Josso,N. (1987) Development 100, 43-55.(3) Taketo, T., Merchant-Larios, H. and Koide, S.S.(1984) Proc. Soc. expo Biol. Med. 176, 148-153.(4) Behringer, R., Cate, R.L., Froelick, GJ., Palmiter,R.D. and Brinster, R.L. (1990) Nature 345,167-170.99 100


Thyroid gland development in <strong>the</strong> brushtail.possum,Trichosurus vulpecula.W. Buaboocha and R. T. GemmellDepartment of Anatomical Sciences, University of Queensland•EARLY PREGNANCY FACfOR (EPF): PURIFICATION TO HOMOGENEITY FROMHUMAN PLATELETS AND IDENTIFICATION AS CHAPERONIN 10.Alice C Cavanagh!, Peter L KayrJ and Halle Morton!.IDept of Surgery, 2Dept of Physiology and Pharmacology, University of Queensland, Brisbane.INTRODUCTIONThe initiation of thyroid gland function usually occursprenatally or at birth in eu<strong>the</strong>rian mammals. Inmarsupials. this endocrine gland is nOt present at birth.The gland becomes functional during <strong>the</strong> latter half ofpouch life. In this smdy <strong>the</strong> ultrastrUcwre ·of <strong>the</strong> thyroidgland will be correlated with <strong>the</strong> plasma concenrrationsof both thyroxine (TJ and rri-iodo thyronine (T 3 ) in <strong>the</strong>developing brushrail possum to determine whe<strong>the</strong>r <strong>the</strong>increase in thyroid hormones in mid-lactation areproduced by me young or are passed across in <strong>the</strong> milkfrom me mo<strong>the</strong>r.~fATERIAL AND METHODSThyroid tissue from seven possums aged 40. 65. 96.113. 115. 129 days POSt partupl and an adult wereexamined \vith me light and <strong>the</strong> electron microscope.The body weights and <strong>the</strong> weights of excised thyroidglands from <strong>the</strong> seven animals were also recorded.Blood samples were obtained weekly via hean puncmrefrom pouch young and <strong>the</strong> plasma concenrrations ofboth T 4 and T:; were determined by radioimmunoassayusing ICN Diagnostics RIA kits (ICN Biomedicals. Inc..Diagnostics Division. Costa Mesa. USA).RESULTS:The weight of thyroid tissue in <strong>the</strong> possums increasedwith age and body weight (Table 1). Lysosomesrequired <strong>for</strong> <strong>the</strong> conversion of thyroglobulin to thyroxinewere first observed within <strong>the</strong> thyroid follicular cells of<strong>the</strong> 96 days old possum. The plasma thyroid hormonesconcentrations varied in <strong>the</strong> young possum during <strong>the</strong>firSt 150 days after birth. Plasma concentration of T..rose from less than 10 ng/ml prior to day 60~ to 12ngiml on day 90 and [0 a peak of 45 ngiml on day 120post panum. Thereafter. <strong>the</strong> values tended to diminishco below 10 ng/ml by day 160 post parwm (Figs. 1 and2). Plasma T;; concenrrations were similar in profilewith a peak concentration of 0.25 ng/ml beingsdetected on day 120 post panum.CONCLUSION:The plasma concenrrations of T.. and T 3 in <strong>the</strong> youngpossum do not correlate with <strong>the</strong> morphologicalevidence <strong>for</strong> <strong>the</strong> initiation of activity of <strong>the</strong> thyroidgland of <strong>the</strong> young. The thyroid hormones in <strong>the</strong>plasma of <strong>the</strong> young, prior to day 96, are presumablyproduced in <strong>the</strong> mo<strong>the</strong>r and are transpOrted to <strong>the</strong> youngvia <strong>the</strong> milk (1). The activation of <strong>the</strong> thyroid glandcoincides with <strong>the</strong> reported increase in growth rate of<strong>the</strong> young possum on day 96 (2).Table 1. Weights of thyroid glands and body weightsof possums.Age Thyroiddays gm (paired)40 0.001465 0.003596 0.0086113 0.0184115 0.0314129 0.0400adult 0.3520EOic504030lDCX0 20~10EOic:00so r40I30 ~lD.S x I>.I-=10 L II0 20 r250 1 0 25Bodygm12.4124.0170.0087.0080.00275.002249.007575,e.,... ~,. .~.% thyroid weightto body weight0.01130.01460.01230.02110.03930.01450.0157" ..,...:.' .II ~I.... I. .... . ... I. e ........:- .I. I. I • .'I • • a ~ •••• I, •5050100 125 150 175 200days post partum100 125 150 175 200days post partum.....Fig. 1. The plasma concemration of thyroxine (T 4) in<strong>the</strong> developing brushtail possum.Fig. 2. The weekly plasma concentration of thyroxine(T 4) in two developing possums.REFERENCES1. Gemmell. R.T.. and Sernia. C. (1992) CompoBiochem. Physiol. 103A:541-543.2. Gemmell. R.T.. and Hendrikz. J.K..(1993) Aust. J.Zoo1. 41: 141-149.101INTRODUCTIONEarly pregnancy factor (EPF), detectable in maternal serumwithin 24 h of fertilisation in all mammalian species tested. isindicative of and necessary <strong>for</strong> embryonic well-being. EPF hasimmunosuppressive properties and is important also as agrowth regulator of transfonned, neoplastic and nonnal cells;its presence in platelets implies a role in inflammation andwound healing. A molecule possessing this exceptionalcombination of properties has great biological significance but,until now, <strong>the</strong> strocture of <strong>the</strong> biologically active molecule hasremained elusive.MATERIALS AND METHODSAll major classes of source materials with activity in <strong>the</strong> EPFbioassay, <strong>the</strong> rosette inhibition test. including pregnancy serum(1.2), were fractionated by a 6-step scheme including reversedphaseHPLC(1). Each purified active protein was treated withan immobilised anti-EPF monoclonal antibody: activity ofbound and unbound fractions was tested (l). Platelet-derivedEPF was analysed by SDS-PAGE (15% Tris-Tricine gels),electrospray mass spectrophotometry and amino acidsequencing of both <strong>the</strong> whole molecule and proteolyticfragments (l).RESULTSFor all source materials. activity followed an identical patternthroughout fractionation and each purified active protein wasspecifically recognised by a monoclonal antibody to EPF (datanot shown)(l). These biochemical and immunologicalsimilarities indicate that we are dealing with a single substanceor closely related family of substances, acting in diversebiological situations.Only human platelets provided enough active protein <strong>for</strong>stroctural studies (45 pg/300 units - 150 litre blood). On SDS­PAGE (Fig Ia), EPF migrated as a single band coincident withbio-activity; thus <strong>the</strong> protein, not a small associate, is active.Even more compelling evidence of homogeneity was providedby mass spectrometry, which also detennined an accuratemolecular size of 10843 (Fig 1b). The amino acid sequence of-70% of <strong>the</strong> molecule was detennined. This was identical torat liver mitochondrial chaperonin 10 (cpnlO). except <strong>for</strong> asingle residue (Fig Ic). Fur<strong>the</strong>r experiments using EPF. cpnlOand its functional associate cpn60 demonstrated that thisstroctural relationship extends to functional identity. Briefly,rat cpnlO was active in <strong>the</strong> EPF bioassay and this activitycould be neutralised by a monoclonal anti-EPFantibody. Inaddition, platelet-derived" EPF fonned an ATP dependent stablecomplex with cpn60 while imrnobilised cpn60. with ATP,could remove all EPF-activity from pregnancy serum; activitycould be recovered by removing ATP from <strong>the</strong> complex (datanot shown)(l). This requirement <strong>for</strong> ATP demonstrates <strong>the</strong>specificity of <strong>the</strong> interaction between cpn60 and <strong>the</strong> activemoiety and illustrates that, in pregnancy serum as in platelets.cpnlO is <strong>the</strong> molecular entity initiating response in <strong>the</strong> EPFbioassay.CONCLUSIONOur studies identify EPF as an extracellular fonn of cpnlO, amember of <strong>the</strong> highly conserved heat-shock family ofmolecules, which functions as a molecular chaperone. This is<strong>the</strong> first example of such a molecule perfonning anextracellular role. We are now investigating extracellular targetmolecules to determine whe<strong>the</strong>r EPF influences o<strong>the</strong>rregulatory molecules (as might be expected <strong>for</strong> a molecularchaperone) or whe<strong>the</strong>r it exerts its effects more directly.C102BIHuman EPl' 1AIIo o \---14.,( IIIJto 15SlIc.c noBOO "00 ICOO 1100 1200 0(10 1400mIlIC.


CP·PLATELET ACTIVATING FACTOR STIMULATES CELL-CYCLE PROGRESSION IN 2-CELL MURINE EMBRYOSC.O'NeillHuman Reproduction Unit, Royal North Shore Hospital, St. Leonards, N.S.W. 2065•IN VITROAMPLIFICATION OF AN OVINE OESTRUS-ASSOCIATED OVIDUCTALGLYCOPROTEIN eDNAINTRODUCTIONWe have previously demonstrated that culture of embryos invitro compromised <strong>the</strong> release of embryo-derived plateletactivatingfactor (PAF) (1), while supplementation of mediawith syn<strong>the</strong>tic PAF resulted in increased metabolism ofcarbohydrate substrates and increased implantation, pregnancyand birth rates in humans and mice following embryo transfer(2,3). It was also demonstrated that 8-cell embryos andblastocysts had a greater mitotic index when treated with PAFthan did controls (4). This latter observation could possibly beconfounded by <strong>the</strong> substantial asynchrony of <strong>the</strong> cell-cycle in<strong>the</strong> later stage preimplantation embryos. In this study weexamined <strong>the</strong> effect of embryo density and PAFsupplementation ofmedia on <strong>the</strong> rate of cell-cycle progressionof2-cell mouse embryos which were synchronised by exposureto a reversible mitotic spindle poison at <strong>the</strong> I-cell stage.MATERIALS AND METIIODSQs female mice were superovulated and mated with fertilemales. 30h after hCG zygotes were collected from <strong>the</strong>reproductive tract. Zygotes were cultured in modHTFM(contains glutamine and EDTA)+3mg BSA/ml and observed attwo-hourly intervals to determine <strong>the</strong> kinetics of <strong>the</strong> first celldivision.Since prolonged culture in nocodazole is detrimentalto development, a 4 hour period was chosen to capture <strong>the</strong>maximum number of I-cells in <strong>the</strong> shortest period of time(Figure 1). In all experiments synchronisation was achieved byculturing zygotes <strong>for</strong> several hours, followed by transfer to 5~M nocodazole in modHTFM <strong>for</strong> 4 h. Upon removal fromnocodazole <strong>the</strong>y were washed in 5 changes of HepesHTFM toremove all Nocodazole and <strong>the</strong>n placed in modHTFM <strong>for</strong> 90min. At <strong>the</strong> end of this time all embryos which had divided to2-cell embryos were collected (synchronised 2-cells). Culturewas in 1O1l1 drops of modHTFMlBSA under paraffm oil andsupplemented with 1.86~M PAF/ml or vehicle (Control).Embryos were allocated at random at a density of 1 or 10/drop.After 20h culture embryos were observed at regular intervalsand <strong>the</strong> proportion progressing to <strong>the</strong> 4-cell stage recorded. Allexperiments were repeated at least 3 times.RESULTSFollowing synchronisation <strong>the</strong> first 4-cell embryos wererecorded at 24 hours. When embryos were cultured as groupsof 10 all embryos capable ofcleaving had done so by 38 hourswith 50% cleaving by 29h. When embryos were culturedindividually, cell-cycle progression was delayed, with <strong>the</strong> 50%progression occurring at 33h (4 hour delay, P < 0.001, logisticregression analysis). The addition of PAF to medium had nosignificant effect on <strong>the</strong> development of embryos in groups of10, but significantly shortened by 2.2h (p


... »THE ROLE OF AN ESTROGEN,DEPENDENT GLYCOPROTEIN, OVIDUCTIN, INFERTILIZATION IN THE GOLDEN HAMSTER D.E. Boatman and G.E. Magnoni,Dept. ofAnimal Health & Biomedical Sciences, University ofWisconsin, Madison, U.S.A.INTRODUCTION• Fertilizationin vivo is more efficient compared tothat in vitro: fewer sperm per egg are requiredand sperm interact <strong>for</strong> a shorter time with <strong>the</strong>zona pellucida prior to penetration.• Oviductal ampullary molecules may increase invivo fertilization efficiency by interacting withsperm or eggs or both to [1]:a. alter binding affinities ofcapacitated sperm<strong>for</strong> <strong>the</strong> zona pellucida.b. increase efficiency ofacrosome reactions(ARs) ofsperm on <strong>the</strong> zona pellucida.c. alter biophysical properties ofzonaepellucidae.• We now present evidence that oviductin (OVN), amammalian member of<strong>the</strong> chitinase family [2],is an oviductal fertilization factor that binds tosperm as well as to eggs and embryos [3].MATERIALS AND METHODS• Isolate OVN from oviducts ofPMSG primedhamsters using Helix pomatia lectin affinitychromatography (no. ofisolations=4) [3,4].• Bioassay OVN fractions <strong>for</strong> follicular eggpenetration enhancing activity (1 h pulse withpre-eapacitated sperm) [4].• Add ± OVN to sperm at two capacitation times.a. capacitate cauda epididymal sperm 0 and 5.5h in TALP (BSA= 3mg/rnl, 37 0 C, 5% CO 2 ),b. add ± OVN to sperm (1.6 X 106/rnl) 30 min.c. layer sperm suspensions onto 2-step Ficollgradients; centrifuge to wash and fix (1 %para<strong>for</strong>maldehyde on bottom).• Assess ARs by phase and bright field microscopy.• Assess binding ofOVN to sperm (immunolocalizationusing anti-OVN MAb, I C4, andgoat anti-mouse IgG FITC conjugate).RESULTS• Follicular egg penetration: OVN fractions,increased penetration 2.9 fold (OVN= 49±2.7 %,vs. BSA alone= 17±4.0) [4].• ARs: ARs increased between 0.5 and 6 h, butOVN (effective in increasing egg penetration) didnot increase ARs at ei<strong>the</strong>r time (Fig. I).• OVN binding to AR intact sperm: at 0.5 h OVNbound to <strong>the</strong> acrosomal crescent (82.5%labelled); at 6 h OVN bound over <strong>the</strong> equatorialand post-acrosomal regions of<strong>the</strong> heads (60.4%labelled) (Fig. 2).105CONCLUSIONS• We conclude that OVN increases penetration andfertilization by increasing sperm binding affinity<strong>for</strong> eggs, because:a. OVN i penetration offollicular eggs [4].b. as a result ofcapacitation, binding ofOVN tosperm shifts regionally and temporally from<strong>the</strong> acrosomal crescent to <strong>the</strong> equatorialsegment.c. sperm binding affinity to oviductal comparedto follicular eggs is increased [1].d. ARs are not stimulated by OVN.Fig. 1 Effect of OVN Added at TwoCapacitation Times on Acrosome ReactionsUJa:


»THE ROLE OF AN ESTROGEN.DEPENDENT GLYCOPROTEIN, OVIDUCTIN, INFERTILIZATION IN THE GOLDEN HAMSTER D.E. Boatman and G.E. Magnoni,Dept. ofAnimal Health & Biomedical Sciences, University ofWisconsin, Madison, U.S.A.INTRODUCTION• Fertilization in vivo is more efficient compared tothat in vitro: fewer sperm per egg are requiredand sperm interact <strong>for</strong> a shorter time with <strong>the</strong>zona pellucida prior to penetration.• Oviductal ampullary molecules may increase invivo fertilization efficiency by interacting withsperm or eggs or both to [1]:a. alter binding affinities ofcapacitated sperm<strong>for</strong> <strong>the</strong> zona pellucida.b. increase efficiency ofacrosome reactions(ARs) ofsperm on <strong>the</strong> zona pellucida.c. alter biophysical properties ofzonaepellucidae.• We now present evidence that oviductin (OVN), amammalian member of<strong>the</strong> chitinase family [2],is an oviductal fertilization factor that binds tosperm as well as to eggs and embryos [3].MATERIALS AND METHODS• Isolate OVN from oviducts ofPMSG primedhamsters using Helix pomatia lectin affInitychromatography (no. ofisolations=4) [3,4].• Bioassay OVN fractions <strong>for</strong> follicular eggpenetration enhancing activity (1 h pulse withpre-eapacitated sperm) [4].• Add ± OVN to sperm at two capacitation times.a. capacitate cauda epididymal sperm 0 and 5.5hinTALP (BSA= 3mg/ml, 37 0 C, 5% CO 2 ),b. add ± OVN to sperm (1.6 X 106/ml) 30 min.c. layer sperm suspensions onto 2-step Ficollgradients; centrifuge to wash and fix (1 %para<strong>for</strong>maldehyde on bottom).• Assess ARs by phase and bright field microscopy.• Assess binding ofOVN to sperm (immunolocalizationusing anti-OVN MAb, I C4, andgoat anti-mouse IgG FITC conjugate).RESULTS• Follicular egg penetration: OVN fractions,increased penetration 2.9 fold (OVN= 49±2.7 %,vs. BSA alone= 17±4.0) [4].• ARs: ARs increased between 0.5 and 6 h, butOVN (effective in increasing egg penetration) didnot increase ARs at ei<strong>the</strong>r time (Fig. I).• OVN binding to AR intact sperm: at 0.5 h OVNbound to <strong>the</strong> acrosomal crescent (82.5%labelled); at 6 h OVN bound over <strong>the</strong> equatorialand post-acrosomal regions of<strong>the</strong> heads (60.4%labelled) (Fig. 2).105CONCLUSIONS• We conclude that OVN increases penetration andfertilization by increasing sperm binding affinity<strong>for</strong> eggs, because:a. OVN i penetration offollicular eggs [4].b. as a result ofcapacitation, binding ofOVN tosperm shifts regionally and temporally from<strong>the</strong> acrosomal crescent to <strong>the</strong> equatorialsegment.c. sperm binding affinity to oviductal comparedto follicular eggs is increased [1].d. ARs are not stimulated by OVN.Fig. 1 Effect of OVN Added at TwoCapacitation Times on Acrosome ReactionsCI.Ia:«-c:a>o L-a>ll..50. r-------------,40302010oco 5.5Time of Oviductin Addition (hr)OVN:o minuso plusFig. 2 Temporal and Regional Shift inOVN Binding to Acrosome-Intact Sperm• OhD 5.5h!EmREFERENCES5.5 h1) Boatman DE et al. (1994) Molec Reprod Dev(in press).2) Hakala BE et al. (1993) J BioI Chern 264:25803.3) Kan FWK et al. (1990) Anat Rec 226:37-47.4) Boatman DE & Magnoni GE. (1994) BioI Reprod51(SuppI1): 129 (abs 297).Proteinases important in implantation are down-regulatedin par<strong>the</strong>nogenetic mouse blastocystsMark B. Haroe:i, Mayi Arcellana-Panlilio 2 , XingQi Zhang 3 , Dylan R. Edwards 4 , and Gilbert A. Schultz 2School of Life Science, Queensland University of Technology, Brisbane, Qld 1 , Deparbnents of Medical Biochemisn-y-2,Obstetrics & Gynaecologf and Pharmacology & Therapeutics 4 , University of Calgary, Calgary, Alberta, CanadaINTRODUCTIONMuch of <strong>the</strong> embryo's activity in <strong>the</strong> implantation process comesfrom <strong>the</strong> trophectodermal component of <strong>the</strong> blastocyst in whic<strong>hth</strong>e uterine tissue is invaded by trophoblast cells. Human andmouse trophoblasts produce two types of proteinases, gelatinaseB (MMP-9) and urokinase-type plasminogen activator (uPA), thathave been implicated in <strong>the</strong> process of implantation becauseinvasion is blocked by neutralizing antibodies or inhibitors.Par<strong>the</strong>nogenetic mouse embryos cleave and develop to <strong>the</strong>blastocyst stage but show implantation defects. (Pedersen et al.,1993). Because of this/ we examined par<strong>the</strong>nogenetic blastocysts, and blastocyst outgrowths in comparison to normal fertilizedembryos in terms of <strong>the</strong>ir ability to express <strong>the</strong> genes and geneproducts<strong>for</strong> uPA and MMP-9.MATERIALS AND METHODSPar<strong>the</strong>nogenetic embryos obtained by ethanol activation of CD1mouse oocytes were cultured to <strong>the</strong> blastocyst stage andsubsequently transferred to 50pl culture droplets pre-treated withfibronectin (50-100 embryos/droplet). After 48 hr culture in TCM199 plus Img/ml, <strong>the</strong> medium was changed and embryos thathad not attached discarded. After a fur<strong>the</strong>r 24 hr culture (day 8 ofdevelopment), <strong>the</strong> conditioned media droplets from normal andpar<strong>the</strong>nogenetic embryos were collected and assayed. <strong>for</strong> (uPA)activity by a chromogenic assay and (MMP) activity byzymography. To investigate <strong>the</strong> expression of <strong>the</strong> genes encoding<strong>the</strong>se proteinases in embryos, RNA was extracted frompar<strong>the</strong>nogenetic and normal blastocyst outgrowths on day 8 ofdevelopment and subjected to RT-PCR analysis <strong>for</strong> detection ofuPA and MMP-9 transcripts.RESULTSAfter 24 hr culture of blastocysts (day 6 of development), 11±8%of par<strong>the</strong>nogentic blastocysts had attached compared to 18±3% <strong>for</strong>normal counterparts (Fig 1). After ano<strong>the</strong>r 24 hr culture (day 7)only half <strong>the</strong> number of par<strong>the</strong>nogenotes had attached comparedto normal blastocysts (37±14% vs 77±2%, P


wEarly pregnancy factor Is reqJ,Jired at implantation to optimize embryonic development.Athanasas-Platsis, S/ Kaye PL} Cavanagh, A.C./ and Morton H. iThe University of Queensland, 1Dept of Surgery, Clinical Sciences Building, RBH 4029; 2Dept ofPhysiology and Phannacology, Brisbane 4072, Qld.INTRODUCTIONEarly pregnancy factor (EPF) has a role in earlydevelopment of <strong>the</strong> embryo. Passive immunization ofmated mice with anti-EPF antibodies (Abs) on d 1 and 2post coitum (pc,d l=day of mating) demonstrated a failureto maintain pregnancy, when examined at d 7 pc (1);embryos flushed from treated mice on d 3 pc showed asignificant retardation at <strong>the</strong> 1-2 cell stage of development(2). This effect apparent in vivo was indirect, asdevelopment proceeded uninterrupted to <strong>the</strong> blastocyst stagewhen embryos were cultured in vitro in <strong>the</strong> presence of anti­EPF Abs (2). The aim of <strong>the</strong> study presented here was toextend <strong>the</strong>se findings, to determine if, as well as beingrequired <strong>for</strong> preimplantation development, EPF is alsorequired by <strong>the</strong> embryo <strong>for</strong> <strong>the</strong> processes of trophoblastoutgrowth and implantation.MATERIALS AND METHODSConfirmed-mated Quackenbush mice were passivelyimmunized with rabbit anti-EPF Abs #816 (1,2) on d 3 and4 pc (500 JIg IgG x 2 daily); control groups received normalrabbit IgG or 0.9% NaCI (saline). On d 7, implantationsites and corpora lutea were counted. Oviducts and uterinehorns were flushed and development of unimplantedembryos recorded. In in vitro experiments, morulae werecollected from untreated mice on d 3 pc, washed andcultured under oil in Eagle's medium containing 10% foetalcalf serum and anti-EPF monoclonal antibodies (rnAbs;l) orcontrol rnAbs 1Dy (see Table 1) After 168 h., <strong>the</strong>development of trophoblast outgrowths was recorded. In <strong>the</strong>lowest concentration of anti-EPF mAbs tested (2.5 pg/ml),in which % blastocysts which outgrew was not significantlydecreased, <strong>the</strong> surface area of outgrowths was measured.RESULTSFollowing passive immunization on d 3 and 4 pc, only 30%embryos (ie embryos as a % of corpora lutea) had implantedby d 7 compared with 90% and 100% in <strong>the</strong> control Absand saline groups respectively (anti-EPF Abs v. control Abs,p < 0.001, t test, Fig 1). Unimplanted embryos in <strong>the</strong>control Abs group (10%) were hatched whereas unimplantedembryos in <strong>the</strong> anti-EPF group (25%) were not; 45%embryos in <strong>the</strong> latter group were not accounted <strong>for</strong>.Examination of embryos after 24-48 hours in culture withanti-EPF mAbs 5/341 revealed a progression of <strong>the</strong> majorityof morulae to <strong>the</strong> blastocyst stage. However developmentwas interrupted after this time. Culture of embryos <strong>for</strong> 168h with 25 or 250 pg/ml anti-EPF rnAbs significantly~ffected <strong>the</strong> development of blastocysts to <strong>the</strong> outgrowthstage when compared to control mAbs IDy (Table 1). Of<strong>the</strong> affected embryos, 29 or 34% were dead after treatmentwith 25 or 250 pg/ml anti-EPF mAbs respectively comparedwith only 0 or 13% after treatment with 25 or 250 pglmlcontrol mAbs IDy respectively (p < 0.001, '1.. 2 test). Cultureof embryos with 2.5 pglml anti-EPF rnAbs did not affectdevelopment to <strong>the</strong> outgrowth stage (Table 1) but did inhibit<strong>the</strong> surface area of <strong>the</strong>se outgrowths. Average area ofoutgrowths (x 10- 3 mm 2 ± sem) with 2.5 pglml anti-EPFmAbs was 64 ± 11.8 (n = 13) compared with 110 ± 8.1 (n= 54) and 108 ± 16.7 (n = 15) in <strong>the</strong> control medium and2.5pglml control mAbs IDy respectively (p < 0.05,Student's t test).CONCLUSIONThe observation that development of morulae to <strong>the</strong>outgrowth stage was inhibited when cultured with anti­EPF mAbs toge<strong>the</strong>r with <strong>the</strong> fact that administration ofanti-EPF to pregnant mice on days 3 and 4 disruptedimplantation, suggests a role <strong>for</strong> EPF as a growthfactor. The inhibition of trophoblast outgrowth or, in ahigh proportion of cases, death of <strong>the</strong> blastocystcoincides with embryonic production of EPF,suggesting that it is autocrine growth factor at thisstage of development.10080~ - a-EPF IgG§33 - control rabbll IgGITIJ - 0.9% NaCI:58 CELLCOMPACTED BLASTOCYSTS IMPLANTEDMORULAEEMBRYOSFig 1: Stage of development of embryos on d 7 pcafter passive immunization of mated mice, at 56, 61,80 and 85 h pc with 500 pg rabbit anti-EPF Abs #816, control Abs or 0.9% NaCl/ injection. Anti-EPFAbs v control Abs, p < 0.001 '1.. 2 test).Table 1: Effect of anti-EPF mAbs 5/341 ontrophoblast outgrowth in vitro.rnAbs (pg/ml)" No of No ofembryos outgrowths %b p*Control medium 72 54 (75)Control rnAbs 1Dy2.5 17 15 (88)25 23 22 (96)250 30 23 (77)Anti-EPF rnAbs 5/3412.5 18 13 (72) NS25 28 15 (53)


.. •THE IDENTIFICATION OF EARLY PREGNANCY FACTOR IN RED DEER (CERVUS ELAPHUS)Gendie Lash and Mike LeggeDepartment of Biochemistry, University of Otago, PO Box 56, Dunedin, New Zealand.INTRODUCTIONEarly pregnancy factor (EPF) is one of <strong>the</strong> firstbiochemical indicators of pregnancy. It is producedby <strong>the</strong> mo<strong>the</strong>r in response to a signal from <strong>the</strong>zygote on fertilization. The aim of this study was todetermine whe<strong>the</strong>r farmed red deer (Ce rvuselaphus) syn<strong>the</strong>sised EPF.MATERIALS AND :METHODSFarmed red deer hinds (n=28) from an ovulationstudy (1) were bled 13 times at 3-4 day intervalsfrom 11 to 15 days after cloprostenol administration(ie 24 to 26 days after <strong>the</strong> initial oestrussynchronization) via jugular venepuncture. Theblood was allowed to clot, serum removed andfrozen at -20°C until required <strong>for</strong> analysis.Pregnancy was diagnosed by rectal ultrasonography25 to 31 and 45 to 51 days after cloprostenoltreatment. The date of calving was determined bydaily observation, and <strong>the</strong> date of conceptioncalculated on a gestation period of 233 days (2).EPF was assayed <strong>for</strong> using <strong>the</strong> rosette inhibition testaccording to Rolfe et al. (1984)(3).RESULTSFour major results came out of this study.1. All hinds which calved (n=20) gave an EPFresponse (Figure 1), dilution range of 10- 3 to 10- 5 .2. One hind of <strong>the</strong> <strong>twenty</strong> which calved had twins,and had a corresponding change in <strong>the</strong> EPF doseresponse curve, dilution range of 10- 5 to 10- 7 (Figure2). Two o<strong>the</strong>r hinds who had singleton births werealso identified as twin pregnancies at around day 25pc but gave normal EPF activity after day 30 pc.r.:Ia:Eo-E:z0 20E:9:c~ 10r.:II: r.:I~0 a:10-1 10-2 10-3 ID-4 10-5 10-6SAMPLE DILUTION• day 9pcFigure 1. Changes in dose response curve duringblastocyst <strong>for</strong>mation in red deer at approximatelyday 8pc.3. Of <strong>the</strong> eight hinds which did not calve three wereconsidered to have had a fertilization event asobserved by a positive EPF result in <strong>the</strong> first serumsample. No subsequent samples were positive.4. As is seen in Figure 1 <strong>the</strong> EPF dose responsecurve changed around day 9 pc. This correspondswith blastocyst <strong>for</strong>mation (4). No fur<strong>the</strong>r changeswere observed over a time course up to day 45 to 46pc. The twin pregnancies did not change until day15 pc (10- 3 to 10- 4 day 15pc).3010-1 10-2 10-3 ID-4 10-5 10-6 10-7 10-8SAMPLE DILUTION• Non-pregnant hindsII Single pregnancyra Multiple pregnancyFigure 2. Changes in <strong>the</strong> dose response curvebetween nonpregnant hinds and those with single ormultiple pregnancies.DISCUSSIONThis study has shown that EPF is detectable in reddeer and that EPF can be used to distinguishbetween a twin and a singleton pregnancy. It hasalso been shown that a change in <strong>the</strong> EPF doseresponse curve corresponds with blastocyst<strong>for</strong>mation, where <strong>the</strong>re is ei<strong>the</strong>r a down regulationof <strong>the</strong> inhibitor or <strong>the</strong> embryo itself starts to produceEPF, and not with implantation at day 40 pc (5). Itwas surprising to find <strong>the</strong> change in <strong>the</strong> twinpregnancies was much later than <strong>the</strong> singletons.This result can not be commented on at present asso little is currently known ab.out deer earlyembryology.REFERENCES(1) Fisher M W, Asher G W, Meikle LM andNewman R E (1994) Animal Reproduction ScienceIn Press(2) Guinness F, Lincoln G A and Short R V(1971) Journal of Reproduction and Fertility 27427-438(3) Rolfe B, Cavanagh A, Forde C, Bastin F, ChenC and Morton H (1984) Journal ofImmunologicalMethods 70 1-11(4) Berg D (1993) Proceedings of<strong>the</strong> sixth annualgeneral meeting of<strong>the</strong> Australasian Association ofAnimal Arificial Breeders (Abstract) 22-25(5) McMahon C D (1989) BSc (Hons) Thesis,University of Otago, Dunedin, New Zealand.COMPARISON OF CERVICAL, TRANSCERVICAL AND LAPAROSCOPIC INSEMINATION OFEWES WITH CHILLED STORED RAM SEMEN.IF. Smith, J. Parr, S. Beaumont, 1. Oliver and G.C. UpretiAgResearch, Ruakura Agricultural Research Centre, P B 3123, Hamilton, N. Z.INTRODUCTIONA technique <strong>for</strong> transcervical artificial insemination (TAI) of<strong>the</strong> ewe has been reported as an alternative to laparoscopicintrauterine insemination (LAl) (1). This study evaluated <strong>the</strong>techniques under New Zealand conditions.METHODRomney ewes (365) were treated in February <strong>for</strong> 14 dayswith CIDR® devices and inseminated (50-56 h) after deviceremoval. Semen was diluted in a milk based diluent toconcentrations of 800xl0 6 /ml <strong>for</strong> cervical insemination (CAl)(0.2 ml) and 200xl0 6 /ml <strong>for</strong> TAl and LAl (0.25 ml straws)and cooled to 15°C until insemination ei<strong>the</strong>r 12 - 18 hr CD0) or 36 - 42 hr CD 1) after collection. Frozen semen(100x10 6 sperm; 50% motile, in 0.25 ml straws) from <strong>the</strong>same rams was used via <strong>the</strong> TAl and LAl methods. Allinseminations were on a time basis about 52h after CIDRremoval. Oestrous status of ewes at Al and <strong>the</strong> depth ofTA! were recorded. Pregnancy was determined byultrasonic scanning 50 days post insemination. Twoadditional trials were conducted in Nov-Dec (214 ewes) andMarch (115 ewes) in which TAl (D 0 semen) and LAl werecompared.RESULTSThe pregnancy rates <strong>for</strong> <strong>the</strong> main trial are shown in Figure1. LA! was superior to TAl with all semen types (P


•IN VITRO AND IN VIVO FERTILITY OF LIQUID-STORED GOAT SPERMATOZOAJ Epplestorfl, C. C. Pomares'J, T. Stojanovb and W.M C. MaxwellbaDep~rtment ofWool and Animal Science, Uruversity ofNSW, Sydney, 2052.bDepartment ofAnimal Science, University ofSydney, NSW, 2006.INTRODUCTIONThere are several reports on <strong>the</strong> liquid storage of ramspermatozoa and its fertility after AI (1) but <strong>the</strong>re islittle in<strong>for</strong>mation available on <strong>the</strong> fertility of liquidstoredgoat spermatozoa. The use of in-vitrofertilisation (IVF), if reliable, would simplify fertilityassessment and facilitate improvements in semenstorage procedures. This study reports both <strong>the</strong> in vitroand in vivo fertility of goat spermatozoa liquid storedin a Tris-based diluent at 5 0 C.MATERIALS AND :METHODSSemen collected by artificial vagina from 4 bucks waspooled and diluted 5;.fold with a Tris diluent (300 mMTris, 27.75 mM fructose, 94.7 mM citric acid, 2% eggyolk)previously bubbled with nitrogen (N). Thediluted semen was stored at 5 0 c under N in cappedtest tubes.The fertility of goat spermatozoa stored <strong>for</strong> 0, 2, 4 or 8days was determined by both IVF and after AI. Thesemen used <strong>for</strong> <strong>the</strong> in vitro and in vivo fertilityassessments were from different batches but werecollected from <strong>the</strong> same bucks and were treatedidentically. The fresh semen (Day 0) was treatedidentically to <strong>the</strong> stored semen except that it was notcooled.For IVF, sheep oocytes collected from an abattoir andmatured in TCM-I99 <strong>for</strong> 22-27 hr, were cultured infertilisation media (bicarbonate-buffered SOF + 20%sheep serum) with Percoll treated buck spermatozoa ata concentration of Ix10 6 /ml <strong>for</strong> 18 hr. Ova wereclassified as fertilised if, after fixing and staining (1 %aceto-orcein), <strong>the</strong>y contained 2 pronuclei and a spermtail.For AI, adult Cashmere does were treated in Marchwith progestogen sponges (Chronogest 45 mg,Intervet) <strong>for</strong> 19 days and with 200 iu PMSG(pregnecol, Horizon) given 24 hours be<strong>for</strong>e spongeremoval. The does were inseminated 50-58 hr aftersponge removal by ei<strong>the</strong>r <strong>the</strong> cervical or laparoscopicmethod using 150 or 45 x 10 6 sperm, respectively. Theproportion of does pregnant was determined byultrasound 50 days later.RESULTSFertility was maintained when semen stored <strong>for</strong> up to4 days was deposited into <strong>the</strong> uterus, but declined after·only 2 days storage when cervical insemination wasused (Table 1). The proportion of ova fertilised invitroalso declined after 2 days storage.The fertility of fresh semen was lower than semenstored <strong>for</strong> 2 days when inseminated into <strong>the</strong> cervix(P


CHARGED LIPOPROTEIN FRACTIONS OF EGG YOLK IN BOVINE SEMEN DILUENTSE. Prendergast*, R. Vishwanath, P. Shannon and P. Molan*Livestock Improvement Corporation Limited, New Zealand Dairy Board, Hamilton, and*Department ofBiological Sciences, University ofWaikato, Hamilton, NewZealand.•MOTILITY CHARACTERISTICS OF SEMEN FROM RAMS SUBJECTED TO SCROTALINSULATION, AFTER STORAGE AT CHILLED TEMPERATURES FOR ONE WEEKChairussyuhur, A., Sanchez-Partida, L.G., Zupp, J.L., Maddocks, S. and Setchell, B.P.Department ofAnimal Sciences, Waite Agricultural Research Institute,University of Adelaide, Glen Osmond, S.A., 5064INTRODUCTIONA bovine semen diluent containing positively chargedlipoprotein material (WTM) has previously been shown tobe superior to a 5% egg yolk control diluent in survivaland fertility studies. This superiority has been attributedto both <strong>the</strong> charged and lipoprotein nature of <strong>the</strong> WTMmolecule (1). The charged protein moiety of <strong>the</strong> lipoproteinis believed to bind to <strong>the</strong> sperm membrane whilst<strong>the</strong> lipid portion blankets <strong>the</strong> membrane providing<strong>the</strong>rmal protection. Fractionation ofWTM by chloro<strong>for</strong>m:water (2: 1 v/v) produced three extracts. Extract 1 was anaqueous extract and Extracts 2 and 3 organic extracts.Both Extract 1 and Extract 2 showed multiple anioniccomponents. The extracts were incorporated into diluentsand tested <strong>for</strong> <strong>the</strong>ir ability to protect bovine sperm againstcold shock damage.METHODS100 ml WTM was mixed with 400 ml chloro<strong>for</strong>m:water(2:1 v/v) <strong>for</strong> 4 hr, on standing at 4°C <strong>for</strong> 24 hours threedistinct layers were visible, <strong>the</strong> middle layer underwenttwo fur<strong>the</strong>r extractions. The corresponding layers fromeach extraction were pooled and concentrated by rotaryevaporation. Protein concentration was determined byei<strong>the</strong>r <strong>the</strong> Biuret method or a modified Biuret method <strong>for</strong>lipid-rich samples (2). The amount of lipid present wasdetermined by <strong>the</strong> method of Foulkes (3) and <strong>the</strong>proportion of neutral and phospholipid by silicic acidchromatography. Isoelectric points were determined byei<strong>the</strong>r IEF using <strong>the</strong> Phastsystem or cellulose acetateelectrophoresis. Semen collected from 3 bulls was dilutedto 100 million sperm/ml in 14GC buffer containing 5%egg yolk or one of <strong>the</strong> EX1Iacts. Cold shocking of spermoccurred at 1°C <strong>for</strong> 20 minutes.RESULTSThe results from protein and lipid determinations areshown in Table 1. Extract 2 contained <strong>the</strong> bulk of<strong>the</strong> lipidand protein that was extracted, Extract 3 contained justlipid and Extract 1 protein. The lipid composition isshown in Table 2. The relative abilities of <strong>the</strong> extracts toprotect sperm against cold shock are shown in Table 3.The diluents containing <strong>the</strong> Extract 1 fraction were foundto af<strong>for</strong>d slight protective action to <strong>the</strong> sperm against coldshock, <strong>the</strong> Extract 2 fraction was as good as <strong>the</strong> 5% eggyolk control,but superior after 24 h incubation at 37°C.Extract 3 was found to have no protective ability. Fur<strong>the</strong>rexperimental work with <strong>the</strong> Extract 2 diluent showed it tomaintain sperm survival over time at both ambienttemperature and 37°C as well as <strong>the</strong> 5% egg yolk diluent,but not <strong>for</strong> as long as sperm in <strong>the</strong> original WTM.CONCLUSIONThe results from this study have shown that organicextraction of <strong>the</strong> cationic lipoprotein W1M producesanionic lipoprotein material that can provide bovinesperm with protection against cold shock and canmaintain sperm survival over time as well as a 5% eggyolk diluent. The changes in <strong>the</strong> lipid and proteinproportions and overall charge have not affected <strong>the</strong>ability to protect against cold shock, but may have affected·<strong>the</strong> ability to sustain survival at 37°C over time comparedwith WTM. This may be due to <strong>the</strong> loss ofhomogeneity of<strong>the</strong> lipoprotein molecule due to <strong>the</strong> disruption of its nativestate as a result of<strong>the</strong> extraction procedure.Table 1. Protein and lipid determinations of<strong>the</strong> extractsofWTMPROTEIN (g) LIPID (g)WTM 3.80 15.2Extract 1 0.41 0Extract 2 3.36 4.48Extract 3 0 8.51Table 2. L' 101'dcompos1uon 0fExtracts 2 and3 0fW1M0/0 0/0 0/0TOTAL PHOSPHO NEUTRALLIPID LIPID LIPIDWTM 79.8 16.7 83.3Extract 2 57.1 83.7 16.3Extract 3 100 13.8 86.2Table 3. Ability of <strong>the</strong> Extracts of WTM to protect spermagainst cold shock. Values are means ± se (n=3). Meanswithin columns with <strong>the</strong> same letter are not significantlydifferentBEFORE AFTER SURVIVALCOLD COLD (after CS)SHOCK SHOCK 24 H/37°CExtract 1 80.0 ± 2.9 a 25.0 ± 2.9 b 8.3 + 1.7 cExtract 2 80.0 ± 2.9 a 68.3 ± 1.7 a 51.7 ± 6.0 aExtract 3 65.0 ± 2.9 b 16.7 ± 4.4 c 5.7 + 4.7 c50/0 EY 78.3 ± 4.4 a 61.7 + 6.0 a 30.0 + 7.6 b14GC 73.3 ± 6.7 a 10.0 ± 2.9 c 5.0 ± O.OcREFERENCES(1) Vishwanath R, Shannon P, Curson B (1992). Anim.Reprod. Sci. 29: 185-194.(2) Upreti GC, Ratcliffe RA, Riches PC (1988). Anal.Biochem. 168: 421-427.(3) Foulkes JA (1977). J. Reprod. Fert. 49: 277-284.INTRODUCTIONWhen normal ram spermatozoa are stored at 5°C<strong>for</strong> up to 10 days, <strong>the</strong>ir motilty is better when anovel CEQY diluent is used than when aconventional HYCG diluent is used (1). However,summer temperature extremes in <strong>the</strong> field maycompromise normal sperm quality. Increase oftesticular temperatures during summer or byscrotal insulation decreases sperm quality (2,3).The present study was <strong>the</strong>re<strong>for</strong>e undertaken toinvestigate <strong>the</strong> effect of CEQY and HYCGdiluents on <strong>the</strong> motility characteristics of semencollected from rams subjected to scrotalinsulation, after storage at cool (5°C), ra<strong>the</strong>r thanfreezing temperatures, <strong>for</strong> one week.MATERIALS AND METHODSThe techniques of scrotal insulation, semencollection and processing were as previouslydescribed (1,2,4). Semen was collected weekly <strong>for</strong>2 weeks be<strong>for</strong>e, <strong>for</strong> 4 weeks during. and <strong>for</strong> 5weeks after scrotal insulation. Sperm motility(MOT) and progressive motility (pROG) wereassessed using a Hamilton Thorn MotilityAnalyser. After angular trans<strong>for</strong>mation, pooleddata <strong>for</strong> each treatment period were subjected toanalysis ofvariance and differences between meanswere determined by Tukey's test.RESULTSAt all time periods, and <strong>for</strong> both treatmentgroups, <strong>the</strong> percent motility and progressivemotility was significantly higher (p


••. STUDIES ON RAM SPERMATOZOAL AROMATIC AMINO ACID OXIDASEG.C. Upreti, K. Jensen, R. Munday, R. Vishwanath* and IF. SmithAgResearch, Ruakura Agricultural Research Centre, PB 3123,-Hamilton, NZ*Livestock Improvement Corporation Ltd, PB 3016, Hamilton, NZTHE EFFECT OF ANTIOXIDANTS ON THE PREGNANCY RATE AFTERINSEMINATION WITH LIQUID STORED RAM SPERMATOZOAT. Stojanov, S.L. Rhodes, W.M.C. Maxwell and O. EvansDepartment ofAnimal Science, University ofSydney, NSW 2006INTRODUCTIONThe addition of desferal to bovine semen diluentimproved <strong>the</strong> shelf-life of diluted bovine semen (1).Desferal is an iron chelator and hence blocks <strong>the</strong>'lconversion of hydrogen peroxide (H 2 0 2 ), producedby semen aromatic aminoacid oxidase (AAO), to <strong>the</strong>Ohydroxyl radical (OH ) in an iron catalysed nonenzymaticreaction. Surprisingly, desferal did notimprove <strong>the</strong> ram spermatozoal motility in a syn<strong>the</strong>ticdiluent, RSD-1 (2). One possible reason <strong>for</strong> <strong>the</strong> lackof a beneficial effect could be due to low levels ofAAO in ram spermatozoa. We <strong>the</strong>re<strong>for</strong>e investigated<strong>the</strong> AAO activity of ram spermatozoa.[K = constant 4.45 III O 2 Iml, V = Volume (5 ml)]RESULTS:AAO activity in ram (R) and bull (B) spermatozoa(37.8 <strong>for</strong> R; 37.0 <strong>for</strong> B; s.e.d. 4.96; n=3) was similar.The relationship between AAO activity and proportionof dead sperm is shown in Fig. 2 and <strong>the</strong> equations<strong>for</strong> various combinations of D, L, Ram & Bull spermin citrate and in RSD-l are:LB/DBLR/DRLB/DBLB/DRRSD-lRSD-lcitratecitratey = 7.62 + 42.4xY= 7.85 + 57.0xy = -3.33 + 38.9xy = -1.81 + 45.0xINTRODUCTIONSpontaneous lipid peroxidation of mammalianspermatozoa in vitro is associated with loss ofmotility and decreased fertility. In vitro studiesshowed that adding antioxidants, specificscavengers or metal chelators to semen extenderscan improve survival time and motility of bovinespermatozoa (1) and in vitro fertilising ability ofram spermatozoa (2) stored in liquid phase. In thisstudy, <strong>the</strong> antioxidants superoxide disniutase(SOD) and catalase (CAT) were included in <strong>the</strong>diluent in an attempt to maintain <strong>the</strong> fertilisingcapacity ofram spermatozoa during liquid storage.Fig. 2: Effect of storage time and diluent onpregnancy rate (%) after insemination with liquidstoredram semen.IITGY.TGY+SO().j.CATMATERIALS AND METHODS:AAO activity was measured in washed ram (R) andbull (B) spermatozoa. The spermatozoa were killedby freeze-thawing (3x) and O 2consumption wasmeasured in 5ml of 2.9% citrate (pH 6.9) containing30 mM phenylalanine and 250 million ofspermatozoa. The AAO activity of live (L) and dead(D) spermatozoa of both species (R & B) wasmonitored by assaying AAO activity in populationscontaining known proportions of D and L sperm inRSD-l and in 2.9% citrate.AAO activity was calculated from 02 consumptioncurves as shown in Figure 1 (see figure & legend).MO activity (uI02/hr/250 mil. sperm)SO -----------")5040302010oRSD-160 80 100Proportion of dead sperm (%)MATERIAL AND METHODSPooled semen collected by artificial vagina from 4rams was diluted 4-fold with Tris-glucose-yolk(TOY) diluent (Tris-3oo mM, citric acid 94.72roM, glucose 27.78 mM, kanamycin 0.07 mg and20 % egg-yolk) or TOY + SOD (800 i.u.lml) +CAT (200 i.u.lml). The diluted semen was storedat 5°C <strong>for</strong> 9 hr (Day 0, control), 7 and 14 days andassessed <strong>for</strong> progressive motility of spermatozoabe<strong>for</strong>e insemination (Fig. 1).--+-TGY---TGY+SOD+CATo 7 14STORAGE TIME (DAYS)RESULTSChi-square analysis showed that pregnancy rateswere higher after insemination with control sementhan with stored semen (P


4IS PERI-OESTROUS INTRAVAGINAL IMPEDANCE AN AID IN SCREENING FORPROSPECTIVE PREGNANT RECIPIENT COWS?A.R. Lehrer 1 ,* and W.H. McMillan 2lInstitute of Animal Science, Agricultural Research Organization, P.G.B. 6, Bet Dagan, Israel;2AgResearch Ruakura, P.B. 3123, Hamilton, New ZealandINTRODUCTIONConception rate of inseminated cows (l) and embryo yieldand quality in donor cows (2), are inversely related withperi-oestrous intravaginal impedance (IVI). No suchin<strong>for</strong>mation exists on recipient cows. The main objectiveof this study was to determine <strong>the</strong> pregnancy rate (PR) ofrecipient cows relative to <strong>the</strong>ir IVI during peri-oestrus andon <strong>the</strong> day of embryo transfer (ET).EXPERIMENTAL PROTOCOLOestrus was synchronised in 36 mixed-age, non-lactatingbeef cows using a CIDR® (3). CIDR® removal was on d 0and IVI was measured once daily on d 0, 1, 4 and 9, andtwice daily on d 2 and 3. A probe developed andmanufactured at <strong>the</strong> first author's Institute was used <strong>for</strong> IVImeasurements. On d 9, a fresh single in-vitro producedembryo was transferred to all oestrous cows (4). PR wasdetermined using return oestrus and ultrasonography ond 35.RESULTS AND DISCUSSIONTwenty eight cows (78%) were in oestrus on d 2-4, with21 (75%) detected on <strong>the</strong> morning of d 2. PR was 13/28(46%). No IV! differences between pregnant and nonpregnantrecipients were significant, o<strong>the</strong>r than IVI on <strong>the</strong>afternoon ofd 2, i.e. -55 h after CIDR® removal, which waslower in pregnant compared with non-pregnant cows(36.1±1.8 vs 41.7±1.4 ohms, P


.'1"4AUTHOR INDEXAlmahbobi,G 84Anderson,S.T. . 5Ankersmit,A.E.L. . 17, 30Arcellana-Panlilio,M 106Armstrong,D.T 15, 25, 36Ashman,R.J 31, 32, 33Aspden,W.J. . , 10Athanasis-Platsis,S 107Atkins,R.C 70Bailey,J.L 90Baker,H.W.G 20Barker-Gibb,M.L. . 1Beaumont,S 110Bell,E.C 45, 46Bergfeld,E. . · ··· 10Boatman,D.E 105Bonello,N. . 66Boublik,J.H. . 1Bourne,H 20Bradley, M.P ~ 88Brannstr5m, M. . 66Breed,B 98Brennan,A.P. . 26Broomfield,D. . 72Brown,B.W 34Brownlee,A.G 104Buab()()Cha,W. ..··.. ·· ·· 101Camus,M 23Cann,C.H 55Cash,M.P 78Catt,J.W 22, 80Cavanagh,A.C. . 102, 107, 108Chairussyuhur, A 114Cheah,C 33Clarke,I.J. . 1, 6, 7Clay, C.M 14Clements,J. . 57Clulow,J 7.1Cox,D.B 47Craw<strong>for</strong>d,R.J 33Curlewis,J.D. . 2, 3D'Occhio,M.J 10, 73Dai,Y.F 36Day, A.M 74~e Kretser,D.M 7, 63, 65, 82, 84, 85, 86g~~:~i::''',~:::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::: ::J~~~~~~~:A:··:::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::~~g~~~~~~~:~:.::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::: ~~::~!~:;;d~:D:R'" .:::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::j b~Ehrljch,A 57Ekpe,G 87Entwistle,K. . 75AUTHOR INDEX (cont)Eppleston, J. . 111Evans,G 52, 116Fairclough,R.J 38, 55Fawcett,A.A. .. 34Findlay,J.K 4, 8,24,37,50,59,60Fisher,J.S 48Fitzpatrick,L.A. . 12, 75Fordyce,G 75Fry,R.C 24, 61Garcia,S 8Gardner,D.K. . 53Gehring,H 45, 46Gemmell, R.T 101Gidley-Baird, A 88Giliarn,K. . 35Glencross, B.D 79G()()(ien, J.M 9Gow,C.B 55Graharn,J.K. .. 51Grigg,G 87Grupen,C.G 18, 33Hamernick, D.L. . 14Hansen,L.A. . 71Harari,O. .. 20Harding,M 33Hariadi,M 13, 72Harris, M 83Harry,J.L. . 68Hartmann,P.E 47, 79Harvey,M.B 106Hayes,R 70Hayes,T 86Healy,D.L. . 60Hearn,M 65Hedger,M.P. . 70Hemsworth,P.H 11Hillard,M.A ' 5Hillyard,N.C. .. 108Hinds,L.A. . 88Hoogkinson,S.C. . 56Holden,C.A. . 26Hosie,M.J 42Hotzel,M.J 48, 76Irvine, B.J ~ 15Jackson,A.E. .. 77Janssenswillen,C. . 23Jenkin,G 38Jensen,K. . 115Jiang,F.-X 81Jillella,D 73Johnston,W.H. . 20Jones,G.M 20Jones,R.C 71, 89Joris,H 23Kashiwazaki,N 31, 32, 33Kaye,P.L 96, 97, 102, 107Kent,J.C 47Kern, S 67, 69


AUTHOR INDEX (coot)Kim,Y.H 84, 85Kinder,J.E 10King,J.C 80Kleemann,D.O 44Klein,R 6Knott,L.M. . 77Kotaras, P 15Lacham-Kaplan,O. . 21Lane,M 53Lash,G 109Laslett,A 65Lavranos,T.C 64-I..ean, I.J. . 9I..edgard,A.M 56ugge,M 109I..ehrer,A.R. . 117Lohuis,H. . 11Loveland,K 63, 86Lui,J 23Macmillan,K.L. . 49, 74Maddocks, S 67, 69, 87,114Magnoni,G.E 105Mahaworasilpa,T. . 35Marsh,M.M 57, 60Marshall,J.T.A. . 104Martin, G.B 48, 76Martin, L. . 3, 39Massa,H 39Mate,K.E 93, 99Ma<strong>the</strong>ws,P.M. . 20Maxwell,W.M.C 27,51,52,111,116McDonald,M. . 20McDougall,S 49, 74McFarlane, J.R 65, 84, 85, 86McGowan,L.T 16, 17,30Mcllfatrick,S.M. . 33McMillan,W.H. . 19, 117Meier,S : 38Mercer,J.E 4, 8ti~~f:;, ~~~~ ~~~~ ~~ :~:~~: ::~~ ~~ ~~ ::~::::~ :~~: ~~:~:::::::::::::::::::::::::::::::::::::::::::::::~ ~~~~:~:::~ ~~ ::~~:~~::~ ::.~Molan,P. . 113Molinia,F.C 28Morton,H. . 102, 107, 108Morton,M. . 22Moss, G.E 14Munday,R 115Murdoch, R.N 40, 89Murphy,C.R 41, 42, 43Murphy,R.M 108Nagashima,H 18, 31, 32, 33Nancarrow,C.D. . 104Nemaia,F.A 17Niasari-Naslaji,A. . 73Nikolics-Paterson,D.J. . 70Norman,R 66Nottle,M.B 18, 31, 32, 33O'Brien,G.M. . 3AUTHOR INDEX (coot)O'Brien,J.K. . 52O'Connor,A 62O'Grady,J 34O'Neill,C 80, 103O'Shea,T 5Oliver,J 110Owens,P.C 36Owens,R.A 47Pantaleon,M. . 96Parr,J 110Parr,R.A. . 78Pedersen,R.A. . 93, 99Perks,C.M 54Peterson,A.J. .. 19, 56Philip,C 13Pitt,C 112Pomares,C.C. . 111Prendergast,E. . 113Pugh,P.A. . 16, 19, 30Rabiee, A.R. . 9Renfree,M.B 68, 100Rhooes,S.L 116Richings,N.M. . 20Rigby,R.D.G 76Riley,S.C 37, 59Risbridger,G 63, 65Roberts,C. . 98Robertson, D.M 6, 50Robertson,S.A 58, 66Robins,A.J 33Robinson,E.S 99Robinson,S.J. . 51Rooger,J.C. . 28, 83, 91, 92Roogers,R.J 64-Roogers,H.F 64Rowe,J.P. . 15Russell,D.L. . 50Rutledge,J. . 25Ryan,J.P 52, 94, 95Sakoff,J.A. . 40Salamonsen,L.A 37, 57, 59, 60, 61Sanchez-Partida, L.G 27, 114Saunders,D 22SawangjarClen,K. . 2Schultz,G.A ,106Scott,K. . 64-Seamark,A.C 58Seamark,R.F 36, 44, 58Setchell,B.P 27, 87, 114Setiadi,D 91, 92Shannon,P 112, 113Shaw,G 45, 46, 68Shaw,T.J 41Shorey,C.D. . 43Short,R.V 81Simorangkir,D.R 82Simpson,T.L. . 24Sistina,Y 91


AUTHOR INDEX (cont)Smith,J.F ~ 16, 110, 115Smitz,J. . , 23Somooevilla Torres,M. . 108Squires,E.L. . 51Squires,T.J 24Stein-Oakley,A. . 63Stevenson,K.R. . 54Stobart,S.J. . 12Stojanov,T 111, 116Storey, B.T. . 90Sutton,R. . 34, 55Tatham,B. . 35Taufa, V.K 74Temple Smith,P.D 84, 85Terry,V 43Tervit,H.R. . 16, 17, 30, 56Tian,L 80Tilbrook,A.J. . 7, 11Toussaint,J.K 79Trewhella,M.A 38Trigg,T.E 77Trounson,A 21, 35Tshewang,U 77Tuckey,R.C. . 79Turner,A 11Udy,G.B 17Upreti,G.C 110, 115Van Steirteghem,A 23VanKirk, E.L. . 14Vella, C.A 64Villar, A.J 93Vishwanath,R 112, 113, 115Vogiagis,D. . 61Waite,K.M. . 52, 94, 95Walkden-Brown,S.W 48, 76Walker,S.K. . 44Ward,W.G 34Waters,J.M. . 78Wa<strong>the</strong>s,D.C 54Watson,A.J :.25Whitworth,D.J 68, 100Whyte, T.R 10Wong,E 37Wre<strong>for</strong>d,N.G.M 62, 82Wright,P.J 13, 72Xia,P 25Young,S 88Zhang,X.Q 106Zlatic,K 63, 86Zupp, J.L 27, 87, 114A

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

Saved successfully!

Ooh no, something went wrong!