11.07.2015 Views

Epigenetics in Human Disease - Computer Science

Epigenetics in Human Disease - Computer Science

Epigenetics in Human Disease - Computer Science

SHOW MORE
SHOW LESS

Create successful ePaper yourself

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

<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>


This page <strong>in</strong>tentionally left blank


<strong>Epigenetics</strong> <strong>in</strong><strong>Human</strong> <strong>Disease</strong>Edited byTrygve O. TollefsbolDepartment of Biology, University of Alabama at Birm<strong>in</strong>gham, AL, USAAmsterdam Boston Heidelberg London New York OxfordParis San Diego San Francisco S<strong>in</strong>gapore Sydney TokyoAcademic Press is an impr<strong>in</strong>t of Elsevier


Academic Press is an impr<strong>in</strong>t of Elsevier32 Jamestown Road, London NW1 7BY, UK225 Wyman Street, Waltham, MA 02451, USA525 B Street, Suite 1800, San Diego, CA 92101-4495, USAFirst edition 2012Copyright Ó 2012 Elsevier Inc. All rights reserved.No part of this publication may be reproduced, stored <strong>in</strong> a retrieval system or transmitted <strong>in</strong> any form or by any meanselectronic, mechanical, photocopy<strong>in</strong>g, record<strong>in</strong>g or otherwise without the prior written permission of the publisherPermissions may be sought directly from Elsevier’s <strong>Science</strong> & Technology Rights Department <strong>in</strong> Oxford, UK: phone (+ 44) (0) 1865843830; fax (+44) (0) 1865 853333; email: permissions@elsevier.com. Alternatively, visit the <strong>Science</strong> and Technology Bookswebsite at www.elsevierdirect.com/rights for further <strong>in</strong>formationNoticeNo responsibility is assumed by the publisher for any <strong>in</strong>jury and/or damage to persons or property as a matter of productsliability, negligence or otherwise, or from any use or operation of any methods, products, <strong>in</strong>structions or ideas conta<strong>in</strong>ed <strong>in</strong> thematerial here<strong>in</strong>. Because of rapid advances <strong>in</strong> the medical sciences, <strong>in</strong> particular, <strong>in</strong>dependent verification of diagnoses and drugdosages should be madeBritish Library Catalogu<strong>in</strong>g-<strong>in</strong>-Publication DataA catalogue record for this book is available from the British LibraryLibrary of Congress Catalog<strong>in</strong>g-<strong>in</strong>-Publication DataA catalog record for this book is available from the Library of CongressISBN: 978-0-12-388415-2For <strong>in</strong>formation on all Academic Press publicationsvisit our website at elsevierdirect.comTypeset by TNQ Books and Journals Pvt Ltd. www.tnq.co.<strong>in</strong>Pr<strong>in</strong>ted and bound <strong>in</strong> United States of America1213141516 10987654321


CONTENTSPREFACE .................................................................................................................. viiCONTRIBUTORS..........................................................................................................ixCHAPTER 1 <strong>Epigenetics</strong> of <strong>Human</strong> <strong>Disease</strong> ............................................................... 1Trygve O. TollefsbolCHAPTER 2Methods and Strategies to Determ<strong>in</strong>e Epigenetic Variation<strong>in</strong> <strong>Human</strong> <strong>Disease</strong>.................................................................................. 7Yoshihisa Watanabe and Masato MaekawaCHAPTER 3 DNA Methylation Alterations <strong>in</strong> <strong>Human</strong> Cancers .................................... 29Yae Kanai and Eri AraiCHAPTER 4 Alterations of Histone Modifications <strong>in</strong> Cancer...................................... 53Ciro Mercurio, Simon Plyte and Saverio M<strong>in</strong>ucciCHAPTER 5 MicroRNA <strong>in</strong> Oncogenesis .................................................................... 89Niamh Lynam-Lennon, Steven G. Gray and Stephen G. MahervCHAPTER 6 Epigenetic Approaches to Cancer Therapy .......................................... 111Sabita N. Saldanha and Trygve O. TollefsbolCHAPTER 7 Epigenomics <strong>in</strong> Neurobehavioral <strong>Disease</strong>s........................................... 127Cassandra L. Smith and Kai HuangCHAPTER 8Emerg<strong>in</strong>g Role of <strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> NeurodevelopmentalDisorders .......................................................................................... 153K. Naga Mohan and J. Richard ChailletCHAPTER 9 The <strong>Epigenetics</strong> of Alzheimer’s <strong>Disease</strong> .............................................. 175Fabio CoppedèCHAPTER 10 Epigenetic Modulation of <strong>Human</strong> Neurobiological Disorders ................. 193Takeo Kubota, Kunio Miyake, Takae Hirasawa, Tatsushi Onakaand Hidenori YamasueCHAPTER 11 Epigenetic Basis of Autoimmune Disorders <strong>in</strong> <strong>Human</strong>s......................... 205Biola M. Javierre, Lorenzo De La Rica and Esteban BallestarCHAPTER 12 Approaches to Autoimmune <strong>Disease</strong>s Us<strong>in</strong>g Epigenetic Therapy .......... 225Christopher ChangCHAPTER 13 Epigenetic Mechanisms of <strong>Human</strong> Impr<strong>in</strong>t<strong>in</strong>g Disorders....................... 253Richard H. Scott and Gudrun E. Moore


CONTENTSCHAPTER 14 Epigenomic Factors <strong>in</strong> <strong>Human</strong> Obesity ................................................ 273Christopher G. BellCHAPTER 15 Epigenetic Approaches to Control Obesity .......................................... 297Abigail S. Lapham, Karen A. Lillycrop, Graham C. Burdge,Peter D. Gluckman, Mark A. Hanson and Keith M. GodfreyCHAPTER 16 <strong>Epigenetics</strong> of Diabetes <strong>in</strong> <strong>Human</strong>s..................................................... 321Charlotte L<strong>in</strong>g and T<strong>in</strong>a RönnCHAPTER 17 The Potential of Epigenetic Compounds <strong>in</strong> Treat<strong>in</strong>g Diabetes............... 331Steven G. GrayCHAPTER 18 Epigenetic Aberrations <strong>in</strong> <strong>Human</strong> Allergic <strong>Disease</strong>s ............................. 369Manori Amarasekera, David Mart<strong>in</strong>o, Meri K. Tulic,Richard Saffery and Susan PrescottCHAPTER 19 Therapy of Airway <strong>Disease</strong>: Epigenetic Potential ................................. 387Peter J. BarnesCHAPTER 20 The Role of <strong>Epigenetics</strong> <strong>in</strong> Cardiovascular <strong>Disease</strong> .............................. 395Boda Zhou, Andriana Margariti and Q<strong>in</strong>gbo XuCHAPTER 21 <strong>Epigenetics</strong> and <strong>Human</strong> Infectious <strong>Disease</strong>s ....................................... 415Hans Helmut Niller and Janos M<strong>in</strong>arovitsviCHAPTER 22 The <strong>Epigenetics</strong> of Endometriosis....................................................... 443Sun-Wei GuoCHAPTER 23 Aberrant DNA Methylation <strong>in</strong> Endometrial Cancer................................ 471Kenta Masuda, Kouji Banno, Megumi Yanokura,Kosuke Tsuji, Iori Kisu, Arisa Ueki, Yusuke Kobayashi,Hiroyuki Nomura, Akira Hirasawa, Nobuyuki Susumuand Daisuke AokiCHAPTER 24 Stem Cell <strong>Epigenetics</strong> and <strong>Human</strong> <strong>Disease</strong> ......................................... 481Mehdi Shafa and Derrick E. RancourtCHAPTER 25 Non-Cod<strong>in</strong>g RNA Regulatory Networks, <strong>Epigenetics</strong>, and Programm<strong>in</strong>gStem Cell Renewal and Differentiation: Implications for Stem CellTherapy............................................................................................. 503Rajesh C. MirandaCHAPTER 26 Ag<strong>in</strong>g and <strong>Disease</strong>: The Epigenetic Bridge .......................................... 519Andrea FusoCHAPTER 27 Early-Life Epigenetic Programm<strong>in</strong>g of <strong>Human</strong> <strong>Disease</strong> and Ag<strong>in</strong>g .......... 545Alexander M. VaisermanINDEX ................................................................................................................... 569


PREFACENow that epigenetics has moved to the forefront of biological sciences, this book is focused onpractical aspects of this burgeon<strong>in</strong>g field of science and is <strong>in</strong>tended to provide the most recent,pert<strong>in</strong>ent, and comprehensive <strong>in</strong>formation with respect to the role of epigenetics <strong>in</strong> humandisease. Epigenetic diseases consist of the <strong>in</strong>creas<strong>in</strong>g number of human diseases that have atleast part of their basis <strong>in</strong> aberrations of epigenetic processes such as DNA methylation,histone modifications, or non-cod<strong>in</strong>g RNAs. Unlike most genetic defects as a cause for humandisease, epigenetic alterations are potentially reversible. This is perhaps the most importantaspect of epigenetic diseases because their reversibility makes these diseases amenable topharmacological treatment. The goal of this volume is to highlight those diseases or conditionsfor which we have considerable epigenetic knowledge such as cancer, autoimmunedisorders, and ag<strong>in</strong>g as well as those that are yield<strong>in</strong>g excit<strong>in</strong>g breakthroughs <strong>in</strong> epigeneticssuch as diabetes, neurological disorders, and cardiovascular disease.Where applicable, attempts are made not only to detail the role of epigenetics <strong>in</strong> the etiology,progression, diagnosis, and prognosis of these diseases, but also to present novel epigeneticapproaches to the treatment of these diseases. The book is designed such that each featuredhuman disease is first described <strong>in</strong> terms of the underly<strong>in</strong>g role of epigenetics <strong>in</strong> the diseaseand, where possible, followed by a chapter describ<strong>in</strong>g the most recent advances <strong>in</strong> epigeneticapproaches for treat<strong>in</strong>g the disease. This allows basic scientists to readily view how their effortsare currently be<strong>in</strong>g translated to the cl<strong>in</strong>ic and it also allows cl<strong>in</strong>icians to review <strong>in</strong> side-by-sidechapters the epigenetic basis of the diseases they are treat<strong>in</strong>g. In some cases our knowledge ofthe epigenetics of human diseases is more extensive. Therefore, <strong>in</strong> those cases, such as cancer,more than one chapter on the underly<strong>in</strong>g epigenetic causes appears. In other cases, such as forneurological disorders, the epigenetic basis of the diseases can vary somewhat due to thevary<strong>in</strong>g nature of the disorders. Chapters are also presented on the epigenetics of humanimpr<strong>in</strong>t<strong>in</strong>g disorders, respiratory diseases, <strong>in</strong>fectious diseases, and gynecological and reproductivediseases, as well as the epigenetics of stem cells, obesity, and allergic diseases. Althoughag<strong>in</strong>g is not considered to be a human disease per se, there are many age-associated diseases.Moreover, s<strong>in</strong>ce epigenetics plays a major role <strong>in</strong> the ag<strong>in</strong>g process, advances <strong>in</strong> the epigeneticsof ag<strong>in</strong>g are highly relevant to many human diseases. Therefore, this volume closes withchapters on ag<strong>in</strong>g epigenetics and breakthroughs that have been made to delay the ag<strong>in</strong>gprocess through epigenetic approaches.viiThe <strong>in</strong>tended audience for this book is the vast body of students and scientists who are<strong>in</strong>terested <strong>in</strong> either the underly<strong>in</strong>g basis of human diseases and/or novel means to treat humandiseases that are caused by reversible epigenetic processes. This book is tailored for those with<strong>in</strong>terests rang<strong>in</strong>g from basic molecular biology to cl<strong>in</strong>ical therapy and who could benefit froma comprehensive analysis of epigenetics as it applies to human diseases.Trygve O. Tollefsbol


This page <strong>in</strong>tentionally left blank


CONTRIBUTORSManori AmarasekeraSchool of Paediatrics and Child Health, University of Western Australia, Roberts Road,Subiaco, WA 6008, AustraliaDaisuke AokiDepartment of Obstetrics and Gynecology, Keio University School of Medic<strong>in</strong>e,Sh<strong>in</strong>anomachi 35, Sh<strong>in</strong>juku-ku, Tokyo 160-8582, JapanEri AraiDivision of Molecular Pathology, National Cancer Center Research Institute,Tokyo 104-0045, JapanEsteban BallestarChromat<strong>in</strong> and <strong>Disease</strong> Group, Cancer <strong>Epigenetics</strong> and Biology Programme (PEBC),Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat,Barcelona, Spa<strong>in</strong>Kouji BannoDepartment of Obstetrics and Gynecology, Keio University School of Medic<strong>in</strong>e,Sh<strong>in</strong>anomachi 35, Sh<strong>in</strong>juku-ku, Tokyo 160-8582, JapanPeter J. BarnesNational Heart and Lung Institute, Imperial College School of Medic<strong>in</strong>e, Dovehouse St,London SW3 6LY, UKixChristopher G. BellMedical Genomics, UCL Cancer Institute, University College London, 72 Huntley Street,London, WC1 6BT, UKGraham C. BurdgeAcademic Unit of <strong>Human</strong> Development and Health, Faculty of Medic<strong>in</strong>e, University ofSouthampton, Southampton, UKJ. Richard ChailletDepartment of Microbiology and Molecular Genetics, Magee-Womens Research Institute,University of Pittsburgh School of Medic<strong>in</strong>e, Pittsburgh, PA, USAChristopher ChangDivision of Allergy, Asthma and Immunology, Thomas Jefferson University, Nemours/A.I.duPont Hospital for Children, 1600 Rockland Road, Wilm<strong>in</strong>gton, DE 19803, USA.Fabio CoppedèFaculty of Medic<strong>in</strong>e, Section of Medical Genetics, University of Pisa, Pisa, ItalyLorenzo De La RicaChromat<strong>in</strong> and <strong>Disease</strong> Group, Cancer <strong>Epigenetics</strong> and Biology Programme (PEBC),Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat,Barcelona, Spa<strong>in</strong>Andrea FusoDept. of Surgery “P. Valdoni”, Sapienza University of Rome, Via AntonioScarpa, 14 00161, Rome, Italy


CONTRIBUTORSPeter D. GluckmanLigg<strong>in</strong>s Institute, University of Auckland, Auckland, New ZealandKeith M. GodfreyAcademic Unit of <strong>Human</strong> Development and Health, Faculty of Medic<strong>in</strong>e, University ofSouthampton, Southampton, UK; MRC Lifecourse Epidemiology Unit, Southampton, UK;NIHR Nutrition Biomedical Research Unit, Southampton General Hospital, Tremona Road,Southampton SO16 6YD, UKSteven G. GrayDepartment of Cl<strong>in</strong>ical Medic<strong>in</strong>e, Tr<strong>in</strong>ity Centre for Health <strong>Science</strong>s, St James’s Hospital,Dubl<strong>in</strong> 8, IrelandSun-Wei GuoShanghai Obstetrics and Gynecology Hospital, Fudan University, 419 Fangxie Road,Shanghai 200011, Ch<strong>in</strong>aMark A. HansonAcademic Unit of <strong>Human</strong> Development and Health, Faculty of Medic<strong>in</strong>e, University ofSouthampton, Southampton, UKAkira HirasawaDepartment of Obstetrics and Gynecology, Keio University School of Medic<strong>in</strong>e,Sh<strong>in</strong>anomachi 35, Sh<strong>in</strong>juku-ku, Tokyo 160-8582, JapanTakae HirasawaDepartment of <strong>Epigenetics</strong> Medic<strong>in</strong>e, Interdiscipl<strong>in</strong>ary Graduate School of Medic<strong>in</strong>e andEng<strong>in</strong>eer<strong>in</strong>g, University of Yamanashi, Yamanashi, JapanxKai HuangFortiss geme<strong>in</strong>nützige GmbHAn-Institut der TU MünchenGuerickestr. 25, 80805Munich, GermanyBiola M. JavierreChromat<strong>in</strong> and <strong>Disease</strong> Group, Cancer <strong>Epigenetics</strong> and Biology Programme (PEBC),Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, Barcelona,Spa<strong>in</strong>Yae KanaiDivision of Molecular Pathology, National Cancer Center Research Institute,Tokyo 104-0045, JapanIori KisuDepartment of Obstetrics and Gynecology, Keio University School of Medic<strong>in</strong>e,Sh<strong>in</strong>anomachi 35, Sh<strong>in</strong>juku-ku, Tokyo 160-8582, JapanYusuke KobayashiDepartment of Obstetrics and Gynecology, Keio University School of Medic<strong>in</strong>e,Sh<strong>in</strong>anomachi 35, Sh<strong>in</strong>juku-ku, Tokyo 160-8582, JapanTakeo KubotaDepartment of <strong>Epigenetics</strong> Medic<strong>in</strong>e, Interdiscipl<strong>in</strong>ary Graduate School of Medic<strong>in</strong>e andEng<strong>in</strong>eer<strong>in</strong>g, University of Yamanashi, Yamanashi, JapanAbigail S. LaphamAcademic Unit of <strong>Human</strong> Development and Health, Faculty of Medic<strong>in</strong>e, University ofSouthampton, Southampton, UK


CONTRIBUTORSKaren A. LillycropFaculty of Natural and Environmental <strong>Science</strong>s, University of Southampton,Southampton, UKCharlotte L<strong>in</strong>g<strong>Epigenetics</strong> and Diabetes Unit, Lund University Diabetes Centre, Department of Cl<strong>in</strong>ical<strong>Science</strong>s, Lund University, Malmö, SwedenNiamh Lynam-LennonDepartment of Surgery, Tr<strong>in</strong>ity College Dubl<strong>in</strong>, Tr<strong>in</strong>ity Centre for Health <strong>Science</strong>s,St James’s Hospital, Dubl<strong>in</strong> 8, IrelandMasato MaekawaDepartment of Laboratory Medic<strong>in</strong>e, Hamamatsu University School of Medic<strong>in</strong>e, 1-20-1Handayama, Higashi-ku, Hamamatsu 431-3192, JapanStephen G. MaherDepartment of Surgery, Tr<strong>in</strong>ity College Dubl<strong>in</strong>, Tr<strong>in</strong>ity Centre for Health <strong>Science</strong>s,St James’s Hospital, Dubl<strong>in</strong> 8, IrelandAndriana MargaritiCardiovascular Division, K<strong>in</strong>g’s College London BHF Centre, London, UKDavid Mart<strong>in</strong>oDevelopmental <strong>Epigenetics</strong>, Murdoch Children’s Research Institute, Flem<strong>in</strong>gton Road,Parkville, VIC 3052, AustraliaKenta MasudaDepartment of Obstetrics and Gynecology, Keio University School of Medic<strong>in</strong>e,Sh<strong>in</strong>anomachi 35, Sh<strong>in</strong>juku-ku, Tokyo 160-8582, JapanCiro MercurioDAC-Genextra group, Via Adamello 16, 20100 Milan Italy and European Institute ofOncology, IFOM-IEO campus, Via Adamello 16, 20100 Milan ItalyxiJanos M<strong>in</strong>arovitsMicrobiological Research Group, National Center for Epidemiology, H-1529 Budapest,Piheno u. 1. HungarySaverio M<strong>in</strong>ucciEuropean Institute of Oncology, IFOM-IEO campus, Via Adamello 16, 20100 Milan Italyand University of Milan, Via Festa del Perdono 7, 20100 Milan ItalyRajesh C. MirandaTexas A&M Health <strong>Science</strong> Ctr., College of Medic<strong>in</strong>e, Dept. Neuroscience andExperimental Therapeutics, Medical Research and Education Build<strong>in</strong>g, 8447 StateHighway 47, Bryan, TX 77807-3260, USAKunio MiyakeDepartment of <strong>Epigenetics</strong> Medic<strong>in</strong>e, Interdiscipl<strong>in</strong>ary Graduate School of Medic<strong>in</strong>e andEng<strong>in</strong>eer<strong>in</strong>g, University of Yamanashi, Yamanashi, JapanK. Naga MohanCentre for <strong>Human</strong> Genetics, Biotech Park Phase one, Electronic City, Bangalore-560100,India; Department of Biological <strong>Science</strong>s, BITS-Pilani, Hyderabad Campus, Shameerpet,Hyderabad-500078, India and Department of Microbiology and Molecular Genetics,Magee-Women’s Research Institute, University of Pittsburgh School of Medic<strong>in</strong>e,Pittsburgh, PA USA


CONTRIBUTORSGudrun E. MooreCl<strong>in</strong>ical and Molecular Genetics Unit, Institute of Child Health, University College London,London, UKHans Helmut NillerInstitute for Medical Microbiology and Hygiene at the University of Regensburg,Franz-Josef-Straub-Allee 11, D-93053 Regensburg, GermanyHiroyuki NomuraDepartment of Obstetrics and Gynecology, Keio University School of Medic<strong>in</strong>e,Sh<strong>in</strong>anomachi 35, Sh<strong>in</strong>juku-ku, Tokyo 160-8582, JapanTatsushi OnakaDivision of Bra<strong>in</strong> and Neurophysiology, Department of Physiology, School of Medic<strong>in</strong>e, JichiMedical University, Tochigi, JapanSimon PlyteCongenia-Genextra group, Via Adamello 16, 20100 Milan Italy and European Institute ofOncology, IFOM-IEO campus, Via Adamello 16, 20100 Milan ItalySusan PrescottSchool of Paediatrics and Child Health, University of Western Australia, Roberts Road,Subiaco, WA 6008, AustraliaDerrick E. RancourtDepartment of Biochemistry and Molecular Biology, University of Calgary, 3330 HospitalDrive NW, Calgary, AB, Canada, T2N 4N1xiiT<strong>in</strong>a Rönn<strong>Epigenetics</strong> and Diabetes Unit, Lund University Diabetes Centre, Department of Cl<strong>in</strong>ical<strong>Science</strong>s, Lund University, Malmö, SwedenRichard SafferyDevelopmental <strong>Epigenetics</strong>, Murdoch Children’s Research Institute, Flem<strong>in</strong>gton Road,Parkville, VIC 3052, AustraliaSabita N. SaldanhaDepartment of Biology, University of Alabama at Birm<strong>in</strong>gham, 175A Campbell Hall, 1300University Boulevard, University of Alabama at Birm<strong>in</strong>gham, Birm<strong>in</strong>gham, AL 35294-1170and Department of Math and <strong>Science</strong>s, Alabama State University, P. O. Box, 271,Montgomery, AL 36101-0271, USARichard H. ScottCl<strong>in</strong>ical and Molecular Genetics Unit, Institute of Child Health, University College London,London and Department of Cl<strong>in</strong>ical Genetics, Great Ormond Street Hospital, London, UKMehdi ShafaDepartment of Biochemistry and Molecular Biology, University of Calgary, 3330 HospitalDrive NW, Calgary, AB, Canada, T2N 4N1Cassandra L. SmithMolecular Biotechnology Research Laboratory, Boston University, 44 Cumm<strong>in</strong>gton Street,Boston, MA, USANobuyuki SusumuDepartment of Obstetrics and Gynecology, Keio University School of Medic<strong>in</strong>e,Sh<strong>in</strong>anomachi 35, Sh<strong>in</strong>juku-ku, Tokyo 160-8582, Japan


CONTRIBUTORSTrygve O. TollefsbolDepartment of Biology, 175 Campbell Hall, 1300 University Boulevard, Birm<strong>in</strong>gham,AL 35294-1170, USAKosuke TsujiDepartment of Obstetrics and Gynecology, Keio University School of Medic<strong>in</strong>e,Sh<strong>in</strong>anomachi 35, Sh<strong>in</strong>juku-ku, Tokyo 160-8582, JapanMeri K. TulicSchool of Paediatrics and Child Health, University of Western Australia, Roberts Road,Subiaco, WA 6008, AustraliaArisa UekiDepartment of Obstetrics and Gynecology, Keio University School of Medic<strong>in</strong>e,Sh<strong>in</strong>anomachi 35, Sh<strong>in</strong>juku-ku, Tokyo 160-8582, JapanAlexander M. VaisermanLaboratory of <strong>Epigenetics</strong>, Institute of Gerontology, Kiev, Ukra<strong>in</strong>eYoshihisa WatanabeDepartment of Laboratory Medic<strong>in</strong>e, Hamamatsu University School of Medic<strong>in</strong>e, 1-20-1Handayama, Higashi-ku, Hamamatsu 431-3192, JapanQ<strong>in</strong>gbo XuCardiovascular Division, K<strong>in</strong>g’s College London BHF Centre, London, UKHidenori YamasueDepartment of Neuropsychiatry, Graduate School of Medic<strong>in</strong>e, The University of Tokyo,Tokyo, JapanMegumi YanokuraDepartment of Obstetrics and Gynecology, Keio University School of Medic<strong>in</strong>e,Sh<strong>in</strong>anomachi 35, Sh<strong>in</strong>juku-ku, Tokyo 160-8582, JapanxiiiBoda ZhouCardiovascular Division, K<strong>in</strong>g’s College London BHF Centre, London, UK and Department ofPhysiology, Pek<strong>in</strong>g University, Beij<strong>in</strong>g, Ch<strong>in</strong>a


This page <strong>in</strong>tentionally left blank


CHAPTER 1<strong>Epigenetics</strong> of <strong>Human</strong><strong>Disease</strong>Trygve O. TollefsbolUniversity of Alabama at Birm<strong>in</strong>gham, Birm<strong>in</strong>gham, AL, USACHAPTER OUTLINE1.1 Introduction 11.2 Epigenetic Variation Methods 21.3 Cancer <strong>Epigenetics</strong> 21.4 <strong>Epigenetics</strong> of Neurological<strong>Disease</strong> 31.5 Autoimmunity and<strong>Epigenetics</strong> 31.6 <strong>Human</strong> Impr<strong>in</strong>t<strong>in</strong>g Disorders 41.7 <strong>Epigenetics</strong> of Obesity 41.8 Diabetes: The EpigeneticConnection 41.9 <strong>Epigenetics</strong> and AllergicDisorders 41.10 Cardiovascular <strong>Disease</strong> and<strong>Epigenetics</strong> 51.11 <strong>Epigenetics</strong> of <strong>Human</strong> Infectious<strong>Disease</strong>s 51.12 Reproductive Disorders andEpigenetic Aberrations 51.13 Stem Cell <strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong><strong>Disease</strong> 51.14 <strong>Epigenetics</strong> of Ag<strong>in</strong>g and Age-Associated <strong>Disease</strong>s 61.15 Conclusion 6References 611.1 INTRODUCTION<strong>Epigenetics</strong> does not <strong>in</strong>volve changes <strong>in</strong> DNA sequence but is nevertheless able to <strong>in</strong>fluenceheritable gene expression through a number of processes such as DNA methylation, modificationsof chromat<strong>in</strong> and non-cod<strong>in</strong>g RNA. Aberrations <strong>in</strong> DNA methylation are commoncontributors to disease. For example, impr<strong>in</strong>t<strong>in</strong>g diseases such as the Angelman, SilvereRussell,PradereWilli and BeckwitheWiedemann syndromes are often associated with alterations <strong>in</strong>DNA methylation [1]. <strong>Human</strong> diseases attributable to DNA methylation-based impr<strong>in</strong>t<strong>in</strong>gdisorders, however, have not been limited to these genetic diseases as diabetes, schizophrenia,autism and cancer have also been associated with aberrations <strong>in</strong> impr<strong>in</strong>t<strong>in</strong>g. Abnormalities ofthe enzymes that mediate DNA methylation can also contribute to disease as illustrated by therare ImmunodeficiencyeCentromere <strong>in</strong>stabilityeFacial anomalies (ICF) syndrome caused bymutations <strong>in</strong> DNA methyltransferase 3B (DNMT3B). Likewise, Rett syndrome, related tomutations <strong>in</strong> the methyl-b<strong>in</strong>d<strong>in</strong>g doma<strong>in</strong> (MBD) prote<strong>in</strong>, MeCP2, leads to dysregulations <strong>in</strong>gene expression and neurodevelopmental disease [2]. Perhaps most commonly, DNA methylationaberrations can often contribute to cancer either through DNA hypo- or hypermethylation.DNA hypomethylation leads to chromosomal <strong>in</strong>stability and can also contributeT. Tollefsbol (Ed): <strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>. DOI: 10.1016/B978-0-12-388415-2.00001-9Copyright Ó 2012 Elsevier Inc. All rights reserved.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>to oncogene activation, both common processes <strong>in</strong> oncogenesis, and DNA hypermethylationis often associated with tumor suppressor gene <strong>in</strong>activation dur<strong>in</strong>g tumorigenesis.Histone modifications frequently contribute to disease development and progressions andhistone acetylation or deacetylation are the most common histone modifications <strong>in</strong>volved <strong>in</strong>diseases. Aberrations <strong>in</strong> histone modifications can significantly disrupt gene regulation,a common factor <strong>in</strong> disease, and could potentially be transmissible across generations [3].Histone modifications have <strong>in</strong> fact been associated with a number of diseases such as cancerand neurological disorders. Collaborations between DNA methylation and histone modificationscan occur and either or both of these epigenetic processes may lead to disease development[4].Non-cod<strong>in</strong>g RNAs are an emerg<strong>in</strong>g area of epigenetics and alternations <strong>in</strong> these RNAs, especiallymicroRNAs (miRNAs), contribute to numerous diseases. miRNAs can <strong>in</strong>hibit translationof mRNA if the miRNA b<strong>in</strong>ds to the mRNA, a process that leads to its degradation, or themiRNA may partially b<strong>in</strong>d to the 3 0 end of the mRNA and prohibit the actions of transfer RNA[5]. Although miRNAs have been associated with a number of diseases such as Crohn’s disease[6], their role <strong>in</strong> tumorigenesis is now established and is considered to be a frequent epigeneticaberration <strong>in</strong> cancer.Collectively, epigenetic processes are now generally accepted to play a key role <strong>in</strong> humandiseases. As the knowledge of epigenetic mechanisms <strong>in</strong> human diseases expands, it isexpected that approaches to disease prevention and therapy us<strong>in</strong>g epigenetic <strong>in</strong>terventions willalso cont<strong>in</strong>ue to develop and may eventually become ma<strong>in</strong>stays <strong>in</strong> disease management.21.2 EPIGENETIC VARIATION METHODSTechnological advances often serve as a major stimulus for knowledge development and thefield of epigenetics is no exception <strong>in</strong> this regard. Recent advances <strong>in</strong> epigenetic-basedmethods have served as major driv<strong>in</strong>g forces <strong>in</strong> the fasc<strong>in</strong>at<strong>in</strong>g and ever-expand<strong>in</strong>g epigeneticphenomena that have been revealed especially over the past decade. Although genome-widemaps have been developed, there is still a need for maps of the human methylome andhistone modifications <strong>in</strong> healthy and diseased tissues, as discussed <strong>in</strong> Chapter 2. Epigeneticvariation is especially prom<strong>in</strong>ent <strong>in</strong> human diseases and established techniques such asbisulfite genomic methylation sequenc<strong>in</strong>g and chromat<strong>in</strong> immunoprecipitation (ChIP)analyses are reveal<strong>in</strong>g numerous epigenetic aberrations <strong>in</strong>volved <strong>in</strong> disease processes.However, cutt<strong>in</strong>g-edge advances <strong>in</strong> comparative genomic hybridization (CGH) and microarrayanalyses as well as quantitative analysis of methylated alleles (QAMA) and many otherdevelop<strong>in</strong>g technologies are now facilitat<strong>in</strong>g the elucidation of epigenetic alterations <strong>in</strong>disease that were previously unimag<strong>in</strong>ed. Comb<strong>in</strong>ations of epigenetic technologies are alsoemerg<strong>in</strong>g that show promise <strong>in</strong> lead<strong>in</strong>g to new advances <strong>in</strong> understand<strong>in</strong>g the epigenetics ofdisease.1.3 CANCER EPIGENETICSAs mentioned above, DNA methylation is often an important factor <strong>in</strong> cancer developmentand progression. DNA methylation changes can now be readily assessed from body fluids andapplied to cancer diagnosis as well as the prognosis of cancer (Chapter 3). Epigenome referencemaps will likely have an impact on our understand<strong>in</strong>g of many different diseases and maylead the way to breakthroughs <strong>in</strong> the diagnosis, prevention and therapy of human cancers.Histone modifications are frequently altered <strong>in</strong> many human cancers and the development ofa histone modification signature may be developed that will aid <strong>in</strong> the prognosis and treatmentof cancers (Chapter 4). These histone maps may also have potential <strong>in</strong> guid<strong>in</strong>g therapy ofhuman cancers. MicroRNAs (miRNAs) are central to many cellular functions and they arefrequently dysregulated dur<strong>in</strong>g oncogenesis (Chapter 5). In fact, miRNA expression profiles


CHAPTER 1<strong>Epigenetics</strong> of <strong>Human</strong> <strong>Disease</strong>may be more useful than gene expression profiles for cl<strong>in</strong>ical applications s<strong>in</strong>ce there are fewermRNA regulatory molecules. These miRNA profiles may be applicable to identify<strong>in</strong>g variouscancers or to stratify tumors <strong>in</strong> addition to serv<strong>in</strong>g prognostic or therapeutic roles. Epigenetictherapy for cancer is perhaps one of the most excit<strong>in</strong>g and rapidly develop<strong>in</strong>g areas ofepigenetics. As discussed <strong>in</strong> Chapter 6, approaches are available for target<strong>in</strong>g enzymes such asthe DNMTs, histone acetyltransferases (HATs), histone deacetylases (HDACs), histone methyltransferases(HMTs) and histone demethylases (HDMTs). The development of drug-based<strong>in</strong>hibitors of these epigenetic-modify<strong>in</strong>g enzymes could be further improved through drugcomb<strong>in</strong>ations or even natural plant-based products, many of which have been found to harborproperties that can mimic the often more toxic and perhaps less bioavailable epigenetic drugsthat are currently <strong>in</strong> use.1.4 EPIGENETICS OF NEUROLOGICAL DISEASEOne of the newer areas of epigenetics that has been rapidly expand<strong>in</strong>g is its role <strong>in</strong> neurologicaldisorders or diseases. These disorders are not limited to the bra<strong>in</strong> as the disease target,but also often <strong>in</strong>volve nutritional and metabolic factors that contribute as well to conditionssuch as neurobehavioral diseases (Chapter 7). At this po<strong>in</strong>t, however, the number ofneurodevelopmental disorders that have been associated with epigenetic aberrations is notvery extensive. A possible explanation for this is that the pervasive nature of epigeneticprocesses could serve as a negative selective force aga<strong>in</strong>st more localized disease such asneurodevelopmental disorders (Chapter 8). In fact, many neurodevelopmental disorders aredue to partial loss-of-function mutations or are X-chromosomal mosaics with recessiveX-l<strong>in</strong>ked mutations. Neurodegenerative diseases such as Alzheimer’s disease have been<strong>in</strong>creas<strong>in</strong>gly associated with alternations <strong>in</strong> epigenetic processes. Environmental factors suchas diet and exposure to heavy metals may lead to the epigenetic changes often <strong>in</strong>volved <strong>in</strong>Alzheimer’s disease eventually contribut<strong>in</strong>g to <strong>in</strong>creased amyloid b peptide (Chapter 9).These factors may beg<strong>in</strong> early <strong>in</strong> life and manifest as late-onset forms of Alzheimer’s disease.Fortunately, as reviewed <strong>in</strong> Chapter 10, a number of new approaches are currently be<strong>in</strong>gdeveloped that could have translational potential <strong>in</strong> prevent<strong>in</strong>g or treat<strong>in</strong>g many of theepigenetic changes that are be<strong>in</strong>g revealed as an important component of neurobiologicaldisorders.31.5 AUTOIMMUNITY AND EPIGENETICSThere is a strong association between environmental factors, age and the development ofautoimmune disorders. Epigenetic processes are central to ag<strong>in</strong>g and are also an importantmediator between the environment and disease and it is thought that these factors may beimportant <strong>in</strong> the development and progression of numerous autoimmune diseases. Forexample, systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA) are autoimmunedisorders that have frequently been associated with aberrations <strong>in</strong> epigenetic mechanisms(Chapter 11). Often the epigenomic and sequence-specific DNA methylation changes found <strong>in</strong>SLE and RA affect key genes <strong>in</strong> immune function. Two challenges are to <strong>in</strong>crease the use ofhigh-throughput approaches to these diseases to m<strong>in</strong>e for additional gene aberrations and totranslate these epigenetic changes to the cl<strong>in</strong>ic through the development of novel approachesfor prevent<strong>in</strong>g or treat<strong>in</strong>g SLE and RA. Fortunately, there is hope for epigenetic therapy ofautoimmune disorders as reviewed <strong>in</strong> Chapter 12. Much of the current research for drugdevelopment relevant to autoimmune dysfunction is focused on correct<strong>in</strong>g alterations <strong>in</strong> DNAmethylation and histone acetylation. However, recent excit<strong>in</strong>g advances suggest promis<strong>in</strong>gavenues for drug development as applied to miRNAs. For <strong>in</strong>stance, miRNAs or <strong>in</strong>hibitors ofmiRNA to impact DNA methylation may have utility <strong>in</strong> affect<strong>in</strong>g gene transcription <strong>in</strong> immunecells that often lead to the development of SLE.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>1.6 HUMAN IMPRINTING DISORDERSBoth DNA methylation and histone modifications can impact impr<strong>in</strong>t<strong>in</strong>g centers that controlparent-of-orig<strong>in</strong>-specific expression and lead to human impr<strong>in</strong>t<strong>in</strong>g disorders. These disorders,such as Angelman, PradereWilli, SilvereRussell and BeckwitheWiedemann syndromesfrequently <strong>in</strong>volve epigenetic changes that contribute to these disorders and they oftenmanifest at a very young age (Chapter 13). However, both epigenetic and genetic factors areoften important <strong>in</strong> human impr<strong>in</strong>t<strong>in</strong>g disorders and the development of epigenetic therapyapproaches <strong>in</strong> this particular area represents a considerable challenge. Advances are be<strong>in</strong>gmade <strong>in</strong> understand<strong>in</strong>g the epigenetic basis of human impr<strong>in</strong>t<strong>in</strong>g disorders which mayprovide breakthroughs <strong>in</strong> treat<strong>in</strong>g these tragic diseases.1.7 EPIGENETICS OF OBESITYRare obesity-associated impr<strong>in</strong>t<strong>in</strong>g disorders have been described and dietary modulationefforts have suggested an epigenetic component may exist <strong>in</strong> these disorders. In fact, the majorrole of environmental factors <strong>in</strong> obesity strongly suggests a role of epigenetic changes such asthose <strong>in</strong>volv<strong>in</strong>g DNA methylation <strong>in</strong> obesity (Chapter 14). Early-life environmental factorscould be especially important <strong>in</strong> controll<strong>in</strong>g epigenetic aberrations that may contribute toobesity as reviewed <strong>in</strong> Chapter 15. It is likely that <strong>in</strong>creased identification of obesitybiomarkers and their associated epigenetic factors may lead to new advances <strong>in</strong> controll<strong>in</strong>g theextant epidemic <strong>in</strong> childhood obesity <strong>in</strong> many developed countries. It is highly likely thatnutritional or lifestyle <strong>in</strong>terventions either dur<strong>in</strong>g pregnancy or early <strong>in</strong> life could impactprocesses such as DNA methylation and histone modifications that are highly responsive toenvironmental stimuli and lead to means to control obesity at very early ages.41.8 DIABETES: THE EPIGENETIC CONNECTIONSimilar to obesity, environmental factors are also often important <strong>in</strong> the development of type2 diabetes. Non-genetic risk factors such as ag<strong>in</strong>g and a sedentary lifestyle have been associatedwith epigenetic aberrations characteristic of type 2 diabetes (Chapter 16). S<strong>in</strong>ce markers suchas DNA methylation have been shown to vary <strong>in</strong> diabetic versus non-diabetic <strong>in</strong>dividuals, it isvery possible that epigenetic manifestations may have a key role <strong>in</strong> the pathogenesis of type2 diabetes. However, multisystem studies are currently needed to further substantiate thisconcept and additional studies on the prediction and prevention of type 2 diabetes are sorelyneeded. Histone modifications have also been strongly implicated <strong>in</strong> diabetes as reviewed <strong>in</strong>Chapter 17. In fact, HDAC <strong>in</strong>hibitors may have potential <strong>in</strong> treat<strong>in</strong>g diabetes <strong>in</strong> the short term.Nutritional compounds that lead to HDAC <strong>in</strong>hibition may have potential <strong>in</strong> treat<strong>in</strong>g type2 diabetes as well as the development of miRNA-based therapeutics that would have greatertarget<strong>in</strong>g potential.1.9 EPIGENETICS AND ALLERGIC DISORDERSConsistent with many other epigenetic diseases, early environmental factors appear to bea critical component to the development of numerous allergic disorders. For example, exposureto specific factors <strong>in</strong> utero may be associated with epigenetic aberrations that affect geneexpression, immune programm<strong>in</strong>g and the development of allergic maladies <strong>in</strong> the offspr<strong>in</strong>g(Chapter 18). Additionally, this transgenerational component may allow for the transmittanceof epigenetic changes to future generations beyond the offspr<strong>in</strong>g lead<strong>in</strong>g to allergic disorders.Novel early <strong>in</strong>terventions <strong>in</strong>to epigenetic-modify<strong>in</strong>g factors such as maternal diet maycontribute to an eventual decl<strong>in</strong>e <strong>in</strong> allergy-based disorders. Asthma is a common disorder ofthis nature and there is some evidence that corticosteroids exert their anti-<strong>in</strong>flammatory effects<strong>in</strong> part by <strong>in</strong>duc<strong>in</strong>g acetylation of anti-<strong>in</strong>flammatory genes (Chapter 19). The potentialrecruitment of HDAC2 to activated <strong>in</strong>flammatory genes by corticosteroids may be a key


CHAPTER 1<strong>Epigenetics</strong> of <strong>Human</strong> <strong>Disease</strong>mechanism for epigenetic-based therapy of allergic disorders such as asthma. Future efforts arenow be<strong>in</strong>g directed toward modifiers of other epigenetic processes <strong>in</strong> allergic disorders such ashistone phosphorylation and ubiquit<strong>in</strong>ation.1.10 CARDIOVASCULAR DISEASE AND EPIGENETICSAtherosclerosis is a major precipitat<strong>in</strong>g factor <strong>in</strong> cardiovascular diseases and the functions ofsmooth muscle cells (SMCs) and endothelial cells (ECs) are central to the development ofatherosclerosis. Mount<strong>in</strong>g evidence has <strong>in</strong>dicated that epigenetic processes such as DNAmethylation and histone acetylation have critical functions <strong>in</strong> modulat<strong>in</strong>g SMC and EChomeostasis. The SMC and EC proliferation, migration, apoptosis and differentiation not onlycontribute to atherosclerosis, but also cardiomyocyte hypertrophy and heart failure, asreviewed <strong>in</strong> Chapter 20. The role of HDACs <strong>in</strong> cardiovascular disease such as arteriosclerosishas been show<strong>in</strong>g promise, although concerns surround the tissue-specificity of these agents.Given this concern, the development of highly selective and cell type-specific HDAC <strong>in</strong>hibitorsmay have potential <strong>in</strong> epigenetic-based therapies for cardiovascular diseases of varied types.1.11 EPIGENETICS OF HUMAN INFECTIOUS DISEASESA common theme is the environmental impact on the epigenome and its role <strong>in</strong> epigeneticdisease processes. Consistent with this concept, bacterial and viral <strong>in</strong>fections often causeepigenetic changes <strong>in</strong> host cells that lead to pathology as reviewed <strong>in</strong> Chapter 21. Theconsequences of these epigenome-modify<strong>in</strong>g <strong>in</strong>fections are not limited to neoplasia. There are,<strong>in</strong> fact, many other diseases that have an epigenetic basis <strong>in</strong>duced by <strong>in</strong>fectious agents such asdiseases of the oral cavity. Even organisms like protozoa can contribute to host epigeneticdysregulation. Knowledge accumulated regard<strong>in</strong>g epigenetic “<strong>in</strong>vaders” of the genome andtheir pathological consequences will undoubtedly lead to the development of more sophisticatedand novel approaches to controll<strong>in</strong>g and treat<strong>in</strong>g epigenetic-based <strong>in</strong>fectious diseases.51.12 REPRODUCTIVE DISORDERS AND EPIGENETICABERRATIONSEndometriosis, or the presence of functional endometrial-like tissues outside of the uter<strong>in</strong>ecavity, is often secondary to hormonal and immunological aberrations. Most excit<strong>in</strong>g <strong>in</strong> thecontext of epigenetics, however, is that many recent studies have <strong>in</strong>dicated thatendometriosis may have an important epigenetic component that contributes to its pathologicalprogression (Chapter 22). A number of <strong>in</strong>vestigations have <strong>in</strong>dicated HDAC <strong>in</strong>hibitorsmay be effective <strong>in</strong> treat<strong>in</strong>g endometriosis. There is also potential for the development ofepigenetic biomarkers for endometriosis such as changes <strong>in</strong> DNA methylation as well asmiRNA-based biomarkers. Epigenetic processes are also ga<strong>in</strong><strong>in</strong>g <strong>in</strong>creas<strong>in</strong>g importance <strong>in</strong>endometrial cancer (Chapter 23). Damage to the mismatch repair system appears to playa significant role <strong>in</strong> the development of endometrial cancer through the mechanism of hMLH1hypermethylation. These f<strong>in</strong>d<strong>in</strong>gs may have important epigenetic therapeutic implications forendometrial cancer and could also have potential for the prevention, diagnosis and riskassessment of endometrial cancer.1.13 STEM CELL EPIGENETICS IN HUMAN DISEASEStem cell-based therapeutic approaches could lead to powerful means of treat<strong>in</strong>g humandiseases and epigenetic regulatory signals play an important role <strong>in</strong> the ma<strong>in</strong>tenance of stemcell potency (Chapter 24). Chromat<strong>in</strong> modifications and dynamics appear to have animportant role <strong>in</strong> conservation of pluripotency and the differentiation of embryonic stem cellswhich are central factors <strong>in</strong> stem cell-based therapeutics. In fact, several epigenetic disordershave been modeled <strong>in</strong> vitro through the use of <strong>in</strong>duced pluripotent stem cells (iPSCs) from the


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>cells of patients. Understand<strong>in</strong>g the basic epigenetic changes central to these processes mayhave considerable potential <strong>in</strong> the treatment of human epigenetic diseases. Non-cod<strong>in</strong>g RNAsalso participate <strong>in</strong> stem cell renewal and differentiation (Chapter 25). The role of epigeneticsand non-cod<strong>in</strong>g RNAs may provide many useful tools for manipulat<strong>in</strong>g stem cell programm<strong>in</strong>gas applied to therapy of epigenetic-based diseases.1.14 EPIGENETICS OF AGING AND AGE-ASSOCIATED DISEASESFew processes are as pervasive as ag<strong>in</strong>g which impacts not only the entire physiological fitnessof an organism, but also its predisposition to develop<strong>in</strong>g age-related diseases which iscomprised of an ever-grow<strong>in</strong>g list of diseases. It is now apparent that epigenetic processes aremajor components of ag<strong>in</strong>g, which opens many avenues to human diseases (Chapter 26).Although ag<strong>in</strong>g is not considered a disease <strong>in</strong> and of itself, it is perhaps the most frequentcontributor to human disease. Therefore, delay<strong>in</strong>g the epigenetic aberrations associated withag<strong>in</strong>g through epigenetic <strong>in</strong>tervention and treat<strong>in</strong>g epigenetic-based age-associated diseasescould have a tremendous impact on the role of epigenetics <strong>in</strong> human disease. Although theyare on opposite sides of the lifespan spectrum, early developmental processes are likely l<strong>in</strong>kedto later life ag<strong>in</strong>g and age-associated diseases (Chapter 27). The role of nutrition, hormonesand metabolic environment early <strong>in</strong> life can have effects throughout life, <strong>in</strong>fluence epigeneticpathways and markers and manifest <strong>in</strong> the form of ag<strong>in</strong>g and age-related diseases. Considerable<strong>in</strong>terest is now focused on the impact of early life epigenetic impacts and the outcome ofthese effects on the myriad of age-associated diseases which comprise much of the pathologythat forms the basis of human disease.61.15 CONCLUSIONEpigenetic processes not only take many forms, but they also can readily become expressed ashuman diseases. These diseases, that can be loosely grouped under the head<strong>in</strong>g of “epigeneticdiseases”, are vast and the list of diseases that fit <strong>in</strong>to this description is rapidly grow<strong>in</strong>g.Elucidation of the epigenetic aberrations <strong>in</strong> human diseases not only has implications forepigenetic-based therapy, but also for risk assessment, prevention, progression analysis,prognosis and biomarker development. A common theme of many epigenetic-based humandiseases is the role of the environment. This may take varied forms, rang<strong>in</strong>g from maternalnutrition to <strong>in</strong>fectious agents. Excit<strong>in</strong>g advances are rapidly develop<strong>in</strong>g that are contribut<strong>in</strong>gsignificantly toward the management of human diseases through epigenetic <strong>in</strong>tervention. It isanticipated that epigenetic-based preventive and therapeutic strategies will cont<strong>in</strong>ue todevelop at a rapid pace and may assume a role at the forefront of medic<strong>in</strong>e <strong>in</strong> the not toodistant future.References[1] Falls JG, Pulford DJ, Wylie AA, Jirtle RL. Genomic impr<strong>in</strong>t<strong>in</strong>g: implications for human disease. Am J Pathol1999;154:635e47.[2] Yasui DH, Peddada S, Bieda MC, Vallero RO, Hogart A, Nagarajan RP, et al. Integrated epigenomic analyses ofneuronal MeCP2 reveal a role for long-range <strong>in</strong>teraction with active genes. Proc Natl Acad Sci USA2007;104:19416e21.[3] Cavalli G, Paro R. The Drosophila Fab-7 chromosomal element conveys epigenetic <strong>in</strong>heritance dur<strong>in</strong>g mitosisand meiosis. Cell 1998;93:505e18.[4] Esteller M. Cancer epigenomics: DNA methylomes and histone-modification maps. Nat Rev Genet2007;8:286e98.[5] Gibney ER, Nolan CM. <strong>Epigenetics</strong> and gene expression. Heredity (Ed<strong>in</strong>b) 2010;105:4e13.[6] Wu F, Zhang S, Dassopoulos T, Harris ML, Bayless TM, Meltzer SJ, et al. Identification of microRNAs associatedwith ileal and colonic Crohn’s disease. Inflamm Bowel Dis 2010;16:1729e38.


CHAPTER 2Methods and Strategiesto Determ<strong>in</strong>e EpigeneticVariation <strong>in</strong> <strong>Human</strong><strong>Disease</strong>Yoshihisa Watanabe, Masato MaekawaHamamatsu University School of Medic<strong>in</strong>e, Hamamatsu, JapanCHAPTER OUTLINE2.1 Introduction 82.2 DNA Methylation Analysis 82.2.1 Methylation-SensitiveRestriction Enzymes 92.2.2 Bisulfite Conversion ofUnmethylated Cytos<strong>in</strong>es,PCR and Sequenc<strong>in</strong>g 92.2.3 Comparative GenomicHybridization (CGH) andMicroarray Analysis 92.2.4 Bisulfite Treatment and PCRS<strong>in</strong>gle-Strand ConformationPolymorphism (SSCP)(BiPS) 92.2.5 Methylation-Sensitive S<strong>in</strong>gle-Nucleotide PrimerExtension 92.2.6 Comb<strong>in</strong>ed Bisulfite andRestriction Analysis 102.2.7 Quantitative BisulfiteSequenc<strong>in</strong>g us<strong>in</strong>gPyrosequenc<strong>in</strong>gTechnology 102.2.8 MethyLight Technology 102.2.9 Quantitative Analysis ofMethylated Alleles(QAMA) 102.2.10 DNA Methylation Analysis byPyrosequenc<strong>in</strong>g 102.2.11 Matrix-Assisted LaserDesorption Ionization Timeof-FlightMassSpectrometry 102.2.12 New Technologies 112.2.13 Computational Tools 112.3 Histone ModificationAnalysis 112.4 Non-Cod<strong>in</strong>g RNA Analysis:MicroRNA 122.5 Analysis of Genome DNAReplication Program Based onDNA Replication Tim<strong>in</strong>g 142.6 Strategy for EpigenomicInvestigation Based onChromosomal BandStructures 152.7 Overview of Recent Epigeneticgenome-Wide or Bio<strong>in</strong>formaticStudies and Strategies 192.8 General Overview and FuturePerspective 22References 227T. Tollefsbol (Ed): <strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>. DOI: 10.1016/B978-0-12-388415-2.00002-0Copyright Ó 2012 Elsevier Inc. All rights reserved.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>2.1 INTRODUCTION<strong>Epigenetics</strong> is not only one of the most rapidly expand<strong>in</strong>g fields of study <strong>in</strong> biomedicalresearch but is also one of the most excit<strong>in</strong>g and promis<strong>in</strong>g <strong>in</strong> terms of <strong>in</strong>creas<strong>in</strong>g ourunderstand<strong>in</strong>g of disease etiologies and of develop<strong>in</strong>g new treatment strategies. Among therecent landmark events <strong>in</strong> this field are the characterization of the human DNA methylome ats<strong>in</strong>gle nucleotide resolution, the discovery of CpG island shores, the identification of newhistone variants and modifications, and development of genome-wide maps of nucleosomepositions. Much of our <strong>in</strong>creased understand<strong>in</strong>g is the result of technological breakthroughsthat have made it feasible to undertake large-scale epigenomic studies. These new methodologieshave enabled ever f<strong>in</strong>er mapp<strong>in</strong>g of the epigenetic marks, such as DNA methylation,histone modifications and nucleosome position<strong>in</strong>g, that are critical for regulat<strong>in</strong>g theexpression of both genes and noncod<strong>in</strong>g RNAs [1]. In turn, we have a grow<strong>in</strong>g understand<strong>in</strong>gof the consequences of aberrant patterns of epigenetic marks and of mutations <strong>in</strong> theepigenetic mach<strong>in</strong>ery <strong>in</strong> the etiology of disease.8However, there are several aspects of the methods used to analyze epigenetic variation associatedwith disease that present potential problems. First, the tissue used to obta<strong>in</strong> the DNA. Thisdepends to some extent on the nature of the disease, and can <strong>in</strong>fluence the analytical methodsthat are employed. For example, the DNA of some tissues may have a low <strong>in</strong>cidence of moietieswith the diagnostic pattern of methylation, which would limit the choice of analytic methodologiesto those with high sensitivity for these molecular signatures. Second, differentdiseases may require analysis of either regional or genome-wide epigenetic variation, with thechoice depend<strong>in</strong>g on the predicted variation <strong>in</strong> the specific disease. The cont<strong>in</strong>u<strong>in</strong>g <strong>in</strong>crease <strong>in</strong>the number of “epigenetic” diseases means that the list of methods that are practical for thedifferent diseases is also <strong>in</strong>creas<strong>in</strong>g. Third, epigenetic variation can be a consequence or a causeof the disease. Therefore, use of strategies that can differentiate the role, or otherwise, ofepigenetic variation <strong>in</strong> the causality of a disease is fundamental. It might, for example, allowdeterm<strong>in</strong>ation of whether epigenetic variation is a marker of disease progression, a potentialtherapeutic target, or a useful marker for assess<strong>in</strong>g the efficiency of a therapy.Although the new technologies have provided considerable <strong>in</strong>sights <strong>in</strong>to epigenetic aspects ofdisease, there is still considerably more work that needs to be carried out. In particular, there isa great need for detailed descriptions of human DNA methylomes and for maps of histonemodifications and nucleosome positions <strong>in</strong> healthy and diseased tissues. A number of<strong>in</strong>ternational projects and <strong>in</strong>itiatives have been established to meet this need: the NIHRoadmap Epigenomics Program, the ENCODE Project, the AHEAD Project, and theEpigenomics NCBI browser, among others [2,3]. The availability of detailed epigenetic mapswill be of enormous value to basic and applied research and will enable pharmacologicalresearch to focus on the most promis<strong>in</strong>g epigenetic targets.This chapter summarizes some of the contemporary methods used to study epigenetics andhighlights new methods and strategies that have considerable potential for future epigeneticand epigenomic studies.2.2 DNA METHYLATION ANALYSISMethylation of cytos<strong>in</strong>e bases <strong>in</strong> DNA is not only an important epigenetic modification of thegenome but is also crucial to the regulation of many cellular processes. DNA methylation isimportant <strong>in</strong> many eukaryotes for both normal biology and disease etiology [1]. Therefore,identify<strong>in</strong>g which genomic sites DNA are methylated and determ<strong>in</strong><strong>in</strong>g how this epigeneticmark is ma<strong>in</strong>ta<strong>in</strong>ed or lost is vital to our understand<strong>in</strong>g of epigenetics. In recent years, thetechnology used for DNA methylation analysis has progressed substantially: previously,analyses were essentially limited to specific loci, but now, they can be performed on a genomewidescale to characterize the entire "methylome" with s<strong>in</strong>gle-base-pair resolution [4].


CHAPTER 2Methods and Strategies to Determ<strong>in</strong>e Epigenetic Variation <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>The new wealth of profil<strong>in</strong>g techniques raises the challenge of which is the most appropriate toselect for a given experimental purpose. Here, we list different methodologies available foranalyz<strong>in</strong>g DNA methylation and briefly compare their relative strengths and limitations [5].We also discuss important considerations for data analysis.2.2.1 Methylation-Sensitive Restriction EnzymesThe identification of DNA methylation sites us<strong>in</strong>g methylation-sensitive restriction enzymesrequires high-molecular-weight DNA and is limited by the target sequence of the chosenenzyme. The use of restriction enzymes that are sensitive to CpG methylation with<strong>in</strong> theircleavage recognition sites [6] is a relatively low-resolution method, but it can be useful whencomb<strong>in</strong>ed with genomic microarrays [7,8].2.2.2 Bisulfite Conversion of Unmethylated Cytos<strong>in</strong>es, PCR andSequenc<strong>in</strong>gConversion of unmethylated sequences with bisulfite followed by PCR amplification andsequenc<strong>in</strong>g analyses provides an unbiased and sensitive alternative to the use of restrictionenzymes. This approach is therefore generally regarded as the “gold-standard technology” fordetection of 5-methyl cytos<strong>in</strong>e as it enables mapp<strong>in</strong>g of methylated sites at s<strong>in</strong>gle-base-pairresolution [9]. The bisulfite method requires a prolonged <strong>in</strong>cubation of the DNA sample withsodium bisulfite; dur<strong>in</strong>g this period, unmethylated cytos<strong>in</strong>es <strong>in</strong> the s<strong>in</strong>gle-stranded DNA aredeam<strong>in</strong>ated to uracil. However, the modified nucleoside 5-methyl cytos<strong>in</strong>e is immune totransformation and, therefore, any cytos<strong>in</strong>es that rema<strong>in</strong> follow<strong>in</strong>g bisulfite treatment musthave been methylated. This method is currently one of the most popular approaches tomethylation analysis and yields reliable, high-quality data [9,10]. The drawback to the methodis that it is labor-<strong>in</strong>tensive and is not suitable for screen<strong>in</strong>g large numbers of samples.2.2.3 Comparative Genomic Hybridization (CGH) and MicroarrayAnalysisA comb<strong>in</strong>ation of CGH and microarray analysis can overcome the limitations of the bisulfitemethod. This comb<strong>in</strong>ation can enable high-throughput methylation analyses. The variousadvantages and disadvantages of this approach have been reviewed previously [11e13]. Recenthigh-throughput studies have used prote<strong>in</strong> aff<strong>in</strong>ity to enrich for methylated sequences andthen exploited these sequences as probes <strong>in</strong> genomic microarrays. Methylated DNA fragmentscan be aff<strong>in</strong>ity-purified either with an anti-5-methyl cytos<strong>in</strong>e antibody or by us<strong>in</strong>g the DNAb<strong>in</strong>d<strong>in</strong>gdoma<strong>in</strong> of a methyl-CpG-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> [14,15].92.2.4 Bisulfite Treatment and PCR S<strong>in</strong>gle-Strand ConformationPolymorphism (SSCP) (BiPS)The comb<strong>in</strong>ation of bisulfite treatment with PCR-based s<strong>in</strong>gle-strand DNA conformationpolymorphism (SSCP) analysis offers a potentially quantitative assay for methylation [16].This comb<strong>in</strong>ation approach, sometimes referred to as BiPS analysis, can be used for the rapididentification of the methylation status of multiple samples, for the quantification ofmethylation differences, and for the detection of methylation heterogeneity <strong>in</strong> amplified DNAfragments. This technique has been successfully used to <strong>in</strong>vestigate the methylation status ofthe promoter region of the hMLH1, p16, and HIC1 genes <strong>in</strong> several cancer cell l<strong>in</strong>es andcolorectal cancer tissues [17].2.2.5 Methylation-Sensitive S<strong>in</strong>gle-Nucleotide Primer ExtensionMethylation-sensitive s<strong>in</strong>gle-nucleotide primer extension (MS-SNuPE) is a technique that canbe used for rapid quantitation of methylation at <strong>in</strong>dividual CpG sites [18,19]. Treatment ofgenomic DNA with sodium bisulfite is used to convert unmethylated cytos<strong>in</strong>e to uracil while


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>leav<strong>in</strong>g 5-methylcytos<strong>in</strong>e unaltered. Strand-specific PCR is performed to generate a DNAtemplate for quantitative methylation analysis us<strong>in</strong>g MS-SNuPE. This protocol can be carriedus<strong>in</strong>g multiplex reactions, thus enabl<strong>in</strong>g the simultaneous quantification of multiple CpG sites<strong>in</strong> each assay.2.2.6 Comb<strong>in</strong>ed Bisulfite and Restriction AnalysisThe comb<strong>in</strong>ed bisulfite and restriction analysis (COBRA) approach <strong>in</strong>volves comb<strong>in</strong><strong>in</strong>g thebisulfite and restriction analysis protocols [20]. It is relatively simple to use while still reta<strong>in</strong><strong>in</strong>gquantitative accuracy. Although both COBRA and MS-SNuPE are quantitative, they have therestrictions that the former can only analyze a specific sequence because it utilizes restrictionenzymes and the latter is somewhat laborious. MS-SnuPE has also been comb<strong>in</strong>ed withmicroarray analysis to allow parallel detection of DNA methylation <strong>in</strong> cancer cells [19].2.2.7 Quantitative Bisulfite Sequenc<strong>in</strong>g us<strong>in</strong>g Pyrosequenc<strong>in</strong>gTechnologyQuantitaive bisulfite sequenc<strong>in</strong>g us<strong>in</strong>g pyrosequenc<strong>in</strong>g technology (QBSUPT) is based on thelum<strong>in</strong>ometric detection of pyrophosphate release follow<strong>in</strong>g nucleotide <strong>in</strong>corporation [21].The advantage of QBSUPT is that quantitative DNA methylation data are obta<strong>in</strong>ed directlyfrom PCR products, without the need for clon<strong>in</strong>g and sequenc<strong>in</strong>g a large number of clones.However, QBSUPT cannot be used to analyze haplotype-specific DNA methylation patterns.Thus, while very sensitive, this assay may be more suited to laboratory diagnosis.102.2.8 MethyLight TechnologyMethyLight technology provides a tool for the quantitative analysis of methylated DNAsequences via fluorescence detection <strong>in</strong> PCR-amplified samples [22]. This method has twoparticular advantages: first, the fluorescent probe can be designed to detect specific DNAmethylation patterns, not simply to discrim<strong>in</strong>ate methylated from unmethylated sequences;second, it has the potential ability to rapidly screen hundreds or even thousands of samples.2.2.9 Quantitative Analysis of Methylated Alleles (QAMA)QAMA is a quantitative variation of MethyLight that uses TaqMan probes based on m<strong>in</strong>orgroove b<strong>in</strong>der (MGB) technology [23]. QAMA has the ma<strong>in</strong> advantage of be<strong>in</strong>g simple to setup, mak<strong>in</strong>g it suitable for high-throughput methylation analyses.2.2.10 DNA Methylation Analysis by Pyrosequenc<strong>in</strong>gPyrosequenc<strong>in</strong>g is a replication-based sequenc<strong>in</strong>g method <strong>in</strong> which addition of the correctnucleotide to immobilized template DNA is signaled by a photometrically detectable reaction.This method has been adapted to quantify methylation of CpG sites. The template DNA istreated with bisulfite and PCR is used for sequenc<strong>in</strong>g; the ratio of T and C residues is then usedto quantify methylation. Pyrosequenc<strong>in</strong>g offers a high-resolution and quantitatively accuratemeasurement of methylation of closely positioned CpGs [24].2.2.11 Matrix-Assisted Laser Desorption Ionization Time-of-Flight MassSpectrometryTost et al. [25] described a method us<strong>in</strong>g matrix-assisted laser desorption ionization time-offlight(MALDI-TOF) for analysis and quantification of methylation at CpGs. Although themethod requires gene-specific amplification, and should therefore be considered a candidategene method, it is amenable to automation as it can make use of the EpiTYPER platformdeveloped by Sequenom. EpiTYPER can be used to determ<strong>in</strong>e methylation status follow<strong>in</strong>ggene-specific amplification of bisulfite-treated DNA followed by <strong>in</strong> vitro transcription, basespecificRNA cleavage and MALDI-TOF analysis [26]. Although it is not a genome-wide


CHAPTER 2Methods and Strategies to Determ<strong>in</strong>e Epigenetic Variation <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>technology, it is quantitative for multiple CpG d<strong>in</strong>ucleotides for large numbers of gene lociand can be reliably applied to pooled DNA samples to obta<strong>in</strong> group averages for valuablesamples.2.2.12 New TechnologiesSeveral second-generation sequenc<strong>in</strong>g platforms became available <strong>in</strong> 2007 and were furtherdeveloped with the launch of the first s<strong>in</strong>gle-molecule DNA sequencer (Helicos Biosciences)<strong>in</strong> 2008 [27]. These new sequenc<strong>in</strong>g tools have been applied to epigenetic research, forexample, studies on DNA methylation. Undoubtedly, future developments of these technologieshold the tantaliz<strong>in</strong>g prospect of high-throughput sequenc<strong>in</strong>g to identify DNAmethylation patterns across the whole mammalian genome, possibly even open<strong>in</strong>g up theprospect of genotyp<strong>in</strong>g <strong>in</strong>dividual cancers to aid the application of custom-designed cancertherapies [28].2.2.13 Computational ToolsThe development of computational tools and resources for DNA methylation analysis isaccelerat<strong>in</strong>g rapidly [29]. Sequence-based analyses <strong>in</strong>volve alignment to a reference genome,collaps<strong>in</strong>g of clonal reads, read counts or bisulfite-based analysis [30], and further dataanalysis. Comparison of the relative strengths and weaknesses of the various methods forDNA methylation analysis is hampered by their complexity and diversity. Inevitably, choiceof method is based on pragmatic grounds, for example, the number of samples, the qualityand quantity of DNA samples, the desired coverage of the genome, and the requiredresolution.2.3 HISTONE MODIFICATION ANALYSISHistones are abundant, small basic prote<strong>in</strong>s that associate with the DNA <strong>in</strong> the eukaryoticnucleus to form chromat<strong>in</strong>. The four core histones (H2A, H2B, H3 and H4) can showsubstantial modifications of 20e40 N-term<strong>in</strong>al am<strong>in</strong>o acids that are highly conserved despiteplay<strong>in</strong>g no structural role. The modifications are thought to constitute a histone code by whichthe cell encodes various chromat<strong>in</strong> conformations and controls gene expression states. Theanalysis of these modified histones can be used as a model for the dissection of complexepigenetic modification patterns and for <strong>in</strong>vestigation of their molecular functions. In thissection, we review the techniques that have been used to decipher these complex histonemodification patterns.11Posttranslational modification (PTM) of prote<strong>in</strong>s plays a key role <strong>in</strong> regulat<strong>in</strong>g the biologicalfunction of many polypeptides. Initially, analyses of the modification status were performedus<strong>in</strong>g either a specialized gel system or a radioactive precursor molecule followed by completeprote<strong>in</strong> hydrolysis and identification of the labeled am<strong>in</strong>o acid [31e35]. This approachshowed that histones could be modified <strong>in</strong> vivo by acetylation, methylation or phosphorylation[31,36,37]. As most of the modifications occurred at the N-term<strong>in</strong>us of the histone, it wasfeasible to map the site of some modifications us<strong>in</strong>g Edman degradation [38]. However, this isonly possible when histones can be purified <strong>in</strong> sufficient quantities and with a high purity. Thepurification process is labor-<strong>in</strong>tensive and <strong>in</strong>volves multiple steps; this precludes the possibilityof analyz<strong>in</strong>g histone modifications from small numbers of cells or of mapp<strong>in</strong>g posttranslationalmodifications at specific loci.Mass spectrometry is the method of choice for analyz<strong>in</strong>g PTM <strong>in</strong> histones [39e42], as eachmodification adds a def<strong>in</strong>ed mass to the molecule. The high resolution of modern massspectrometers and recent developments <strong>in</strong> soft ionization techniques have facilitated themapp<strong>in</strong>g of posttranslational modifications. As a result, these high-resolution methods haveenabled much faster detection of PTMs and have shown that such modifications are


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>considerably more abundant than expected. The <strong>in</strong>creased complexity of the proteomerevealed by these analyses presents major challenges both for <strong>in</strong>vestigation and for theprocess<strong>in</strong>g of the raw data. The mass spectrometry methods currently used to precisely mapa modified residue are very elaborate and require enrichment of the peptides that carryparticular modifications [43e46]. Different molecules can carry several modifications thatlocalize on a s<strong>in</strong>gle peptide with<strong>in</strong> a protease digest [47e50]. These short stretches of densemodifications have been termed eukaryotic l<strong>in</strong>ear motifs (ELMs) and are thought to playa critical role <strong>in</strong> regulat<strong>in</strong>g the global function of prote<strong>in</strong>s [51]. The high level of sequenceconservation with<strong>in</strong> these short ELMs also supports this idea. Many ELMs conta<strong>in</strong> a numberof am<strong>in</strong>o acids that can be modified and the position of each modification has to be preciselydeterm<strong>in</strong>ed [51]. Identification of each modification at different sites with<strong>in</strong> a highlymodified ELM is laborious and also hampered by the fact that some modifications result <strong>in</strong>similar mass differences.A variety of different methods are available to study complex histone modification patterns;these range from “bottom-up approaches” to produce detailed and quantitative measurementsof particular histone modifications, to “top-down approaches” aimed at elucidat<strong>in</strong>g the<strong>in</strong>teractions of different modifications [52]. The use of a range of methods should greatlyfacilitate analysis of complex modification patterns and provide a greater <strong>in</strong>sight <strong>in</strong>to thebiological roles of these histone modifications. Many of the methods used to analyze histonemodifications can equally be applied to other types of modified prote<strong>in</strong> that can function as<strong>in</strong>tegrators <strong>in</strong> multiple signal<strong>in</strong>g pathways. The <strong>in</strong>formation on epigenomic analyses,<strong>in</strong>clud<strong>in</strong>g histone modifications us<strong>in</strong>g new technology such as next-generation sequenc<strong>in</strong>g(NGS), is reviewed below.122.4 NON-CODING RNA ANALYSIS: MicroRNAThere is <strong>in</strong>creas<strong>in</strong>g evidence that small non-cod<strong>in</strong>g RNAs, such as microRNA, and long noncod<strong>in</strong>gRNAs, such as l<strong>in</strong>cRNA, can regulate gene expression. Mature microRNAs (miRNAs) arevery small molecules, 19e25 nucleotides (nt), which poses a problem for their quantification.As small RNAs are less efficiently precipitated <strong>in</strong> ethanol, it is necessary to avoid resuspension<strong>in</strong> ethanol when us<strong>in</strong>g the standard Trizol protocol for RNA isolation. On the other hand,miRNAs appear to be more stable than longer RNAs and, consequently, <strong>in</strong> degraded samplesit is still possible to obta<strong>in</strong> readable miRNA expression data. miRNAs have been reportedto have greater stability than mRNAs <strong>in</strong> samples obta<strong>in</strong>ed from tissues which were fixedwith formal<strong>in</strong> and paraff<strong>in</strong> embedded [53e55].However, the <strong>in</strong>tr<strong>in</strong>sic characteristics of miRNAs make production of miRNA expressionprofiles very problematic. For example, mature miRNAs lack common sequence features, suchas a poly-A tail or 5 0 cap, that can be used to drive selective purification. As mentioned above,the mature miRNAs are very small, which reduces the effectiveness of most conventionalbiological amplification methods. This problem arises because of poor specificity <strong>in</strong> primerb<strong>in</strong>d<strong>in</strong>g. As a consequence, standard real-time PCR methods can only be applied to miRNAprecursors. Furthermore, sequence heterogeneity among the miRNAs with respect to GCcontent, results <strong>in</strong> a wide range of optimal melt<strong>in</strong>g temperatures for these nucleic acidduplexes and hampers the simultaneous detection of multiple miRNAs. An additionalproblem for the specificity of miRNA detection arises from the close sequence similarity ofmiRNAs of the same family (mature miRNA, pri-miRNA, and pre-miRNA) and of the targetsequence.Currently, various methodologies have been adapted to detect miRNAs, <strong>in</strong>clud<strong>in</strong>g Northernblot analysis with radiolabeled probes [56,57], microarray-based [58] and PCR-based analyses[59], s<strong>in</strong>gle molecule detection <strong>in</strong> a liquid phase, <strong>in</strong> situ hybridization [60,61] and highthroughputsequenc<strong>in</strong>g [62]. However, all of these methods have <strong>in</strong>herent limitations and the


CHAPTER 2Methods and Strategies to Determ<strong>in</strong>e Epigenetic Variation <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>choice of method for miRNA detection depends ma<strong>in</strong>ly on specific experimental conditions.Ideally, an miRNA profil<strong>in</strong>g method should fulfill the follow<strong>in</strong>g requirements: sufficientsensitivity to allow quantitative analysis of miRNA levels, even with small amounts of start<strong>in</strong>gmaterial; sensitive to s<strong>in</strong>gle-nucleotide differences between miRNAs; highly reproducible;capable of process<strong>in</strong>g many samples at one time; and, easy to perform without the need forexpensive reagents or equipment [63].miRNAs were first identified us<strong>in</strong>g Northern blott<strong>in</strong>g [64e66]. Small RNA molecules can bedetected with a modified version of the standard protocol for Northern blott<strong>in</strong>g <strong>in</strong> which highpercentageurea-acrylamide gels are ma<strong>in</strong>ly used; this modified approach can detect small RNAmolecules that are approximately 100 times smaller than the average cod<strong>in</strong>g RNA. There arethree ma<strong>in</strong> techniques for detect<strong>in</strong>g and quantify<strong>in</strong>g miRNA <strong>in</strong> tissue samples: clon<strong>in</strong>g ofmiRNA; PCR-based detection; and, hybridization with selective probes. Initially, clon<strong>in</strong>g wasthe ma<strong>in</strong> approach used as it offers advantages for discovery of new miRNAs not predictedfrom bio<strong>in</strong>formatic analysis and for sequenc<strong>in</strong>g the miRNAs [65,67,68]. However, clon<strong>in</strong>g isless precise than the other methods for quantify<strong>in</strong>g miRNAs. The PCR-based technique is ableto detect low copy numbers with high sensitivity and specificity of both the precursor andmature form of miRNAs [69]. It is relatively <strong>in</strong>expensive, can be used for cl<strong>in</strong>ical samples, andcan work with m<strong>in</strong>ute amounts of RNA. Various hybridization techniques can be used onmiRNAs, namely, Northern blott<strong>in</strong>g, bead-based flow cytometry, <strong>in</strong> situ hybridization andmicroarray [70,71]. Northern blott<strong>in</strong>g us<strong>in</strong>g radioactive probes is very sensitive; however, it isvery time-consum<strong>in</strong>g, is only practical <strong>in</strong> large cl<strong>in</strong>ical studies for detect<strong>in</strong>g expression ofhundreds of miRNAs, and requires large amounts of total RNA from each sample.Follow<strong>in</strong>g their <strong>in</strong>itial discovery, the number of miRNAs quickly <strong>in</strong>creased and they wereshown to be present <strong>in</strong> all eukaryotic species [66,67,72]. In order to analyze a large number ofmiRNAs <strong>in</strong> many patients, it is essential to have a technique that can simultaneously processmultiple miRNAs us<strong>in</strong>g the relatively small amounts of RNA that can be obta<strong>in</strong>ed from eachpatient. Design<strong>in</strong>g probes for miRNAs is complicated by their short length and their lowabundance. As each miRNA is only 19e25 nt long, the probe is almost exclusively determ<strong>in</strong>edby the sequence of the miRNA itself, which necessitates a different anneal<strong>in</strong>g temperature foreach probe and miRNA <strong>in</strong>teraction.13Microarray technology was developed <strong>in</strong> 1995 and has been applied to miRNA quantification[71,73]. In brief, microarrays are based on multiple hybridizations <strong>in</strong> parallel, us<strong>in</strong>g a glass orquartz support where probes have either been spotted or synthesized by photochemicalsynthesis [74e76]. The ability to <strong>in</strong>clude a high density of spots on an array enables a highnumber of genes to be analyzed simultaneously [76,77]. Three approaches are <strong>in</strong> general usefor detect<strong>in</strong>g nucleic acids such as DNA or RNA on an array platform. The first, which iscommon for custom arrays, uses glass slides and is based on the spott<strong>in</strong>g of unmodifiedoligonucleotides over the slide [78]. The second also uses glass slides and is based on thedeposition of probes on the slide. The dist<strong>in</strong>ction is that the 5 0 term<strong>in</strong>us of the probe is crossl<strong>in</strong>kedto the matrix on the glass. This allows the spott<strong>in</strong>g of a much higher number of probeson these slides. In the third method, probes are photochemically synthesized directly ona quartz surface, allow<strong>in</strong>g the number of probes to rise to millions on a small and compact area[75]. Usually, but not always, the first two methods compare two samples on each slide (oneused as reference) that are sta<strong>in</strong>ed <strong>in</strong> different colors. The third method uses s<strong>in</strong>gle-colorhybridization where each slide is hybridized with only one sample.Most microarrays use DNA oligo spott<strong>in</strong>g, a few use locked nucleic acid (LNA) that may enable<strong>in</strong>creased aff<strong>in</strong>ity between probes and miRNAs, thereby achiev<strong>in</strong>g more uniform conditions ofhybridization with different probes. Ideally, microarray-based detection of miRNAs shouldavoid manipulation of the samples, such as enrichment of low-molecular-weight RNA speciesand amplification of miRNAs. Additionally, it is feasible to develop microarrays able todiscrim<strong>in</strong>ate the two predom<strong>in</strong>ant forms of miRNAs (precursor and mature). The


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>International <strong>Human</strong> Epigenome Consortium (IHEC) recommended that the identity andabundance of all non-cod<strong>in</strong>g RNA species <strong>in</strong> a cell type should be determ<strong>in</strong>ed and suggestedthat this should be accomplished by RNA-seq by next-generation DNA sequenc<strong>in</strong>g afterisolation of large or small RNA species.2.5 ANALYSIS OF GENOME DNA REPLICATION PROGRAM BASEDON DNA REPLICATION TIMINGChromosomal DNA replication is essential for normal cellular division and also has a significantrole <strong>in</strong> the ma<strong>in</strong>tenance of genomic <strong>in</strong>tegrity. Genomic <strong>in</strong>stability <strong>in</strong>creases when DNAreplication errors occur and, thus, mistakes <strong>in</strong> replication may be an important factor <strong>in</strong> theetiology of cancers and neuronal disorders. Replication <strong>in</strong> eukaryotes is <strong>in</strong>itiated from discretegenomic regions, termed orig<strong>in</strong>s. The replication program is strict with<strong>in</strong> a cell or tissue typebut can vary among tissues and dur<strong>in</strong>g development. The genetic program that controlsactivation of replication orig<strong>in</strong>s <strong>in</strong> mammalian cells awaits elucidation. Nevertheless, there isevidence that the specification of replication sites and the tim<strong>in</strong>g of replication are responsiveto epigenetic modifications. Over the last decade, many new techniques have been developedand applied to analysis of DNA replication tim<strong>in</strong>g <strong>in</strong> the human genome. These techniqueshave provided significant <strong>in</strong>sights <strong>in</strong>to cell cycle controls, human chromosome structure, andthe role of epigenetic changes to the genome with respect to DNA replication. In this section,we describe the methods that are currently employed for determ<strong>in</strong><strong>in</strong>g the spatiotemporalregulation of DNA replication <strong>in</strong> the human genome (DNA replication tim<strong>in</strong>g).14Two approaches are generally used to <strong>in</strong>vestigate DNA replication tim<strong>in</strong>g, fluorescence <strong>in</strong> situhybridization (FISH) or PCR [79e82]. The FISH method is based on the cytogeneticdiscrim<strong>in</strong>ation of replicated (two double signals for autosomal loci; DD) and unreplicated loci(two s<strong>in</strong>gle signals; SS) us<strong>in</strong>g DNA probes that are labeled with a fluorescent dye [79]. Bycompar<strong>in</strong>g the frequencies of the two types of signal, the relative replication tim<strong>in</strong>g of eachlocus can be determ<strong>in</strong>ed. However, the method is absolutely dependent on the assumptionthat replicated loci will provide DD-type FISH signals, that is, the replicated signals created bypassage of the replication fork will separate sufficiently to be seen as a DD signal.The PCR-based method <strong>in</strong>volves label<strong>in</strong>g cells <strong>in</strong> exponential growth with BrdU for60e90 m<strong>in</strong> and then fractionat<strong>in</strong>g them by flow cytometry. Typically, this allows discrim<strong>in</strong>ationof six cell cycle fractions: G1, four successive S phase stages, and G2/M (mitotic)[80e82]. Samples conta<strong>in</strong><strong>in</strong>g equal numbers of cells from each cell-cycle fraction are collected,and newly replicated DNA labeled with BrdU is extracted and purified from each fraction.Whether or not a locus has commenced or completed replication can be determ<strong>in</strong>ed byquantitative PCR of the newly replicated DNA. This approach has been exploited to providereplication tim<strong>in</strong>gs for sequence tagged sites on human chromosomes 11q and 21q [81,82]and identified Mb-sized zones that replicated early or late <strong>in</strong> S phase (i.e. early/late transitionzones). The early zones were found to be more GC-rich and gene-rich than the late zones, andthe early/late transitions occurred primarily <strong>in</strong> genome regions that showed rapid switches <strong>in</strong>the relative GC content <strong>in</strong> the chromat<strong>in</strong> [81,82].Woodf<strong>in</strong>e et al. [83] performed the first microarray-based analysis to map replication tim<strong>in</strong>g <strong>in</strong>the human genome. They adapted the comparative genomic hybridization technique, whichhad been developed to assess genomic copy-number differences <strong>in</strong> cancer cells. Relativereplication times can be <strong>in</strong>ferred by measur<strong>in</strong>g the relative amounts of different sequences <strong>in</strong>a population of S-phase cells compared to a non-replicat<strong>in</strong>g G1 genome. In this method,S-phase cells <strong>in</strong> an asynchronously grow<strong>in</strong>g human cell culture are isolated and their DNAextracted. The DNA is color-labeled and then mixed with DNA from G1 phase cells that hasbeen differentially color-labeled. The comb<strong>in</strong>ed DNA sample is hybridized to an array ofgenomic sequences and, after normalization of the data, the relative fluorescence <strong>in</strong>tensities


CHAPTER 2Methods and Strategies to Determ<strong>in</strong>e Epigenetic Variation <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>of the S-phase DNA at each array spot (the S to G1 replication tim<strong>in</strong>g ratio) provide a measureof replication tim<strong>in</strong>g. Comparison of the data obta<strong>in</strong>ed by this method with those fromnascent strand quantitative PCR methods described above [81e84], showed that this newapproach provided estimations of replication tim<strong>in</strong>g that were consistent with those obta<strong>in</strong>edearlier [82,83].White et al. [85] modified the experimental approach of Woodf<strong>in</strong>e et al. [83] by compar<strong>in</strong>g therepresentation of genomic sequences <strong>in</strong> newly replicated DNA isolated from early S-phase cellswith that from late S-phase cells. In this way, they obta<strong>in</strong>ed a replication tim<strong>in</strong>g ratio for early Sto late S. Although their measure has a different basis to the S phase to G1 ratio of Woodf<strong>in</strong>eet al. [83], nevertheless, the results provide a similar description of replication tim<strong>in</strong>g. Forexample, a replication profile for chromosome 22 <strong>in</strong> a lymphoblastoid cell l<strong>in</strong>e obta<strong>in</strong>ed byWhite et al. [85] was consistent with that of Woodf<strong>in</strong>e et al. [83]. To date, high-resolutionanalyses have shown a positive correlation between replication tim<strong>in</strong>g and a range of genomicparameters such as GC content, gene density and transcriptional activity [82,83,85].DNA replication errors have been implicated <strong>in</strong> the etiology of many diseases [86e89]. Onepossible mechanism for this relationship is that disease-related reprogramm<strong>in</strong>g of the epigenomemight depend on impaired regulation of replication tim<strong>in</strong>g patterns [90]. Thus, forexample, chromosomal rearrangements <strong>in</strong> cancers have been reported to be associated withreplication tim<strong>in</strong>g changes <strong>in</strong> translocation breakpo<strong>in</strong>ts [91,92]. Likewise, peripheral bloodcells from prostate cancer patients have an altered pattern of replication accompanied byaneuploidy that dist<strong>in</strong>guishes them from <strong>in</strong>dividuals with benign prostate hyperplasia(a common disorder <strong>in</strong> elderly men). These cellular characteristics have been suggested to bea better marker for prostate cancer than use of the blood marker, prostate-specific antigen(PSA) [93,94]. Analyses of changes <strong>in</strong> replication tim<strong>in</strong>g <strong>in</strong> the human genome have shownthat the tumor suppressor gene p53 plays a role <strong>in</strong> its regulation through the control of cellcycle checkpo<strong>in</strong>ts [95]. Thus, <strong>in</strong> cancer cells, the normal order of DNA replication is altered:regions that normally replicate late sometimes replicate early, and vice versa[84,91e93,96e98]. Replication tim<strong>in</strong>g has also been shown to change dur<strong>in</strong>g development,differentiation and tumorigenesis; moreover, the structure of the chromat<strong>in</strong> may also change.The model illustrated <strong>in</strong> Figure 2.1 shows a possible mode of <strong>in</strong>teraction of chromat<strong>in</strong>conformation, replication tim<strong>in</strong>g and the expression of genes, <strong>in</strong>clud<strong>in</strong>g oncogenes, <strong>in</strong> anearly/late-switch region of replication tim<strong>in</strong>g (R/G-chromosome band boundary) [99]. Forexample, the replication tim<strong>in</strong>g environment of an oncogene (or a tumor suppressor gene)located <strong>in</strong> an early/late-switch region of replication tim<strong>in</strong>g may change from <strong>in</strong>termediatereplication, between early and late S phase, to early replication tim<strong>in</strong>g (or late replicationtim<strong>in</strong>g) by an <strong>in</strong>crease (or decrease) <strong>in</strong> the number of early replication orig<strong>in</strong>s at the edge of anearly replication zone (Figure 2.1B). In addition, the chromat<strong>in</strong> environment of such anoncogene (or tumor suppressor gene) may also change from that of an R/G-chromosomeband boundary to an R band (or from that of an R/G-chromosome band boundary to a Gband). Stall<strong>in</strong>g of the replication fork <strong>in</strong> the vic<strong>in</strong>ity of oncogenes might also <strong>in</strong>duce translocationevents, thereby alter<strong>in</strong>g the structure or the local environment of the oncogenes andaffect<strong>in</strong>g their function (Figures 2.1A, 2.1B) [99]. The <strong>in</strong>terrelationship of these various factorssuggests that analysis of replication tim<strong>in</strong>g assays as part of an epigenetics <strong>in</strong>vestigation might,<strong>in</strong> future, allow much earlier cancer detection than is possible today [5,99,100].152.6 STRATEGY FOR EPIGENOMIC INVESTIGATION BASED ONCHROMOSOMAL BAND STRUCTURESThe various methods for genome-wide epigenetic analyses described <strong>in</strong> the above sections aresummarized <strong>in</strong> Table 2.1. The replication tim<strong>in</strong>g of genes along the entire lengths of humanchromosomes 11q and 21q has been described previously; these analyses showed that cancerrelatedgenes, <strong>in</strong>clud<strong>in</strong>g several oncogenes, are concentrated <strong>in</strong> regions show<strong>in</strong>g transition


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>(A)(B)16FIGURE 2.1(A) Characterization of genetically and epigenetically “unstable” region of human genome. Replication orig<strong>in</strong>s (Ori) generally fire bi-directionally. The transitionzone, which is shown by a thick arrow, is a large orig<strong>in</strong>-free region between early and late-replicat<strong>in</strong>g doma<strong>in</strong>s [134,135]. Only the replication fork that starts atthe edge of the early zone is predicted to be able to cont<strong>in</strong>ue replicat<strong>in</strong>g over a period of hours or to pause at specific sites <strong>in</strong> the replication-transition regionuntil it meets the fork <strong>in</strong>itiated from the adjacent later-replicat<strong>in</strong>g zone. A pause dur<strong>in</strong>g replication is known to <strong>in</strong>crease the risk of DNA breaks andrearrangements [105e107]. Therefore, later replication sites with<strong>in</strong> early/late-switch regions are particularly “unstable” regions of human genome [82]. Thepossible structure of the R/G-chromosome band boundary is shown above the orig<strong>in</strong> map. Typical characteristics of early and late replicat<strong>in</strong>g regions are<strong>in</strong>dicated. R- band, G-band, and R/G-band boundaries are shown <strong>in</strong> white, black, and gray, respectively. We propose that R/G chromosome band boundaries,which are transition regions for replication tim<strong>in</strong>g and GC content, are genetically and epigenetically more “unstable” regions of human genome than othergenomic regions [5,82,84,96e98]. (B) Model for epigenetic changes to genes, <strong>in</strong>clud<strong>in</strong>g oncogenes <strong>in</strong> the R/G-chromosome band boundary. Dur<strong>in</strong>gtumorigenesis, chromat<strong>in</strong> structures as well as replicon structures may change. For example, the replication tim<strong>in</strong>g environment of an oncogene located <strong>in</strong> anearly/late-switch region of replication tim<strong>in</strong>g (R/G-chromosome band boundary) may change from <strong>in</strong>termediate replication, between early and late S phase,to early replication tim<strong>in</strong>g by an <strong>in</strong>crease <strong>in</strong> the number of early replication orig<strong>in</strong>s at the edge of an early replication zone. In addition, the chromat<strong>in</strong>environment of such an oncogene may also change from that of an R/G-chromosome band boundary to an R band. Stall<strong>in</strong>g of the replication fork <strong>in</strong> the vic<strong>in</strong>ityof oncogenes could also <strong>in</strong>duce chromosome translocations that alter the structure or the local environment of the oncogenes, and thereby affect their function.The position of the oncogene is <strong>in</strong>dicated by the black circle. (E), early replication zone; (L), late replication zone; (E/L), early/late-switch region; (R), R-band;(G), G-band; (R/G), R/G-band boundary; (Ori 1), orig<strong>in</strong> 1; (Ori 2), orig<strong>in</strong> 2.


CHAPTER 2Methods and Strategies to Determ<strong>in</strong>e Epigenetic Variation <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>TABLE 2.1 Methods of genome-wide epigenetics analysisMethod of Analysis Techniques MicroarrayTechniquesNext-GenerationSequenc<strong>in</strong>gBisulfite reaction treatment BiMP BC-seq, WGSBSDNA methylationConcentration of methylated MeDIP, mDIP, MIRA MeDIP-seq, MIRA-seqanalysisDNA by immunoprecipitationApplication of methylationsensitiveHELP, MIAMIHELP-seqrestriction enzymeHistone modification Chromat<strong>in</strong>ChIP-on-chipChIP-seqanalysisimmunoprecipitation (ChIP)MicroRNA analysis Extraction of RNA MicroRNA-chip MicroRNA-seqDNA replication tim<strong>in</strong>ganalysisConcentration of newlyreplicated DNA byimmunoprecipitationReplicationTim<strong>in</strong>gchipReplicationTim<strong>in</strong>g-seqBC-seq, bisulfite conversion followed by capture and sequenc<strong>in</strong>g; BiMP, bisulfite methylation profil<strong>in</strong>g; ChIP, chromat<strong>in</strong> immunoprecipitation; -chip, followed bymicroarray; HELP, HpaII t<strong>in</strong>y fragment enrichment by ligation-mediated PCR; MeDIP, mDIP, methylated DNA immunoprecipitation; MIAMI, microarray-based<strong>in</strong>tegrated analysis of methylation by isoschizomers; MIRA, methylated CpG island recovery assay; WGSBS, whole-genome shotgun bisulfite sequenc<strong>in</strong>g.from early to late replication tim<strong>in</strong>g [82,84,96,97]. Scrut<strong>in</strong>y of the updated replication tim<strong>in</strong>gmap for human chromosome 11q found that amplicons, gene amplifications associated withcancer, are located <strong>in</strong> the early/late switch regions of replication tim<strong>in</strong>g <strong>in</strong> human cell l<strong>in</strong>es[84]. These transition regions also conta<strong>in</strong> genes related to neural diseases, such as APPassociated with familial Alzheimer’s disease (AD1), and SOD1 associated with familialamyotrophic lateral sclerosis (ALS1) [82]. Several neural disease genes are present <strong>in</strong>chromosomal regions with early/late transitions [82,96]. Interest<strong>in</strong>gly, <strong>in</strong> metaphase and<strong>in</strong>terphase nuclei, early-replicat<strong>in</strong>g zones have a looser chromat<strong>in</strong> structure, whereas latereplicationzones have compact chromat<strong>in</strong> [101e104]. Therefore, transitions <strong>in</strong> chromat<strong>in</strong>compaction co<strong>in</strong>cide with replication transition regions. In term<strong>in</strong>ally differentiated cells,such as neurons, it is expected that the level of chromat<strong>in</strong> compaction established dur<strong>in</strong>g thef<strong>in</strong>al round of DNA replication will be ma<strong>in</strong>ta<strong>in</strong>ed. Transitions <strong>in</strong> chromat<strong>in</strong> compactionwith<strong>in</strong> a gene might lead to reduced genomic stability, and may also <strong>in</strong>crease susceptibility toagents that can <strong>in</strong>fluence gene expression. Thus, the probability of epimutation, such as<strong>in</strong>stability of chromat<strong>in</strong> structures and DNA damage (<strong>in</strong>clud<strong>in</strong>g DNA rearrangements) appearsto be greater <strong>in</strong> replication transition regions than elsewhere <strong>in</strong> the genome[82,84,96,97,105e107] (Figure 2.1A).17It is likely that transition zones are subject to tight regulation, as chang<strong>in</strong>g their positionswould affect the replication tim<strong>in</strong>g patterns of several flank<strong>in</strong>g replicons. There is strongevidence that transition zones are conserved among different ES cell l<strong>in</strong>es [108]. Dur<strong>in</strong>gdevelopment, transition zones may therefore be targets for chromat<strong>in</strong>-modify<strong>in</strong>g enzymesto facilitate rapid reconfiguration and establishment of new replication tim<strong>in</strong>g patterns.Early and late replication zones tend to be located <strong>in</strong> different regions of the nucleus dur<strong>in</strong>gS phase; it is possible that transition regions flank<strong>in</strong>g these replication zones might besubject to dynamic reorganization or relocation dur<strong>in</strong>g replication fork movement. Thetransition zones for replication tim<strong>in</strong>g are known to be associated with genomic <strong>in</strong>stability,which is suspected to be <strong>in</strong>volved <strong>in</strong> the etiology of human diseases such as cancer.Common fragile sites (CFSs) represent the best-known examples of regions of the humangenome that break under replication stress. CFSs are associated with very large genes [109]and are frequently found at R/G band boundaries [110]. The human genome appears to havea large excess of so-called “dormant” or “backup” orig<strong>in</strong>s and these may be used to rescuestalled replication forks. Interest<strong>in</strong>gly, “spare” orig<strong>in</strong>s appear to be absent from R/G bandboundaries [111,112].


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>(A)(B)18FIGURE 2.2(A) Epimutation-sensitive genomic regions on the human genome associated with chromosomal bands. Chromosomal bandboundaries, <strong>in</strong>dicated by gray arrows, are suggested to be “unstable” genomic regions <strong>in</strong> the human genome, which are moreepimutation-sensitive than other genomic regions. (B) Strategy for epigenomic analysis based on chromosomal bandstructures.In conclusion, early/late-switch regions of replication tim<strong>in</strong>g generally correspond withtransitions <strong>in</strong> relative GC content, are correlated with R/G chromosome band boundaries, andare suspected of be<strong>in</strong>g “unstable” genomic regions that have <strong>in</strong>creased susceptibility toepigenetic mutation, as well as DNA damage (Figure 2.2A) [5,99]. There is a clear need forfurther epigenomic analysis on chromosomal band structures, <strong>in</strong> particular, to obta<strong>in</strong> a greaterunderstand<strong>in</strong>g of these epimutation-sensitive regions at the genome sequence level(Figure 2.2B, Table 2.2). Before perform<strong>in</strong>g epigenomic analysis us<strong>in</strong>g DNA methylation andTABLE 2.2 Characterization of Chromosome BandsChromosome Band G/Q Band R Band T Band (R Subgroup)Replication tim<strong>in</strong>g Late Early Very earlyGC% AT-rich Medium GC-richGene density Low High Very highChromat<strong>in</strong> compactness Compact Loose Loose


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>often “masked out” on the arrays. As a consequence, <strong>in</strong>vestigation of heterochromat<strong>in</strong> ormicrosatellites is optimized by use of ChIP-seq. Sequence variations with<strong>in</strong> repeat elementscan be identified and used to align the reads <strong>in</strong> the genome; unique sequences that flankrepeats are similarly helpful [117].The ma<strong>in</strong> disadvantages of ChIP-seq are cost and availability. Several groups have successfullydeveloped and applied their own protocols for library construction, which has substantiallylowered that part of the cost. For high-resolution profil<strong>in</strong>g of an entire large genome, ChIPseqcan already be less expensive than ChIP-chip; however, this depends on the genome sizeand the level of sequenc<strong>in</strong>g detail required; a ChIP-chip experiment on selected regionsus<strong>in</strong>g a customized microarray may yield as much biologically mean<strong>in</strong>gful data. The recentdecrease <strong>in</strong> sequenc<strong>in</strong>g cost per base-pair has not had as large an effect on ChIP-seq as onother applications, s<strong>in</strong>ce the decrease has come as much from <strong>in</strong>creased read lengths as fromthe number of sequenced fragments. The ga<strong>in</strong> <strong>in</strong> the fraction of reads that can be uniquelyaligned to the genome decl<strong>in</strong>es rapidly after 25e35 bp and is marg<strong>in</strong>al beyond 70e100nucleotides [118]. However, as the cost of sequenc<strong>in</strong>g decreases and <strong>in</strong>stitutional support forsequenc<strong>in</strong>g platforms grows, ChIP-seq is likely to become the method of choice for nearly allChIP experiments <strong>in</strong> the near future.20ChIP-seq analyses have been performed on multiple transcription factors with their transcriptionalco-regulators, boundary elements, numerous types of histone modifications,histone variants, nucleosome occupancy, DNA methylation patterns and gene transcription[119]. The data from these analyses are provid<strong>in</strong>g fresh <strong>in</strong>sights <strong>in</strong>to complex transcriptionalregulatory networks. Furthermore, “chromat<strong>in</strong> signatures”, characteristic chromat<strong>in</strong> structures<strong>in</strong> particular genomic regions, enable genome annotation based on predict<strong>in</strong>g histonemodifications and an overall landscape of the epigenome <strong>in</strong> human cells [52,119]. In addition,identification of the specific chromat<strong>in</strong> signatures associated with genomic features such asenhancers, <strong>in</strong>sulators, boundary elements and promoters, will provide another means ofannotat<strong>in</strong>g complex genomes. NGS technologies provide an <strong>in</strong>creas<strong>in</strong>g ability to querymultiple genomic features, which were previously too technically challeng<strong>in</strong>g and costly; this<strong>in</strong>evitably has raised expectations and ambitions, as exemplified by the published goals of theInternational <strong>Human</strong> Epigenome Consortium.Histone PTMs <strong>in</strong>fluence gene expression patterns and genome function by establish<strong>in</strong>g andorchestrat<strong>in</strong>g DNA-based biological processes [120]. PTMs can either directly affect thestructure of chromat<strong>in</strong> or can recruit co-factors that recognize histone marks and therebyadjust local chromat<strong>in</strong> structures and their behavior. A comprehensive and high-resolutionanalysis of histone modifications across the human genome will help our understand<strong>in</strong>g ofthe functional correlation of various PTMs with processes such as transcription, DNA repairand DNA replication [121,122]. Use of modification-specific antibodies <strong>in</strong> ChIP has revolutionizedthe ability to ascribe biological functions to histone modifications. ChIP-on-chiphas allowed a description of the global distribution and dynamics of various histone modifications[123]. However, prior to NGS, it had not been practical to map multiple modifications<strong>in</strong> an unbiased genomic fashion.One of the first applications of ChIP-seq was <strong>in</strong> the analysis of the genome-wide distributionof histone modifications [119]. This study, and others that followed, exemplified thenewfound feasibility and utility of obta<strong>in</strong><strong>in</strong>g collections of comprehensive genomic datasets.Twenty histone methylation sites <strong>in</strong> human T-cells were mapped [124], while five histonemethylation patterns <strong>in</strong> pluripotent and l<strong>in</strong>eage-committed mouse cells were described[125]. Such genome-wide analyses have revealed associations between specific modifiedhistones and gene activity as well as the spatial and comb<strong>in</strong>atorial relationship betweendifferent types of histone modifications. Moreover, dynamic changes <strong>in</strong> histone modificationpatterns dur<strong>in</strong>g cellular differentiation and allele-specific histone modifications wererevealed [125].


CHAPTER 2Methods and Strategies to Determ<strong>in</strong>e Epigenetic Variation <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>These <strong>in</strong>itial ChIP-seq studies, <strong>in</strong> comb<strong>in</strong>ation with more recent analyses exam<strong>in</strong><strong>in</strong>g thedistribution of other types of histone modifications, have revealed that specific genomicfeatures are associated with dist<strong>in</strong>ct types of chromat<strong>in</strong> signatures [126,127]. Such genomewidechromat<strong>in</strong> landscape maps have subsequently been exploited as a tool for def<strong>in</strong><strong>in</strong>g andpredict<strong>in</strong>g novel transcription units, enhancers, promoters, and most recently ncRNAs <strong>in</strong>previously unannotated regions of the human genome [128]. In future, the <strong>in</strong>fluence andutilization of NGS technologies will undoubtedly f<strong>in</strong>d widespread use and relevance <strong>in</strong> manydifferent areas of biology, far beyond the test-bed of epigenetics.Recent studies of the epigenome have shown that many promoters and enhancers havedist<strong>in</strong>ctive chromat<strong>in</strong> signatures. These characteristic motifs can be used as to search and mapthe regulatory elements of the genome. Won et al. [129] used this approach <strong>in</strong> a supervisedlearn<strong>in</strong>g method <strong>in</strong>volv<strong>in</strong>g a tra<strong>in</strong>ed Hidden Markov model (HMM) based on histonemodification data for known promoters and enhancers. They used the tra<strong>in</strong>ed HMMs toidentify promoter or enhancer-like sequences <strong>in</strong> the human genome [129]. In a somewhatsimilar manner, Ernst and Kellis [130] sought to identify biologically mean<strong>in</strong>gful comb<strong>in</strong>ationsof epigenetic comb<strong>in</strong>ations <strong>in</strong> the genome of human T-cells. They def<strong>in</strong>ed thesegenomic regions as hav<strong>in</strong>g “spatially coherent and biologically mean<strong>in</strong>gful chromat<strong>in</strong> markcomb<strong>in</strong>ations”, and applied a multivariate HMM analysis to search for them. Fifty-one dist<strong>in</strong>ctchromat<strong>in</strong> states were identified by the analysis, <strong>in</strong>clud<strong>in</strong>g those associated with promoters,transcription, active <strong>in</strong>tergenic regions, large-scale repressed regions and repetitive chromat<strong>in</strong>.Each chromat<strong>in</strong> state showed specific enrichments for particular sequence motifs, suggest<strong>in</strong>gdist<strong>in</strong>ct biological roles. This approach, therefore, provides a means of annotat<strong>in</strong>g the humangenome with respect to function and describes the locations of regions with diverse classes ofepigenetic function across the genome [130].There is considerable uncerta<strong>in</strong>ty regard<strong>in</strong>g the <strong>in</strong>fluence of variations <strong>in</strong> chromat<strong>in</strong> structureand transcription factor b<strong>in</strong>d<strong>in</strong>g on gene expression, and whether such variations underlie orcontribute to phenotypic differences. To address this question, McDaniell et al. [131] catalogedvariation <strong>in</strong> chromat<strong>in</strong> structure and transcription factor b<strong>in</strong>d<strong>in</strong>g between <strong>in</strong>dividuals andbetween homologous chromosomes with<strong>in</strong> <strong>in</strong>dividuals (allele-specific variation). The analysiswas carried out on lymphoblastoid cells from <strong>in</strong>dividuals with diverse geographical ancestries.They reported that 10% of active chromat<strong>in</strong> sites were specific to <strong>in</strong>dividuals, and a similarproportion was allele-specific. Both <strong>in</strong>dividual-specific and allele-specific sites could betransmitted from parent to child, suggest<strong>in</strong>g that these epigenetic marks are heritable featuresof the human genome. The study highlights the potential importance of heritable epigeneticvariation for phenotypic variation <strong>in</strong> humans [131].21Ernst et al. [132] extended their earlier chromat<strong>in</strong> profil<strong>in</strong>g analysis described above bymapp<strong>in</strong>g n<strong>in</strong>e chromat<strong>in</strong> marks <strong>in</strong> n<strong>in</strong>e different human cell types with the aim of identify<strong>in</strong>gregulatory elements, their cell-type specificities and their functional <strong>in</strong>teractions. By compar<strong>in</strong>gchromat<strong>in</strong> profiles across a range of cell types they were able to def<strong>in</strong>e cell-type-specificpatterns of promoters and enhancers affect<strong>in</strong>g chromat<strong>in</strong> status, gene expression, regulatorymotif enrichment and regulator expression. Us<strong>in</strong>g the profiles, they l<strong>in</strong>ked enhancers toputative target genes and predicted the cell-type-specific activators and repressors with whichthey <strong>in</strong>teracted [132].Computational methods for analyz<strong>in</strong>g data from epigenomic studies are be<strong>in</strong>g cont<strong>in</strong>uallydeveloped and becom<strong>in</strong>g ever more sophisticated; they have been used to identify functionalgenomic elements and to determ<strong>in</strong>e gene structures and cis-regulatory elements. For example,Hon et al. [133] described a statistical program called ChromaSig with the capacity to identifycommonly occurr<strong>in</strong>g chromat<strong>in</strong> signatures from histone modification data. They demonstratedthe potential utility of the algorithm <strong>in</strong> data from HeLa cells by identify<strong>in</strong>g five clusters of chromat<strong>in</strong>signatures associated with transcriptional promoters and enhancers. Thus, through use ofChromaSig, chromat<strong>in</strong> signatures associated with specific biological functions were identified.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>2.8 GENERAL OVERVIEW AND FUTURE PERSPECTIVEOver the last decade, the technologies available to study the mechanisms and consequences ofepigenetic modifications have <strong>in</strong>creased exponentially. The stimulus for this has been the rapid<strong>in</strong>crease <strong>in</strong> our understand<strong>in</strong>g and appreciation of the importance of epigenetic changes onphenotypes and <strong>in</strong> the etiology of diseases. Technological advances now enable large-scaleepigenomic analyses. The first whole-genome, high-resolution maps of epigenetic modificationshave been produced, but there is clearly much more to do. Detailed maps of the humanmethylome, histone modifications and nucleosome positions <strong>in</strong> healthy and diseased tissuesare still needed. This review section has attempted to provide an overview of the currentlyavailable techniques and to discuss some of the advantages and limitations of each technology.With the rapid growth <strong>in</strong> <strong>in</strong>terest <strong>in</strong> understand<strong>in</strong>g the epigenetic regulation of diseasedevelopment, a variety of new and improved methodologies are certa<strong>in</strong> to emerge <strong>in</strong> thecom<strong>in</strong>g years. These technologies will undoubtedly change the scope of epigenetic studies andwill provide valuable new <strong>in</strong>sights <strong>in</strong>to the developmental basis of diseases and <strong>in</strong>to reproductivetoxicology. Particularly, <strong>in</strong> future, the <strong>in</strong>fluence and utilization of NGS technologieswill f<strong>in</strong>d widespread use and relevance <strong>in</strong> many different areas of biology, far beyond the testbedof epigenetics.22Here, we outl<strong>in</strong>e a promis<strong>in</strong>g strategy for epigenome <strong>in</strong>vestigation that comb<strong>in</strong>es several of theepigenetic methods described above (Figures 2.2A, 2.2B). The early/late-switch regions ofreplication tim<strong>in</strong>g generally correspond to chromosomal zones with transitions <strong>in</strong> relative GCcontent; they are also correlated to R/G chromosome band boundaries, and are suspected ofbe<strong>in</strong>g “unstable” genomic regions that have <strong>in</strong>creased susceptibility to epigenetic mutationand DNA damage [5,99]. There is a clear need for further epigenomic analysis on chromosomalband structures, <strong>in</strong> particular, to obta<strong>in</strong> a greater understand<strong>in</strong>g of these epimutationsensitiveregions at the genome sequence level. Before perform<strong>in</strong>g epigenomic analysis us<strong>in</strong>gDNA methylation and histone modification, the direct correspondence between chromosomalband (R-, T-, and G-bands) and genome sequence should be elucidated by analyz<strong>in</strong>g DNAreplication tim<strong>in</strong>g and GC%, etc. (Figures 2.2A, 2.2B). F<strong>in</strong>ally, we suggest that epigenomicanalysis focused on chromosomal band structures, the boundaries of which were identified asepimutation-sensitive genomic regions at the genome sequence level, will provide considerable<strong>in</strong>sights <strong>in</strong>to normal and disease conditions.References[1] Portela A, Esteller M. Epigenetic modifications and human disease. Nat Biotechnol 2010;28:1057e68.[2] Bernste<strong>in</strong> B. The NIH Roadmap Epigenome Mapp<strong>in</strong>g Consortium. Nat Biotechnol 2010;28:1045e8.[3] Satterlee J. Tackl<strong>in</strong>g the epigenome: challenges and opportunities for collaborative efforts. Nat Biotechnol2010;28:1039e44.[4] Laird PW. Pr<strong>in</strong>ciples and challenges of genomewide DNA methylation analysis. Nat Rev Genet2010;11:191e203.[5] Watanabe Y, Maekawa M. Methylation of DNA <strong>in</strong> cancer. Adv Cl<strong>in</strong> Chem 2010;52:145e68.[6] Bird AP. Use of restriction enzymes to study eukaryotic DNA methylation. II: the symmetry of methylated sitessupports semi-conservative copy<strong>in</strong>g of the methylation pattern. J Mol Biol 1978;118:48e60.[7] Khulan B, Thompson RF, Ye K, Fazzari MJ, Suzuki M, Stasiek E, et al. Comparative isoschizomer profil<strong>in</strong>g ofcytos<strong>in</strong>e methylation: the HELP assay. Genome Res 2006;16:1046e55.[8] Schumacher A, Kapranov P, Kam<strong>in</strong>sky Z, Flanagan J, Assadzadeh A, Yau P, et al. Microarray-based DNAmethylation profil<strong>in</strong>g: technology and applications. Nucleic Acids Res 2006;34:528e42.[9] Zhang Y, Rohde C, Tierl<strong>in</strong>g S, Stamerjohanns H, Re<strong>in</strong>hardt R, Walter J, et al. DNA methylation analysis bybisulfite conversion, clon<strong>in</strong>g, and sequenc<strong>in</strong>g of <strong>in</strong>dividual clones. Methods Mol Biol 2009;507:177e87.[10] Derks S, Lentjes MH, Hellebrekers DM, de Bruïne AP, Herman JG, van Engeland M. Methylation-specific PCRunraveled. Cell Oncol 2004;26:291e9.


CHAPTER 2Methods and Strategies to Determ<strong>in</strong>e Epigenetic Variation <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>[11] Liu ZJ, Maekawa M. Polymerase cha<strong>in</strong> reaction-based methods of DNA methylation analysis. Anal Biochem2003;317:259e65.[12] Fazzai MJ, Greally JM. Epigenomics: beyond CpG islands. Nat Rev Genet 2004;5:446e55.[13] Hou P, Ji M, Li S, He N, Lu Z. High-throughput method for detect<strong>in</strong>g DNA methylation. J Biochem BiophysMethods 2004;60:139e50.[14] Cross SH, Charlton JA, Nan X, Bird AP. Purification of CpG islands us<strong>in</strong>g a methylated DNA b<strong>in</strong>d<strong>in</strong>g column.Nat Genet 1994;6:236e44.[15] Fraga MF, Ballestar E, Montoya G, Taysavang P, Wade PA, Esteller M. The aff<strong>in</strong>ity of different MBD prote<strong>in</strong>sfor a specific methylated locus depends on their <strong>in</strong>tr<strong>in</strong>sic b<strong>in</strong>d<strong>in</strong>g properties. Nucleic Acids Res2003;31:1765e74.[16] Maekawa M, Sugano K, Kashiwabara H, Ushiama M, Fujita S, Yoshimori M, et al. DNA methylation analysisus<strong>in</strong>g bisulfite treatment and PCR-s<strong>in</strong>gle-strand conformation polymorphism <strong>in</strong> colorectal cancer show<strong>in</strong>gmicrosatellite <strong>in</strong>stability. Biochem Biophys Res Commun 1999;262:671e6.[17] Maekawa M, Sugano K, Ushiama M, Fukayama N, Nomoto K, Kashiwabara H, et al. Heterogeneity of DNAmethylation status analyzed by bisulfite-PCR-SSCP and correlation with cl<strong>in</strong>ico-pathological characteristics <strong>in</strong>colorectal cancer. Cl<strong>in</strong> Chem Lab Med 2001;39:121e8.[18] Gonzalgo ML, Liang G. Methylation-sensitive s<strong>in</strong>gle-nucleotide primer extension (Ms-SNuPE) for quantitativemeasurement of DNA methylation. Nat Protoc 2007;2:1931e6.[19] Wu Z, Luo J, Ge Q, Lu Z. Microarray-based Ms-SNuPE: near-quantitative analysis for a high-throughput DNAmethylation. Biosens Bioelectron 2008;23:1333e9.[20] Xiong Z, Laird PW. COBRA: a sensitive and quantitative DNA methylation assay. Nucleic Acids Res1997;25:2532e4.[21] Colella S, Shen L, Baggerly KA, Issa JP, Krahe R. Sensitive and quantitative universal Pyrosequenc<strong>in</strong>g methylationanalysis of CpG sites. Biotechniques 2003;35:146e50.[22] Eads CA, Danenberg KD, Kawakami K, Saltz LB, Blake C, Shibata D, et al. MethyLight: a high-throughput assayto measure DNA methylation. Nucleic Acids Res 2000;28:E32.[23] Zeschnigk M, Böhr<strong>in</strong>ger S, Price EA, Onadim Z, Masshöfer L, Lohmann DR. A novel real-time PCR assay forquantitative analysis of methylated alleles (QAMA): analysis of the ret<strong>in</strong>oblastoma locus. Nucleic Acids Res2004;32:e125.[24] Tost J, Gut IG. DNA methylation analysis by pyrosequenc<strong>in</strong>g. Nat Protoc 2007;2:2265e75.[25] Tost J, Schatz P, Schuster M, Berl<strong>in</strong> K, Gut IG. Analysis and accurate quantification of CpG methylation byMALDI mass spectrometry. Nucleic Acids Res 2003;31:e50.[26] Ehrich M, Nelson MR, Stanssens P, Zabeau M, Liloglou T, X<strong>in</strong>arianos G, et al. Quantitative high-throughputanalysis of DNA methylation patterns by base-specific cleavage and mass spectrometry. Proc Natl Acad Sci USA2005;102:15785e90.[27] Gupta PK. S<strong>in</strong>gle-molecule DNA sequenc<strong>in</strong>g technologies for future genomics research. Trends Biotechnol2008;26:602e11.[28] Zilberman D, Henikoff S. Genome-wide analysis of DNA methylation patterns. Development2007;134:3959e65.[29] Bock C, Lengauer T. Computational epigenetics. Bio<strong>in</strong>formatics 2008;24:1e10.[30] Lister R, Pelizzola M, Dowen RH, Hawk<strong>in</strong>s RD, Hon G, Tonti-Filipp<strong>in</strong>i J, et al. <strong>Human</strong> DNA methylomes atbase resolution show widespread epigenomic differences. Nature 2009;462:315e22.[31] Allfrey VG, Faulkner R, Mirsky AE. Acetylation and methylation of histones and their possible role <strong>in</strong> theregulation of RNA synthesis. Proc Natl Acad Sci USA 1964;51:786e94.[32] Waterborg JH, Fried SR, Matthews HR. Acetylation and methylation sites <strong>in</strong> histone H4 from Physarum polycephalum.Eur J Biochem 1983;136:245e52.[33] Waterborg JH, Matthews HR. Patterns of histone acetylation <strong>in</strong> the cell cycle of Physarum polycephalum.Biochemistry 1983;22:1489e96.[34] Waterborg JH. Dynamic methylation of alfalfa histone H3. J Biol Chem 1993;268:4918e21.[35] Waterborg JH. Histone synthesis and turnover <strong>in</strong> alfalfa. Fast loss of highly acetylated replacement histonevariant H3.2. J Biol Chem 1993;268:4912e7.[36] Tidwell T, Allfrey VG, Mirsky AE. The methylation of histones dur<strong>in</strong>g regeneration of the liver. J Biol Chem1968;243:707e15.[37] Johnson EM, Vidali G, Littau VC, Allfrey VG. Modulation by exogenous histones of phosphorylation of nonhistonenuclear prote<strong>in</strong>s <strong>in</strong> isolated rat liver nuclei. J Biol Chem 1973;248:7595e600.[38] Sobel RE, Cook RG, Perry CA, Annunziato AT, Allis CD. Conservation of deposition-related acetylation sites <strong>in</strong>newly synthesized histones H3 and H4. Proc Natl Acad Sci USA 1995;92:1237e41.23


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>24[39] Wilk<strong>in</strong>s MR, Gasteiger E, Gooley AA, Herbert BR, Molloy MP, B<strong>in</strong>z PA, et al. High-throughput mass spectrometricdiscovery of prote<strong>in</strong> post-translational modifications. J Mol Biol 1989;289:645e57.[40] Sickmann A, Mreyen M, Meyer HE. Identification of modified prote<strong>in</strong>s by mass spectrometry. IUBMB Life2002;54:51e7.[41] Mann M, Jensen ON. Proteomic analysis of post-translational modifications. Nat Biotechnol 2003;21:255e61.[42] Sickmann A, Mreyen M, Meyer HE. Mass spectrometry: a key technology <strong>in</strong> proteome research. Adv BiochemEng Biotechnol 2003;83:141e76.[43] Posewitz MC, Tempst P. Immobilized gallium(III) aff<strong>in</strong>ity chromatography of phosphopeptides. Anal Chem1999;71:2883e92.[44] Stensballe A, Andersen S, Jensen ON. Characterization of phosphoprote<strong>in</strong>s from electrophoretic gels bynanoscale Fe(III) aff<strong>in</strong>ity chromatography with off-l<strong>in</strong>e mass spectrometry analysis. Proteomics2001;1:207e22.[45] Zhou H, Watts JD, Aebersold R. A systematic approach to the analysis of prote<strong>in</strong> phosphorylation. NatBiotechnol 2001;19:375e8.[46] Schlosser A, Vanselow JT, Kramer A. Mapp<strong>in</strong>g of phosphorylation sites by a multi-proteaseapproach with specific phosphopeptide enrichment and NanoLC-MS/MS analysis. Anal Chem 2005;77:5243e50.[47] Wisniewski JR, Szewczuk Z, Petry I, Schwanbeck R, Renner U. Constitutive phosphorylation of the acidic tailsof the high mobility group 1 prote<strong>in</strong>s by case<strong>in</strong> k<strong>in</strong>ase II alters their conformation, stability, and DNA b<strong>in</strong>d<strong>in</strong>gspecificity. J Biol Chem 1999;274:20116e22.[48] Appella E, Anderson CW. Signal<strong>in</strong>g to p53: break<strong>in</strong>g the posttranslational modification code. Pathol Biol(Paris) 2000;48:227e45.[49] Puri PL, Sartorelli V. Regulation of muscle regulatory factors by DNA-b<strong>in</strong>d<strong>in</strong>g, <strong>in</strong>teract<strong>in</strong>g prote<strong>in</strong>s, and posttranscriptionalmodifications. J Cell Physiol 2000;185:155e73.[50] Jenuwe<strong>in</strong> T, Allis CD. Translat<strong>in</strong>g the histone code. <strong>Science</strong> 2001;293:1074e80.[51] Puntervoll P, L<strong>in</strong>d<strong>in</strong>g R, Gemünd C, Chabanis-Davidson S, Matt<strong>in</strong>gsdal M, Cameron S, et al. ELM server: a newresource for <strong>in</strong>vestigat<strong>in</strong>g short functional sites <strong>in</strong> modular eukaryotic prote<strong>in</strong>s. Nucleic Acids Res2003;31:3625e30.[52] Schones DE, Zhao K. Genome-wide approaches to study<strong>in</strong>g chromat<strong>in</strong> modifications. Nat Rev Genet2008;9:179e91.[53] Nelson PT, Baldw<strong>in</strong> DA, Scearce LM, Oberholtzer JC, Tobias JW, Mourelatos Z. Microarray-based, highthroughputgene expression profil<strong>in</strong>g of microRNAs. Nat Methods 2004;1:155e61.[54] Wang H, Ach RA, Curry B. Direct and sensitive miRNA profil<strong>in</strong>g from low-<strong>in</strong>put total RNA. RNA2007;13:151e9.[55] Tricoli JV, Jacobson JW. MicroRNA: Potential for Cancer Detection, Diagnosis, and Prognosis. Cancer Res2007;67:4553e5.[56] Sempere LF, Freemantle S, Pitha-Rowe I, Moss E, Dmitrovsky E, Ambros V. Expression profil<strong>in</strong>g of mammalianmicroRNAs uncovers a subset of bra<strong>in</strong>-expressed microRNAs with possible roles <strong>in</strong> mur<strong>in</strong>e and humanneuronal differentiation. Genome Biol 2004;5:R13.[57] Válóczi A, Hornyik C, Varga N, Burgyán J, Kaupp<strong>in</strong>en S, Havelda Z. Sensitive and specific detection ofmicroRNAs by northern blot analysis us<strong>in</strong>g LNA-modified oligonucleotide probes. Nucleic Acids Res2004;32:e175.[58] Castoldi M, Benes V, Hentze MW, Muckenthaler MU. miChip: a microarray platform for expression profil<strong>in</strong>gof microRNAs based on locked nucleic acid (LNA) oligonucleotide capture probes. Methods2007;43:146e52.[59] Chen C, Ridzon DA, Broomer AJ, Zhou Z, Lee DH, Nguyen JT, et al. Real-time quantification of microRNAs bystem-loop RT-PCR. Nucleic Acids Res 2005;33:e179.[60] Kloosterman WP, Wienholds E, de Bruijn E, Kaupp<strong>in</strong>en S, Plasterk RH. In situ detection of miRNAs <strong>in</strong> animalembryos us<strong>in</strong>g LNA-modified oligonucleotide probes. Nat Methods 2006;3:27e9.[61] de Planell-Saguer M, Rodicio MC, Mourelatos Z. Rapid <strong>in</strong> situ codetection of noncod<strong>in</strong>g RNAs and prote<strong>in</strong>s <strong>in</strong>cells and formal<strong>in</strong>-fixed paraff<strong>in</strong>-embedded tissue sections without protease treatment. Nat Protoc2010;5:1061e73.[62] Schulte JH, Marschall T, Mart<strong>in</strong> M, Rosenstiel P, Mestdagh P, Schlierf S, et al. Deep sequenc<strong>in</strong>g revealsdifferential expression of microRNAs <strong>in</strong> favorable versus unfavorable neuroblastoma. Nucleic Acids Res2010;38:5919e28.[63] Takada S, Mano H. Profil<strong>in</strong>g of microRNA expression by mRAP. Nat Protoc 2007;2:3136e45.[64] Lee RC, Fe<strong>in</strong>baum RL, Ambros V. The C. elegans heterochronic gene l<strong>in</strong>-4 encodes small RNAs with antisensecomplementarity to l<strong>in</strong>-14. Cell 1993;75:843e54.


CHAPTER 2Methods and Strategies to Determ<strong>in</strong>e Epigenetic Variation <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>[65] Lau NC, Lim LP, We<strong>in</strong>ste<strong>in</strong> EG, Bartel DP. An abundant class of t<strong>in</strong>y RNAs with probable regulatory roles <strong>in</strong>Caenorhabditis elegans. <strong>Science</strong> 2001;294:858e62.[66] Lee RC, Ambros V. An extensive class of small RNAs <strong>in</strong> Caenorhabditis elegans. <strong>Science</strong> 2001;294:862e4.[67] Lagos-Qu<strong>in</strong>tana M, Rauhut R, Lendeckel W, Tuschl T. Identification of novel genes cod<strong>in</strong>g for small expressedRNAs. <strong>Science</strong> 2001;294:853e8.[68] Grishok A, Pasqu<strong>in</strong>elli AE, Conte D, Li N, Parrish S, Ha I, et al. Genes and mechanisms related to RNA<strong>in</strong>terference regulate expression of the small temporal RNAs that control C. elegans developmental tim<strong>in</strong>g. Cell2001;106:23e34.[69] Jiang J, Lee EJ, Gusev Y, Schmittgen TD. Real-time expression profil<strong>in</strong>g of microRNA precursors <strong>in</strong> humancancer cell l<strong>in</strong>es. Nucleic Acids Res 2005;33:5394e403.[70] Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, et al. MicroRNA expression profiles classify humancancers. Nature 2005;435:834e8.[71] Liu CG, Cal<strong>in</strong> GA, Meloon B, Gamliel N, Sevignani C, Ferrac<strong>in</strong> M, et al. An oligonucleotide microchipfor genome-wide microRNA profil<strong>in</strong>g <strong>in</strong> human and mouse tissues. Proc Natl Acad Sci USA2004;101:9740e4.[72] Rhoades MW, Re<strong>in</strong>hart BJ, Lim LP, Burge CB, Bartel B, Bartel DP. Prediction of plant microRNA targets. Cell2002;110:513e20.[73] Schena M, Shalon D, Davis RW, Brown PO. Quantitative monitor<strong>in</strong>g of gene expression patterns witha complementary DNA microarray. <strong>Science</strong> 1995;270:467e70.[74] Kreil DP, Russell RR, Russell S. Microarray oligonucleotide probes. Methods Enzymol 2006;410:73e98.[75] Wolber PK, Coll<strong>in</strong>s PJ, Lucas AB, De Witte A, Shannon KW. The Agilent <strong>in</strong> situ-synthesized microarray platform.Methods Enzymol 2006;410:28e57.[76] Dalma-Weiszhausz DD, Warr<strong>in</strong>gton J, Tanimoto EY, Miyada CG. The affymetrix GeneChip platform: anoverview. Methods Enzymol 2006;410:3e28.[77] Gershon D. DNA microarrays: more than gene expression. Nature 2005;437:1195e8.[78] Hughes TR, Hiley SL, Saltzman AL, Babak T, Blencowe BJ. Microarray analysis of RNA process<strong>in</strong>g andmodification. Methods Enzymol 2006;410:300e16.[79] Selig S, Okumura K, Ward DC, Cedar H. Del<strong>in</strong>eation of DNA replication time zones by fluorescence <strong>in</strong> situhybridization. EMBO J 1992;11:1217e25.[80] Hansen RS, Canfield TK, Lamb MM, Gartler SM, Laird CD. Association of fragile X syndrome with delayedreplication of the FMR1 gene. Cell 1993;73:1403e9.[81] Watanabe Y, Tenzen T, Nagasaka Y, Inoko H, Ikemura T. Replication tim<strong>in</strong>g of the human X-<strong>in</strong>activation center(XIC) region: correlation with chromosome bands. Gene 2000;252:163e72.[82] Watanabe Y, Fujiyama A, Ichiba Y, Hattori M, Yada T, Sakaki Y, et al. Chromosome-wide assessment ofreplication tim<strong>in</strong>g for human chromosomes 11q and 21q: disease-related genes <strong>in</strong> tim<strong>in</strong>g-switch regions. HumMol Genet 2002;11:13e21.[83] Woodf<strong>in</strong>e K, Fiegler H, Beare DM, Coll<strong>in</strong>s JE, McCann OT, Young BD, et al. Replication tim<strong>in</strong>g of the humangenome. Hum Mol Genet 2004;13:191e202.[84] Watanabe Y, Ikemura T, Sugimura H. Amplicons on human chromosome 11q are located <strong>in</strong> the early/lateswitchregions of replication tim<strong>in</strong>g. Genomics 2004;84:796e805.[85] White EJ, Emanuelsson O, Scalzo D, Royce T, Kosak S, Oakeley EJ, et al. DNA replication-tim<strong>in</strong>g analysis ofhuman chromosome 22 at high resolution and different developmental states. Proc Natl Acad Sci USA2004;101:17771e6.[86] Mirk<strong>in</strong> EV, Mirk<strong>in</strong> SM. Replication fork stall<strong>in</strong>g at natural impediments. Microbiol Mol Biol Rev2007;71:13e35.[87] Sequeira-Mendes J, Díaz-Uriarte R, Apedaile A, Huntley D, Brockdorff N, Gómez M. Transcription <strong>in</strong>itiationactivity sets replication orig<strong>in</strong> efficiency <strong>in</strong> mammalian cells. PLoS Genet 2009;5:e1000446.[88] Bacolla A, Wells RD. Non-B DNA conformations as determ<strong>in</strong>ants of mutagenesis and human disease. MolCarc<strong>in</strong>og 2009;48:273e85.[89] Hiratani I, Gilbert DM. Replication tim<strong>in</strong>g as an epigenetic mark. <strong>Epigenetics</strong> 2009;4:93e7.[90] Göndör A, Ohlsson R. Replication tim<strong>in</strong>g and epigenetic reprogramm<strong>in</strong>g of gene expression: a two-wayrelationship? Nat Rev Genet 2009;10:269e76.[91] State MW, Greally JM, Cuker A, Bowers PN, Henegariu O, Morgan TM, et al. Epigenetic abnormalities associatedwith a chromosome 18(q21eq22) <strong>in</strong>version and a Gilles de la Tourette syndrome phenotype. Proc NatlAcad Sci USA 2003;100:4684e9.[92] D’Antoni S, Matt<strong>in</strong>a T, Di Mare P, Federico C, Motta S, Saccone S. Altered replication tim<strong>in</strong>g of the HIRA/Tuple1 locus <strong>in</strong> the DiGeorge and Velocardiofacial syndromes. Gene 2004;333:111e9.25


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>26[93] Dotan ZA, Dotan A, Ramon J, Avivi L. Altered mode of allelic replication accompanied by aneuploidy <strong>in</strong>peripheral blood lymphocytes of prostate cancer patients. Int J Cancer 2004;111:60e6.[94] Dotan ZA, Dotan A, Ramon J, Avivi L. Aberrant allele-specific replication, <strong>in</strong>dependent of parental orig<strong>in</strong>, <strong>in</strong>blood cells of cancer patients. BMC Cancer 2008;8:390.[95] Watanabe Y, Shibata K, Sugimura H, Maekawa M. p53-dependent change <strong>in</strong> replication tim<strong>in</strong>g of the humangenome. Biochem Biophys Res Commun 2007;364:289e93.[96] Watanabe Y, Shibata K, Ikemura T, Maekawa M. Replication tim<strong>in</strong>g of extremely large genes on humanchromosomes 11q and 21q. Gene 2008;421:74e80.[97] Watanabe Y, Abe T, Ikemura T, Maekawa M. Relationships between replication tim<strong>in</strong>g and GC content ofcancer-related genes on human chromosomes 11q and 21q. Gene 2009;433:26e31.[98] Göndör A, Ohlsson R. Replication tim<strong>in</strong>g and epigenetic reprogramm<strong>in</strong>g of gene expression: a two-wayrelationship? Nat Rev Genet 2009;10:269e76.[99] Watanabe Y, Maekawa M. Spatiotemporal regulation of DNA replication <strong>in</strong> the human genome and itsassociation with genomic <strong>in</strong>stability and disease. Curr Med Chem 2010;17:222e33.[100] Maekawa M, Watanabe Y. <strong>Epigenetics</strong>: relations to disease and laboratory f<strong>in</strong>d<strong>in</strong>gs. Curr Med Chem2007;14:2642e53.[101] Holmquist G, Gray M, Porter T, Jordan J. Characterization of Giemsa dark- and light-band DNA. Cell1982;31:121e9.[102] Holmquist GP. Evolution of chromosome bands: molecular ecology of noncod<strong>in</strong>g DNA. J Mol Evol1989;28:469e86.[103] Bernardi G. The isochore organization of the human genome. Annu Rev Genet 1989;23:637e61.[104] Craig JM, Bickmore WA. Chromosome bandsdflavours to savour. Bioessays 1993;15:349e54.[105] Bierne H, Michel B. When replication forks stop. Mol Microbiol 1994;13:17e23.[106] Verbovaia LV, Raz<strong>in</strong> SV. Mapp<strong>in</strong>g of replication orig<strong>in</strong>s and term<strong>in</strong>ation sites <strong>in</strong> the Duchenne musculardystrophy gene. Genomics 1997;45:24e30.[107] Rothste<strong>in</strong> R, Michel B, Gangloff S. Replication fork paus<strong>in</strong>g and recomb<strong>in</strong>ation or ‘gimme a break’. Genes Dev2000;14:1e10.[108] Hiratani I, Ryba T, Itoh M, Yokochi T, Schwaiger M, Chang CW, et al. Global reorganization of replicationdoma<strong>in</strong>s dur<strong>in</strong>g embryonic stem cell differentiation. PLoS Biol 2008;6:e245.[109] Helmrich A, Stout-Weider K, Hermann K, Schrock E, Heiden T. Common fragile sites are conservedfeatures of human and mouse chromosomes and relate to large active genes. Genome Res 2006;16:1222e30.[110] Debatisse M, El Achkar E, Dutrillaux B. Common fragile sites nested at the <strong>in</strong>terfaces of early and latereplicat<strong>in</strong>gchromosome bands: cis act<strong>in</strong>g components of the G2/M checkpo<strong>in</strong>t? Cell Cycle2006;5:578e81.[111] Ge XQ, Jackson DA, Blow JJ. Dormant orig<strong>in</strong>s licensed by excess Mcm2-7 are required for human cells tosurvive replicative stress. Genes Dev 2007;21:3331e41.[112] Ibarra A, Schwob E, Méndez J. Excess MCM prote<strong>in</strong>s protect human cells from replicative stress by licens<strong>in</strong>gbackup orig<strong>in</strong>s of replication. Proc Natl Acad Sci USA 2008;105:8956e61.[113] Rakyan VK, Down TA, Bald<strong>in</strong>g DJ, Beck S. Epigenome-wide association studies for common human diseases.Nat Rev Genet 2011;12:529e41.[114] Mardis ER. Next-generation DNA sequenc<strong>in</strong>g methods. Annu Rev Genomics Hum Genet 2008;9:387e402.[115] Kim TH, Barrera LO, Zheng M, Qu C, S<strong>in</strong>ger MA, Richmond TA, et al. A high-resolution map of activepromoters <strong>in</strong> the human genome. Nature 2005;436:876e80.[116] Alekseyenko AA, Peng S, Larschan E, Gorchakov AA, Lee OK, Kharchenko P, et al. A sequence motifwith<strong>in</strong> chromat<strong>in</strong> entry sites directs MSL establishment on the Drosophila X chromosome. Cell2008;134:599e609.[117] Rozowsky J, Euskirchen G, Auerbach RK, Zhang ZD, Gibson T, Bjornson R, et al. PeakSeq enables systematicscor<strong>in</strong>g of ChIPeseq experiments relative to controls. Nature Biotech 2009;27:66e75.[118] Whiteford N, Haslam N, Weber G, Prügel-Bennett A, Essex JW, Roach PL, et al. An analysis of the feasibility ofshort read sequenc<strong>in</strong>g. Nucleic Acids Res 2005;33:e171.[119] Park PJ. ChIP-seq: advantages and challenges of a matur<strong>in</strong>g technology. Nat Rev Genet 2009;10:669e80.[120] Kouzarides T. Chromat<strong>in</strong> modifications and their function. Cell 2007;128:693e705.[121] Li B, Carey M, Workman JL. The role of chromat<strong>in</strong> dur<strong>in</strong>g transcription. Cell 2007;128:707e19.[122] Groth A, Rocha W, Verreault A, Almouzni G. Chromat<strong>in</strong> challenges dur<strong>in</strong>g DNA replication and repair. Cell2007;128:721e33.[123] Rando OJ. Global patterns of histone modifications. Curr Op<strong>in</strong> Genet Dev 2007;17:94e9.


CHAPTER 2Methods and Strategies to Determ<strong>in</strong>e Epigenetic Variation <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>[124] Barski A, Cuddapah S, Cui K, Roh TY, Schones DE, Wang Z, et al. High-resolution profil<strong>in</strong>g of histonemethylations <strong>in</strong> the human genome. Cell 2007;129:823e37.[125] Mikkelsen TS, Ku M, Jaffe DB, Issac B, Lieberman E, Giannoukos G, et al. Genome-wide maps of chromat<strong>in</strong>state <strong>in</strong> pluripotent and l<strong>in</strong>eage-committed cells. Nature 2007;448:553e60.[126] Wang Z, Zang C, Rosenfeld JA, Schones DE, Barski A, Cuddapah S, et al. Comb<strong>in</strong>atorial patterns of histoneacetylations and methylations <strong>in</strong> the human genome. Nat Genet 2008;40:897e903.[127] Robertson AG, Bilenky M, Tam A, Zhao Y, Zeng T, Thiessen N, et al. Genome-wide relationship betweenhistone H3 lys<strong>in</strong>e 4 mono- and tri-methylation and transcription factor b<strong>in</strong>d<strong>in</strong>g. Genome Res2008;18:1906e17.[128] Guttman M, Amit I, Garber M, French C, L<strong>in</strong> MF, Feldser D, et al. Chromat<strong>in</strong> signature reveals overa thousand highly conserved large non-cod<strong>in</strong>g RNAs <strong>in</strong> mammals. Nature 2009;458:223e7.[129] Won KJ, Chepelev I, Ren B, Wang W. Prediction of regulatory elements <strong>in</strong> mammalian genomes us<strong>in</strong>gchromat<strong>in</strong> signatures. BMC Bio<strong>in</strong>formatics 2008;9:547.[130] Ernst J, Kellis M. Discovery and characterization of chromat<strong>in</strong> states for systematic annotation of the humangenome. Nat Biotechnol 2010;28:817e25.[131] McDaniell R, Lee BK, Song L, Liu Z, Boyle AP, Erdos MR, et al. Heritable <strong>in</strong>dividual-specific and allele-specificchromat<strong>in</strong> signatures <strong>in</strong> humans. <strong>Science</strong> 2010;328:235e9.[132] Ernst J, Kheradpour P, Mikkelsen TS, Shoresh N, Ward LD, Epste<strong>in</strong> CB, et al. Mapp<strong>in</strong>g and analysis ofchromat<strong>in</strong> state dynamics <strong>in</strong> n<strong>in</strong>e human cell types. Nature 2011;473:43e9.[133] Hon G, Ren B, Wang W. ChromaSig: a probabilistic approach to f<strong>in</strong>d<strong>in</strong>g common chromat<strong>in</strong> signatures <strong>in</strong>the human genome. PLoS Comput Biol 2008;4:e1000201.[134] Ermakova OV, Nguyen LH, Little RD, Chevillard C, Riblet R, Ashouian N, et al. Evidence that a s<strong>in</strong>glereplication fork proceeds from early to late replicat<strong>in</strong>g doma<strong>in</strong>s <strong>in</strong> the Igh locus <strong>in</strong> a non-B cell l<strong>in</strong>e. Mol Cell1999;3:321e30.[135] Karnani N, Taylor C, Malhotra A, Dutta A. Pan-S replication patterns and chromosomal doma<strong>in</strong>s def<strong>in</strong>ed bygenome-til<strong>in</strong>g arrays of ENCODE genomic areas. Genome Res 2007;17:865e76.27


This page <strong>in</strong>tentionally left blank


CHAPTER 3DNA MethylationAlterations <strong>in</strong> <strong>Human</strong>CancersYae Kanai, Eri AraiNational Cancer Center Research Institute, Tokyo, JapanCHAPTER OUTLINE3.1 Introduction: Biological Roles ofDNA Methylation 293.2 DNA Methylation Alterations <strong>in</strong><strong>Human</strong> Cancers 313.3 Aberrant DNA Methylation <strong>in</strong>Precancerous ConditionsAssociated with ChronicInflammation, Persistent ViralInfection and Smok<strong>in</strong>g 313.4 Abnormal Expression of DNMTs <strong>in</strong><strong>Human</strong> Cancers 333.5 Mutations, Polymorphism andSplic<strong>in</strong>g Alterations of DNMTsand <strong>Human</strong> Cancers 343.6 Signal Pathways Affect<strong>in</strong>g DNAMethylation Status Dur<strong>in</strong>gTumorigenesis 353.7 DNA Methylation and HistoneModifications 363.8 Subclassification of <strong>Human</strong>Cancers Based on DNAMethylation Profil<strong>in</strong>g 373.9 Diagnosis of Cancers <strong>in</strong> BodyFluids and Biopsy SpecimensBased on DNA MethylationProfiles 403.10 Carc<strong>in</strong>ogenetic Risk EstimationBased on DNA MethylationProfiles 423.11 Personalized Medic<strong>in</strong>e Based onDNA Methylation Profiles:Prognostication of Patients withCancers and Prediction ofResponse to Chemotherapy 443.12 New Technologies for DNAMethylation Analysis and FutureDirections 47References 47293.1 INTRODUCTION: BIOLOGICAL ROLES OF DNA METHYLATIONEpigenetic processes, i.e. alterations to biological <strong>in</strong>formation without changes <strong>in</strong> the DNAsequences that are mitotically and/or meiotically heritable, go beyond DNA-stored <strong>in</strong>formationand are essential for packag<strong>in</strong>g and <strong>in</strong>terpretation of the genome [1]. The modulation ofepigenetic profiles contributes significantly to embryonic development, differentiation, andtransition from a stem cell to a l<strong>in</strong>eage-committed cell, and underlies responses to environmentalsignals such as hormones, nutrients and <strong>in</strong>flammation [2]. DNA methylation is a keyelement of epigenetic mechanisms that <strong>in</strong>clude histone-modification, position<strong>in</strong>g of histonevariants, nucleosome remodel<strong>in</strong>g, and non-cod<strong>in</strong>g RNA. DNA methylation is a covalentT. Tollefsbol (Ed): <strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>. DOI: 10.1016/B978-0-12-388415-2.00003-2Copyright Ó 2012 Elsevier Inc. All rights reserved.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>chemical modification result<strong>in</strong>g <strong>in</strong> addition of a methyl (CH 3 ) group at the carbon 5 positionof the cytos<strong>in</strong>e r<strong>in</strong>g of CpG d<strong>in</strong>ucleotides. CpG sites are concentrated either <strong>in</strong> repetitivesequences or CpG islands <strong>in</strong> promoter regions.The C-term<strong>in</strong>al catalytic doma<strong>in</strong> of DNA (cytos<strong>in</strong>e-5-)-methyltransferases (DNMTs) transfersmethyl groups from S-adenosyl-L-methion<strong>in</strong>e (AdoMet) to cytos<strong>in</strong>es. Dietary folate,vitam<strong>in</strong>s B6 and B12, methion<strong>in</strong>e and chol<strong>in</strong>e can critically affect the synthesis of AdoMet[3]. The C-term<strong>in</strong>al catalytic doma<strong>in</strong> of DNMTs is composed of five conserved am<strong>in</strong>o acidmotifs, namely I, IV, VI, IX and X [4,5]. Motifs I and X are filed together to form the b<strong>in</strong>d<strong>in</strong>gsite for AdoMet. Motif IV conta<strong>in</strong>s the prolylcyste<strong>in</strong>yl dipeptide that provides the thiolate atthe active site. Motif VI conta<strong>in</strong>s the glutamyl residue that protonates the 3 position of thetarget cytos<strong>in</strong>e. Motif IX forms the target recognition doma<strong>in</strong>. The N-term<strong>in</strong>al regulatorydoma<strong>in</strong> of DNMT1 conta<strong>in</strong>s a PCNA (proliferat<strong>in</strong>g cell nuclear antigen)-b<strong>in</strong>d<strong>in</strong>g doma<strong>in</strong>,a cyste<strong>in</strong>e-rich ATRX (alpha thalassemia/mental retardation syndrome X-l<strong>in</strong>ked) z<strong>in</strong>c f<strong>in</strong>gerDNA-b<strong>in</strong>d<strong>in</strong>g motif, and a polybromo homology doma<strong>in</strong> target<strong>in</strong>g DNMT1 to the replicationfoci. The preference of DNMT1 for hemimethylated over unmethylated substrates<strong>in</strong> vitro and its target<strong>in</strong>g of replication foci are believed to allow copy<strong>in</strong>g of the methylationpattern of the parental strand to the newly synthesized daughter DNA strand. Thus, DNMT1has been recognized as the “ma<strong>in</strong>tenance” DNMT, whereas DNMT3A and DNMT3B show denovo DNA methylation activity <strong>in</strong> vitro [6]. However, s<strong>in</strong>ce de novo methylation of CpGislands has actually been observed <strong>in</strong> human fibroblasts overexpress<strong>in</strong>g DNMT1, DNMT1 iscapable of de novo DNA methylation activity <strong>in</strong> vivo as well as hav<strong>in</strong>g a ma<strong>in</strong>tenancefunction [3]. DNA methylation profiles <strong>in</strong> vivo may be determ<strong>in</strong>ed on the basis ofcooperation between DNMT1 and the DNMT3 family. DNMT3L lacks conserved motifs ofthe catalytic doma<strong>in</strong> and cooperates with the DNMT3 family to establish an impr<strong>in</strong>t<strong>in</strong>gpattern [7].30DNA methylation plays critical roles <strong>in</strong> the ma<strong>in</strong>tenance of chromat<strong>in</strong> <strong>in</strong>tegrity and regulationof gene expression [8]: (a) repetitive and parasitic sequences, such as retrotransposons andendogenous retroviral elements, are usually repressed due to DNA methylation and (b)methylation of CpGs islands can directly impede the b<strong>in</strong>d<strong>in</strong>g of transcription factors to theirtarget sites, thus prohibit<strong>in</strong>g the transcription of specific genes. Moreover, methylation of CpGislands normally promotes a highly condensed heterochromat<strong>in</strong> structure, where activetranscription does not occur. Weber et al. reported that approximately 70% of human genes arel<strong>in</strong>ked to promoter CpG islands and about 4% of CpG island promoters are methylated <strong>in</strong>somatic cells [9]. Methylation of CpG islands naturally takes place dur<strong>in</strong>g X chromosome<strong>in</strong>activation and impr<strong>in</strong>t<strong>in</strong>g, though the majority of CpG islands rema<strong>in</strong> unmethylated dur<strong>in</strong>gdevelopment and differentiation. Extensive changes <strong>in</strong> DNA methylation dur<strong>in</strong>g the processesof differentiation are known to take place at CpG island shores, regions of comparatively lowCpG density close to CpG islands [10].On the other hand, DNA demethylation is a process <strong>in</strong>volv<strong>in</strong>g removal of a methyl group froma nucleotide <strong>in</strong> DNA. Although passive demethylation occurs <strong>in</strong> the absence of methylation ofnewly synthesized DNA strands by “ma<strong>in</strong>tenance” DNMT dur<strong>in</strong>g replication rounds, activeremoval of cytos<strong>in</strong>e methylation has long rema<strong>in</strong>ed a mystery. Recently, it has been proved that5-methylcytos<strong>in</strong>e can be converted to 5-hydroxymethylcytos<strong>in</strong>e, an <strong>in</strong>termediate formpotentially <strong>in</strong>volved <strong>in</strong> active demethylation, by the 2-oxoglutarate and Fe (II)-dependentoxygenases TET1, TET2 and TET3 [11,12]. MLL (myeloid/lymphoid or mixed-l<strong>in</strong>eageleukemia)eTET1 translocations have been found <strong>in</strong> patients with acute lymphoblasticleukemia [13] and deletions or mutations at the TET2 locus have been reported <strong>in</strong> myelodysplasticsyndrome and acute myeloid leukemia [14], <strong>in</strong>dicat<strong>in</strong>g that impairment of theconversion of 5-methylcytos<strong>in</strong>e to 5-hydroxymethylcytos<strong>in</strong>e may also participate <strong>in</strong>tumorigenesis [15]. Further <strong>in</strong>vestigation will be needed to elucidate the significance ofconversion to 5-hydroxymethylcytos<strong>in</strong>e.


CHAPTER 3DNA Methylation Alterations <strong>in</strong> <strong>Human</strong> Cancers3.2 DNA METHYLATION ALTERATIONS IN HUMAN CANCERSHeterozygosity of the Dnmt1 gene, <strong>in</strong> conjunction with treatment us<strong>in</strong>g the DNMT <strong>in</strong>hibitor5-aza-deoxycytid<strong>in</strong>e, reduces the average number of <strong>in</strong>test<strong>in</strong>al adenomas <strong>in</strong> ApcM<strong>in</strong> mice [16].On the other hand, genomic hypomethylation <strong>in</strong> p53þ/e mice due to the <strong>in</strong>troduction ofa hypomorphic allele of Dnmt 1 <strong>in</strong>duces sarcomas at an earlier age <strong>in</strong> comparison withlittermates possess<strong>in</strong>g normal levels of DNMT1 activity [17,18]. Increased loss of heterozygosity(LOH) accompanied by activation of endogenous retroviral elements has been observed<strong>in</strong> Dnmt1 hypomorphic mice [19]. These observations of genetically eng<strong>in</strong>eered animalsclearly demonstrate a causal relationship between alterations of DNA methylation and humancancers.In fact, human cancer cells obta<strong>in</strong>ed from cl<strong>in</strong>ical tissue specimens frequently show genomewideDNA hypomethylation and region-specific DNA hypermethylation [20]. DNA hypomethylation<strong>in</strong>duces a higher probability of translocation of parasitic sequences to othergenomic regions, and chromosomal rearrangement result<strong>in</strong>g <strong>in</strong> chromosomal <strong>in</strong>stability [21].Furthermore, aberrant DNA hypomethylation can also <strong>in</strong>duce activation of oncogenes andloss of impr<strong>in</strong>t<strong>in</strong>g. However, a more widely recognized epigenetic change <strong>in</strong> human cancers isDNA hypermethylation at the CpG islands of promoters that silences specific genes, <strong>in</strong>clud<strong>in</strong>gtumor-suppressor genes [20] such as CDKN2A (cycl<strong>in</strong>-dependent k<strong>in</strong>ase <strong>in</strong>hibitor 2A),CDKN2B (cycl<strong>in</strong>-dependent k<strong>in</strong>ase <strong>in</strong>hibitor 2B), TP73 (tumor prote<strong>in</strong> p73), MLH1 (mutLhomolog 1), Apc (adenomatosis polyposis coli), BRCA1 (breast cancer 1), MGMT(O-6-methylguan<strong>in</strong>e-DNA methyltransferase), VHL (von Hippel-L<strong>in</strong>dau tumor-suppressor),GSTP1 (glutathione S-transferase pi 1), CDH1 (cadher<strong>in</strong> 1) and DAPK1 (death-associatedprote<strong>in</strong> k<strong>in</strong>ase 1). DNA hypermethylation of tumor-suppressor genes frequently becomes thesecond hit for driver events <strong>in</strong> accordance with the two-hit theory [22]. Moreover, some tumorsuppressorgenes, such as TIMP3 (tissue <strong>in</strong>hibitor of metalloprote<strong>in</strong>ase 3), SFRP1 (secretedfrizzled-related prote<strong>in</strong> 1), SFRP2, SFRP4, SFRP5 and RASSF1 (Ras association (RalGDS/AF-6)doma<strong>in</strong> family member 1), are seldom mutated, or their mutations have never been reported<strong>in</strong> human cancers [23]. Therefore, <strong>in</strong>tensive screen<strong>in</strong>g of genes that are methylated <strong>in</strong> humancancers may be a strategy for identification of tumor-related genes that have potential astherapeutic targets. In some <strong>in</strong>stances, genes can be silenced simultaneously due to a process oflong-range epigenetic silenc<strong>in</strong>g, and the spread<strong>in</strong>g of silenc<strong>in</strong>g seems to affect neighbor<strong>in</strong>gunmethylated genes through repressive chromat<strong>in</strong> [24].31MiRNAs are the best-known class of short non-cod<strong>in</strong>g RNAs, which are typically around 21nucleotides <strong>in</strong> length, imperfectly aligned with the 3 0 UTR of target mRNAs, and <strong>in</strong>duce theirtranslational repression. Observations of silenc<strong>in</strong>g due to DNA hypermethylation haveexpanded to tumor-suppressive microRNAs (25), such as miR-34a and 34b/c, miR-124, miR-137, miR-152, miR-193a, miR-200, miR-203, miR-205, miR-218 and miR-345. In addition totheir tumor-suppressor function, miRNAs can also serve as oncogenes to promote cancergrowth. B-cell <strong>in</strong>tegration cluster (BIC)/miR-155 is the first miRNA shown to have such tumorpromot<strong>in</strong>gactivity. miR-10b is another oncogene highly associated with cancer metastasis.Transcription of miR-10b is regulated by the transcription factor Twist, and the downstreamtargets of miR-10b <strong>in</strong>clude homeobox D10. Other miRNAs with oncogene function <strong>in</strong>cludemiR-17 clusters, miR-21, and miR-373 and miR-520c as metastasis-promot<strong>in</strong>g miRNAs [26].3.3 ABERRANT DNA METHYLATION IN PRECANCEROUSCONDITIONS ASSOCIATED WITH CHRONIC INFLAMMATION,PERSISTENT VIRAL INFECTION AND SMOKINGDNA methylation alterations are frequently observed even <strong>in</strong> precancerous conditions andearly-stage cancers, suggest<strong>in</strong>g that epigenetic alterations may precede the classical transform<strong>in</strong>gevents, such as mutations of tumor-suppressor genes, amplification of oncogenes and


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>chromosomal <strong>in</strong>stability. Environmental factors <strong>in</strong>fluence health, and epigenetic profiles areknown to be responses to environmental signals. Thus, aberrant DNA methylation participatesespecially <strong>in</strong> precancerous conditions associated with chronic <strong>in</strong>flammation, persistent viral<strong>in</strong>fection and smok<strong>in</strong>g [27,28]. For example, <strong>in</strong> the 1990s, although LOH on chromosome 16was frequently detected by classical Southern blott<strong>in</strong>g <strong>in</strong> hepatocellular carc<strong>in</strong>omas (HCCs)associated with metastasis, the molecular events occurr<strong>in</strong>g <strong>in</strong> non-cancerous liver tissueshow<strong>in</strong>g chronic hepatitis or liver cirrhosis, which are widely considered to be precancerousconditions, were unknown. When we exam<strong>in</strong>ed the DNA methylation status on chromosome16 us<strong>in</strong>g Southern blott<strong>in</strong>g with a DNA methylation-sensitive restriction enzyme, DNAmethylation alterations at multiple loci were frequently revealed even <strong>in</strong> chronic hepatitis orliver cirrhosis, compared with normal liver tissue, <strong>in</strong>dicat<strong>in</strong>g that DNA methylation alterationsare a very early event dur<strong>in</strong>g multistage hepatocarc<strong>in</strong>ogenesis [29]. This was one of the earliestreports of DNA methylation alterations at the precancerous stage.We then exam<strong>in</strong>ed whether aberrant DNA methylation precedes chromosomal <strong>in</strong>stabilitydur<strong>in</strong>g hepatocarc<strong>in</strong>ogenesis. Bisulfite modification, which converts unmethylated cytos<strong>in</strong>eresidues to uracil, leav<strong>in</strong>g methylated cytos<strong>in</strong>e residues unchanged, was applied to microdissectedspecimens obta<strong>in</strong>ed from lobules, pseudo-lobules or regenerative nodules <strong>in</strong> noncancerousliver tissue from patients with HCCs. Although no degree of DNA methylation ofany of the exam<strong>in</strong>ed C-type CpG islands, which are generally methylated <strong>in</strong> a cancer-specificbut not age-dependent manner, was ever detected <strong>in</strong> normal liver tissue from patients withoutHCCs, DNA hypermethylation of such islands was frequently found even <strong>in</strong> microdissectedspecimens of non-cancerous liver tissue show<strong>in</strong>g no remarkable histological changes obta<strong>in</strong>edfrom patients with HCCs <strong>in</strong> which LOH was never detected by PCR us<strong>in</strong>g multiple microsatellitemarkers. Thus it was directly confirmed that aberrant DNA methylation is an earlierevent preced<strong>in</strong>g chromosomal <strong>in</strong>stability dur<strong>in</strong>g hepatocarc<strong>in</strong>ogenesis [30].32DNA hypermethylation around the promoter region of the CDH1 gene at 16q22.1 [31], whichencodes a Ca 2þ -dependent cellecell adhesion molecule [32], has been detected even <strong>in</strong>samples of non-cancerous liver tissue show<strong>in</strong>g chronic hepatitis or cirrhosis [33]. HeterogeneousE-cadher<strong>in</strong> expression <strong>in</strong> such non-cancerous liver tissue, which is associated with smallfocal areas of hepatocytes show<strong>in</strong>g only slight E-cadher<strong>in</strong> immunoreactivity, might be due, atleast partly, to DNA hypermethylation [33]. Reduction of E-cadher<strong>in</strong> expression due to DNAhypermethylation around the promoter region may participate even <strong>in</strong> the very early stage ofhepatocarc<strong>in</strong>ogenesis through loss of <strong>in</strong>tercellular adhesiveness and destruction of tissuemorphology.In addition to the chronic hepatitis and liver cirrhosis stages result<strong>in</strong>g from <strong>in</strong>fection withhepatitis B virus (HBV) and/or hepatitis C virus (HCV) [30,34,35], DNA methylation alterationsare frequently found at the precancerous stage <strong>in</strong> various organs, especially <strong>in</strong> associationwith chronic <strong>in</strong>flammation and/or persistent <strong>in</strong>fection with viruses. Epste<strong>in</strong>eBarr virus(EBV) <strong>in</strong>fection <strong>in</strong> stomach cancers is significantly associated with marked accumulation ofDNA hypermethylation of C-type CpG islands [36,37], and viral latent membrane prote<strong>in</strong> 2Aup-regulates DNMT1 <strong>in</strong> cultured cancer cells [38]. Helicobacter pylori <strong>in</strong>fection, another etiologicfactor that is believed to be <strong>in</strong>volved <strong>in</strong> stomach carc<strong>in</strong>ogenesis, has also been reported tostrongly promote regional DNA hypermethylation [39]. Cervical <strong>in</strong>traepithelial neoplasia(CIN) is a precursor lesion for squamous cell carc<strong>in</strong>oma of the uter<strong>in</strong>e cervix closely associatedwith human papillomavirus (HPV) <strong>in</strong>fection. DNMT1 prote<strong>in</strong> expression is <strong>in</strong>creased even <strong>in</strong>low-grade CINs relative to normal squamous epithelia, and is further <strong>in</strong>creased <strong>in</strong> higher-gradeCINs and squamous cell carc<strong>in</strong>omas of the uter<strong>in</strong>e cervix [40]. HPV-16 E7 prote<strong>in</strong> has beenreported to associate directly with DNMT1 and stimulate the methyltransferase activity ofDNMT1 <strong>in</strong> vitro [41].DNA hypermethylation at the HIC1 (hypermethylated <strong>in</strong> cancer 1) locus has been observed <strong>in</strong>non-cancerous lung tissues, which may conta<strong>in</strong> progenitor cells for cancers, obta<strong>in</strong>ed from


CHAPTER 3DNA Methylation Alterations <strong>in</strong> <strong>Human</strong> Cancerspatients with non-small-cell lung cancers, and <strong>in</strong> the correspond<strong>in</strong>g non-small-cell lungcancers [42]. HIC1 is a growth-regulatory and tumor-repressor gene [43] that was first identified<strong>in</strong> the commonly methylated chromosomal region <strong>in</strong> human cancer cells [44]. The<strong>in</strong>cidence of DNA hypermethylation at this locus was significantly associated with poorerdifferentiation of lung adenocarc<strong>in</strong>omas. The <strong>in</strong>cidence of DNA hypermethylation <strong>in</strong> samplesof both non-cancerous lung tissue and non-small-cell lung cancer from patients who werecurrent smokers was significantly higher than <strong>in</strong> patients who had never smoked [42]. The<strong>in</strong>cidence of DNA hypermethylation <strong>in</strong> non-cancerous lung tissue from patients with nonsmall-celllung cancers was significantly correlated with the extent of pulmonary anthracosis,as an <strong>in</strong>dex of the cumulative effects of smok<strong>in</strong>g [27]. Cigarette smok<strong>in</strong>g seems to be anotherbackground factor associated with alterations of DNA methylation dur<strong>in</strong>g multistagecarc<strong>in</strong>ogenesis.3.4 ABNORMAL EXPRESSION OF DNMTS IN HUMAN CANCERSAt least a proportion of DNA methylation alterations <strong>in</strong> human cancers may beattributable to abnormalities of DNMTs. In fact, altered expression of DNMTs has beenreported <strong>in</strong> human cancers. For example, the levels of DNMT1 mRNA expression aresignificantly higher <strong>in</strong> samples of non-cancerous liver tissue show<strong>in</strong>g chronic hepatitis orcirrhosis than <strong>in</strong> normal liver tissue, and are even higher <strong>in</strong> HCCs [45,46]. The <strong>in</strong>cidenceof DNMT1 overexpression <strong>in</strong> HCCs is significantly correlated with poorer tumor differentiationand portal ve<strong>in</strong> tumor <strong>in</strong>volvement. Moreover, DNMT1 overexpression <strong>in</strong>tumors is <strong>in</strong>versely correlated with the recurrence-free and overall survival rates ofpatients with HCCs [47].Ductal adenocarc<strong>in</strong>omas of the pancreas frequently develop after chronic damage due topancreatitis. At least a proportion of peripheral pancreatic ductal epithelia with an <strong>in</strong>flammatorybackground may be at the precancerous stage. The <strong>in</strong>cidence of DNMT1 prote<strong>in</strong>expression <strong>in</strong>creases with progression from peripheral pancreatic ductal epithelia with an<strong>in</strong>flammatory background, to another precancerous lesion, pancreatic <strong>in</strong>traductal neoplasia(PanIN), to well-differentiated ductal adenocarc<strong>in</strong>oma, and f<strong>in</strong>ally to poorly differentiatedductal adenocarc<strong>in</strong>oma, <strong>in</strong> comparison with normal peripheral pancreatic duct epithelia [48].DNMT1 overexpression <strong>in</strong> ductal adenocarc<strong>in</strong>omas of the pancreas is significantly correlatedwith the extent of <strong>in</strong>vasion to surround<strong>in</strong>g tissue, an advanced stage, and poorer patientoutcome [48]. The average number of methylated CpG islands of exam<strong>in</strong>ed tumor-suppressorgenes <strong>in</strong> microdissected specimens of peripheral pancreatic ductal epithelia with an <strong>in</strong>flammatorybackground, PanIN and ductal adenocarc<strong>in</strong>oma was significantly correlated with thelevel of DNMT1 prote<strong>in</strong> expression demonstrated immunohistochemically <strong>in</strong> preciselymicrodissected areas [49].33When the human DNMT3A and DNMT3B genes were first cloned, the expression levels ofDNMT1, 3A and 3B were reported <strong>in</strong> ten paired samples of normal and cancerous tissueobta<strong>in</strong>ed from various organs. Robertson et al. observed 2-fold overexpression of DNMT3A<strong>in</strong> five of ten samples, DNMT1 <strong>in</strong> six of ten samples, and DNMT3B <strong>in</strong> eight of ten samples, andDNMT3B clearly show<strong>in</strong>g the largest fold <strong>in</strong>creases among the three enzymes [50]. On theother hand, the cancer phenotype associated with accumulation of DNA methylation onC-type CpG islands is def<strong>in</strong>ed as the CpG-island methylator phenotype (CIMP) [51], and suchaccumulation is generally associated with frequent silenc<strong>in</strong>g of tumor-related genes due toDNA hypermethylation only, or a two-hit mechanism <strong>in</strong>volv<strong>in</strong>g DNA hypermethylation andLOH <strong>in</strong> human cancers of various organs [52]. Expression levels of DNMT1 mRNA and prote<strong>in</strong>are significantly correlated with poorer differentiation and CIMP <strong>in</strong> stomach cancers, but nosuch association has been observed for the expression of DNMT2, DNMT3A or DNMT3B [53].EBV <strong>in</strong>fection <strong>in</strong> stomach cancers is significantly associated with marked accumulation of DNAmethylation on C-type CpG islands and overexpression of DNMT1 prote<strong>in</strong>, although


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>Helicobacter pylori <strong>in</strong>fection, another etiologic factor strongly promot<strong>in</strong>g regional DNAhypermethylation, was not correlated with DNMT1 expression levels.Urothelial carc<strong>in</strong>omas (UCs) of the ur<strong>in</strong>ary bladder are cl<strong>in</strong>ically remarkable because of theirmulticentricity and tendency to recur due to a “field effect”. Even non-cancerous urotheliashow<strong>in</strong>g no remarkable histological changes obta<strong>in</strong>ed from patients with UCs can beconsidered precancerous, because they may have been exposed to carc<strong>in</strong>ogens <strong>in</strong> the ur<strong>in</strong>e.Our immunohistochemical exam<strong>in</strong>ations have clearly revealed that the <strong>in</strong>cidence of nuclearDNMT1 immunoreactivity is already higher <strong>in</strong> non-cancerous urothelia show<strong>in</strong>g no remarkablehistological changes obta<strong>in</strong>ed from patients with UCs, where the PCNA label<strong>in</strong>g <strong>in</strong>dexhad not yet <strong>in</strong>creased, compared to that <strong>in</strong> normal urothelia from patients without UCs,<strong>in</strong>dicat<strong>in</strong>g that DNMT1 overexpression was not a secondary result of <strong>in</strong>creased cell proliferativeactivity, but <strong>in</strong> fact preceded such activity [54]. The <strong>in</strong>cidence of nuclear DNMT1immunoreactivity showed a progressive <strong>in</strong>crease <strong>in</strong> dysplastic urothelia, and dur<strong>in</strong>g transitionto UCs, be<strong>in</strong>g significantly correlated with accumulation of DNA methylation on C-type CpGislands [55].34With respect to the mechanisms regulat<strong>in</strong>g the expression levels of DNMTs [56], the membersof the miR-29 family, <strong>in</strong>clud<strong>in</strong>g miR-29a, miR-29b and mir-29c, have been shown to directlytarget DNMT3A and DNMT3B [57]. Enforced expression of miR-29s <strong>in</strong> lung cancer cell l<strong>in</strong>esrestores the normal patterns of DNA methylation, <strong>in</strong>duces re-expression of methylationsilencedtumor-suppressor genes, and <strong>in</strong>hibits tumorigenicity <strong>in</strong> vitro and <strong>in</strong> vivo [57].Enforced expression of miR-29b <strong>in</strong> acute myeloid leukemia cells resulted <strong>in</strong> markedly reducedexpression of DNMT1, DNMT3A, and DNMT3B at both the RNA and prote<strong>in</strong> levels [58].Although down-regulation of DNMT3A and DNMT3B was the result of direct <strong>in</strong>teraction ofmiR-29b with the 3 0 UTRs of these genes, miR-29b down-regulates DNMT1 <strong>in</strong>directly bytarget<strong>in</strong>g Sp1, a transactivator of the DNMT1 gene [58]. miR-148 has been observed to b<strong>in</strong>d tothe cod<strong>in</strong>g region, outside the usual 3 0 UTR, of DNMT3B and to <strong>in</strong>duce splic<strong>in</strong>g alteration ofDNMT3B <strong>in</strong> human cancer cells [59]. DNMT1 may also be directly regulated by miR-148 [60]and miR-126 [61]. Down-regulated miR-152 <strong>in</strong>duces aberrant DNA methylation <strong>in</strong> HCC cellsby target<strong>in</strong>g DNMT1 [62]. In addition to miRNAs, Hu-antigen R (HuR) prote<strong>in</strong>s b<strong>in</strong>d to targetmRNAs and modify their levels of expression by alter<strong>in</strong>g their stability. HuR prote<strong>in</strong>s target the3 0 UTR of DNMT3B <strong>in</strong> human colon cancer cells, result<strong>in</strong>g <strong>in</strong> DNA hypermethylation of itstarget genes [63].3.5 MUTATIONS, POLYMORPHISM AND SPLICING ALTERATIONSOF DNMTS AND HUMAN CANCERSEven though our previous screen<strong>in</strong>g <strong>in</strong>dicated that mutations of DNMT1 are not the majorevent dur<strong>in</strong>g carc<strong>in</strong>ogenesis <strong>in</strong> the liver and stomach [64], recent massively parallel DNAsequenc<strong>in</strong>g has identified somatic mutations <strong>in</strong>clud<strong>in</strong>g missense mutations, frameshifts,splice-site mutations and large deletions, which were predicted to affect DNMT3A translation<strong>in</strong> acute myeloid leukemia cells [65]. The overall survival of patients show<strong>in</strong>g DNMT3Amutations was significantly shorter than that of patients without such mutations. Mutations ofthe DNMT3A gene, which reduce its enzymatic activity and alter the DNA methylation profiles,have also been reported <strong>in</strong> acute monocytic leukemia [66]. These observations add to theevidence for participation of aberrant DNMT activity <strong>in</strong> the pathogenesis of malignancies.DNMT3A gene polymorphism can affect transcriptional levels of DNMT3A and susceptibilityto cancers. The effect of a s<strong>in</strong>gle nucleotide polymorphism, A/G, <strong>in</strong> the DNMT3A promoterregion on transcriptional activity has been evaluated us<strong>in</strong>g a luciferase assay. Carriage of the Aallele conferred significantly higher promoter activity <strong>in</strong> comparison with the G allele, and AAhomozygotes had a six-fold <strong>in</strong>creased risk of gastric cancer [67]. Similarly, a marked associationbetween DNMT3B6 promoter C/T polymorphism and overall survival of patients


CHAPTER 3DNA Methylation Alterations <strong>in</strong> <strong>Human</strong> Cancerswith head and neck squamous cell carc<strong>in</strong>oma has been reported [68]: the homozygotes(CC-genotype and TT-genotype) survived significantly longer than the heterozygotes(CT-type). Such polymorphism may affect the gene expression profiles through dist<strong>in</strong>ct DNAmethylation patterns.Pericentromeric satellite regions are considered to be one of the specific targets of DNMT3B,s<strong>in</strong>ce Dnmt3Be/e mice lack DNA methylation <strong>in</strong> such regions and die <strong>in</strong> utero [6]. DNAhypomethylation <strong>in</strong> pericentromeric satellite regions is known to result <strong>in</strong> centromericdecondensation and enhanced chromosome recomb<strong>in</strong>ation. In fact, germl<strong>in</strong>e mutations ofthe DNMT3B gene have been reported <strong>in</strong> patients with immunodeficiency, centromeric<strong>in</strong>stability, and facial anomalies (ICF) syndrome, a rare recessive autosomal disorder characterizedby DNA hypomethylation of pericentromeric satellite regions [69]. In HCCs [70] andUCs [71], DNA hypomethylation of these regions is correlated with copy number alterationson chromosomes 1 and 9, respectively, where satellite regions are rich. The major splice variantof DNMT3B <strong>in</strong> normal liver tissue samples is DNMT3B3, which possesses the conservedcatalytic doma<strong>in</strong>s. DNMT activity of human DNMT3B3 has been confirmed <strong>in</strong> vitro [72]. Onthe other hand, DNMT3B4 lacks the conserved catalytic doma<strong>in</strong>s, although it reta<strong>in</strong>s theN-term<strong>in</strong>al doma<strong>in</strong> required for target<strong>in</strong>g to heterochromat<strong>in</strong> sites. Samples of normal livertissue show only a trace level of DNMT3B4 expression. The levels of DNMT3B4 mRNAexpression and the ratio of DNMT3B4 mRNA to DNMT3B3 <strong>in</strong> samples of non-cancerous livertissue obta<strong>in</strong>ed from patients with HCCs, and <strong>in</strong> HCCs themselves, are significantly correlatedwith the degree of DNA hypomethylation <strong>in</strong> pericentromeric satellite regions [73]. DNAdemethylation on satellite 2 has been observed <strong>in</strong> DNMT3B4-transfected human epithelial293 cells [73]. S<strong>in</strong>ce DNMT3B4 lack<strong>in</strong>g DNMT activity competes with DNMT3B3 for target<strong>in</strong>gto pericentromeric satellite regions, DNMT3B4 overexpression may lead to chromosomal<strong>in</strong>stability through <strong>in</strong>duction of DNA hypomethylation <strong>in</strong> such regions.As another molecular mechanism <strong>in</strong>volved <strong>in</strong> site-specific DNA methylation alterations,<strong>in</strong>teraction between DNMT3A and c-myc has been reported. This <strong>in</strong>teraction promotes thesite-specific methylation of CpG d<strong>in</strong>ucleotides localized <strong>in</strong> c-myc boxes <strong>in</strong> the promoterregions of the CDKN2a, CCND1 and TIMP2 genes [74]. The <strong>in</strong>validation of c-myc revealsthat c-myc allows recruitment of DNMT 3A on the c-myc box of c-myc-regulated genes.Monitor<strong>in</strong>g transcription factor arrays have identified transcription factors <strong>in</strong>teract<strong>in</strong>g withDNMT3A and DNMT3B (such as CREB and FOS), those <strong>in</strong>teract<strong>in</strong>g with DNMT 3A (suchas AP2alpha and p53) and those <strong>in</strong>teract<strong>in</strong>g with DNMT 3B (such as SP1 and SP4) [74].Thus, direct <strong>in</strong>teraction between DNMT 3A and/or DNMT 3B and transcription factorsprovides a rational molecular explanation for the mechanism of targeted DNAmethylation.353.6 SIGNAL PATHWAYS AFFECTING DNA METHYLATION STATUSDURING TUMORIGENESISMolecular l<strong>in</strong>ks between the major signal<strong>in</strong>g pathways <strong>in</strong>volved <strong>in</strong> tumorigenesis andepigenetic events have been reported [75]. For example, correlations between the phosphatidyl<strong>in</strong>ositol3-k<strong>in</strong>ase (PI3K)/AKT pathway and epigenetic events <strong>in</strong> tumorigenesis andprogression have been attract<strong>in</strong>g attention. It has been reported that PTEN methylationbecomes progressively higher from benign thyroid adenoma to follicular thyroid cancer and toaggressive anaplastic thyroid cancer, which harbors activat<strong>in</strong>g genetic alterations <strong>in</strong> the PI3K/AKT pathway that correspond to a progressively higher prevalence [76]. An association ofPTEN methylation with PIK3CA alterations and ras mutations has been reported <strong>in</strong> thyroidtumors [76]. Aberrant methylation and hence silenc<strong>in</strong>g of the PTEN gene, which coexists withactivat<strong>in</strong>g genetic alterations of the PI3K/AKT pathway, may enhance the signal<strong>in</strong>g of thispathway and contribute to tumor progression.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>With regard to BRAF-MEK signal<strong>in</strong>g, BRAF is highly expressed <strong>in</strong> neurons. Expression of MAP2,a neuron-specific microtubule-associated prote<strong>in</strong> that b<strong>in</strong>ds and stabilizes dendritic microtubules,is expressed <strong>in</strong> cutaneous primary melanomas and <strong>in</strong>versely associated with melanomaprogression. Ectopic expression of MAP2 <strong>in</strong> metastatic melanoma cells <strong>in</strong>hibits cell growth by<strong>in</strong>duc<strong>in</strong>g mitotic sp<strong>in</strong>dle defects and apoptosis [77]. Levels of MAP2 promoter activity <strong>in</strong>melanoma cell l<strong>in</strong>es are correlated with activat<strong>in</strong>g mutation <strong>in</strong> BRAF: hyperactivation ofBRAF-MEK signal<strong>in</strong>g activates MAP2 expression <strong>in</strong> melanoma cells through promoterdemethylation or down-regulation of the neuronal transcription repressor HES1 [77]. Thus,BRAF oncogene levels can regulate the neuronal differentiation and tumor progression ofmelanoma. Genome-wide DNA methylation analysis after shRNA knockdown of BRAF V600E<strong>in</strong> thyroid cancer cells has revealed numerous methylation targets <strong>in</strong>clud<strong>in</strong>g hyper- orhypo-methylated genes with metabolic and cellular functions [78]. Among such genes, theHMGB2 gene plays a role <strong>in</strong> thyroid cancer cell proliferation, and the FDG1 gene <strong>in</strong> cell <strong>in</strong>vasion[78]. A prom<strong>in</strong>ent epigenetic mechanism through which BRAF V600E can promotetumorigenesis is alteration of the expression of numerous important genes through DNAmethylation alterations.36The Ras signal<strong>in</strong>g pathway also regulates DNA methylation status. Forced expression ofa cDNA encod<strong>in</strong>g human GAP120 (hGAP), a down-modulator of Ras activity, or delta 9-Jun,a transdom<strong>in</strong>ant negative mutant of Jun, <strong>in</strong> adrenocortical tumor Y1 cells causes transformedcells to revert to their orig<strong>in</strong>al morphology, result<strong>in</strong>g <strong>in</strong> a reduced level of DNA methylationthrough a reduction of both mRNA expression and the enzymatic activity of DNMTs [79].Introduction of oncogenic Ha-ras <strong>in</strong>to GAP transfectants has been found to <strong>in</strong>crease the levelsof DNA methylation and DNMT activity. Moreover, transient transfection CAT assays havedemonstrated that the DNMT promoter <strong>in</strong> Y1 cells is activated by AP-1 and <strong>in</strong>hibited by downregulatorsof Ras signal<strong>in</strong>g [79]. In addition to Y1 cells, it has been reported that overexpressionof unmutated Ha-ras <strong>in</strong> human T cells causes an <strong>in</strong>crease <strong>in</strong> DNMT expression, andthat DNMT is decreased by <strong>in</strong>hibitory signal<strong>in</strong>g via the ras-MAPK pathway [80].The apoptosis-promot<strong>in</strong>g prote<strong>in</strong> Par-4 has been shown to be down-regulated <strong>in</strong> RastransformedNIH 3T3 fibroblasts through the Raf/MEK/ERK MAPK pathway. The par-4promoter is methylated <strong>in</strong> Ras-transformed cells through a MEK-dependent pathway, andtreatment with a DNMT <strong>in</strong>hibitor restores the levels of both the Par-4 mRNA transcript andprote<strong>in</strong>, suggest<strong>in</strong>g that the Ras-mediated down-regulation of Par-4 occurs through promotermethylation [81]. In fact, it has been revealed that Ras transformation is associated with upregulationof DNMT1 and DNMT3 expression [81].3.7 DNA METHYLATION AND HISTONE MODIFICATIONSDNA methylation determ<strong>in</strong>es chromat<strong>in</strong> configuration and regulates the expression levels ofgenes <strong>in</strong> cooperation with histone modifications [82,83]. Covalent histone modificationsmark active promoters (methylation of lys<strong>in</strong>e 4 of histone H3 [H3K4] and acetylation ofhistone H3 lys<strong>in</strong>e 27 [H3K27]), active enhancers (H3K4 methylation, H3K27 acetylation),actively transcribed genes (H3K36 methylation), or heterochromat<strong>in</strong> regions (H3K9 methylation,H3K27 methylation) [82,83]. When methyl-CpG-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s, such as MeCP2 andMBD2, b<strong>in</strong>d to methylated CpG d<strong>in</strong>ucleotide, their transcriptional repression doma<strong>in</strong> recruitsa co-repressor complex conta<strong>in</strong><strong>in</strong>g histone deacetylases (HDACs) [84]. On the other hand,histone methyltransferases, such as G9A and SUV39H1, are required to recruit DNMTs [85].Transcriptionally repressive chromat<strong>in</strong> modifications with<strong>in</strong> the promoters of tumorsuppressorgenes silenced by DNA methylation are known to resemble the chromat<strong>in</strong> modificationsof these genes <strong>in</strong> normal embryonic stem cells, e.g. polycomb (PcG) complex b<strong>in</strong>d<strong>in</strong>gand H3K27 methylation. These genes also have an active marker, H3K4 methylation, <strong>in</strong>normal stem cells, and this bivalent state is converted to a primary active or repressive


CHAPTER 3DNA Methylation Alterations <strong>in</strong> <strong>Human</strong> Cancerschromat<strong>in</strong> conformation after differentiation cues have been received [86]. Dur<strong>in</strong>g carc<strong>in</strong>ogenesis,such modifications may render the genes vulnerable to errors, result<strong>in</strong>g <strong>in</strong> aberrantDNA methylation. These PcG complexes have been shown to directly <strong>in</strong>teract with DNMTs,and possibly to promote cancer-specific gene silenc<strong>in</strong>g. EZH2, the PcG prote<strong>in</strong>s <strong>in</strong> the polycombrepressive complex 2/3 (PRC2/3) that catalyzes the trimethylation of H3K27, may bea key player [87]. Overexpression of EZH2 is correlated with tumor progression and poorerprognosis <strong>in</strong> various cancers [88,89]. Depletion of EZH2 <strong>in</strong> cancer cells leads to growth arrest[90]. CBX7, another PcG prote<strong>in</strong>, is a constituent of PRC1, and has also been shown toread the repressive histone marks, H3K9me3 and H3K27me3 [91]. Similarly to EZH2, CBX7 isable to recruit DNA methylation mach<strong>in</strong>ery to gene promoters and facilitate gene silenc<strong>in</strong>gdur<strong>in</strong>g the development of cancers.It has long been known that <strong>in</strong>dividual cancers each consist of heterogeneous cell populations.The recently proposed cancer stem cell hypothesis has emphasized that only certa<strong>in</strong>subpopulations, known as cancer stem cells, cancer-<strong>in</strong>itiat<strong>in</strong>g cells or tumor-propagat<strong>in</strong>g cells,have tumorigenic potential. These cancer-<strong>in</strong>itiat<strong>in</strong>g cells are usually resistant to chemotherapyand radiotherapy, lead<strong>in</strong>g to treatment failure. Moreover, they may be capable of form<strong>in</strong>gmetastatic foci <strong>in</strong> distant organs. Despite the existence of such subpopulations, the cancer stemcell hypothesis cont<strong>in</strong>ues to generate controversy. S<strong>in</strong>ce the PcG complex targets similar sets ofgenes <strong>in</strong> embryonic stem cells and cancer cells, much effort should be focused on howepigenetic mechanisms participate <strong>in</strong> the generation of cancer-<strong>in</strong>itiat<strong>in</strong>g cells [20,23].3.8 SUBCLASSIFICATION OF HUMAN CANCERS BASED ON DNAMETHYLATION PROFILINGAlmost all cancers are heterogeneous diseases composed of dist<strong>in</strong>ct cl<strong>in</strong>icopathologicalsubtypes. DNA methylation profiles may, at least partly, represent the molecular basis of eachsubtype [92,93]. Recently, analysis on a genome-wide scale has become possible us<strong>in</strong>g DNAmethylation-sensitive restriction enzyme-based or anti-methyl-cytos<strong>in</strong>e antibody aff<strong>in</strong>itytechniques that enrich the methylated and unmethylated fractions of genomic DNA [94].These fractions can then be hybridized to DNA microarrays. Such DNA methylation profil<strong>in</strong>gmay provide new <strong>in</strong>sight <strong>in</strong>to disease entities and help to provide more accurate classificationsof human cancers [23]. Such subclassification may yield clues for clarification of dist<strong>in</strong>ctmechanisms of carc<strong>in</strong>ogenesis <strong>in</strong> various organs, and identify possible target molecules forprevention and therapy <strong>in</strong> patients belong<strong>in</strong>g to specific clusters.37For example, progressive accumulation of genetic and epigenetic abnormalities has been bestdescribed <strong>in</strong> colon cancers. Cluster<strong>in</strong>g analyses based on either epigenetic (DNA methylationof multiple CpG island promoter regions) profil<strong>in</strong>g or a comb<strong>in</strong>ation of genetic (mutations ofBRAF, KRAS, and p53 and microsatellite <strong>in</strong>stability [MSI]) and epigenetic profil<strong>in</strong>g haverevealed dist<strong>in</strong>ct molecular signatures. Colon cancers were clustered <strong>in</strong>to CIMP1, CIMP2, andCIMP-negative groups based on DNA methylation data [95]. CIMP1 is characterized by MSIand BRAF mutations and rare KRAS and p53 mutations. CIMP2 is associated with KRASmutations and rare MSI, BRAF, or p53 mutations. CIMP-negative cases have a high rate of p53mutation and lower rates of MSI or mutation of BRAF or KRAS. Together, the data show thatcolon cancers can be grouped <strong>in</strong>to three molecularly dist<strong>in</strong>ct disease subclasses [95]. Thesethree groups also differ cl<strong>in</strong>ically: CIMP1 and CIMP2 are more often proximal, CIMP1 hasa good prognosis because it consists mostly of MSI-high cancers, and CIMP2 has a poorprognosis. Moreover, these groups may have dist<strong>in</strong>ct precancerous lesions that can bediagnosed endoscopically, such as serrated adenomas for CIMP1, and villous adenomasfor CIMP2.We focused on renal carc<strong>in</strong>ogenesis and exam<strong>in</strong>ed the DNA methylation status of C-type CpGislands of multiple tumor-related genes us<strong>in</strong>g bisulfite conversion. Even <strong>in</strong> non-cancerous


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>renal tissue show<strong>in</strong>g no remarkable histological changes obta<strong>in</strong>ed from patients withconventional-type clear cell renal cell carc<strong>in</strong>omas (RCCs), the average number of methylatedCpG islands was significantly higher than <strong>in</strong> normal renal tissue obta<strong>in</strong>ed from patientswithout any primary renal tumor, regardless of patient age [96]. Stepwise accumulation ofDNA methylation on CpG islands has been clearly shown to progress from normal renaltissue, to non-cancerous renal tissue show<strong>in</strong>g no remarkable histological changes obta<strong>in</strong>edfrom patients with RCCs, and to RCCs. S<strong>in</strong>ce it has not been possible to observe any histologicalchange <strong>in</strong> non-cancerous renal tissue obta<strong>in</strong>ed from patients with RCCs, and RCCsusually develop from backgrounds without chronic <strong>in</strong>flammation or persistent viral <strong>in</strong>fection,precancerous conditions <strong>in</strong> the kidney have been rarely described. However, from the viewpo<strong>in</strong>tof altered DNA methylation, we have shown that it is possible to recognize the presenceof precancerous conditions even <strong>in</strong> the kidney [96]. In other words, regional DNA methylationalterations may participate <strong>in</strong> the early and precancerous stage of multistage renal carc<strong>in</strong>ogenesis.Surpris<strong>in</strong>gly, the average number of methylated CpG islands <strong>in</strong> non-cancerous renaltissues obta<strong>in</strong>ed from patients with RCCs show<strong>in</strong>g higher histological grades was significantlyhigher than that <strong>in</strong> equivalent tissue obta<strong>in</strong>ed from patients with low-grade RCCs, suggest<strong>in</strong>gthat precancerous conditions show<strong>in</strong>g regional DNA hypermethylation may generate moremalignant RCCs [96].38In order to further clarify the significance of DNA methylation alterations dur<strong>in</strong>g renal carc<strong>in</strong>ogenesis,we performed genome-wide DNA methylation analysis us<strong>in</strong>g BAC array-basedmethylated CpG island amplification (BAMCA), which may be suitable, not for focus<strong>in</strong>gon specific promoter regions or <strong>in</strong>dividual CpG sites, but for overview<strong>in</strong>g the DNA methylationtendency of <strong>in</strong>dividual large regions among all chromosomes [92,93], <strong>in</strong> tissue samples. Theaverage numbers of BAC clones show<strong>in</strong>g DNA hypo- or hypermethylation <strong>in</strong> non-tumorousrenal tissue obta<strong>in</strong>ed from patients with chromophobe RCCs and oncocytomas were significantlylower than the average number <strong>in</strong> non-tumorous renal tissue obta<strong>in</strong>ed from patientswith clear cell RCCs [97]. In non-tumorous renal tissue from all exam<strong>in</strong>ed patients with renaltumors (clear cell RCCs, papillary RCCs, chromophobe RCCs and oncocytomas), biphasicaccumulation of DNA methylation alterations was evident. Among such patients, therecurrence-free survival rate of patients show<strong>in</strong>g DNA hypo- or hypermethylation on more BACclones <strong>in</strong> their non-tumorous renal tissue was significantly lower than that of patients show<strong>in</strong>gDNA hypo- or hypermethylation on fewer BAC clones [97]. Significant DNA methylationprofiles determ<strong>in</strong><strong>in</strong>g the histological subtype (chromophobe RCCs and oncocytomas vs clearcell RCCs) of future develop<strong>in</strong>g renal tumors and/or patient outcome (favorable outcome vspoorer outcome) may already be established at the precancerous stage.We performed two-dimensional unsupervised hierarchical cluster<strong>in</strong>g analysis based on thegenome-wide DNA methylation status (signal ratios obta<strong>in</strong>ed by BAMCA) of samples of noncancerousrenal tissue. On the basis of the DNA methylation profiles of these samples, thepatients with clear cell RCCs were clustered <strong>in</strong>to two subclasses, Clusters KA N and KB N [98].The correspond<strong>in</strong>g clear cell RCCs of patients <strong>in</strong> Cluster KB N showed more frequent macroscopicallyevident mult<strong>in</strong>odular growth, vascular <strong>in</strong>volvement and renal ve<strong>in</strong> tumor thrombi,and higher pathological tumor-node-metastasis (TNM) stages than those <strong>in</strong> Cluster KA N . OurClusters KA N and KB N <strong>in</strong> precancerous tissue can be considered cl<strong>in</strong>icopathologically valid: theoverall survival rate of patients <strong>in</strong> Cluster KB N was significantly lower than that of patients <strong>in</strong>Cluster KA N . DNA methylation alterations at the precancerous stage may even determ<strong>in</strong>e theoutcome of patients with clear cell RCCs.Two-dimensional unsupervised hierarchical cluster<strong>in</strong>g analysis based on BAMCA data for clearcell RCCs themselves was able to group patients <strong>in</strong>to two subclasses, Clusters KA T and KB T [98].Clear cell RCCs <strong>in</strong> Cluster KB T showed more frequent vascular <strong>in</strong>volvement and renal ve<strong>in</strong>tumor thrombi, and also higher pathological TNM stages than those <strong>in</strong> Cluster KA T . Theoverall survival rate of patients <strong>in</strong> Cluster KB T was significantly lower than that of patients <strong>in</strong>


CHAPTER 3DNA Methylation Alterations <strong>in</strong> <strong>Human</strong> CancersCluster KA T . Multivariate analysis revealed that our cluster<strong>in</strong>g was a predictor of recurrence andwas <strong>in</strong>dependent of histological grade, macroscopic configuration, vascular <strong>in</strong>volvement orpresence of renal ve<strong>in</strong> tumor thrombi.When we compared the DNA methylation profiles of non-cancerous renal tissue and those ofthe correspond<strong>in</strong>g clear cell RCC, Cluster KB N was completely <strong>in</strong>cluded <strong>in</strong> Cluster KB T .BACclones, of which DNA methylation status significantly discrim<strong>in</strong>ated Cluster KB N from ClusterKA N , also discrim<strong>in</strong>ated Cluster KB T from Cluster KA T without exception. When we exam<strong>in</strong>edeach of the representative BAC clones characteriz<strong>in</strong>g both Clusters KB N and KB T , the BAMCAsignal ratio <strong>in</strong> the non-cancerous renal tissue was at almost the same level as that <strong>in</strong> thecorrespond<strong>in</strong>g clear cell RCC develop<strong>in</strong>g <strong>in</strong> each <strong>in</strong>dividual patient [98]. Accord<strong>in</strong>gly, weconcluded that the genome-wide DNA methylation profiles of non-cancerous renal tissue arebasically <strong>in</strong>herited by each correspond<strong>in</strong>g clear cell RCC [99].The average number of exam<strong>in</strong>ed methylated C-type CpG islands was significantly higher <strong>in</strong>Cluster KB T than <strong>in</strong> Cluster KA T . The frequency of CIMP <strong>in</strong> Cluster KB T was significantly higherthan that <strong>in</strong> Cluster KA T . Genome-wide DNA methylation alterations consist<strong>in</strong>g of both hypoandhypermethylation of DNA revealed by BAMCA <strong>in</strong> Cluster KB T are associated with regionalDNA hypermethylation on CpG islands. Moreover, a subclass of Cluster KB N and KB T based onBAMCA data showed particularly marked accumulation of copy number alterations [100]:specific DNA methylation profiles at the precancerous stage may be closely related to, or maybe prone to, chromosomal <strong>in</strong>stability. DNA methylation alterations <strong>in</strong> precancerous conditions,which do not occur randomly but are prone to further accumulation of epigenetic andgenetic alterations, can generate more malignant cancers and even determ<strong>in</strong>e the outcome of<strong>in</strong>dividual patients [92] (Figure 3.1).With respect to urothelial carc<strong>in</strong>ogenesis, unsupervised hierarchical cluster<strong>in</strong>g of UCs basedon array comparative genomic hybridization (CGH) data clustered UCs <strong>in</strong>to threesubclasses, Clusters UA, UB 1 ,andUB 2 [101] (Figure 3.2). In Cluster UA, copy numberalterations, especially chromosomal ga<strong>in</strong>s, revealed by array CGH analysis, and DNA39FIGURE 3.1DNA methylation profiles <strong>in</strong> precancerous conditions andrenal cell carc<strong>in</strong>omas (RCCs). Two-dimensionalunsupervised hierarchical cluster<strong>in</strong>g analysis based on BACarray-based methylated CpG island amplification (BAMCA)data for non-cancerous renal tissue samples clusteredpatients with clear cell RCCs <strong>in</strong>to two subclasses, ClustersKA N and KB N [98]. On the basis of the DNA methylationprofiles of clear cell RCCs themselves, the patients with clearcell RCCs were divided <strong>in</strong>to Clusters KA T and KB T [98].Patients with more malignant RCCs and show<strong>in</strong>g a pooreroutcome were accumulated <strong>in</strong> Clusters KB N and KB T. TheDNA methylation profile of Cluster KB N was <strong>in</strong>herited bypatients with RCCs belong<strong>in</strong>g to Cluster KB T. Regional DNAhypermethylation of C-type CpG islands and copy numberalterations were accumulated <strong>in</strong> Cluster KB T . DNAmethylation alterations <strong>in</strong> precancerous conditions, such asthe DNA methylation profile correspond<strong>in</strong>g to Cluster KB N ,may be prone to further accumulation of epigenetic andgenetic alterations, thus generat<strong>in</strong>g more malignantcancers, such as the RCCs <strong>in</strong> patients belong<strong>in</strong>g to ClusterKB T . This figure is reproduced <strong>in</strong> the color plate section.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>FIGURE 3.2Hierarchical cluster<strong>in</strong>g analysis of urothelial carc<strong>in</strong>omas(UCs) based on array comparative genomic hybridization(CGH) data. In Cluster UA, copy number alterations,especially chromosomal ga<strong>in</strong>s, revealed by array CGHanalysis, and DNA hypomethylation revealed by BAMCAwere both accumulated <strong>in</strong> a genome-wide manner [109].Cluster UB 1 showed accumulation of regional DNAhypermethylation on C-type CpG islands [109]. In Cluster B 2 ,the number of BAC clones show<strong>in</strong>g both DNA hypo- andhypermethylation by BAMCA was rather high, and thenumber of probes show<strong>in</strong>g loss or ga<strong>in</strong> by array CGH wasrather low, <strong>in</strong> comparison to Cluster UB 1 [109]. Genetic andepigenetic events appear to accumulate <strong>in</strong> a complexmanner dur<strong>in</strong>g the developmental stage of <strong>in</strong>dividual tumors.This figure is reproduced <strong>in</strong> the color plate section.40hypomethylation revealed by BAMCA, were both accumulated <strong>in</strong> a genome-wide manner,suggest<strong>in</strong>g that DNA hypomethylation may result <strong>in</strong> chromosomal <strong>in</strong>stability throughchanges <strong>in</strong> chromat<strong>in</strong> configuration and enhancement of chromosomal recomb<strong>in</strong>ation[101]. Cluster UB1 showed accumulation of regional DNA hypermethylation on C-type CpGislands. Silenc<strong>in</strong>g of tumor-related genes due to DNA hypermethylation and chromosomallosses may be critical for the development of UCs belong<strong>in</strong>g to Cluster UB1 [101]. In ClusterUB2, the number of BAC clones shown by BAMCA to have both DNA hypo- and hypermethylationwas rather high, and the number of probes shown by array CGH to have loss orga<strong>in</strong> was rather low, <strong>in</strong> comparison to Cluster UB1 [101]. In addition to copy numberalterations, genome-wide DNA methylation alterations may also participate <strong>in</strong> the developmentof UCs belong<strong>in</strong>g to Cluster UB 2 . Taken together, the data suggest that genetic andepigenetic events accumulate <strong>in</strong> a complex manner dur<strong>in</strong>g the developmental stage of<strong>in</strong>dividual UCs (Figure 3.2).3.9 DIAGNOSIS OF CANCERS IN BODY FLUIDS AND BIOPSYSPECIMENS BASED ON DNA METHYLATION PROFILESThe <strong>in</strong>cidence of DNA methylation alterations is generally high <strong>in</strong> human cancers derivedfrom various organs. Therefore, DNA methylation alterations are applicable as biomarkers forearly diagnosis of patients with cancers [102]. Cancer diagnosis based on DNA methylationalterations was <strong>in</strong>itially attempted us<strong>in</strong>g body fluids, such as ur<strong>in</strong>e, that can be collected non<strong>in</strong>vasively.For example, DNA hypermethylation of regulatory sequences at the GSTP1 genelocus is present <strong>in</strong> the majority of primary prostate carc<strong>in</strong>omas, but not <strong>in</strong> normal prostatictissue or other normal tissues. Matched specimens of primary tumor, peripheral bloodlymphocytes, and simple voided ur<strong>in</strong>e were collected from patients with prostate cancers atvarious cl<strong>in</strong>ical stages, and the DNA methylation status of GSTP1 was exam<strong>in</strong>ed us<strong>in</strong>gmethylation-specific PCR [102]. Decod<strong>in</strong>g of the results <strong>in</strong>dicated that ur<strong>in</strong>e from prostatecancer patients conta<strong>in</strong>ed shed cancer cells or debris. Furthermore, there was no case whereur<strong>in</strong>e-sediment DNA harbored methylation when the correspond<strong>in</strong>g tumor was negative,suggest<strong>in</strong>g the feasibility of molecular diagnosis us<strong>in</strong>g DNA methylation status as an <strong>in</strong>dicatorof prostatic cancer cells <strong>in</strong> ur<strong>in</strong>e [103].Quantitative analysis has been <strong>in</strong>troduced for cancer diagnosis based on DNA methylationalterations. For example, quantitative fluorogenic real-time PCR assay has been used to


CHAPTER 3DNA Methylation Alterations <strong>in</strong> <strong>Human</strong> Cancersexam<strong>in</strong>e primary tumor DNA and ur<strong>in</strong>e sediment DNA from patients with UCs of theur<strong>in</strong>ary bladder for promoter hypermethylation of multiple genes <strong>in</strong> order to identifypotential biomarkers for bladder cancer [104]. The promoter methylation pattern <strong>in</strong> ur<strong>in</strong>egenerally matched that <strong>in</strong> the primary tumors. A selected gene panel <strong>in</strong>clud<strong>in</strong>g CDKN2A,MGMT, and GSTP1 was validated <strong>in</strong> ur<strong>in</strong>e-sediment DNA samples from an additionalvalidation cohort of patients with UCs of various stages and grades, and from additionalage-matched control subjects [104]. Test<strong>in</strong>g of such a gene panel us<strong>in</strong>g quantitativemethylation-specific PCR assay has been shown to be a powerful non-<strong>in</strong>vasive approach fordetection of cancers.DNA methylation may become an alternative biomarker which can compensate for thedemerits of conventional diagnostic techniques. Gastro<strong>in</strong>test<strong>in</strong>al endoscopy followed bypathological diagnosis of biopsy specimens is useful for diagnosis of stomach cancers.However, the diagnostic power depends on the technical skill of the endoscopist. Endoscopicbiopsy is a topical procedure whereby only a small portion of the lesion is removed. Moreover,gastro<strong>in</strong>test<strong>in</strong>al endoscopy is neither comfortable nor risk-free for patients, and is associatedwith frequent morbidity. Therefore, a method for sensitive and specific detection of earlygastric cancer has been established us<strong>in</strong>g DNA methylation analysis of gastric washes [105].This revealed a close correlation between the DNA methylation level of the MINT 25 locus <strong>in</strong>tumor biopsy specimens and that <strong>in</strong> gastric washes. MINT25 methylation had high sensitivity,specificity, and area under the receiver operat<strong>in</strong>g characteristic curve for tumor cell detection <strong>in</strong>gastric washes [105]. In addition, even when compared with potential prote<strong>in</strong> or mRNAbiomarkers <strong>in</strong> gastric washes, DNA methylation <strong>in</strong> such samples may be optimal because of itsstability and amplifiability.In general, pancreatic biopsy yields only a small amount of tissue, and <strong>in</strong> specimens ofpancreatic juice the cellular morphology is not well preserved due to degeneration. We appliedthe BAMCA method to normal pancreatic tissue obta<strong>in</strong>ed from patients without ductaladenocarc<strong>in</strong>omas, non-cancerous pancreatic tissue obta<strong>in</strong>ed from patients with ductaladenocarc<strong>in</strong>omas, and cancerous tissue. The results of BAMCA for normal pancreatic tissuesamples reflected the DNA methylation profiles of normal peripheral pancreatic duct epithelia(the orig<strong>in</strong> of ductal adenocarc<strong>in</strong>omas), ac<strong>in</strong>ar cells and islet cells. In samples of noncancerouspancreatic tissue obta<strong>in</strong>ed from patients with ductal adenocarc<strong>in</strong>omas, BAMCArevealed DNA hypo- or hypermethylation on many BAC clones <strong>in</strong> comparison to normalpancreatic tissue samples. Microscopic observation of non-cancerous pancreatic tissue samplesobta<strong>in</strong>ed from patients with ductal adenocarc<strong>in</strong>omas revealed lymphocytes and fibroblastsassociated with various degrees of chronic pancreatitis, which is considered to be one of theprecancerous conditions for ductal adenocarc<strong>in</strong>omas (Figure 3.3). Our previous studies us<strong>in</strong>gmicrodissection and immunohistochemistry revealed accumulation of DNA hypermethylationof tumor-related genes associated with DNMT1 overexpression, even <strong>in</strong> peripheralpancreatic duct epithelia at the precancerous stage [48,49]. Therefore, the results ofBAMCA for samples of non-cancerous pancreatic tissue from patients with ductal adenocarc<strong>in</strong>omasmay reflect the DNA methylation profiles of peripheral pancreatic duct epithelia atthe precancerous stage, lymphocytes, fibroblasts, ac<strong>in</strong>ar cells, and islet cells. In order todiagnose ductal adenocarc<strong>in</strong>omas <strong>in</strong> tissue samples, cancer-specific DNA methylation profilesshould be discrim<strong>in</strong>ated from those of normal or precancerous peripheral pancreatic ductepithelia, lymphocytes, fibroblasts, ac<strong>in</strong>ar cells, and islet cells. Therefore, we identified 12 BACclones whose DNA methylation status was able to discrim<strong>in</strong>ate cancerous tissue samples fromboth normal pancreatic tissue and non-cancerous pancreatic tissue samples obta<strong>in</strong>ed frompatients with ductal adenocarc<strong>in</strong>omas <strong>in</strong> the learn<strong>in</strong>g cohort with a specificity of 100%. Us<strong>in</strong>gthe criteria that comb<strong>in</strong>ed these 12 BAC clones, cancerous tissues were precisely diagnosedwith 100% sensitivity and specificity <strong>in</strong> both the learn<strong>in</strong>g and validation cohorts [106]. Ourdiagnostic criteria may be advantageous for support<strong>in</strong>g the histological and cytologicalassessment of pancreatic cancers (Figure 3.3).41


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>42FIGURE 3.3Diagnostic criteria based on DNA methylation profiles for ductal adenocarc<strong>in</strong>omas of the pancreas. In order to diagnose ductal adenocarc<strong>in</strong>omas <strong>in</strong> tissuesamples, cancer-specific DNA methylation profiles should be discrim<strong>in</strong>ated from those of normal and precancerous peripheral pancreatic duct epithelia,<strong>in</strong>flammatory cells, fibroblasts, ac<strong>in</strong>ar cells, and islet cells. Therefore, we identified 12 BAC clones for which the DNA methylation status was able todiscrim<strong>in</strong>ate cancerous tissue (T) samples from both normal pancreatic tissue obta<strong>in</strong>ed from patients without pancreatic cancers (C) and non-cancerouspancreatic tissue from patients with cancers (N). Us<strong>in</strong>g criteria that comb<strong>in</strong>ed these 12 BAC clones, cancerous tissue samples were precisely diagnosed with100% sensitivity and specificity <strong>in</strong> both the learn<strong>in</strong>g and validation cohorts [106]. In general, pancreatic biopsy yields only a small amount of tissue, and <strong>in</strong>specimens of pancreatic juice the cellular morphology is not well preserved due to degeneration. Our diagnostic criteria may be advantageous for support<strong>in</strong>g thehistological and cytological assessment of such specimens. This figure is reproduced <strong>in</strong> the color plate section.3.10 CARCINOGENETIC RISK ESTIMATION BASED ON DNAMETHYLATION PROFILESDNA methylation alterations play a role even <strong>in</strong> the early and precancerous stage dur<strong>in</strong>gmultistage carc<strong>in</strong>ogenesis. S<strong>in</strong>ce even subtle alterations of DNA methylation profiles at theprecancerous stage are stably preserved on DNA double strands by covalent bonds, they maybe better <strong>in</strong>dicators for risk estimation than mRNA and prote<strong>in</strong> expression profiles, which canbe easily affected by the microenvironment of precursor cells. Personalized prevention byelim<strong>in</strong>ation of <strong>in</strong>flammatory conditions, viruses, and other microorganisms, together withprohibition of smok<strong>in</strong>g, which causes DNA methylation alterations, may be applicable topatients with precancerous conditions.S<strong>in</strong>ce HCC usually develops <strong>in</strong> liver already affected by chronic hepatitis or liver cirrhosisassociated with HBV and/or HCV <strong>in</strong>fection, the prognosis of patients with HCC is deemedpoor unless the cancer is diagnosed at an early stage. Therefore, surveillance at the precancerousstage becomes a priority. In cl<strong>in</strong>ical practice, especially <strong>in</strong>tensive surveillance shouldbe performed for patients at high risk of HCC development, even if the patients areasymptomatic. Therefore, we applied the BAMCA method to samples of liver tissue. Wilcoxontest showed that 25 BAC clones, whose DNA methylation status was <strong>in</strong>herited by HCCs from


CHAPTER 3DNA Methylation Alterations <strong>in</strong> <strong>Human</strong> Cancersnon-cancerous liver tissue <strong>in</strong> patients with HCCs, were able to discrim<strong>in</strong>ate such noncancerousliver tissue from normal liver tissue obta<strong>in</strong>ed from patients without HCCs. Thecriteria for carc<strong>in</strong>ogenetic risk estimation that comb<strong>in</strong>ed the 25 BAC clones allowed diagnosisof non-cancerous liver tissue from patients with HCCs <strong>in</strong> the learn<strong>in</strong>g cohort as be<strong>in</strong>g at highrisk of carc<strong>in</strong>ogenesis with 100% sensitivity and specificity [107]. In the validation cohort,these criteria allowed such discrim<strong>in</strong>ation with 96% sensitivity and specificity [107]. Inpatients with HCCs, there were no significant differences <strong>in</strong> DNA methylation status <strong>in</strong> these25 BAC clones between samples of non-cancerous liver tissue show<strong>in</strong>g chronic hepatitis andthose show<strong>in</strong>g cirrhosis, <strong>in</strong>dicat<strong>in</strong>g that the criteria we employed were not associated with<strong>in</strong>flammation or fibrosis. In addition, the average number of BAC clones satisfy<strong>in</strong>g thesecriteria was significantly lower <strong>in</strong> liver tissue from patients with HBV or HCV <strong>in</strong>fection butwithout HCCs than <strong>in</strong> non-cancerous liver tissue from patients with HCCs. DNA methylationstatus <strong>in</strong> these 25 BAC clones does not simply depend on hepatitis virus <strong>in</strong>fection butmay actually reflect the risk of carc<strong>in</strong>ogenesis itself. Therefore, our criteria not onlydiscrim<strong>in</strong>ate non-cancerous liver tissue from patients with HCCs from normal liver tissues,but may be capable of discrim<strong>in</strong>at<strong>in</strong>g patients who may or may not develop HCCs fromamong those who are be<strong>in</strong>g followed up for HBV or HCV <strong>in</strong>fections, chronic hepatitis, orcirrhosis.Next, to precisely identify the CpG sites hav<strong>in</strong>g the largest diagnostic impact on each of the25 BAC clones and to improve the sensitivity and specificity of carc<strong>in</strong>ogenetic risk estimation,we quantitatively evaluated the DNA methylation status of 203 Sma I sites on these 25 BACclones us<strong>in</strong>g highly quantitative pyrosequenc<strong>in</strong>g of tissue specimens. In order to overcomePCR bias, we optimized the PCR conditions for each pyrosequenc<strong>in</strong>g primer set. It wasrevealed that 30 regions <strong>in</strong>clud<strong>in</strong>g 45 CpG sites had the largest diagnostic impact. Us<strong>in</strong>g these30 regions, we then established criteria revised on the basis of pyrosequenc<strong>in</strong>g for estimationof carc<strong>in</strong>ogenetic risk [108]. The revised criteria allowed diagnosis of all samples of noncancerousliver tissue obta<strong>in</strong>ed from HCC patients <strong>in</strong> the validation cohort as be<strong>in</strong>g at high riskof carc<strong>in</strong>ogenesis, with improved sensitivity and specificity [108]. It is feasible that only oneCpG site <strong>in</strong> the promoter region was <strong>in</strong>cluded <strong>in</strong> the revised criteria, because DNA methylationstatus <strong>in</strong> genomic regions, which do not directly participate <strong>in</strong> gene silenc<strong>in</strong>g, may be altered atthe precancerous stage before alterations <strong>in</strong> the promoter regions themselves occur. Many CpGsites with evident diagnostic impact are located with<strong>in</strong> non-CpG islands, gene bodies, andnon-cod<strong>in</strong>g regions that have been overlooked as DNA methylation biomarkers. Meticulousexam<strong>in</strong>ation of such regions may be important for identify<strong>in</strong>g optimal <strong>in</strong>dicators of carc<strong>in</strong>ogeneticrisk.43Dur<strong>in</strong>g the surveillance period, <strong>in</strong> order to clarify the basel<strong>in</strong>e liver histology, liver biopsy isperformed <strong>in</strong> patients with HBV or HCV <strong>in</strong>fection prior to <strong>in</strong>terferon therapy. Therefore,carc<strong>in</strong>ogenetic risk estimation us<strong>in</strong>g such liver biopsy specimens will be advantageous for closefollow-up of patients who are at high risk of HCC development. We have confirmed thatcarc<strong>in</strong>ogenetic risk estimation us<strong>in</strong>g pyrosequenc<strong>in</strong>g is applicable to rout<strong>in</strong>e formal<strong>in</strong>-fixed,paraff<strong>in</strong>-embedded liver biopsy specimens. Our next step is to validate the reliability of suchrisk estimation prospectively us<strong>in</strong>g liver biopsy specimens obta<strong>in</strong>ed prior to <strong>in</strong>terferon therapyfrom a large cohort of patients with HBV or HCV <strong>in</strong>fection.As mentioned above, UC is cl<strong>in</strong>ically remarkable because of its multicentricity due to the“field effect”. Even non-cancerous urothelia show<strong>in</strong>g no remarkable histological changesobta<strong>in</strong>ed from patients with UCs can be considered to be at the precancerous stage,because they may have been exposed to carc<strong>in</strong>ogens <strong>in</strong> the ur<strong>in</strong>e. In fact, pr<strong>in</strong>cipalcomponent analysis based on BAMCA data have revealed progression of DNA methylationalterations from normal urothelia to non-cancerous urothelia obta<strong>in</strong>ed from patients withUCs, and to UCs themselves. Unsupervised hierarchical cluster<strong>in</strong>g analysis of patients withUCs based on the DNA methylation status of their non-cancerous urothelia showed that


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>the DNA methylation profiles of non-cancerous urothelia were significantly correlated withthe <strong>in</strong>vasiveness of UCs develop<strong>in</strong>g <strong>in</strong> <strong>in</strong>dividual patients, suggest<strong>in</strong>g that DNA methylationalterations at the precancerous stage may generate more malignant cancers [109]. Thecomb<strong>in</strong>ation of DNA methylation status on 83 BAC clones was able to completelydiscrim<strong>in</strong>ate between non-cancerous urothelia from patients with UCs and normalurothelia, and allowed diagnosis of non-cancerous urothelia from patients with UCs ashav<strong>in</strong>g a high risk of carc<strong>in</strong>ogenesis, with 100% sensitivity and specificity [109]. Differences<strong>in</strong> DNA methylation profiles between muscle-<strong>in</strong>vasive UCs and non-<strong>in</strong>vasive UCs have alsobeen extensively exam<strong>in</strong>ed: frequent DNA hypermethylation of the HOXB2 [110] andRASSF1A [111] genes is known to be associated with <strong>in</strong>vasiveness of UCs.443.11 PERSONALIZED MEDICINE BASED ON DNA METHYLATIONPROFILES: PROGNOSTICATION OF PATIENTS WITH CANCERS ANDPREDICTION OF RESPONSE TO CHEMOTHERAPYS<strong>in</strong>ce DNA methylation alterations frequently correlate with cl<strong>in</strong>icopathological parameters ofcancers, they can be used as prognostic <strong>in</strong>dicators <strong>in</strong> patients with cancers. For example, basedon BAMCA data, 41 BAC clones, whose DNA methylation status was able to discrim<strong>in</strong>ate HCCpatients who survived more than 4 years after hepatectomy from patients who sufferedrecurrence with<strong>in</strong> 6 months and died with<strong>in</strong> a year after hepatectomy, have been identified[107]. The DNA methylation status of these 41 BAC clones was correlated with the cancer-freesurvival rate of HCC patients <strong>in</strong> the validation cohort. Prognostication based on our criteriamay be promis<strong>in</strong>g for supportive use dur<strong>in</strong>g follow-up after surgical resection, s<strong>in</strong>ce multivariateanalysis revealed that our criteria are able to predict overall patient outcome <strong>in</strong>dependentlyof parameters observed <strong>in</strong> hepatectomy specimens, such as the degree ofhistological differentiation, presence of portal ve<strong>in</strong> tumor thrombi, <strong>in</strong>trahepatic metastasisand multicentricity, which are already known to have a prognostic impact. Such prognosticationus<strong>in</strong>g liver biopsy specimens obta<strong>in</strong>ed before transarterial embolization, transarterialchemoembolization, and radiofrequency ablation may be advantageous even for patients whoundergo such therapies.Even when surgery is performed with curative <strong>in</strong>tent for patients with pancreatic cancers,therateofrecurrenceisveryhigh.Althoughpreviousstudieshavesuggestedtheefficacyofadjuvant chemotherapy, it needs to be carried out carefully, pay<strong>in</strong>g close attention toadverse reactions. In order to decide the <strong>in</strong>dications for such adjuvant chemotherapy,prognostic criteria should be explored. We have identified 11 BAC clones whose DNAmethylation status was able to discrim<strong>in</strong>ate patients show<strong>in</strong>g early relapse from thosewithout relapse <strong>in</strong> the learn<strong>in</strong>g cohort with 100% specificity, and this was correlated withthe recurrence-free and overall survival rates <strong>in</strong> the validation cohort [106]. Multivariateanalysis revealed that satisfy<strong>in</strong>g the prognostic criteria us<strong>in</strong>g these 11 BAC clones wasa parameter <strong>in</strong>dependent of surgical marg<strong>in</strong> positivity and lymph node metastasis at thetime of surgery [106].The quality of life of patients with ur<strong>in</strong>ary bladder cancers is generally poor after totalcystectomy. In general, therefore, after therapeutic diagnosis of UC tumors obta<strong>in</strong>ed by transurethralresection, patients are followed-up by repeat cystoscopy exam<strong>in</strong>ations. In patientsshow<strong>in</strong>g sudden prom<strong>in</strong>ent malignant progression, it is difficult to determ<strong>in</strong>e the appropriatetim<strong>in</strong>g of total cystectomy. Therefore, prognostic <strong>in</strong>dicators need to be explored. Thecomb<strong>in</strong>ation of DNA methylation status on 20 BAC clones selected by Wilcoxon test was ableto completely discrim<strong>in</strong>ate patients who suffered recurrence after surgery from patients whodid not [109]. DNA methylation profil<strong>in</strong>g may thus provide optimal <strong>in</strong>dicators for prognostication<strong>in</strong> patients with UCs. Other recently published DNA methylation alterations <strong>in</strong>


CHAPTER 3DNA Methylation Alterations <strong>in</strong> <strong>Human</strong> CancersTABLE 3.1 DNA methylation alterations <strong>in</strong> human cancers that are correlated with the outcome ofpatients and can be used as prognostic <strong>in</strong>dicatorsTumor Gene DNA Methylation Status ReferencesGlioma MGMT DNA hypermethylation PLoS One 2011; 6: e23332J Neurooncol 2011; 102: 311e16LINE-1 DNA hypomethylation PLoS One 2011; 6: e23332POTEH DNA hypomethylation Bra<strong>in</strong> Res 2011; 1391: 125e31Head and neckcancermiRNA-137 DNA hypermethylation Cancer 2011; 117: 1454e62ESR1 DNA hypermethylation Cl<strong>in</strong> <strong>Epigenetics</strong> 2010; 1: 61e69HIC1 DNA hypermethylation Cl<strong>in</strong> <strong>Epigenetics</strong> 2010; 1: 61e69LATS2 DNA hypomethylation BMC Cancer 2010; 10: 538p16 DNA hypermethylation Oral Oncol 2010; 46: 734e9Salivary gland RUNX3 DNA hypermethylation Cancer Sci 2011; 102: 492e7cancerLung cancer RASSF1A DNA hypermethylation Carc<strong>in</strong>ogenesis 2011; 32: 411e16.Esophagealcancerp 14 DNA hypermethylation J Cl<strong>in</strong> Pathol 2011; 64: 246e51p 15 DNA hypermethylation J Cl<strong>in</strong> Pathol 2011; 64: 246e51p 16 DNA hypermethylation J Cl<strong>in</strong> Pathol 2011; 64: 246e51p 21 DNA hypermethylation J Cl<strong>in</strong> Pathol 2011; 64: 246e51p 27 DNA hypermethylation J Cl<strong>in</strong> Pathol 2011; 64: 246e51p 57 DNA hypermethylation J Cl<strong>in</strong> Pathol 2011; 64: 246e51p 73 DNA hypermethylation J Cl<strong>in</strong> Pathol 2011; 64: 246e51PAX6 DNA hypermethylation Ann Surg Oncol 2011; 18: 1185e94ENST00000363328 DNA hypermethylation Ann Surg Oncol 2011; 18: 1185e94Stomach cancer Claud<strong>in</strong>-4 DNA hypomethylation Lab Invest 2011; 91: 1652e67BNIP3 DNA hypermethylation Oncol Rep 2011; 25: 513e18DAPK DNA hypermethylation Oncol Rep 2011; 25: 513e18S100A6 DNA hypomethylation Am J Pathol 2010; 177: 586e97EphA1 DNA hypermethylation Oncol Rep 2010; 24: 1577e84Colorectal cancer fibul<strong>in</strong>-3b DNA hypermethylation Neoplasma 2011; 58: 441e8p16 DNA hypermethylation Anticancer Res 2011; 31: 1643e6Oncol Rep 2011; 25: 789e94LINE-1 DNA hypomethylation Cancer 2011; 117: 1847e54RASSF1A DNA hypermethylation J Cell Physiol 2011; 226: 1934e9SFRP2 DNA hypermethylation Cl<strong>in</strong> Invest Med 2011; 34: E88e95DSC3 DNA hypermethylation Br J Cancer 2011; 104: 1013e19IGFBP3 DNA hypermethylation Cl<strong>in</strong> Cancer Res 2011; 17: 1535e45EVL DNA hypermethylation Cl<strong>in</strong> Cancer Res 2011; 17: 1535e45hMLH1 DNA hypermethylation Oncol Rep 2011; 25: 789e94PPARG DNA hypermethylation PLoS One 2010; 5: e14229MGMT DNA hypermethylation Cancer Causes Control 2011; 22: 301e9IGF2 DNA hypomethylation Gastroenterology 2010; 139: 1855e64RARb2 DNA hypermethylation Tumour Biol 2010; 31: 503e11Gastro<strong>in</strong>test<strong>in</strong>alstromal tumorHepatocellularcarc<strong>in</strong>omaRenal cellcarc<strong>in</strong>omaREC8 DNA hypermethylation Gut 2012; 61: 392e401PAX3 DNA hypermethylation Gut 2012; 61: 392e401p16 DNA hypermethylation Gut 2012; 61: 392e401RASSF1A DNA hypermethylation Asian Pac J Cancer Prev 2010; 11: 1677e81CADM1 DNA hypermethylation Oncol Rep 2011; 25: 1053e62WIF-1 DNA hypermethylation Tumour Biol 2011; 32: 233e40RELN DNA hypermethylation Ann Surg Oncol 2011; 18: 572e9HOXA5 DNA hypermethylation Pathol Int 2010; 60: 661e6MSH2 DNA hypermethylation Pathol Int 2010; 60: 661e6hsa-miR-9 DNA hypermethylation Oncogene 2010; 29: 5724e8Neuroblastoma CASP8 DNA hypermethylation Mol Carc<strong>in</strong>og 2011; 50: 153e62TMS1 DNA hypermethylation Mol Carc<strong>in</strong>og 2011; 50: 153e62APAF1 DNA hypermethylation Mol Carc<strong>in</strong>og 2011; 50: 153e6245Cont<strong>in</strong>ued


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>TABLE 3.1 dcont<strong>in</strong>uedTumor Gene DNA Methylation Status ReferencesBreast cancer Endogl<strong>in</strong> DNA hypermethylation Oncogene 2011; 30: 1046e58RASSF1A DNA hypermethylation Breast Cancer Res Treat 2011; 129: 1e9CDO1 DNA hypermethylation BMC Cancer 2010; 10: 247Cervical cancer APC1A DNA hypermethylation Int J Oncol 2011; 39: 683e8Endometrioid CDH1 DNA hypermethylation Cancer Invest 2011; 29: 86e92cancerOvarian cancer GREB1 DNA hypomethylation Oncology 2011; 80: 12e20TGIF DNA hypomethylation Oncology 2011; 80: 12e20TOB1 DNA hypomethylation Oncology 2011; 80: 12e20TMCO5 DNA hypermethylation Oncology 2011; 80: 12e20PTPRN DNA hypermethylation Oncology 2011; 80: 12e20GUCY2C DNA hypermethylation Oncology 2011; 80: 12e20HERV-K DNA hypomethylation Int J Gynecol Cancer 2011; 21: 51e7Trophoblastic ASPP1 DNA hypermethylation Mod Pathol 2011; 24: 522e32tumorMelanoma LINE-1 DNA hypomethylation J Transl Med 2011; 9: 78Acute myeloid CEBPA DNA hypermethylation Leukemia 2011; 25: 32e40leukemiaMultiple myeloma p16 DNA hypermethylation Ann Hematol 2011; 90: 73e9human cancers that are correlated with patient outcome and can be used as prognostic<strong>in</strong>dicators are summarized <strong>in</strong> Table 3.1.46In addition, DNA methylation profiles may be predictive <strong>in</strong>dicators of response to chemotherapy.One such example is silenc<strong>in</strong>g of the mitotic checkpo<strong>in</strong>t gene CHFR (checkpo<strong>in</strong>t withforkhead and r<strong>in</strong>g f<strong>in</strong>ger doma<strong>in</strong>s) <strong>in</strong> gastric cancers. Mitotic checkpo<strong>in</strong>ts prevent errors <strong>in</strong>chromosome segregation that can lead to neoplasia, and it is notable that gastric cancers oftenshow impaired checkpo<strong>in</strong>t function. CHFR expression was silenced by DNA methylation ofthe 5 0 region of the gene <strong>in</strong> tested gastric cancer cell l<strong>in</strong>es and primary gastric cancers;expression was restored by treatment with 5-aza-2 0 -deoxycytid<strong>in</strong>e. In addition, histones H3and H4 were found to be deacetylated <strong>in</strong> cell l<strong>in</strong>es show<strong>in</strong>g aberrant methylation. Cells notexpress<strong>in</strong>g CHFR showed impaired checkpo<strong>in</strong>t function, lead<strong>in</strong>g to nuclear localization ofcycl<strong>in</strong> B1 after treatment with microtubule <strong>in</strong>hibitors such as docetaxel or paclitaxel. Absenceof CHFR appears to be associated with the sensitivity of cells to mitotic stress caused bymicrotubule <strong>in</strong>hibition, and restoration of CHFR expression by 5-aza-2 0 -deoxycytid<strong>in</strong>e oradenoviral gene transfer restores the checkpo<strong>in</strong>t. By affect<strong>in</strong>g mitotic checkpo<strong>in</strong>t function,CHFR <strong>in</strong>activation likely plays a key role <strong>in</strong> gastric cancer tumorigenesis [112]. Moreover,aberrant methylation of CHFR appears to be a good molecular marker with which to predictthe sensitivity of gastric cancers to microtubule <strong>in</strong>hibitors.Another example is MGMT, a DNA repair prote<strong>in</strong>, which reverses the addition of alkyl groupsto the guan<strong>in</strong>e base of DNA. Silenc<strong>in</strong>g of MGMT due to DNA methylation <strong>in</strong> glioma is a usefulpredictor of response to alkylat<strong>in</strong>g agents such as carmust<strong>in</strong>e or temozolomide [113]. Similarly,methylation of a mismatch repair gene, hMLH1, <strong>in</strong> ovarian and colon cancer cell l<strong>in</strong>esconfers chemoresistance to many chemotherapeutic agents. Treatment with a DNA demethylat<strong>in</strong>gagent, 5-aza-2 0 -deoxycytid<strong>in</strong>e, can reactivate hMLH1 and reverse the chemoresistance.Likewise, silenc<strong>in</strong>g of APAF-1 (apoptotic peptidase activat<strong>in</strong>g factor-1),a proapoptotic gene, confers chemoresistance to melanoma and leukemia cells by mediat<strong>in</strong>gresistance to cytochrome c-dependent apoptosis [114]. These f<strong>in</strong>d<strong>in</strong>gs demonstrate thepotential cl<strong>in</strong>ical utility of DNA methylation markers for <strong>in</strong>dividualized therapy of cancerpatients.


CHAPTER 3DNA Methylation Alterations <strong>in</strong> <strong>Human</strong> Cancers3.12 NEW TECHNOLOGIES FOR DNA METHYLATION ANALYSISAND FUTURE DIRECTIONSCurrently available forms of screen<strong>in</strong>g technology, such as s<strong>in</strong>gle-base-pair resolution wholegenomeDNA methylation analysis us<strong>in</strong>g second-generation sequencers, and <strong>in</strong>ternationalefforts aimed at determ<strong>in</strong><strong>in</strong>g reference epigenome profiles, are now open<strong>in</strong>g new avenues ofepigenome therapy for cancer patients. Although broad DNA methylation profil<strong>in</strong>g was<strong>in</strong>itially performed on the basis of two-dimensional gel electrophoresis, adaptation ofmicroarray hybridization techniques used <strong>in</strong> gene expression and genome studies to theprofil<strong>in</strong>g of DNA methylation patterns opened the door to the era of the epigenome. Enzymebasedand aff<strong>in</strong>ity enrichment-based DNA methylation analysis techniques have been provedsuitable for exam<strong>in</strong>ation of human tissue samples us<strong>in</strong>g hybridization arrays [115]. Currentlyavailable high-throughput DNA sequenc<strong>in</strong>g technologies us<strong>in</strong>g second-generation sequencersare now capable of s<strong>in</strong>gle-base-pair resolution for whole-genome DNA methylation analysis.Although projects <strong>in</strong>volv<strong>in</strong>g analysis of large numbers of human tissue samples will still relyon array-based approaches for several more years, the trend will be towards bisulfite shotgunsequenc<strong>in</strong>g [94]. Nanopore sequenc<strong>in</strong>g provides s<strong>in</strong>gle-molecule detection and avoids anybias <strong>in</strong>troduced by differential amplification of methylation-derived states [116]. Moreover,third-generation sequencers for real-time sequenc<strong>in</strong>g can directly detect 5-methylcytos<strong>in</strong>ewithout bisulfite conversion [117]. In addition, genome-wide analysis of histone modificationand non-cod<strong>in</strong>g RNA is also be<strong>in</strong>g robustly performed. Thus, high-throughput mapp<strong>in</strong>g of theepigenome, i.e. an overview of DNA methylation, histone modification, non-cod<strong>in</strong>g RNA, andchromat<strong>in</strong> accessibility <strong>in</strong> normal, precursor and cancer cells, is now highly reproducible andstandardized.Importantly, changes <strong>in</strong> the epigenome are potentially reversible by drug treatments. This hassignificant implications for the prevention and therapy of human cancers. Indeed, several<strong>in</strong>hibitors of chromat<strong>in</strong>-modify<strong>in</strong>g enzymes, <strong>in</strong>clud<strong>in</strong>g DNMT <strong>in</strong>hibitors, as well as HDAC<strong>in</strong>hibitors, have been approved by the US Food and Drug Adm<strong>in</strong>istration and the EU, and arenow be<strong>in</strong>g used <strong>in</strong> cl<strong>in</strong>ical practice [118,119]. However, to maximize the potential of suchtherapeutic approaches, a more comprehensive characterization of the epigenome changesthat occur dur<strong>in</strong>g normal development and adult cell renewal should be accomplished by<strong>in</strong>ternational consortia.47Scientists and representatives of major fund<strong>in</strong>g agencies have decided to launch the International<strong>Human</strong> Epigenome Consortium (IHEC) [120]. Just as the <strong>Human</strong> Genome Projectprovided a reference “normal” sequence for study<strong>in</strong>g human disease, high-resolutionreference epigenome maps consist<strong>in</strong>g of the various epigenetic layers of detailed DNAmethylation as well as histone modification, nucleosome occupancy and correspond<strong>in</strong>gcod<strong>in</strong>g, and non-cod<strong>in</strong>g RNA expression <strong>in</strong> different normal cell types will be provided byIHEC. Such a reference human epigenome will be available to the worldwide researchcommunity. Information on the methods utilized by IHEC members will be useful forproduc<strong>in</strong>g large epigenomic datasets related to health and diseases <strong>in</strong> humans. It will becomepossible to compare profiles of different human populations, thereby help<strong>in</strong>g to evaluate theimpact of environment and nutrition on the epigenome. Epigenome reference maps willhave an immediate impact on our understand<strong>in</strong>g of cancers as well as diabetes, cardiopulmonarydiseases, neuropsychiatric disorders, impr<strong>in</strong>t<strong>in</strong>g disorders, <strong>in</strong>flammation, andautoimmune diseases, and will hopefully lead to breakthroughs <strong>in</strong> the prevention, diagnosis,and therapy of human cancers.References[1] Issa JP, Just W. <strong>Epigenetics</strong>. FEBS Lett 2011;585:1993.[2] Sk<strong>in</strong>ner MK. Environmental epigenetic transgenerational <strong>in</strong>heritance and somatic epigenetic mitotic stability.<strong>Epigenetics</strong> 2011;6:838e42.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>48[3] Siedlecki P, Zielenkiewicz P. Mammalian DNA methyltransferases. Acta Biochim Pol 2006;53:245e56.[4] Cheng X, Blumenthal RM. Mammalian DNA methyltransferases: a structural perspective. Structure2008;16:341e50.[5] Hermann A, Gowher H, Jeltsch A. Biochemistry and biology of mammalian DNA methyltransferases. Cell MolLife Sci 2004;61:2571e87.[6] Okano M, Bell DW, Haber DA, Li E. DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novomethylation and mammalian development. Cell 1999;99:247e57.[7] Bourc’his D, Xu GL, L<strong>in</strong> CS, Bollman B, Bestor TH. Dnmt3L and the establishment of maternal genomicimpr<strong>in</strong>ts. <strong>Science</strong> 2001;294:2536e9.[8] Deaton AM, Bird A. CpG islands and the regulation of transcription. Genes Dev 2011;25:1010e22.[9] Weber M, Hellmann I, Stadler MB, Ramos L, Pääbo S, Rebhan M, et al. Distribution, silenc<strong>in</strong>g potential andevolutionary impact of promoter DNA methylation <strong>in</strong> the human genome. Nat Genet 2007;39:457e66.[10] Irizarry RA, Ladd-Acosta C, Wen B, Wu Z, Montano C, Onyango P, et al. The human colon cancer methylomeshows similar hypo- and hypermethylation at conserved tissue-specific CpG island shores. Nat Genet2009;41:178e86.[11] Tahiliani M, Koh KP, Shen Y, Pastor WA, Bandukwala H, Brudno Y, et al. Conversion of 5-methylcytos<strong>in</strong>e to 5-hydroxymethylcytos<strong>in</strong>e <strong>in</strong> mammalian DNA by MLL partner TET1. <strong>Science</strong> 2009;324:930e5.[12] Veron N, Peters AH. <strong>Epigenetics</strong>: Tet prote<strong>in</strong>s <strong>in</strong> the limelight. Nature 2011;473:293e4.[13] Burmeister T, Meyer C, Schwartz S, Hofmann J, Molkent<strong>in</strong> M, Kowarz E, et al. The MLL recomb<strong>in</strong>ome of adultCD10-negative B-cell precursor acute lymphoblastic leukemia: results from the GMALL study group. Blood2009;113:4011e5.[14] Delhommeau F, Dupont S, Della Valle V, James C, Trannoy S, Masse A, et al. Mutation <strong>in</strong> TET2 <strong>in</strong> myeloidcancers. N Engl J Med 2009;360:2289e301.[15] Langemeijer SM, Aslanyan MG, Jansen JH. TET prote<strong>in</strong>s <strong>in</strong> malignant hematopoiesis. Cell Cycle 2009;8:4044e8.[16] Laird PW, Jackson-Grusby L, Fazeli A, Dick<strong>in</strong>son SL, Jung WE, Li E, et al. Suppression of <strong>in</strong>test<strong>in</strong>al neoplasia byDNA hypomethylation. Cell 1995;81:197e205.[17] Eden A, Gaudet F, Waghmare A, Jaenisch R. Chromosomal <strong>in</strong>stability and tumors promoted by DNA hypomethylation.<strong>Science</strong> 2003;300:455.[18] Gaudet F, Hodgson JG, Eden A, Jackson-Grusby L, Dausman J, Gray JW, et al. Induction of tumors <strong>in</strong> mice bygenomic hypomethylation. <strong>Science</strong> 2003;300:489e92.[19] Howard G, Eiges R, Gaudet F, Jaenisch R, Eden A. Activation and transposition of endogenous retroviralelements <strong>in</strong> hypomethylation <strong>in</strong>duced tumors <strong>in</strong> mice. Oncogene 2008;27:404e8.[20] Sharma S, Kelly TK, Jones PA. <strong>Epigenetics</strong> <strong>in</strong> cancer. Carc<strong>in</strong>ogenesis 2010;31:27e36.[21] Jones PA, Bayl<strong>in</strong> SB. The epigenomics of cancer. Cell 2007;128:683e92.[22] Lewandowska J, Bartoszek A. DNA methylation <strong>in</strong> cancer development, diagnosis and therapydmultipleopportunities for genotoxic agents to act as methylome disruptors or remediators. Mutagenesis 2011;26:475e87.[23] Tsai HC, Bayl<strong>in</strong> SB. Cancer epigenetics: l<strong>in</strong>k<strong>in</strong>g basic biology to cl<strong>in</strong>ical medic<strong>in</strong>e. Cell Res 2011;21:502e17.[24] Clark SJ. Action at a distance: epigenetic silenc<strong>in</strong>g of large chromosomal regions <strong>in</strong> carc<strong>in</strong>ogenesis. Hum MolGenet 2007;16(Spec No 1):R88e95.[25] Saito Y, Jones PA. Epigenetic activation of tumor suppressor microRNAs <strong>in</strong> human cancer cells. Cell Cycle2006;5:2220e2.[26] Li M, Mar<strong>in</strong>-Muller C, Bharadwaj U, Chow KH, Yao Q, Chen C. MicroRNAs: control and loss of control <strong>in</strong>human physiology and disease. World J Surg 2009;33:667e84.[27] Kanai Y, Hirohashi S. Alterations of DNA methylation associated with abnormalities of DNA methyltransferases<strong>in</strong> human cancers dur<strong>in</strong>g transition from a precancerous to a malignant state. Carc<strong>in</strong>ogenesis2007;28:2434e42.[28] Kanai Y. Alterations of DNA methylation and cl<strong>in</strong>icopathological diversity of human cancers. Pathol Int2008;58:544e58.[29] Kanai Y, Ushijima S, Tsuda H, Sakamoto M, Sugimura T, Hirohashi S. Aberrant DNA methylation on chromosome16 is an early event <strong>in</strong> hepatocarc<strong>in</strong>ogenesis. Jpn J Cancer Res 1996;87:1210e7.[30] Kondo Y, Kanai Y, Sakamoto M, Mizokami M, Ueda R, Hirohashi S. Genetic <strong>in</strong>stability and aberrant DNAmethylation <strong>in</strong> chronic hepatitis and cirrhosisdA comprehensive study of loss of heterozygosity and microsatellite<strong>in</strong>stability at 39 loci and DNA hypermethylation on 8 CpG islands <strong>in</strong> microdissected specimens frompatients with hepatocellular carc<strong>in</strong>oma. Hepatology 2000;32:970e9.[31] Yoshiura K, Kanai Y, Ochiai A, Shimoyama Y, Sugimura T, Hirohashi S. Silenc<strong>in</strong>g of the E-cadher<strong>in</strong><strong>in</strong>vasion-suppressor gene by CpG methylation <strong>in</strong> human carc<strong>in</strong>omas. Proc Natl Acad Sci USA1995;92:7416e9.


CHAPTER 3DNA Methylation Alterations <strong>in</strong> <strong>Human</strong> Cancers[32] Hirohashi S, Kanai Y. Cell adhesion system and human cancer morphogenesis. Cancer Sci 2003;94:575e81.[33] Kanai Y, Ushijima S, Hui AM, Ochiai A, Tsuda H, Sakamoto M, et al. The E-cadher<strong>in</strong> gene is silenced by CpGmethylation <strong>in</strong> human hepatocellular carc<strong>in</strong>omas. Int J Cancer 1997;71:355e9.[34] Kanai Y, Hui AM, Sun L, Ushijima S, Sakamoto M, Tsuda H, et al. DNA hypermethylation at the D17S5 locus andreduced HIC-1 mRNA expression are associated with hepatocarc<strong>in</strong>ogenesis. Hepatology 1999;29:703e9.[35] Kanai Y, Ushijima S, Tsuda H, Sakamoto M, Hirohashi S. Aberrant DNA methylation precedes loss ofheterozygosity on chromosome 16 <strong>in</strong> chronic hepatitis and liver cirrhosis. Cancer Lett 2000;148:73e80.[36] Etoh T, Kanai Y, Ushijima S, Nakagawa T, Nakanishi Y, Sasako M, et al. Increased DNA methyltransferase 1(DNMT1) prote<strong>in</strong> expression correlates significantly with poorer tumor differentiation and frequent DNAhypermethylation of multiple CpG islands <strong>in</strong> gastric cancers. Am J Pathol 2004;164:689e99.[37] Ryan JL, Jones RJ, Kenney SC, Rivenbark AG, Tang W, Knight ER, et al. Epste<strong>in</strong>eBarr virus-specific methylationof human genes <strong>in</strong> gastric cancer cells. Infect Agent Cancer 2010;5:27.[38] H<strong>in</strong>o R, Uozaki H, Murakami N, Ushiku T, Sh<strong>in</strong>ozaki A, Ishikawa S, et al. Activation of DNA methyltransferase1 by EBV latent membrane prote<strong>in</strong> 2A leads to promoter hypermethylation of PTEN gene <strong>in</strong> gastric carc<strong>in</strong>oma.Cancer Res 2009;69:2766e74.[39] Maekita T, Nakazawa K, Mihara M, Nakajima T, Yanaoka K, Iguchi M, et al. High levels of aberrant DNAmethylation <strong>in</strong> Helicobacter pylori-<strong>in</strong>fected gastric mucosae and its possible association with gastric cancerrisk. Cl<strong>in</strong> Cancer Res 2006;12:989e95.[40] Sawada M, Kanai Y, Arai E, Ushijima S, Ojima H, Hirohashi S. Increased expression of DNA methyltransferase 1(DNMT1) prote<strong>in</strong> <strong>in</strong> uter<strong>in</strong>e cervix squamous cell carc<strong>in</strong>oma and its precursor lesion. Cancer Lett 2007;251:211e9.[41] Laurson J, Khan S, Chung R, Cross K, Raj K. Epigenetic repression of E-cadher<strong>in</strong> by human papillomavirus 16E7 prote<strong>in</strong>. Carc<strong>in</strong>ogenesis 2010;31:918e26.[42] Eguchi K, Kanai Y, Kobayashi K, Hirohashi S. DNA hypermethylation at the D17S5 locus <strong>in</strong> non-small celllung cancers: its association with smok<strong>in</strong>g history. Cancer Res 1997;57:4913e5.[43] Chen W, Cooper TK, Zahnow CA, Overholtzer M, Zhao Z, Ladanyi M, et al. Epigenetic and genetic loss of Hic1function accentuates the role of p53 <strong>in</strong> tumorigenesis. Cancer Cell 2004;6:387e98.[44] Wales MM, Biel MA, el Deiry W, Nelk<strong>in</strong> BD, Issa JP, Cavenee WK, et al. p53 activates expression of HIC-1,a new candidate tumour suppressor gene on 17p13.3. Nat Med 1995;1:570e7.[45] Sun L, Hui AM, Kanai Y, Sakamoto M, Hirohashi S. Increased DNA methyltransferase expression is associatedwith an early stage of human hepatocarc<strong>in</strong>ogenesis. Jpn J Cancer Res 1997;88:1165e70.[46] Saito Y, Kanai Y, Sakamoto M, Saito H, Ishii H, Hirohashi S. Expression of mRNA for DNA methyltransferasesand methyl-CpG-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s and DNA methylation status on CpG islands and pericentromeric satelliteregions dur<strong>in</strong>g human hepatocarc<strong>in</strong>ogenesis. Hepatology 2001;33:561e8.[47] Saito Y, Kanai Y, Nakagawa T, Sakamoto M, Saito H, Ishii H, et al. Increased prote<strong>in</strong> expression of DNAmethyltransferase (DNMT) 1 is significantly correlated with the malignant potential and poor prognosis ofhuman hepatocellular carc<strong>in</strong>omas. Int J Cancer 2003;105:527e32.[48] Peng DF, Kanai Y, Sawada M, Ushijima S, Hiraoka N, Kosuge T, et al. Increased DNA methyltransferase 1(DNMT1) prote<strong>in</strong> expression <strong>in</strong> precancerous conditions and ductal carc<strong>in</strong>omas of the pancreas. Cancer Sci2005;96:403e8.[49] Peng DF, Kanai Y, Sawada M, Ushijima S, Hiraoka N, Kitazawa S, et al. DNA methylation of multiple tumorrelatedgenes <strong>in</strong> association with overexpression of DNA methyltransferase 1 (DNMT1) dur<strong>in</strong>g multistagecarc<strong>in</strong>ogenesis of the pancreas. Carc<strong>in</strong>ogenesis 2006;27:1160e8.[50] Robertson KD, Uzvolgyi E, Liang G, Talmadge C, Sumegi J, Gonzales FA, et al. The human DNA methyltransferases(DNMTs) 1, 3a and 3b: coord<strong>in</strong>ate mRNA expression <strong>in</strong> normal tissues and overexpression <strong>in</strong>tumors. Nucleic Acids Res 1999;27:2291e8.[51] Toyota M, Ahuja N, Ohe-Toyota M, Herman JG, Bayl<strong>in</strong> SB, Issa JP. CpG island methylator phenotype <strong>in</strong>colorectal cancer. Proc Natl Acad Sci USA 1999;96:8681e6.[52] Issa JP, Shen L, Toyota M. CIMP, at last. Gastroenterology 2005;129:1121e4.[53] Kanai Y, Ushijima S, Kondo Y, Nakanishi Y, Hirohashi S. DNA methyltransferase expression and DNAmethylation of CPG islands and peri-centromeric satellite regions <strong>in</strong> human colorectal and stomach cancers.Int J Cancer 2001;91:205e12.[54] Nakagawa T, Kanai Y, Saito Y, Kitamura T, Kakizoe T, Hirohashi S. Increased DNA methyltransferase 1 prote<strong>in</strong>expression <strong>in</strong> human transitional cell carc<strong>in</strong>oma of the bladder. J Urol 2003;170:2463e6.[55] Nakagawa T, Kanai Y, Ushijima S, Kitamura T, Kakizoe T, Hirohashi S. DNA hypermethylation on multipleCpG islands associated with <strong>in</strong>creased DNA methyltransferase DNMT1 prote<strong>in</strong> expression dur<strong>in</strong>g multistageurothelial carc<strong>in</strong>ogenesis. J Urol 2005;173:1767e71.[56] Denis H, Ndlovu MN, Fuks F. Regulation of mammalian DNA methyltransferases: a route to new mechanisms.EMBO Rep 2011;12:647e56.49


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>50[57] Fabbri M, Garzon R, Cimm<strong>in</strong>o A, Liu Z, Zanesi N, Callegari E, et al. MicroRNA-29 family reverts aberrantmethylation <strong>in</strong> lung cancer by target<strong>in</strong>g DNA methyltransferases 3A and 3B. Proc Natl Acad Sci USA2007;104:15805e10.[58] Garzon R, Liu S, Fabbri M, Liu Z, Heaphy CE, Callegari E, et al. MicroRNA-29b <strong>in</strong>duces global DNA hypomethylationand tumor suppressor gene reexpression <strong>in</strong> acute myeloid leukemia by target<strong>in</strong>g directlyDNMT3A and 3B and <strong>in</strong>directly DNMT1. Blood 2009;113:6411e8.[59] Duursma AM, Kedde M, Schrier M, le Sage C, Agami R. miR-148 targets human DNMT3b prote<strong>in</strong> cod<strong>in</strong>gregion. Rna 2008;14:872e7.[60] Braconi C, Huang N, Patel T. MicroRNA-dependent regulation of DNA methyltransferase-1 and tumorsuppressor gene expression by <strong>in</strong>terleuk<strong>in</strong>-6 <strong>in</strong> human malignant cholangiocytes. Hepatology2010;51:881e90.[61] Zhao S, Wang Y, Liang Y, Zhao M, Long H, D<strong>in</strong>g S, et al. MicroRNA-126 regulates DNA methylation <strong>in</strong> CD4þ Tcells and contributes to systemic lupus erythematosus by target<strong>in</strong>g DNA methyltransferase 1. Arthritis Rheum2011;63:1376e86.[62] Huang J, Wang Y, Guo Y, Sun S. Down-regulated microRNA-152 <strong>in</strong>duces aberrant DNA methylation <strong>in</strong>hepatitis B virus-related hepatocellular carc<strong>in</strong>oma by target<strong>in</strong>g DNA methyltransferase 1. Hepatology2010;52:60e70.[63] Lopez de Silanes I, Gorospe M, Taniguchi H, Abdelmohsen K, Srikantan S, Alam<strong>in</strong>os M, et al. The RNAb<strong>in</strong>d<strong>in</strong>gprote<strong>in</strong> HuR regulates DNA methylation through stabilization of DNMT3b mRNA. Nucleic Acids Res2009;37:2658e71.[64] Kanai Y, Ushijima S, Nakanishi Y, Sakamoto M, Hirohashi S. Mutation of the DNA methyltransferase (DNMT)1 gene <strong>in</strong> human colorectal cancers. Cancer Lett 2003;192:75e82.[65] Ley TJ, D<strong>in</strong>g L, Walter MJ, McLellan MD, Lamprecht T, Larson DE, et al. DNMT3A mutations <strong>in</strong> acute myeloidleukemia. N Engl J Med 2010;363:2424e33.[66] Yan XJ, Xu J, Gu ZH, Pan CM, Lu G, Shen Y, et al. Exome sequenc<strong>in</strong>g identifies somatic mutations of DNAmethyltransferase gene DNMT3A <strong>in</strong> acute monocytic leukemia. Nat Genet 2011;43:309e15.[67] Fan H, Liu D, Qiu X, Qiao F, Wu Q, Su X, et al. A functional polymorphism <strong>in</strong> the DNA methyltransferase-3A promoter modifies the susceptibility <strong>in</strong> gastric cancer but not <strong>in</strong> esophageal carc<strong>in</strong>oma. BMC Med2010;8:12.[68] Wang L, Rodriguez M, Kim ES, Xu Y, Bekele N, El-Naggar AK, et al. A novel C/T polymorphism <strong>in</strong> the corepromoter of human de novo cytos<strong>in</strong>e DNA methyltransferase 3B6 is associated with prognosis <strong>in</strong> head andneck cancer. Int J Oncol 2004;25:993e9.[69] Hansen RS, Wijmenga C, Luo P, Stanek AM, Canfield TK, Weemaes CM, et al. The DNMT3B DNA methyltransferasegene is mutated <strong>in</strong> the ICF immunodeficiency syndrome. Proc Natl Acad Sci USA1999;96:14412e7.[70] Wong N, Lam WC, Lai PB, Pang E, Lau WY, Johnson PJ. Hypomethylation of chromosome 1 heterochromat<strong>in</strong>DNA correlates with q-arm copy ga<strong>in</strong> <strong>in</strong> human hepatocellular carc<strong>in</strong>oma. Am J Pathol2001;159:465e71.[71] Nakagawa T, Kanai Y, Ushijima S, Kitamura T, Kakizoe T, Hirohashi S. DNA hypomethylation on pericentromericsatellite regions significantly correlates with loss of heterozygosity on chromosome 9 <strong>in</strong> urothelialcarc<strong>in</strong>omas. J Urol 2005;173:243e6.[72] Soejima K, Fang W, Roll<strong>in</strong>s BJ. DNA methyltransferase 3b contributes to oncogenic transformation <strong>in</strong>duced bySV40T antigen and activated Ras. Oncogene 2003;22:4723e33.[73] Saito Y, Kanai Y, Sakamoto M, Saito H, Ishii H, Hirohashi S. Overexpression of a splice variant of DNAmethyltransferase 3b, DNMT3b4, associated with DNA hypomethylation on pericentromeric satellite regionsdur<strong>in</strong>g human hepatocarc<strong>in</strong>ogenesis. Proc Natl Acad Sci USA 2002;99:10060e5.[74] Hervouet E, Vallette FM, Cartron PF. Dnmt3/transcription factor <strong>in</strong>teractions as crucial players <strong>in</strong> targeted DNAmethylation. <strong>Epigenetics</strong> 2009;4:487e99.[75] Simon JA, Lange CA. Roles of the EZH2 histone methyltransferase <strong>in</strong> cancer epigenetics. Mutat Res2008;647:21e9.[76] Hou P, Ji M, X<strong>in</strong>g M. Association of PTEN gene methylation with genetic alterations <strong>in</strong> the phosphatidyl<strong>in</strong>ositol3-k<strong>in</strong>ase/AKT signal<strong>in</strong>g pathway <strong>in</strong> thyroid tumors. Cancer 2008;113:2440e7.[77] Maddodi N, Bhat KM, Devi S, Zhang SC, Setaluri V. Oncogenic BRAFV600E <strong>in</strong>duces expression of neuronaldifferentiation marker MAP2 <strong>in</strong> melanoma cells by promoter demethylation and down-regulation of transcriptionrepressor HES1. J Biol Chem 2010;285:242e54.[78] Hou P, Liu D, X<strong>in</strong>g M. Genome-wide alterations <strong>in</strong> gene methylation by the BRAF V600E mutation <strong>in</strong>papillary thyroid cancer cells. Endocr Relat Cancer 2011;18:687e97.[79] MacLeod AR, Rouleau J, Szyf M. Regulation of DNA methylation by the Ras signal<strong>in</strong>g pathway. J Biol Chem1995;270:11327e37.


CHAPTER 3DNA Methylation Alterations <strong>in</strong> <strong>Human</strong> Cancers[80] Deng C, Yang J, Scott J, Hanash S, Richardson BC. Role of the ras-MAPK signal<strong>in</strong>g pathway <strong>in</strong> the DNAmethyltransferase response to DNA hypomethylation. Biol Chem 1998;379:1113e20.[81] Pruitt K, Ulkü AS, Frantz K, Rojas RJ, Muniz-Med<strong>in</strong>a VM, Rangnekar VM, et al. Ras-mediated loss of the proapoptoticresponse prote<strong>in</strong> Par-4 is mediated by DNA hypermethylation through Raf-<strong>in</strong>dependent and Rafdependentsignal<strong>in</strong>g cascades <strong>in</strong> epithelial cells. J Biol Chem 2005;280:23363e70.[82] Young NL, Dimaggio PA, Garcia BA. The significance, development and progress of high-throughputcomb<strong>in</strong>atorial histone code analysis. Cell Mol Life Sci 2010;67:3983e4000.[83] Gardner KE, Allis CD, Strahl BD. Operat<strong>in</strong>g on chromat<strong>in</strong>, a colorful language where context matters. J MolBiol 2011;409:36e46.[84] Bogdanovic O, Veenstra GJ. DNA methylation and methyl-CpG b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s: developmental requirementsand function. Chromosoma 2009;118:549e65.[85] Miremadi A, Oestergaard MZ, Pharoah PD, Caldas C. Cancer genetics of epigenetic genes. Hum Mol Genet2007;16(Spec No 1):R28e49.[86] Ohm JE, McGarvey KM, Yu X, Cheng L, Schuebel KE, Cope L, et al. A stem cell-like chromat<strong>in</strong> pattern maypredispose tumor suppressor genes to DNA hypermethylation and heritable silenc<strong>in</strong>g. Nat Genet2007;39:237e42.[87] Vire E, Brenner C, Deplus R, Blanchon L, Fraga M, Didelot C, et al. The Polycomb group prote<strong>in</strong> EZH2directly controls DNA methylation. Nature 2006;439:871e4.[88] D<strong>in</strong>g L, Kleer CG. Enhancer of Zeste 2 as a marker of preneoplastic progression <strong>in</strong> the breast. Cancer Res2006;66:9352e5.[89] Bryant RJ, Cross NA, Eaton CL, Hamdy FC, Cunliffe VT. EZH2 promotes proliferation and <strong>in</strong>vasiveness ofprostate cancer cells. Prostate 2007;67:547e56.[90] Fussbroich B, Wagener N, Macher-Goepp<strong>in</strong>ger S, Benner A, Falth M, Sultmann H, et al. EZH2 depletionblocks the proliferation of colon cancer cells. PLoS One 2011;6:e21651.[91] Mohammad HP, Cai Y, McGarvey KM, Easwaran H, Van Neste L, Ohm JE, et al. Polycomb CBX7 promotes<strong>in</strong>itiation of heritable repression of genes frequently silenced with cancer-specific DNA hypermethylation.Cancer Res 2009;69:6322e30.[92] Kanai Y. Genome-wide DNA methylation profiles <strong>in</strong> precancerous conditions and cancers. Cancer Sci2010;101:36e45.[93] Arai E, Kanai Y. DNA methylation profiles <strong>in</strong> precancerous tissue and cancers: carc<strong>in</strong>ogenetic risk estimationand prognostication based on DNA methylation status. Epigenomics 2010;2:467e81.[94] Laird PW. Pr<strong>in</strong>ciples and challenges of genomewide DNA methylation analysis. Nat Rev Genet 2010;11:191e203.[95] Shen L, Toyota M, Kondo Y, L<strong>in</strong> E, Zhang L, Guo Y, et al. Integrated genetic and epigenetic analysis identifiesthree different subclasses of colon cancer. Proc Natl Acad Sci USA 2007;104:18654e9.[96] Arai E, Kanai Y, Ushijima S, Fujimoto H, Mukai K, Hirohashi S. Regional DNA hypermethylation and DNAmethyltransferase (DNMT) 1 prote<strong>in</strong> overexpression <strong>in</strong> both renal tumors and correspond<strong>in</strong>g nontumorousrenal tissues. Int J Cancer 2006;119:288e96.[97] Arai E, Wakai-Ushijima S, Fujimoto H, Hosoda F, Shibata T, Kondo T, et al. Genome-wide DNA methylationprofiles <strong>in</strong> renal tumors of various histological subtypes and non-tumorous renal tissues. Pathobiology2011;78:1e9.[98] Arai E, Ushijima S, Fujimoto H, Hosoda F, Shibata T, Kondo T, et al. Genome-wide DNA methylation profiles<strong>in</strong> both precancerous conditions and clear cell renal cell carc<strong>in</strong>omas are correlated with malignant potentialand patient outcome. Carc<strong>in</strong>ogenesis 2009;30:214e21.[99] Arai E, Kanai Y. Genetic and epigenetic alterations dur<strong>in</strong>g renal carc<strong>in</strong>ogenesis. Int J Cl<strong>in</strong> Exp Pathol 2011;4:58e73.[100] Arai E, Ushijima S, Tsuda H, Fujimoto H, Hosoda F, Shibata T, et al. Genetic cluster<strong>in</strong>g of clear cell renal cellcarc<strong>in</strong>oma based on array-comparative genomic hybridization: its association with DNA methylation alterationand patient outcome. Cl<strong>in</strong> Cancer Res 2008;14:5531e9.[101] Nishiyama N, Arai E, Nagashio R, Fujimoto H, Hosoda F, Shibata T, et al. Copy number alterations <strong>in</strong>urothelial carc<strong>in</strong>omas: their cl<strong>in</strong>icopathological significance and correlation with DNA methylation alterations.Carc<strong>in</strong>ogenesis 2011;32:462e9.[102] Sepulveda AR, Jones D, Og<strong>in</strong>o S, Samowitz W, Gulley ML, Edwards R, et al. CpG methylation analysisecurrentstatus of cl<strong>in</strong>ical assays and potential applications <strong>in</strong> molecular diagnostics: a report of theAssociation for Molecular Pathology. J Mol Diagn 2009;11:266e78.[103] Cairns P, Esteller M, Herman JG, Schoenberg M, Jeronimo C, Sanchez-Cespedes M, et al. Molecular detectionof prostate cancer <strong>in</strong> ur<strong>in</strong>e by GSTP1 hypermethylation. Cl<strong>in</strong> Cancer Res 2001;7:2727e30.[104] Hoque MO, Begum S, Topaloglu O, Chatterjee A, Rosenbaum E, Van Criek<strong>in</strong>ge W, et al. Quantitation ofpromoter methylation of multiple genes <strong>in</strong> ur<strong>in</strong>e DNA and bladder cancer detection. J Natl Cancer Inst2006;98:996e1004.51


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>52[105] Watanabe Y, Kim HS, Castoro RJ, Chung W, Estecio MR, Kondo K, et al. Sensitive and specific detection ofearly gastric cancer with DNA methylation analysis of gastric washes. Gastroenterology 2009;136:2149e58.[106] Gotoh M, Arai E, Wakai-Ushijima S, Hiraoka N, Kosuge T, Hosoda F, et al. Diagnosis and prognostication ofductal adenocarc<strong>in</strong>omas of the pancreas based on genome-wide DNA methylation profil<strong>in</strong>g by bacterialartificial chromosome array-based methylated CpG island amplification. J Biomed Biotechnol2011;2011:780836.[107] Arai E, Ushijima S, Gotoh M, Ojima H, Kosuge T, Hosoda F, et al. Genome-wide DNA methylation profiles <strong>in</strong>liver tissue at the precancerous stage and <strong>in</strong> hepatocellular carc<strong>in</strong>oma. Int J Cancer 2009;125:2854e62.[108] Nagashio R, Arai E, Ojima H, Kosuge T, Kondo Y, Kanai Y. Carc<strong>in</strong>ogenetic risk estimation based on quantificationof DNA methylation levels <strong>in</strong> liver tissue at the precancerous stage. Int J Cancer 2011;129:1170e9.[109] Nishiyama N, Arai E, Chihara Y, Fujimoto H, Hosoda F, Shibata T, et al. Genome-wide DNA methylationprofiles <strong>in</strong> urothelial carc<strong>in</strong>omas and urothelia at the precancerous stage. Cancer Sci 2010;101:231e40.[110] Marsit CJ, Houseman EA, Christensen BC, Gagne L, Wrensch MR, Nelson HH, et al. Identification ofmethylated genes associated with aggressive bladder cancer. PLoS One 2010;5:e12334.[111] Jarmalaite S, Jankevicius F, Kurgonaite K, Suziedelis K, Mutanen P, Husgafvel-Pursia<strong>in</strong>en K. Promoterhypermethylation <strong>in</strong> tumour suppressor genes shows association with stage, grade and <strong>in</strong>vasiveness ofbladder cancer. Oncology 2008;75:145e51.[112] Satoh A, Toyota M, Itoh F, Sasaki Y, Suzuki H, Ogi K, et al. Epigenetic <strong>in</strong>activation of CHFR and sensitivity tomicrotubule <strong>in</strong>hibitors <strong>in</strong> gastric cancer. Cancer Res 2003;63:8606e13.[113] Hegi ME, Liu L, Herman JG, Stupp R, Wick W, Weller M, et al. Correlation of O6-methylguan<strong>in</strong>e methyltransferase(MGMT) promoter methylation with cl<strong>in</strong>ical outcomes <strong>in</strong> glioblastoma and cl<strong>in</strong>ical strategies tomodulate MGMT activity. J Cl<strong>in</strong> Oncol 2008;26:4189e99.[114] Campioni M, Sant<strong>in</strong>i D, Ton<strong>in</strong>i G, Murace R, Dragonetti E, Spugn<strong>in</strong>i EP, et al. Role of Apaf-1, a key regulatorof apoptosis, <strong>in</strong> melanoma progression and chemoresistance. Exp Dermatol 2005;14:811e8.[115] Estecio MR, Issa JP. Tackl<strong>in</strong>g the methylome: recent methodological advances <strong>in</strong> genome-wide methylationprofil<strong>in</strong>g. Genome Med 2009;1:106.[116] Branton D, Deamer DW, Marziali A, Bayley H, Benner SA, Butler T, et al. The potential and challenges ofnanopore sequenc<strong>in</strong>g. Nat Biotechnol 2008;26:1146e53.[117] Eid J, Fehr A, Gray J, Luong K, Lyle J, Otto G, et al. Real-time DNA sequenc<strong>in</strong>g from s<strong>in</strong>gle polymerasemolecules. <strong>Science</strong> 2009;323:133e8.[118] Kelly TK, De Carvalho DD, Jones PA. Epigenetic modifications as therapeutic targets. Nat Biotechnol2010;28:1069e78.[119] Boumber Y, Issa JP. <strong>Epigenetics</strong> <strong>in</strong> cancer: what’s the future? Oncology (Williston Park) 2011;25:220e6, 228.[120] International <strong>Human</strong> Epigenome Consortium [homepage on the <strong>in</strong>ternet] Cited. Available from: http://www.ihec-epigenomes.org/; February 2012.


CHAPTER 4Alterations of HistoneModifications <strong>in</strong> CancerCiro Mercurio 1,3 , Simon Plyte 2,3 , Saverio M<strong>in</strong>ucci 3,41 DAC-Genextra Group, Milan, Italy2 Congenia-Genextra Group, Milan, Italy3 European Institute of Oncology, Milan, Italy4 University of Milan, Milan ItalyCHAPTER OUTLINE4.1 Introduction 534.2 Chromat<strong>in</strong> Organization 544.3 Histone Modifications 554.3.1 Histone Acetylation 554.3.2 Histone Methylation 574.3.3 Histone Phosphorylation 614.3.4 Histone Ubiquit<strong>in</strong>ation 624.3.5 Mode of Action of HistoneModifications 634.3.6 Histone Cross-Talk 654.4 Histone Modifications andCancer 664.4.1 Alterations <strong>in</strong> the Pattern ofHistone H4Modifications 664.4.2 Alterations <strong>in</strong> the Pattern ofHistone H3Modifications 684.5 Mechanisms Underly<strong>in</strong>g HistoneAlterations <strong>in</strong> Cancer 694.5.1 Alteration of the HistoneAcetylation Network (HATs,HDACs and Sirtu<strong>in</strong>s) 704.5.2 Alteration of the HistoneMethylation Network (HMTs,PRMTs and HDMs) 714.5.3 Alterations of HistoneK<strong>in</strong>ases, HistoneUbiquit<strong>in</strong>at<strong>in</strong>g andDeubiquit<strong>in</strong>at<strong>in</strong>gEnzymes 734.5.4 Other Mechanisms 744.6 Conclusions 74References 75534.1 INTRODUCTIONDynamic changes <strong>in</strong> chromat<strong>in</strong> structure, which permit local decondensation and remodel<strong>in</strong>gand are necessary for the role of chromat<strong>in</strong> <strong>in</strong> processes as gene transcription, DNA replicationand repair, are achieved by modification of the chromat<strong>in</strong> and <strong>in</strong> particular by posttranslationalmodifications of the histone component [1,2].The post-translational modifications of histones identified so far <strong>in</strong>clude acetylation, phosphorylation,methylation, monoubiquit<strong>in</strong>ation, sumoylation, and ADP ribosylation [2]. Thereversibility of these modifications is what confers the necessary dynamicity of the chromat<strong>in</strong>remodel<strong>in</strong>g events and these are tightly controlled by the oppos<strong>in</strong>g activity of enzymesresponsible for add<strong>in</strong>g or remov<strong>in</strong>g the modifications (for example, histone acetyltransferasesT. Tollefsbol (Ed): <strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>. DOI: 10.1016/B978-0-12-388415-2.00004-4Copyright Ó 2012 Elsevier Inc. All rights reserved.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>and histone deacetylates for histone acetylation and histone methyltransferases and histonedemethylase for histone methylation). It is worth not<strong>in</strong>g that those enzymes responsible forhistone modifications can also modify non-histone prote<strong>in</strong>s: while this observation hasenormous implications, we have chosen here to restrict our analysis to the study of histonemodifications and to the action of those enzymes on histones. We will first <strong>in</strong>troduce theknown molecular and biochemical properties of the different types of histone posttranslationalmodifications, concentrat<strong>in</strong>g primarily on acetylation, methylation, phosphorylation,and ubiquit<strong>in</strong>ation. Then, we will summarize the current knowledge regard<strong>in</strong>g therelevance of histone modifications <strong>in</strong> cancer, with a particular emphasis on the description ofglobal changes to the pattern of histone modifications <strong>in</strong> cancer cells and their potential role asprognostic factors. F<strong>in</strong>ally we will discuss the molecular mechanisms that are potentially<strong>in</strong>volved <strong>in</strong> the generation of these altered patterns <strong>in</strong> cancer cells.54The various histone modifications act <strong>in</strong> a coord<strong>in</strong>ate and ordered manner to control theconformation of chromat<strong>in</strong> [3]. A further level of complexity is present, due to the <strong>in</strong>terplaybetween the different histone modifications, DNA methylation, and ATP-dependent chromat<strong>in</strong>remodel<strong>in</strong>g components [3]. Remarkable progress has been made <strong>in</strong> recent years <strong>in</strong>the identification of these histone modifications, their genome-wide distribution and thelevel of <strong>in</strong>terconnection between them and other relevant events such as DNA methylation.The<strong>in</strong>creasedknowledgeand<strong>in</strong>terest<strong>in</strong>theroleofepigeneticmodifications<strong>in</strong>cancerhasbeen re<strong>in</strong>forced by the identification of a deregulated pattern of histone modification <strong>in</strong>several cancer types. The reversibility of histone modification and the identification of themolecular mach<strong>in</strong>ery that governs these modifications have made histone-modify<strong>in</strong>genzymes attractive new targets for anticancer therapy. In addition, a clear role for the patternof histone modification as a predictor of prognosis <strong>in</strong> several cancers has emerged, althoughthe use of such “histone modification signature” as predictor of therapeutic response is still atan <strong>in</strong>itial stage.4.2 CHROMATIN ORGANIZATIONThe structural and functional unit of chromat<strong>in</strong> is the nucleosome, which consists of a discshapedoctamer composed of two copies of each histone prote<strong>in</strong> (H2A, H2B, H3, and H4),around which 147 base-pairs of DNA are wrapped twice (Figure 4.1) [4e7]. Electronmicroscopy studies revealed that organization of nucleosomal arrays is constituted by a seriesof “beads on a str<strong>in</strong>g”, with the “beads” be<strong>in</strong>g the <strong>in</strong>dividual nucleosomes and the “str<strong>in</strong>g”be<strong>in</strong>g the l<strong>in</strong>ker DNA [4,5]. L<strong>in</strong>ker histones, such as histone H1 and other non-histoneprote<strong>in</strong>s <strong>in</strong>teract with the nucleosomal arrays to further package the nucleosomes to formhigher-order chromat<strong>in</strong> structures [7,8].Histones are high evolutionarily conserved prote<strong>in</strong>s with flexible N and C term<strong>in</strong>al doma<strong>in</strong>s anda conserved related globular doma<strong>in</strong> which mediates histoneehistone <strong>in</strong>teractions with<strong>in</strong> theoctamer (Figure 4.1) [9e11]. There are two small doma<strong>in</strong>s protrud<strong>in</strong>g from the globular doma<strong>in</strong>:an am<strong>in</strong>oterm<strong>in</strong>al doma<strong>in</strong> constituted by 20e35 residues rich <strong>in</strong> basic am<strong>in</strong>o acids and a shortprotease accessible carboxyterm<strong>in</strong>al doma<strong>in</strong> [9e11]. Histone H2A is unique among the histoneshav<strong>in</strong>g an additional 37 am<strong>in</strong>o acids carboxy-term<strong>in</strong>al doma<strong>in</strong> that protrudes from the nucleosome[11]. Additional histone variants have also been identified and tend to have specialized roles[12]. The N-term<strong>in</strong>al tail of histones, as well as more recently def<strong>in</strong>ed positions <strong>in</strong> the globulardoma<strong>in</strong>, are subject to eight different classes of post-translational modification <strong>in</strong>volv<strong>in</strong>g morethan 60 dist<strong>in</strong>ct modification sites: lys<strong>in</strong>e acetylation, ubiquit<strong>in</strong>ation and sumolylation, ser<strong>in</strong>e,threon<strong>in</strong>e and thyros<strong>in</strong>e phosphorylation, lys<strong>in</strong>e and arg<strong>in</strong><strong>in</strong>e methylation, glutamate poly-ADPribosylation, arg<strong>in</strong><strong>in</strong>e deim<strong>in</strong>ation, and prol<strong>in</strong>e isomerization (Figure 4.1) [2,11]. The comb<strong>in</strong>ationof these histone modifications, the <strong>in</strong>terplay between them and DNA methylation and ATPchromat<strong>in</strong> remodel<strong>in</strong>g prote<strong>in</strong>s, dynamically regulates chromat<strong>in</strong> structure and <strong>in</strong> so do<strong>in</strong>g,


CHAPTER 4Alterations of Histone Modifications <strong>in</strong> CancerH3N-terH2AN-terPS1 AcK5 AcK9 AcMeK12 AcK15AcK36K99Ub MeK119P Me P PH2AT3 R8 AcS10C-terT120T11K4K18 MePR2K9 K14 R17 R26MeMe Ac Me AcPAcK23PMePS28 Y41 T45 K79Me AcK56 MeK27K36Me Ac K37 Me AcMe Ac MeMe AcAcAcK5Ac K12Ac Me PPAc MeAcK15S36 S32 K28 K27K116K108 K85 K43 S14K20PUb AcUb AcK120H2BC-terMe AcR92AcK20MeMeK77AcK91S47K12K79 K59K5AcP K16 K8 AcAc MeAcAcR3MeS1PN-terH2BH4N-terChromat<strong>in</strong> Residues modified Function regulatedModificationAcetylation Lys<strong>in</strong>e Transcription, DNA repair,replication and condensationMethylation (Lys<strong>in</strong>e) Lys<strong>in</strong>e me1, me2, me3 Transcription, DNA repairMethylation (Arg<strong>in</strong><strong>in</strong>e) Arg<strong>in</strong><strong>in</strong>e-me1, Arg<strong>in</strong><strong>in</strong>e-me2a TranscriptionArg<strong>in</strong><strong>in</strong>e-me2sPhosphorylation Ser<strong>in</strong>e, Threon<strong>in</strong>e, Tyros<strong>in</strong>e Transcription, DNA repair andcondensationUbiqut<strong>in</strong>ationLys<strong>in</strong>eTranscription, DNA repairSumoylation Lys<strong>in</strong>e TranscriptionADP ribosylation Glutamic TranscriptionDeim<strong>in</strong>ation Arg<strong>in</strong><strong>in</strong>e TranscriptionProl<strong>in</strong>e isomerization P-cis, P-trans TranscriptionDifferent classes of histone modificationsAcMePUbAcetylationMethylationPhosphorylationUbiqut<strong>in</strong>ationFIGURE 4.1Mammalian core histone modifications. N- and C-term<strong>in</strong>al histone tails extend from the globular doma<strong>in</strong>s of histones H2A, H2B, H3, and H4. DNA is wrappedaround the nucleosome octamer made up of two H2AeH2B dimers and a H3eH4 tetramer. Post-translational covalent modifications <strong>in</strong>clude acetylation,methylation, phosphorylation, and ubiquitylation. <strong>Human</strong> histone tail am<strong>in</strong>o acid sequences are shown. Lys<strong>in</strong>e positions 56 and 79 on histone H3 are locatedwith<strong>in</strong> the globular doma<strong>in</strong> of the histone. This figure is reproduced <strong>in</strong> the color plate section.55coord<strong>in</strong>ates several relevant cellular processes <strong>in</strong>clud<strong>in</strong>g transcription, DNA replication, DNArepair, and genomic stability [13e16].4.3 HISTONE MODIFICATIONSWe will now look at the various histone modifications available.4.3.1 Histone AcetylationHistone acetylation is a reversible covalent modification, occurr<strong>in</strong>g at specific lys<strong>in</strong>es residues<strong>in</strong> the histone tails. This modification is able to neutralize the positive charge of the targetedlys<strong>in</strong>e, weaken<strong>in</strong>g the histone-DNA [2,16,17] or nucleosomeenucleosome <strong>in</strong>teractions andconsequently <strong>in</strong>duc<strong>in</strong>g conformational changes result<strong>in</strong>g <strong>in</strong> an open chromat<strong>in</strong> architecture[16]. Furthermore, histone acetylation represents a “histone mark” recognized by specificprote<strong>in</strong>s such as bromo doma<strong>in</strong>-conta<strong>in</strong><strong>in</strong>g prote<strong>in</strong>s, whose <strong>in</strong>teraction with the modifiedchromat<strong>in</strong> leads to a cascade of additional modifications often culm<strong>in</strong>at<strong>in</strong>g <strong>in</strong> <strong>in</strong>creasedtranscriptional activity [2e17]. The steady state level of histone lys<strong>in</strong>e acetylation is determ<strong>in</strong>edby the oppos<strong>in</strong>g activity of two different types of enzymes: histone acetyltransferases(HATs), which use acetyl CoA to transfer an acetyl group to the ε-am<strong>in</strong>o group of the Nterm<strong>in</strong>al of histone tails, and histone deacetylases (HDACs) which reverse this modification(Table 4.1) [18].HATs are often part of large multiprote<strong>in</strong> complexes, and can be divided <strong>in</strong>to two ma<strong>in</strong> groups:type A and type B based on their cellular localization [19]. The type A HATs are nuclear


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>TABLE 4.1 Histone Modify<strong>in</strong>g Enzymes: Histone Acetyltransferases and HistoneDeacetylasesCategory Gene Histone SpecificityHistone AcetyltransferasesGNC5/PCAF family HAT1 H4Gcn5H3/H4PCAFH3/H4MYST family MOF (MYST1) H4K16HBO1 (MYST2)H4>H3MOZ (MYST3) KAT6aH3MORF (MYST4) KAT6bH3Tip60H4 H2aP300/CBP family P300 H2A/H2B/H3/H4CBPH2A/H2B/H3/H4Histone DeacetylasesHDAC class I HDAC1,HDAC2,HDAC3,HDAC8 dHDAC class IIA HDAC4,HDAC5,HDAC7,HDAC9 dHDAC class IIB HDAC6,HDAC10 dHDAC class IV HDAC11 dHDAC Class III (Sirtu<strong>in</strong>)Sirtu<strong>in</strong> class I SIRT1,SIRT2,SIRT3 dSirtu<strong>in</strong> class II SIRT4 dSirtu<strong>in</strong> class III SIRT5 dSirtu<strong>in</strong> class IV SIRT6,SIRT7 d56prote<strong>in</strong>s acetylat<strong>in</strong>g both nucleosomal histones and other chromat<strong>in</strong>-associated prote<strong>in</strong>s,whereas type B HATs are prevalently cytoplasmatic, and acetylate newly synthesized histonesnot yet deposited <strong>in</strong>to chromat<strong>in</strong> [19,20].Type A HATs may be further grouped <strong>in</strong>to at least three different sub-groups based on theirsequence homology and conformational structure: GNATs (Gcn5-related N-acetyl transferases)related HATs, MYST-related HATs (MOZ (monocytic leukaemia z<strong>in</strong>c-f<strong>in</strong>ger prote<strong>in</strong>), Ybf2 (yeastb<strong>in</strong>d<strong>in</strong>g factor 2)/Sas3 (someth<strong>in</strong>g about silenc<strong>in</strong>g 3), Sas2, Tip60 (Tat <strong>in</strong>teractive prote<strong>in</strong>-60)),and p300/CBP family (CREB (cAMP response element b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>)-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>) [19].The type B HATs share sequence homology with scHat1 [20], the found<strong>in</strong>g member of this typeof HATs and have a role <strong>in</strong> the deposition of histones <strong>in</strong>to the nucleosome through theacetylation of newly synthesized histone H4 at lys<strong>in</strong>e 5 (K5) and lys<strong>in</strong>e 12 (K12) (as well ascerta<strong>in</strong> sites with<strong>in</strong> H3) [20]. HATs generally conta<strong>in</strong> multiple subunits and their catalyticactivity depends, <strong>in</strong> part, on the context of the other subunits <strong>in</strong> those complexes [19].HDACs are also part of large multiprote<strong>in</strong> complexes and comprise a family of 18 genessubdivided <strong>in</strong>to four classes on the basis of their sequence homology to ortholog yeastprote<strong>in</strong>s, subcellular localization and enzymatic activities (Table 4.1) [21,22]. Of these classes,class I, II, and IV have <strong>in</strong> common a z<strong>in</strong>c (Zn)-dependent enzymatic activity [23], while class III(or Sirtu<strong>in</strong>s from the homology with the yeast Sir2) constitutes a separate, structurally unrelated,nicot<strong>in</strong>amide aden<strong>in</strong>e d<strong>in</strong>ucleotide (NAD)-dependent subfamily [24,25]. HDAC class I(HDAC1, 2, 3, and 8) are nuclear prote<strong>in</strong>s with homology to the yeast RPD3 prote<strong>in</strong> and areubiquitously expressed <strong>in</strong> various human cell l<strong>in</strong>es and tissues [21]. HDAC class II is constitutedby large prote<strong>in</strong>s with homology to the Hda1 yeast prote<strong>in</strong> and shuttle between thecytoplasm and the nucleus. They can be subdivided <strong>in</strong>to class IIa (HDAC4, 5, 7, and 9) and


CHAPTER 4Alterations of Histone Modifications <strong>in</strong> Cancerclass IIb (HDAC6 and 10) on the basis of the presence of a double deacetylase doma<strong>in</strong> typicalof HDAC6 and HDAC10 [26]. Class IV is represented by a s<strong>in</strong>gle prote<strong>in</strong>: HDAC11 which ischaracterized by a deacetylase doma<strong>in</strong> shar<strong>in</strong>g homology with both HDAC class I and class IIdoma<strong>in</strong>s [27].HDACs, <strong>in</strong> opposition to the enzymatic activity of HATs, reverse the acetylation of lys<strong>in</strong>e and <strong>in</strong>so do<strong>in</strong>g, restore the positive charge of the lys<strong>in</strong>e residue. In general, HDACs, as well as theHATs, have relatively low substrate specificity by themselves. In addition, the determ<strong>in</strong>ation oftheir specificity is complicated by the fact that these enzymes are present <strong>in</strong> multiple complexesoften <strong>in</strong>clud<strong>in</strong>g other HDAC family members [21]. For <strong>in</strong>stance, HDAC1 is found togetherwith HDAC2 with<strong>in</strong> the NuRD, S<strong>in</strong>3a, and Co-REST complexes and this may lead to changes<strong>in</strong> their activity/specificities [21]. Removal of acetylation tends to stabilize the chromat<strong>in</strong>structure and leads to a more closed chromat<strong>in</strong> conformation which suggests a ma<strong>in</strong>, but notexclusive, function of HDACs as transcriptional repressors.Histone acetylation is almost <strong>in</strong>variably associated with transcriptional activation [19] andalthough most of the acetylation sites fall with<strong>in</strong> the N-term<strong>in</strong>al tail of the histones, which aremore accessible for modification, acetylation with<strong>in</strong> the core doma<strong>in</strong> of H3 at lys<strong>in</strong>e 56(H3K56ac) has also been reported [28]. With respect to the specific role of histone acetylationon gene transcription, gene-specific and global effects can be dist<strong>in</strong>guished [16]. Histoneacetylation targeted at specific promoters regulates the transcription of specific genes, whereas,histone acetylation over large regions of chromat<strong>in</strong>, <strong>in</strong>clud<strong>in</strong>g cod<strong>in</strong>g regions and nonpromoterregions, affects global gene expression levels. A characteristic enrichment of histoneacetylation at enhancer elements, and particularly <strong>in</strong> gene promoters, where they presumablyfacilitate the transcription factor access, has been recently reported [29]. However, genomewidemapp<strong>in</strong>g of HATs and HDACs revealed that the above scenario, l<strong>in</strong>k<strong>in</strong>g histone acetylationto transcriptional activation and histone deacetylation to transcriptional repression, ismuch too simplistic [30]. The study conducted by Zhao and colleagues also revealeda surpris<strong>in</strong>gly strong association of HDACs with active genes. This observation suggests thatthere are two dist<strong>in</strong>ct functions for HDACs on active or primed genes. On active genes, byremov<strong>in</strong>g the acetyl group added dur<strong>in</strong>g transcription, HDACs would reset the elongation ofthe chromat<strong>in</strong> for subsequent transcription rounds. In contrast, on primed genes, HDACswould exert a repressive action by ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g low levels of acetylation, which <strong>in</strong> turn couldprevent pol II b<strong>in</strong>d<strong>in</strong>g and gene transcription [30]. The suppression of spurious <strong>in</strong>ternaltranscription <strong>in</strong>itiation is an additional function of HDACs that is thought to be important toassure accurate gene transcription and ma<strong>in</strong>ta<strong>in</strong> chromat<strong>in</strong> <strong>in</strong>tegrity follow<strong>in</strong>g transcription[31e33]. Sem<strong>in</strong>al studies conducted <strong>in</strong> yeast have demonstrated the relevant role of Rpd3Sdeacetylase <strong>in</strong> <strong>in</strong>hibit<strong>in</strong>g the assembly of transcription factors at <strong>in</strong>appropriate or “cryptic”sites with<strong>in</strong> genes and <strong>in</strong> the suppression of cryptic transcription <strong>in</strong>itiation [31e33].57The role of histone acetylation is not limited to the regulation of gene transcription butextended to additional processes such as nucleosomal assembly, chromat<strong>in</strong> fold<strong>in</strong>g,chromosome condensation, genome stability, DNA duplication and repair [16,28,34]. For<strong>in</strong>stance, a specific role of H4K16 acetylation (H4K16ac) <strong>in</strong> chromosome condensation [34]and a role of H3K56 acetylation (H3K56ac) <strong>in</strong> DNA repair and genome stability have beenreported [16,28]. The correlation between histone tail acetylation and the tim<strong>in</strong>g of replicationalso strongly supports a critical role of this histone modification <strong>in</strong> DNA replication [35]. Thisis further supported by the essential role of ING5eHBO1 acetyltransferase complex <strong>in</strong> DNAreplication [36].4.3.2 Histone MethylationHistone methylation is a reversible modification ma<strong>in</strong>ly occurr<strong>in</strong>g on the side cha<strong>in</strong>s of bothlys<strong>in</strong>es and arg<strong>in</strong><strong>in</strong>es [37e40]. Methylation is unique among the histone post-translationalmodifications because up to three methyl groups can be added to a s<strong>in</strong>gle lys<strong>in</strong>e residue,


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>creat<strong>in</strong>g a total of four different methyl states: un-methylated, mono-, di-, or trimethylatedstates. Furthermore, arg<strong>in</strong><strong>in</strong>e residues can undergo both monomethylation and dimethylation,with the latter <strong>in</strong> a symmetric or asymmetrical configuration [37e40]. Histone methylation, <strong>in</strong>contrast to acetylation and phosphorylation, does not alter the charge of the histone tails but<strong>in</strong>fluences the basicity and hydrophobicity and consequently the aff<strong>in</strong>ity of certa<strong>in</strong> moleculessuch as transcription factors toward DNA [38,41,42]. Histone methylation on lys<strong>in</strong>e andarg<strong>in</strong><strong>in</strong>e is mediated by histone lys<strong>in</strong>e methyltransferases (HMTs) (Table 4.2) [38, 43] andprote<strong>in</strong> arg<strong>in</strong><strong>in</strong>e methyltransferases (PRMTs) (Table 4.2) [37,44], respectively, and theseenzymes transfer a methyl group, from the cofactor S-adenosyl-L-methion<strong>in</strong>e to either theε-am<strong>in</strong>o group of lys<strong>in</strong>e or to the guanid<strong>in</strong>o group of arg<strong>in</strong><strong>in</strong>e.HMTs have been grouped <strong>in</strong> two ma<strong>in</strong> different classes: lys<strong>in</strong>e-specific SET doma<strong>in</strong> conta<strong>in</strong><strong>in</strong>ghistone methyltransferases, characterized by a 130-am<strong>in</strong>o-acid catalytic doma<strong>in</strong> known asSET (Su (var), Enhancer of Zeste, and Trithorax), and the non-SET-conta<strong>in</strong><strong>in</strong>g lys<strong>in</strong>emethyltransferases [45]. Remarkably, all the known HMTs have a SET doma<strong>in</strong> harbor<strong>in</strong>g theenzymatic activity with the exception of DOT1, that methylates lys<strong>in</strong>e-79 with<strong>in</strong> the coredoma<strong>in</strong> of H3 only <strong>in</strong> nucleosomal substrates and not <strong>in</strong> free histones [46]. The SET doma<strong>in</strong>conta<strong>in</strong><strong>in</strong>g HMTs catalyze mono-, di-, and trimethylation of their target lys<strong>in</strong>e residuelocalized on histone tails and are classified <strong>in</strong>to six subfamilies: SET1, SET2, SUV39, EZH,SMYD, and PRDM. Of note, a small number of SET-conta<strong>in</strong><strong>in</strong>g HMTs that do not fall <strong>in</strong>to theabove six subfamilies due to an absence of conserved sequences flank<strong>in</strong>g their SET doma<strong>in</strong>s<strong>in</strong>clude Set8/PR-Set7, SUV4-20H1, and SUV4-20H2, Set7/9, as well as MLL5, RIZ (ret<strong>in</strong>oblastomaprote<strong>in</strong>-<strong>in</strong>teract<strong>in</strong>g z<strong>in</strong>c-f<strong>in</strong>ger) and SMYD3 (SET- and MYND-doma<strong>in</strong> conta<strong>in</strong><strong>in</strong>gprote<strong>in</strong> 3) [43].58HMTs, <strong>in</strong> contrast to HATs and HDACs, which can be promiscuous <strong>in</strong> their histone substratespecificity, typically show a high degree of specificity toward their histone targets (Figure 4.2)[38,43] and are able to recognize and modulate different degrees of methylation on the samelys<strong>in</strong>e (i.e. mono- vs. di- and tri-methyaltion) [2,42].PRMTs prote<strong>in</strong>s, characterized by a common catalytic methyltransferase doma<strong>in</strong> and uniqueN-term<strong>in</strong>al and C-term<strong>in</strong>al regions, are divided <strong>in</strong>to two different classes: type I and type II[37,44]. The type II enzymes (PRMT 5, 7, and 9) catalyze mono- and di-symmetric methylationof the arg<strong>in</strong><strong>in</strong>es (R), whereas, the formation of mono- and di-asymmetric tails is achievedby type I enzymes (PRMT 1e4, 6, and 8). All the PRMTs transfer methyl group to severalsubstrates but PRMT1, 4, 5, and 6 are the most relevant enzymes with respect to histonearg<strong>in</strong><strong>in</strong>e methylation [37,44].In addition, with<strong>in</strong> the family of PR doma<strong>in</strong>-conta<strong>in</strong><strong>in</strong>g prote<strong>in</strong>s (PRDMs) [47], PRDM2(RIZ1) and PRDM6 have been shown to have <strong>in</strong>tr<strong>in</strong>sic methyltransferase activity toward lys<strong>in</strong>eresidues, such as lys<strong>in</strong>e 9 of histone H3 (H3K9) and lys<strong>in</strong>e 20 of histone H4 (H4K20) [48,49].Prior to recent identification of numerous prote<strong>in</strong>s <strong>in</strong>volved <strong>in</strong> histone demethylation, a s<strong>in</strong>glemechanism of arg<strong>in</strong><strong>in</strong>e deim<strong>in</strong>ation has been described. This mechanism is catalyzed bypeptidylarg<strong>in</strong><strong>in</strong>e deim<strong>in</strong>ase 4 (PAD4) [50,51] and characterized by the conversion of thearg<strong>in</strong><strong>in</strong>e to citrull<strong>in</strong>e via a deim<strong>in</strong>ation reaction [50,51]. The identification of this mechanismwas the first demonstration of the reversibility of the histone methylation mark, althoughrepresent<strong>in</strong>g deim<strong>in</strong>ation (that is, a further modification of the methylated residue) ratherthan a direct reversion of methylation [51]. Over the last few years several “bona fide” histonedemethylases, able to revert both lys<strong>in</strong>e and arg<strong>in</strong><strong>in</strong>e methylation, have been identified(Table 4.2) [51e54].Lys<strong>in</strong>e demethylat<strong>in</strong>g enzymes have been subdivided <strong>in</strong>to two ma<strong>in</strong> families: KDM1 (lys<strong>in</strong>e(K) demethylase 1) family which are FAD-dependent am<strong>in</strong>e oxidases, act<strong>in</strong>g only on monoanddimethylated lys<strong>in</strong>e [55] ,and the JMJC doma<strong>in</strong> conta<strong>in</strong> prote<strong>in</strong>s which are Fe(II) and 2-oxoglutarate-dependent enzymes able to remove all methylation states (Table 4.2) [54]. The


CHAPTER 4Alterations of Histone Modifications <strong>in</strong> CancerTABLE 4.2 Histone Modify<strong>in</strong>g Enzymes: Histone Lys<strong>in</strong>e Methyltransferases, HistoneLys<strong>in</strong>e Demethylases and Histone Arg<strong>in</strong><strong>in</strong>e MethyltransferasesCategory Gene Histone SpecificityHistone Lys<strong>in</strong>e MethyltransferasesHistone Lys<strong>in</strong>e DemethylasesSUV39-H1/KMT1A H3K9me 2,3SUV39-H2/KMT1B H3K9me 2,3G9a/KTM1C H3K9me 1,2EuHMTase/GLP/KMT1D H3K9me 1,2ESET/SETDB1/KMT1E H3K9me 2,3CLL8/KMT1FH3K9meMLL1/KMT2AH3K4me1,2,3MLL2/KMT2BH3K4me1,2,3MLL3/KMT2CH3K4me1,2,3MLL4/KMT2DH3K4me1,2,3MLL5/KMT2EH3K4mehSET1A/KMT2FH3K4me1,2,3hSET1B/KMT2GH3K4me1,2,3ASH1/KMT2HH3K4SET2/KMT3AH3K4NSD1/KMT3B H3K3me6 2,3NSD2H3K36me3NSD3 -SMYD1 -SMYD2/KMT3CH3K36me 2, H3K4meSMYD3 H3K4me 2,3DOT1L/KMT4H3K79PR-SET7e8/KMT5ASUV4e20H1/KMT5B H4K20me 2,3SUV4e20H2/KMT5C H4K20me 2,3EZH2/KMT6 H3K27me 2,3EZH1 H3K27me 2,3SET7e9/KMT7 H3K4 1RIZ1/KMT8H3K9meLSD1/KDM1AH3K4me1/2, H3K9me1/2LSD2/KDM1BH3K4me1/2, H3K9me1/2KDM2 cluster FBXL11A/JHDM1A/KDM2A H3K36me1/2FBXL10B/JHDM1B/ KDM2BH3K36me1/2KDM3 cluster JMJD1A,JHDM2A/ KDM3A H3K9me1/2JMJD1B/JHDM2B/KDM3BH3K9meKDM4 cluster JMJD2A/JHDM3A/ KDM4A H3K9/ K36me2/3JMJD2B/ KDM4BH3K9/ K36me2/3JMJD2C/GASC1/KDM4CH3K9/ K3K36me2/3JMJD2D/KDM4DH3K9me2/3KDM5 cluster JARID1A/RBP2/KDM5A H3K4me2/3JARID1B/PLU1/KDM5BH3K4me1/2/3JARID1C/SMCX /KDM5CH3K4me2/3JARID1D/SMCY/KDM5DH3K4me2/3KDM6 cluster UTX/KDM6A H3K27me2/3JMJD3/KDM6BH3K27me2/3KDM7H3K9me1/2, H3K27me1/2KDM8H3K36me2PFH8H3K9me1/2, H4K20meArg<strong>in</strong><strong>in</strong>e MethyltransferasesPRMT type IPRMT1, PRMT3, PRMT4,H4 mono di-symmetricPRMT6, PRMT8PRMT type II PRMT5, PRMT7, PRMT9 H4 mono di-symmetricNot def<strong>in</strong>edPRMT2, PRMT10, PRMT1159


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>KDM5A/JARID2AKDM5B/JARID2BKDM5C/JARID2CKDM5D/JARID2DKDM1A/LSD1KDM1B/LSD2H3MeK4KDM1A/LSD1KDM1B/LSD2KDM3A/JHDM2AKDM3B/JHDM2BKDM4A/JMJD2AKDM4B/JMJD2BKDM4C/JMJD2CKDM4D/JMJD2DKDM7SMYD3KMT7/Set7-9KMT2A/MLL1KMT2B/MLL2KMT2C/MLL3KMT2D/MLL4KMT2E/MLL5KMT2F/hSET1AKMT2G/hSet1BKMT2H/ASH1K9MeKMT1A/SUV39H1KMT1B/SUV39H2KMT1C/G9aKMT1D/GLPKMT1E/SETDB1KMT1F/CLL8KMT8/RIZ1KDM7KDM6A/UTXKDM6B/JMJD3MeK27K36MeKDM2A/JHDM1AKDM2B/JHDM1BKDM5A/JARID2AKDM5B/JARID2BKDM5C/JARID2CKDM8EZH1EZH2K79MeKMT3B/NSD1NSD2SMYD2/KTM3CKMT4/DOT1LK20MePHF8KMT5B/SUV4-20H1KMT5C/SUV4-20H2KMT5A/Pr-Set7/8Histone lys<strong>in</strong>e methyltransferasesH4Histone lys<strong>in</strong>e demethylasesFIGURE 4.2Histone lys<strong>in</strong>e methylases and demethylases. Histone H3 and H4 tails with known lys<strong>in</strong>e methylases (green box) and demethylases (red box). Lighter shades<strong>in</strong>dicate monomethylation by HMTs or demethylation of mono- and di-methylated lys<strong>in</strong>e residues by HDMs. Darker shade <strong>in</strong>dicates mono-, di-, and trimethylationby HMTs or demethylation of di- and tri-methylated lys<strong>in</strong>e residues by HDMs. This figure is reproduced <strong>in</strong> the color plate section.60JMJC doma<strong>in</strong>-conta<strong>in</strong><strong>in</strong>g prote<strong>in</strong>s, compris<strong>in</strong>g 25 prote<strong>in</strong>s of which 15 have confirmeddemethylase activity, are divided <strong>in</strong>to subfamilies based on sequence similarity: KDM2 (FBXL),KDM3 (JMJD1), KDM4 (JMJD2/JHDM3), KDM5 (Jarid1), KDM6(UTX/JMJD6), and Jumonjiclusters [54].The jumonji prote<strong>in</strong> JMJD6 is, at the moment, the only known enzyme able to demethylatearg<strong>in</strong><strong>in</strong>e (R) 2 and 3 specifically on histones H3 (H3R2) and H4 (H4R3) [56]. However, due tothe low catalytic rate observed for this enzyme, it is currently unclear whether this reactionactually occurs physiologically or whether there are, as yet, uncharacterized arg<strong>in</strong><strong>in</strong>e demethylasesthat perform this function <strong>in</strong> cells. As with the lys<strong>in</strong>e methyltransferases, the histonedemethylases possess a high level of substrate specificity with respect to their target lys<strong>in</strong>e andappear sensitive to the degree of lys<strong>in</strong>e methylation (Figure 4.2). The substrate specificity ofhistone demethylase can be further <strong>in</strong>fluenced by the association of additional prote<strong>in</strong>s. Forexample the specificity of KDM1A for H3K9 when <strong>in</strong> complex with steroid receptors changes toH3K4 when the prote<strong>in</strong> is <strong>in</strong> a complex with Co-Rest [57]. Interest<strong>in</strong>gly, this substrate switch<strong>in</strong>galso correlates with a switch<strong>in</strong>g of the role of KDM1A from activator to repressor of genetranscription [57].Histone methylation represents a mechanism of “mark<strong>in</strong>g” the histone <strong>in</strong> order to recruitseveral effector prote<strong>in</strong>s with recognition doma<strong>in</strong>s specific for different methylated lys<strong>in</strong>eresidues [58]. For <strong>in</strong>stance, plant homeodoma<strong>in</strong> (PHD) of bromodoma<strong>in</strong>-PHD transcriptionfactor(BPTF) b<strong>in</strong>ds to H3K4 trimethylated/dimethylated (H3K4me3/me2) and recruits thenucleosome remodel<strong>in</strong>g factor (NURF) complex to the target gene lead<strong>in</strong>g to gene activation[59]. In contrast, the chromodoma<strong>in</strong> of heterochromat<strong>in</strong> prote<strong>in</strong> 1 (HP1) b<strong>in</strong>ds H3K9trimethylated (H3K9me3) lead<strong>in</strong>g to heterochromat<strong>in</strong> formation and gene silenc<strong>in</strong>g [60].In contrast to acetylation, which is generally associated with transcriptional activation, histonelys<strong>in</strong>e methylation correlates with either an activation or a repression of transcription


CHAPTER 4Alterations of Histone Modifications <strong>in</strong> Cancerdepend<strong>in</strong>g both on the methylated site and the degree of methylation [41,42]. A betterunderstand<strong>in</strong>g of the organization and the complexity of histone methlyation and acetylationhas come from the generation of high-resolution, genome-wide maps of the distribution ofhistone lys<strong>in</strong>e and arg<strong>in</strong><strong>in</strong>e methylations [61] as well as from the analysis of comb<strong>in</strong>atorialpattern of histone acetylation and methylation [29]. These works [29,61] led to the identificationof dist<strong>in</strong>ct and comb<strong>in</strong>atorial patterns of histone marks at different genomic regions<strong>in</strong>clud<strong>in</strong>g promoters, <strong>in</strong>sulators, enhancers, and transcribed regions. More importantly, theseanalyses have highlighted the cooperative manner <strong>in</strong> which diverse modifications can act toglobally regulate gene expression. For <strong>in</strong>stance, these studies clearly demonstrated the associationof methylation of histones H3K9, H3K27, and H4K20 with gene silenc<strong>in</strong>g, as well asthe l<strong>in</strong>k between active gene transcription and H3K4, H3K36, and H3K79 methylation.Interest<strong>in</strong>gly, a strict equilibrium between methylation of H3K4 which activates transcriptionand methylation of H3K27, which represses transcription, was recently reported to beimportant <strong>in</strong> the activity of “stemness” transcription patterns to ma<strong>in</strong>ta<strong>in</strong> pluripotency ofembryonic stem cells [62].4.3.3 Histone PhosphorylationPhosphorylation of histones, like the other histone modifications, is a highly dynamic processspecifically characterized by the addition of a phosphate group from ATP to the hydroxyl group ofthe target am<strong>in</strong>o acid side cha<strong>in</strong> of several and different residues with<strong>in</strong> histone tails. The additionof phosphate and hence negative charge is able to modify the chromat<strong>in</strong> structure and <strong>in</strong> so do<strong>in</strong>g,is able to <strong>in</strong>fluence <strong>in</strong>teractions between transcription factors and other chromat<strong>in</strong> components[63,64]. Histone phosphorylation takes place on ser<strong>in</strong>e (S), tyros<strong>in</strong>e (Y), and threon<strong>in</strong>e (T), withthe large majority of histone phosphorylation sites be<strong>in</strong>g found with<strong>in</strong> the N-term<strong>in</strong>al tails andonly a very few examples, such as H3Y41, are found with<strong>in</strong> the histone core [63,65].Dist<strong>in</strong>ct phosphorylation patterns of histones have been l<strong>in</strong>ked to several cellular processes[63,64]. The contribution and <strong>in</strong>terdependency of cross-talk between histone phosphorylationand other histone modifications is important <strong>in</strong> def<strong>in</strong><strong>in</strong>g the role of histone phosphorylation.This is clearly seen <strong>in</strong> the <strong>in</strong>terdependency of histone acetylation and methylation on phosposhorylationof histone H3 and vice versa [66]. A well-characterized case of this<strong>in</strong>terdependency is the phosphorylation of H3 at S10 which, to facilitate gene transcription,enhances H3K14 acetylation and H3K4 methylation and simultaneously <strong>in</strong>hibits H3K9methylation [66]. Conversely, the methylation of H3K9 <strong>in</strong>terferes with H3S10 phosphorylation[66]. As with acetylation and methylation, histone phosphorylation represents a histone markrecognized by specific ancillary prote<strong>in</strong>s which <strong>in</strong> this case comprise the 14-3-3 prote<strong>in</strong> family[67,68]. Several dist<strong>in</strong>ct histone k<strong>in</strong>ases and histone phosphatases have been identified (Table4.3) [63,64]. For example, phosphorylation of histone H2AX (a variant of histone H2A),<strong>in</strong>duced by a DNA-damage signal<strong>in</strong>g pathway is dependent on phosphatidyl<strong>in</strong>ositol-3-OHk<strong>in</strong>ases (PI3Ks) such as ATM, ATR, and DNA-PK [69]. Histone H2B phosphorylation at S14,catalyzed by Mst1 (mammalian sterile-20-like k<strong>in</strong>ase), has a role <strong>in</strong> the <strong>in</strong>duction of apoptosis[70]. Phosphorylation of histone H3 at S10 and S28, associated with the seem<strong>in</strong>gly contrast<strong>in</strong>gfunctions of chromat<strong>in</strong> condensation and transcriptional activation, is due to the catalyticactivity of aurora k<strong>in</strong>ase family and primarily to aurora-B activity [71e73]. Other k<strong>in</strong>asesmediat<strong>in</strong>g the phosphorylation of histone H3S10 and regulat<strong>in</strong>g gene expression have beenidentified <strong>in</strong> the MSK/RSK/Jil-1 family [73]. Hasp<strong>in</strong> has recently been identified as the k<strong>in</strong>aseresponsible for the phosphorylation at H3T3, an event required for normal metaphasechromosome alignment [74], while Dlk/ZIP k<strong>in</strong>ase (Dlk: Death-associated prote<strong>in</strong> (DAP)-likek<strong>in</strong>ase, ZIP: Zipper <strong>in</strong>teract<strong>in</strong>g prote<strong>in</strong> k<strong>in</strong>ase) is the enzyme responsible for phosphorylation ofH3T11 dur<strong>in</strong>g mitosis [75]. Recently, Chk1 has been identified as another histone k<strong>in</strong>ase thatregulates DNA damage-<strong>in</strong>duced transcriptional repression through the phosphorylation ofH3T11 [76]. Ribosomal S6 k<strong>in</strong>ase 2 (RSK2) appears to be the ma<strong>in</strong> k<strong>in</strong>ase that catalyzes thephosphorylation of H2BS32 and H2AX [77,78]. JAK2 is the k<strong>in</strong>ase responsible for61


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>TABLE 4.3 Histone-Modify<strong>in</strong>g Enzymes: K<strong>in</strong>ases, Phosphatases, and Ubiquitilases62Category Gene Histone SpecificityK<strong>in</strong>asesAURORA-BH3S10,H3S28MST1H2BS14ATM, ATR, DNAPK.H2AXMSK-RSKH3S10HASPINH3T3DLK/ZIPH3T11CHK1H3T11JAK2H3Y41PhosphatasesPP1H3S10,H3S28PP2AH2AXPPgH3T11UbiquitilasesE2 and E3 ligases hHR6A/hHR6B (E2 ligase) H2BK120RNF20 (E3 ligase)H2BK120RING1B , 2A-HUBH2AK119(E3 ligases)RNF8 (E3 ligase)H2AK119Dubs USP22 H2AK119USP16H2BK120MYSM1H2BK120USP3H2BK120USP7H2AK119BRCC36H2AXphosporylation of H3Y41 [65] and <strong>in</strong> do<strong>in</strong>g so determ<strong>in</strong>es the release of the transcriptionalrepressor heterochromat<strong>in</strong> prote<strong>in</strong> 1a (HP1a) from chromat<strong>in</strong> [65].Little is known about the role of prote<strong>in</strong> phosphatases <strong>in</strong> regulat<strong>in</strong>g the dephosphorylation ofhistones. Prote<strong>in</strong> phosphatase 1 (PP1) appears to be <strong>in</strong>volved <strong>in</strong> the dephosphorylation ofH3S10 and H3S28 [73,74]. Prote<strong>in</strong> phosphatase 2 (PP2A) is responsible for the dephosphorylationof H2AX after DNA repair [79,80] and Prote<strong>in</strong> phosphatase g (PPg) is <strong>in</strong>volved<strong>in</strong>to dephosphorylation of H3T11 after DNA damage [81].4.3.4 Histone Ubiquit<strong>in</strong>ationHistone ubiquit<strong>in</strong>ation differs substantially from the other modifications, s<strong>in</strong>ce it is a verylarge modification with the ubiquit<strong>in</strong> moiety compris<strong>in</strong>g of a 76-am<strong>in</strong>o-acid polypeptide [82].Histone ubiquit<strong>in</strong>ation is a reversible modification whose steady state is determ<strong>in</strong>ed by twoenzymatic activities <strong>in</strong>volved <strong>in</strong> addition and removal of the ubiquit<strong>in</strong> moiety from histones[39]. Histone ubiquit<strong>in</strong>ation, occurr<strong>in</strong>g <strong>in</strong> human ma<strong>in</strong>ly on histone H2A at lys<strong>in</strong>e 119(H2AK119ub1) and histone H2B at lys<strong>in</strong>e 120 (H2BK120ub1), is catalyzed by the formationof an isopeptide bond between the carboxy-term<strong>in</strong>al glyc<strong>in</strong>e of ubiquit<strong>in</strong> and the ε-group ofa lys<strong>in</strong>e residue on carboxyterm<strong>in</strong>al tail of the histones. This bond is the result of the sequentialcatalytic actions of E1 activat<strong>in</strong>g, E2 conjugat<strong>in</strong>g, and E3 ligase enzymes with the E3 ligasesresponsible for specific recognition and ligation of ubiquit<strong>in</strong> to its substrates [39,82,83].Substrates can be both poly- and monoubiquit<strong>in</strong>ated. Polyubiquit<strong>in</strong>ation creates an irreversiblesignal for proteosomal-mediated degradation whilst monoubiquit<strong>in</strong>ation generatesa regulatory signal which can be reversed by the action of ubiquit<strong>in</strong>-specific proteases called


CHAPTER 4Alterations of Histone Modifications <strong>in</strong> Cancerdeubiquit<strong>in</strong>at<strong>in</strong>g enzymes [39,82,83]. Histone ubiquit<strong>in</strong>ation occurs largely <strong>in</strong> the monoubiqut<strong>in</strong>atedform and correlates with active and open chromat<strong>in</strong>, although histone ubiquit<strong>in</strong>ationhas been l<strong>in</strong>ked with both transcriptional activation and silenc<strong>in</strong>g depend<strong>in</strong>g on thegenomic context [39,84,85]. Furthermore, a role of monoubiquit<strong>in</strong>ation at histone H2Al<strong>in</strong>ked to DNA repair mechanism has been reported [86]. Interest<strong>in</strong>gly, conjugation of a s<strong>in</strong>gleubiquit<strong>in</strong> moiety to histone H2A results <strong>in</strong> a significantly different outcome when compared tothe addition of ubiquit<strong>in</strong> to H2B. H2A ubiquit<strong>in</strong>ation, be<strong>in</strong>g associated predom<strong>in</strong>antly withtranscriptional repression, may be considered a repressive mark whilst H2B ubiquit<strong>in</strong>ationappears be <strong>in</strong>volved both <strong>in</strong> transcriptional activation and gene silenc<strong>in</strong>g [84e90]. The role ofhistone ubiquit<strong>in</strong>ation <strong>in</strong> transcriptional control is due to both a direct effect of histone H2Bubiquit<strong>in</strong>ation on RNA polymerase II transcription elongation [85e87] and to the cross-talkwith other histone modifications [39,84,88e90]. Several data have demonstrated the ability ofhistone H2B ubiquit<strong>in</strong>ation to directly promote accurate and efficient RNA pol II transcriptionaffect<strong>in</strong>g nucleosomal dynamics [85e87]. Further, <strong>in</strong> the context of trans-histone cross-talk, itis well documented that the monoubiquit<strong>in</strong>ation of H2B is required for lys<strong>in</strong>e methylation ofhistone H3K4 whilst this methylation is <strong>in</strong>hibited by the monoubiquit<strong>in</strong>ation of H2A[39,84,88e90]. The possible molecular mechanisms l<strong>in</strong>k<strong>in</strong>g histone ubiquit<strong>in</strong>ation to transcriptionalregulation are at least two. One mechanism envisages that the addition of a largemacromolecule, such as ubiquit<strong>in</strong>, to a histone tail would lead to a modification of the highorderchromat<strong>in</strong> structure. The other one suggests that ubiquit<strong>in</strong>ation represents a signal forsuccessive histone modifications, and/or a signal for recruitment of other prote<strong>in</strong>s to thechromat<strong>in</strong>. For example, recent evidence obta<strong>in</strong>ed us<strong>in</strong>g chemically modified histones,demonstrated that H2B monoubiquit<strong>in</strong>ation, <strong>in</strong> contrast to H2A ubiquit<strong>in</strong>ation, had a clearimpact on chromat<strong>in</strong> organization by <strong>in</strong>hibit<strong>in</strong>g both nucleosome-array fold<strong>in</strong>g as well as<strong>in</strong>terfiber oligomerization [90].Table 4.3 lists some of the enzymes <strong>in</strong>volved <strong>in</strong> this modification. The E2 conjugat<strong>in</strong>g enzymeshHR6A/hHR6B and the E3 ligases, RNF20, are responsible of H2B ubiquit<strong>in</strong>ation <strong>in</strong> cells[91e94]. R<strong>in</strong>g1B and 2A-HUB are the E3 ligase responsible of H2A ubiquit<strong>in</strong>ation [95,96],whilst RNF8 is E3 ligase responsible for monoubiquit<strong>in</strong>ation of H2A dur<strong>in</strong>g DNA repair[97,98]. The deubiquit<strong>in</strong>at<strong>in</strong>g enzyme (Dubs) USP22, as a component of hSAGA complex, is<strong>in</strong>volved <strong>in</strong> the deubiquit<strong>in</strong>ation of both H2B and H2A [99e101]. USP22 is required for thetranscription of cell cycle genes and target genes regulated by the Myc oncoprote<strong>in</strong> whereby itis recruited to specific target gene loci [99e101]. In addition, Ubp-M (USP16) and 2A-DUB(MYSM1) have also been implicated <strong>in</strong> the deubiquit<strong>in</strong>ation of H2B [102], whilst recentlyUSP3 [103] and USP7 [104] have been suggested to be <strong>in</strong>volved <strong>in</strong> the deubiquit<strong>in</strong>ation ofboth H2B and H2A <strong>in</strong> human cells. BRCA1-conta<strong>in</strong><strong>in</strong>g complex (BRCC36) appears to be theDUB responsible for the deubiquit<strong>in</strong>ation of H2AX [105].634.3.5 Mode of Action of Histone ModificationsThe molecular mechanisms underly<strong>in</strong>g the function of each <strong>in</strong>dividual histone modification canbe generalized <strong>in</strong>to two categories: “cis”and“trans”’ mechanisms [106]. Thecis mechanism, forwhich histone acetylation and phosphorylation represent the best examples, corresponds toalterations of <strong>in</strong>tra- and <strong>in</strong>ternucleosomal contacts via changes of steric or charge <strong>in</strong>teractions,<strong>in</strong>fluenc<strong>in</strong>g chromat<strong>in</strong> structure [106].Thetrans mechanism is characterized by the <strong>in</strong>volvementof non-histone prote<strong>in</strong> “readers” that b<strong>in</strong>d to specific histone modifications giv<strong>in</strong>g rise tofunctional consequences [107]. Asanticipated,thecis mechanism is responsible for a directstructural perturbation of the chromat<strong>in</strong>. Explicative of this is the case of acetylation that, byreduc<strong>in</strong>g the positive charge of histones, disrupts the electrostatic <strong>in</strong>teraction between histonesand DNA, and <strong>in</strong> so do<strong>in</strong>g <strong>in</strong>fluences chromat<strong>in</strong> organization [2,106].The enrichment of multiple histone acetylation sites on regions <strong>in</strong>volved <strong>in</strong> active transcription,such as gene promoters, represents a strik<strong>in</strong>g example of the cis mechanism [29]. The


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>fundamental role of the H4K16 acetylation <strong>in</strong> the control of the chromat<strong>in</strong> structuredemonstrates that a s<strong>in</strong>gle modification site can also have a strong impact on chromat<strong>in</strong>organization and reveals that the presence of multiple acetylation sites is not necessary to<strong>in</strong>voke gross structural changes <strong>in</strong> chromat<strong>in</strong> [31]. Similarly, conjugation with ubiquit<strong>in</strong> cancause direct structural perturbations <strong>in</strong> chromat<strong>in</strong>. The ubiquit<strong>in</strong>ation of histone H2B hasbeen shown to disrupt compaction of local and higher-order chromat<strong>in</strong> [90]. In the transmechanism, histone modifications represent a mark for the recruitment of so-called “chromat<strong>in</strong>readers” [108e110]. These prote<strong>in</strong>s are characterized by recogniz<strong>in</strong>g specific histonemodifications and <strong>in</strong>clude bromodoma<strong>in</strong> prote<strong>in</strong>s that recognize lys<strong>in</strong>e acetylation [111], the“Royal super family” compris<strong>in</strong>g chromodoma<strong>in</strong>, Tudor, PWWP, and MBT doma<strong>in</strong> thatrecognize histone lys<strong>in</strong>e methylation [108e113], and PHD f<strong>in</strong>gers prote<strong>in</strong> [111]. Additionalhistone modification “readers” <strong>in</strong>clude the 14-3-3 sigma prote<strong>in</strong> which recognized histonephosphorylation [68] and MDC1 which conta<strong>in</strong>s tandem BRCT doma<strong>in</strong>s that b<strong>in</strong>d to gH2AX,the DSB-<strong>in</strong>duced phosphorylated H2A variant [114].64Interest<strong>in</strong>gly, with<strong>in</strong> the group of methyl lys<strong>in</strong>e b<strong>in</strong>ders the same modified site can berecognized by different doma<strong>in</strong>s. For example, trimethylation of H3K4 b<strong>in</strong>ds both thetandem chromodoma<strong>in</strong>s with<strong>in</strong> CHD1 and the tandem Tudor doma<strong>in</strong>s with<strong>in</strong> JMJD2A[115,116]. In some cases, the chromat<strong>in</strong> reader prote<strong>in</strong>s can also simultaneously b<strong>in</strong>ddifferent histone modifications, as <strong>in</strong> the case of the L3MBTL1 prote<strong>in</strong> simultaneously b<strong>in</strong>dsto mono/dimethylated lys<strong>in</strong>e 20 of histone H4 (H4K20me1/2) and mono/dimethylatedlys<strong>in</strong>e 26 of histone H1B (H1BK26me1/2), and <strong>in</strong> so do<strong>in</strong>g compacts nucleosomal arraysbear<strong>in</strong>g the two histone modifications [117]. Not only can histone modifications generatea platform for “reader” recruitment but they can also disrupt <strong>in</strong>teractions between histonesand “readers”. For example, H3K4me3 can prevent the NuRD complex from b<strong>in</strong>d<strong>in</strong>g to theH3 N-term<strong>in</strong>al tail [118], as well as prevent b<strong>in</strong>d<strong>in</strong>g of the PHD f<strong>in</strong>ger of DNMT3L to the H3tail [119].The functional consequences of histone modifications can be of two different types: establishmentof global chromat<strong>in</strong> environments and orchestration of DNA-based biological taskssuch as transcription, chromosome condensation, DNA replication, and repair [106,107].Histone modifications contribute to the establishment of the global chromat<strong>in</strong> environmentby arrang<strong>in</strong>g the genome <strong>in</strong>to dist<strong>in</strong>ct doma<strong>in</strong>s. For example, euchromat<strong>in</strong>, where DNA is kept‘‘accessible’’ for transcription and heterochromat<strong>in</strong>, where chromat<strong>in</strong> is ma<strong>in</strong>ta<strong>in</strong>ed ‘‘<strong>in</strong>accessible’’for transcription [106,107,120]. Euchromat<strong>in</strong> is typically enriched <strong>in</strong> acetylatedhistones and methylation of H3K4, H3K36, and H3K79, whilst heterochromat<strong>in</strong> is characterizedby histone hypoacetylation, a high level of methylation on H3K9, H3K27, and H4K20and association of heterochromat<strong>in</strong> prote<strong>in</strong>-1 (HP1) [121,122]. Histone modifications coord<strong>in</strong>atechromat<strong>in</strong> fold<strong>in</strong>g to facilitate the execution of specific functions [106,107]. Generally,for transcription, histone modifications can be divided <strong>in</strong>to those correlat<strong>in</strong>g with activationand those correlat<strong>in</strong>g with repression. However, under specific conditions, certa<strong>in</strong> types ofmodification have the potential to either activate or repress transcription [2,106,107]. Forexample, acetylation, methylation, phosphorylation, and ubiquit<strong>in</strong>ation have all beenimplicated <strong>in</strong> activation of transcription [123], whereas methylations and ubiquit<strong>in</strong>ation havealso been implicated <strong>in</strong> repression [123]. These functions can be both local, as <strong>in</strong> the case ofregulation of specific gene transcription, and genome wide as <strong>in</strong> the case of DNA replication,and chromosome condensation [2,104].An important feature is that histone modifications have both short- and long-term functionaleffects [123]. An example of the short-term effect can be seen by the rapid and cyclic changes <strong>in</strong>histone modifications associated with transcription <strong>in</strong> response to external stimulation [124].In this case, histone modifications on chromat<strong>in</strong> are the endpo<strong>in</strong>t of a signal<strong>in</strong>g pathway thatcorresponds to a mechanism through which the genome responds to external stimuli. Anexample of the longer-term effects of histone modification on genomic function is evident <strong>in</strong>


CHAPTER 4Alterations of Histone Modifications <strong>in</strong> Cancerthe def<strong>in</strong>ition and ma<strong>in</strong>tenance of chromat<strong>in</strong> structures throughout the cell cycle [125].Histone modifications hav<strong>in</strong>g the longest effect are related to modification of heterochromat<strong>in</strong>.Constitutive heterochromat<strong>in</strong> is characterized by a specific pattern of histone modifications<strong>in</strong>clud<strong>in</strong>g an enrichment of trimethylation of H3K9 and H4K20 and a depletion ofoverall acetylation [121,122]. Similarly, facultative heterochromat<strong>in</strong>, as observed <strong>in</strong> the <strong>in</strong>activeX chromosome of females, is characterized by the loss of H3K4 methylation and an<strong>in</strong>crease <strong>in</strong> H3K27 methylation [126].4.3.6 Histone Cross-TalkHistone modifications do not necessarily act on a stand-alone basis and frequently crosstalk<strong>in</strong>g<strong>in</strong>teractions are <strong>in</strong> place as a mechanism of signal<strong>in</strong>g <strong>in</strong>tegration [2,107,127]. Histonecross-talk occurs on s<strong>in</strong>gle and multiple histone tails and between histones with<strong>in</strong> the same or<strong>in</strong> different nucleosomes [127e129]. A first level of cross-talk can be identified <strong>in</strong> the mutuallyexclusive antagonism between different types of modifications, such as acetylation andmethylation, occurr<strong>in</strong>g on the same lys<strong>in</strong>e residue. Another level corresponds to the <strong>in</strong>terdependencybetween different modifications. A good example of histone trans-tail cross-talk,reported for the first time <strong>in</strong> yeast [130,131], is represented by the prerequisite of ubiquit<strong>in</strong>ationof H2BK123 for H3K4 methylation, lead<strong>in</strong>g to the silenc<strong>in</strong>g of genes located nearchromosome telomers [130,131]. An additional example of this trans-histone tail cross-talk isthe dependency of H3K4 methylation by sCOMPASS and H3K79 methylation by scDot1 onthe ubiquitylation of H2BK123 by scRad6/Bre1 [132]. Interest<strong>in</strong>gly, this mechanism isconserved <strong>in</strong> humans [133]. Furthermore, adjacent modifications can <strong>in</strong>fluence each other as<strong>in</strong> the case of HP1 b<strong>in</strong>d<strong>in</strong>g to H3K9 methylation be<strong>in</strong>g affected by the presence of an adjacentphosphorylated H3S10 [134].Additionally, the catalytic activity of an enzyme could be <strong>in</strong>fluenced by modification of itssubstrate recognition site, for example the isomerization of H3P38 can <strong>in</strong>fluence the ability ofSet2 to methylate H3K36 [135].65Similarly, the efficiency of substrate recognition by an enzyme could be different <strong>in</strong> the contextof an additional modification as demonstrated by higher efficiency of H3 recognition byGCN5 acetyltransferase <strong>in</strong> the presence of H3S10 phosphorylation [136]. A further level<strong>in</strong>volves a cooperative mechanism between some modifications to recruit specific factors, anexample of which is the <strong>in</strong>volvement of H3K9 and H3K14 acetylation <strong>in</strong> the recruitment ofPHF8 to methylated H3K4 [110].Equally important <strong>in</strong> the f<strong>in</strong>e tun<strong>in</strong>g control of chromat<strong>in</strong> organization is the <strong>in</strong>terplaybetween the histone modifications, DNA methylation [137,138] and ATP-dependent chromat<strong>in</strong>remodel<strong>in</strong>g [139]. The large number of histone modifications and the possible<strong>in</strong>terplay between them led to the proposition of the so-called “histone code hypothesis” <strong>in</strong>which “multiple histone modifications, act<strong>in</strong>g <strong>in</strong> a comb<strong>in</strong>atorial or sequential fashion onone or multiple histone tails, specify unique downstream functions” [140,141]. Thishypothesis led the scientific community to adopt some metaphors to describe it such that thecode is written by some enzymes (“writers”), removed by others (“erasers”), and is readilyrecognized by prote<strong>in</strong>s (“readers”) recruited to modifications through the b<strong>in</strong>d<strong>in</strong>g of specificdoma<strong>in</strong>s.Independently of the debate on the accuracy of consider<strong>in</strong>g the cross-talk between histonemodifications as a histone code rather than an epigenetic code or a language of histone crosstalk[142,143], it is evident that the dynamic plasticity of chromat<strong>in</strong>, necessary for the controlof cellular processes, represents the end po<strong>in</strong>t of a f<strong>in</strong>e-tun<strong>in</strong>g mechanism that <strong>in</strong>volves theconcerted action of the histone modifications together with the actions of DNA and chromat<strong>in</strong>remodel<strong>in</strong>g prote<strong>in</strong>s.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>4.4 HISTONE MODIFICATIONS AND CANCEREpigenetic alterations have been known to occur <strong>in</strong> cancer cells for decades. In fact, aberrantDNA methylation was described as an early event <strong>in</strong> tumorigenesis, although only recently wasrecognized to play a causal role, and not just an unwanted molecular consequence of thetransformation process. With this appreciation, it became clear that other epigenetic modifications,such as histone post-translational changes, are also altered <strong>in</strong> cancer cells. The firstexamples of altered patterns of histone modifications <strong>in</strong> cancer [144,145] were found us<strong>in</strong>gdifferent methodologies (liquid chromatographyeelectrospray mass spectrometry (LC-ES/MS) [146] and immunohistochemical (IHC) analysis [145]). One study showed the globallevel of trimethylation of H4K20 (H4K20me3) and acetylation of H4K16 (H4K16ac) <strong>in</strong> severaltypes of cancer cells [144], while another reported the global level of the dimethylation ofH3K4 (H3K4me2) and H3R3 (H3R3me2) as well as the level of acetylation of H3K9, H3K18,and H4K12 <strong>in</strong> primary prostate cancer tissues [145]. An impressive set of data/publications hasconfirmed and extended those <strong>in</strong>itial studies. A comprehensive analysis of all of the alterations<strong>in</strong> the histone modification patterns found <strong>in</strong> cancer cells is prohibitive and beyond the scopeof our work. However, we will try to review several cases of well-documented alterations <strong>in</strong>histone modifications <strong>in</strong> cancer and discuss their mechanistical implications. For clarity, wewill describe separately histone H4 and histone H3 alterations.664.4.1 Alterations <strong>in</strong> the Pattern of Histone H4 ModificationsFraga and colleagues [144] compared histone H4 modifications <strong>in</strong> normal lymphocytesaga<strong>in</strong>st several types of cancer cells and reported, for the first time, a globally lower level ofboth trimethylation of H4K20 (H4K20me3) and acetylation of H4K16 (H4K16ac) <strong>in</strong> cancertissues, which was further validated <strong>in</strong> an analysis of primary tumors versus their normalcounterpart [144]. Interest<strong>in</strong>gly, the reduction of both histone modifications correlated withDNA hypomethylation at repetitive DNA sequences and with silenc<strong>in</strong>g of the promoter oftumor suppressor genes such as CDKN2A, BRCA1, and MLH1 [144]. In the same study, us<strong>in</strong>ga well-recognized model of tumor progression (the mouse multistage sk<strong>in</strong> carc<strong>in</strong>ogenesismodel), the authors reported that the loss of those two histone modifications occurredprogressively from the first stage of carc<strong>in</strong>ogenesis, represented by benign papilloma, to themost malignant stage [144]. The progressive loss of H4K20me3 has been subsequentlyobserved <strong>in</strong> additional animal models of carc<strong>in</strong>ogenesis [146], <strong>in</strong>clud<strong>in</strong>g estradiol-<strong>in</strong>ducedmammary carc<strong>in</strong>ogenesis <strong>in</strong> rats [147], and then re<strong>in</strong>forced by several studies performed ontissues derived from different cancer patients [148e152]. An aberrant pattern of histone H4modifications, characterized by hypoacetylation of H4K12/H4K16, a loss of H4K20me3, andhyperacetylation of H4K5/H4K8, has been reported <strong>in</strong> a study conducted on non-small-celllung cancer (NSCL) patients [148]. Loss of H4K20me3 <strong>in</strong> this case also represents an earlyevent <strong>in</strong> tumorigenesis that was already present <strong>in</strong> early lesions and that becomes more evidentdur<strong>in</strong>g the sequential progression of disease mov<strong>in</strong>g from cell hyperplasia to metaplasia,dysplasia, and then to carc<strong>in</strong>oma <strong>in</strong> situ [148]. Reduction of H4K20me3 was more frequent <strong>in</strong>squamous cell carc<strong>in</strong>omas (67%) compared to adenocarc<strong>in</strong>omas (27%), whilst H4K16ac wasmore homogeneously reduced <strong>in</strong> the two histological types [148]. In lung adenocarc<strong>in</strong>omas,the observed down-regulation of H4K20me3 correlated with prognosis and permitted theidentification of two populations of stage I tumor samples with dist<strong>in</strong>ct cl<strong>in</strong>ical outcomewhere a longer survival was observed <strong>in</strong> patients hav<strong>in</strong>g higher levels of H4K20me3 [148].Interest<strong>in</strong>gly, loss of H4K20me3 correlated with decreased expression of a specific H4K20trimethyltransferase, Suv4-20h2 [148]. Similar f<strong>in</strong>d<strong>in</strong>gs were also obta<strong>in</strong>ed <strong>in</strong> an experimentalmodel of hepatocarc<strong>in</strong>ogenesis <strong>in</strong>duced by methyl deficiency <strong>in</strong> rats, strengthen<strong>in</strong>g the l<strong>in</strong>kbetween the two events [153]. In ovarian cancer, a significant correlation between H4K20me3loss and an <strong>in</strong>crease <strong>in</strong> malignancy has been observed by an IHC analysis conducted onnormal ovarian epithelium, ovarian adenomas, and ovarian epithelial carc<strong>in</strong>omas [149]. Inaddition, <strong>in</strong> breast cancer, reduced levels of both H4K20me3 and H4K16ac correlated well


CHAPTER 4Alterations of Histone Modifications <strong>in</strong> Cancerwith tumor grade, progression of disease, and with worse prognosis [150]. Similarly,a progressive loss of H4K16ac and H4K20me3 has been reported from low- to high-grade lungneuroendocr<strong>in</strong>e tumors, reflect<strong>in</strong>g both the degree of differentiation and the proliferation rateof the tumors [151]. In bladder cancer [152], IHC analysis of the global expression levels ofdifferent histone modifications (H3K4me1, H3K4me3, H4K20me1, H4K20me2, andH4K20me3), generally confirmed a correlation between cancer progression and a progressivedecrease of histone modification levels. However, <strong>in</strong>creased H4K20me3 correlated with worseprognosis [152]. Therefore, changes <strong>in</strong> H4K20 methylation levels appear to be frequentlyassociated with chromat<strong>in</strong> alterations <strong>in</strong> cancer cells, but the precise significance of this f<strong>in</strong>d<strong>in</strong>gis not necessarily consistent from cancer to cancer, exclud<strong>in</strong>g a simple <strong>in</strong>terpretation of thisphenomenon.Other histone H4 modifications have been found associated with cancer cells. As mentionedabove, H4K16 hypoacetylation correlates with worse prognosis <strong>in</strong> breast cancer and medulloblastoma[150,154]. In breast cancer, a study conducted on a very large dataset of patientsrevealed low or absent acetylation of H4K16 <strong>in</strong> the majority of analyzed cases and a strongcorrelation with cl<strong>in</strong>ico-histological features such as tumor grade, vascular <strong>in</strong>vasion, andprognosis [150]. In medulloblastoma patients, a concurrent analysis of H4K16 acetylation andof the acetylase responsible for the modification (hMOF), demonstrated that the hypoacetylationof H4K16 <strong>in</strong> primary medulloblastomas, compared to normal tissues, correlatedwell with a reduction <strong>in</strong> hMOF and poor prognosis [154].H4K12 acetylation (H4K12ac) is another histone H4 modification found altered <strong>in</strong> cancer[145,148,155,156]. A good correlation between hypoacetylation of H4K12, tumor grade, andcancer recurrence has been reported <strong>in</strong> prostate cancer patients [145]. In this cancer type, theprognostic value of H4K12ac was <strong>in</strong>dependent of tumor stage. If measured together with H3K9and H3K18 acetylation, H4K12 acetylation permitted the cluster<strong>in</strong>g of low-grade prostatecancer cases (Gleason 6 or less) <strong>in</strong>to two prognostically separate groups [145]. This f<strong>in</strong>d<strong>in</strong>ghighlights another important pr<strong>in</strong>ciple (see also below): it will require an <strong>in</strong>tegrated analysisof the different histone modifications to reveal complex histone patterns that will lead toa more consistent “epigenetic” classification of cancer types rather than a s<strong>in</strong>gle histonemodification which will only provide partial <strong>in</strong>formation.67A general decrease <strong>in</strong> H4K12ac has been reported <strong>in</strong> lung cancer, predom<strong>in</strong>antly <strong>in</strong> adenocarc<strong>in</strong>omapatients [148]. In addition, a correlation between H4K12 hypoacetylation andtumor grade has been reported for colorectal cancer [155]. Hypoacetylation of H4K12 is alsoobserved <strong>in</strong> aggressive breast carc<strong>in</strong>omas <strong>in</strong>clud<strong>in</strong>g basal carc<strong>in</strong>omas and HER-2-positivetumors [150]. Histone H4 and tubul<strong>in</strong> acetylation has been analyzed <strong>in</strong> breast cancer patientsat different stages with a lower level of H4K12 acetylation be<strong>in</strong>g observed <strong>in</strong> both ductalcarc<strong>in</strong>omas <strong>in</strong> situ (DCIS) and <strong>in</strong>vasive ductal carc<strong>in</strong>oma (IDC) relative to synchronousnormal breast epithelium [156]. A greater reduction <strong>in</strong> acetylation was seen <strong>in</strong> high-grade IDCversus low/<strong>in</strong>termediate-grade, ER-negative versus ER-positive, and PR-negative versusPR-positive tumors. Unexpectedly, the observed gradual loss of H4K12ac occurred <strong>in</strong> thecontext of a parallel reduction <strong>in</strong> the expression of HDAC1, 2 and HDAC6 [156]. Though thisobservation does not have an explanation so far, it does underl<strong>in</strong>e the difficulties <strong>in</strong> draw<strong>in</strong>gmechanistical conclusions at this stage (discussed below).F<strong>in</strong>ally, we note that other technical approaches have been attempted to study histonemodifications <strong>in</strong> cancer cells, and may also provide further <strong>in</strong>sights. Cuomo and colleaguescomprehensively analyzed post-translational modifications of histone H4 <strong>in</strong> a panel of breastcancer cell l<strong>in</strong>es, compared to normal epithelial mammary cells comb<strong>in</strong><strong>in</strong>g high-resolutionmass spectrometry analysis of histones with stable isotope label<strong>in</strong>g with am<strong>in</strong>o acids <strong>in</strong> cellculture (SILAC) [157]. Besides confirm<strong>in</strong>g the presence of known alterations <strong>in</strong> histone H4modifications (H4K16 hypoacetylation and loss of H4K20me3), a novel alteration wasidentified <strong>in</strong> the levels of H4K20me1 [157]. Although, at this stage, this method cannot be


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>applied to rout<strong>in</strong>e cl<strong>in</strong>ical samples, it may provide <strong>in</strong>formation that can be used for test<strong>in</strong>gwith different approaches (such as standard IHC) new candidate markers <strong>in</strong> patient samples.684.4.2 Alterations <strong>in</strong> the Pattern of Histone H3 ModificationsA pioneer study by Seligston and colleagues (discussed above for histone H4) highlighted thepossibility of us<strong>in</strong>g the study of histone modifications as prognostic predictors of cl<strong>in</strong>icaloutcome [145]. The work characterized by IHC analysis on primary prostate cancer tissue, thelevels of acetylation of H3K9, H3K18, and H4K12, and of the dimethylation of H4R3 andH3K4. This work revealed a clear difference <strong>in</strong> the pattern of modification on histone H3 <strong>in</strong>tumor versus normal prostate tissue. While no s<strong>in</strong>gle histone modification analyzed waspredictive per se, a more complex pattern obta<strong>in</strong>ed comb<strong>in</strong><strong>in</strong>g global histone modifications atmultiple sites was able to def<strong>in</strong>e the cl<strong>in</strong>ical outcome of the analyzed patients: lower levels ofmodified histones characterized patients with poorer prognosis and with <strong>in</strong>creased risk oftumor recurrence after removal of primary tumor [145]. These observations have beensubsequently confirmed and expanded by a larger study report<strong>in</strong>g low levels of H3K4monomethylation (H3K4me1), H3K9 dimethylation (H3K9me2), H3K9 trimethylation(H3K9me3), H3 and H4 acetylation <strong>in</strong> prostate cancer compared to non-malignant prostatetissue [158]. H3K4 dimethylation (H3K4me2) and H3K18 acetylation (H3K18ac), identifiedas the most predictive histone modifications <strong>in</strong> prostate cancer, have been further analyzedand their prognostic power has been confirmed <strong>in</strong> different cancer types [150,159e166]. Lowlevels of H3K4me2 and H3K18ac correlate with worse prognosis and survival <strong>in</strong> lung andkidney cancer [159]. In the same study, H3K9me2 alone predicts a poorer prognosis <strong>in</strong>prostate and kidney cancer [159]. The f<strong>in</strong>d<strong>in</strong>gs <strong>in</strong> kidney cancer have been confirmed byanother study, <strong>in</strong> which the analysis of several histone modifications (H3K9ac, H3K18ac, totalH3ac, and H4ac) led to the identification of a strong correlation between global level of H3K18and H3K9 acetylation with cancer progression and worse prognosis [160]. Low levels ofH3K4me2, H3K9me2, or H3K18ac have also been identified as significant and <strong>in</strong>dependentpredictors of poor survival <strong>in</strong> pancreatic adenocarc<strong>in</strong>oma patients [161]. In this patientpopulation, the comb<strong>in</strong>ation of low levels of H3K4me2 and H3K18ac was identified as themost significant predictor of overall survival [161]. Importantly, <strong>in</strong> this study there wasa significant correlation between low levels of H3K4me2 and/or H3K8me2 and a worse overallsurvival <strong>in</strong> the subgroup of patients treated with 5-FU, but not <strong>in</strong> the subgroup of patientstreated with Gemcitab<strong>in</strong>e: though not conclusive, this <strong>in</strong>formation poses the relevant questionabout the l<strong>in</strong>k between epigenetic pattern of the tumor and the response to treatment[161,162]. The largest correlative study of several histone modifications (H3K9ac, H3K18ac,H4K12ac, and H4K16ac, H3K4me2, and H4K20me3, and H4R3me2), cl<strong>in</strong>icopathologicfeatures and prognosis has been conducted <strong>in</strong> breast cancer [150]. From this study, beside thedata perta<strong>in</strong><strong>in</strong>g to the histone H4 modifications discussed above, an additional f<strong>in</strong>d<strong>in</strong>g was thecorrelation between low levels of H3K9 and H3K18 acetylation with high tumor grade andwith biological markers such as the absence of steroid receptor expression [150]. In the studythere was a strong correlation between low levels of H3K9 and H3K18 acetylation and breastcarc<strong>in</strong>oma with poorer prognosis <strong>in</strong>clud<strong>in</strong>g basal carc<strong>in</strong>oma and Her2-positive tumors [150].In the case of NSCLC patients of early stage (stages I to III), the most significant observationwas a correlation between low levels of acetylation of lys<strong>in</strong>e 5 of histone H2A (H2AK5ac) andworse survival for patients with tumor stage II, as well as a correlation between low levels ofH3K4me2 and poor prognosis for patients with stage I [163]. In contrast to the data availablefor prostate, breast, and pancreatic cancer, where there was a poor prognosis [145,150,151],the study showed a correlation between low levels of H3K9 acetylation and better survival[163]. Further, contrast<strong>in</strong>g data for H3K18 acetylation (H3K18ac) have been reported foresophageal and glioma cancer patients [170,171]. Nonetheless, for glioma, the analysis ofseveral histone modifications (H3K9Ac, H3K18Ac, H3K4me2, H4K12Ac, and H4R3Me)confirmed the predictive power of low levels of H3K4me2 for worse survival [165]. In relation


CHAPTER 4Alterations of Histone Modifications <strong>in</strong> Cancerto the modification of additional sites on histone H3, an <strong>in</strong>creased level of H3K56 acetylationcorrelated with higher grade <strong>in</strong> many cancer types <strong>in</strong>clud<strong>in</strong>g thyroid, laryngo-pharynx, coloncancer, and astrocytomas [166]. H3K27 methylation plays an important regulatory role <strong>in</strong>gene transcription and is found frequently altered <strong>in</strong> cancer cells compared to normal tissue,although not consistently among the different cancer types [164,167e170].Low levels of H3K27 trimethylation (H3K27me3) have been reported <strong>in</strong> breast, ovarian, andpancreatic cancers [167]. In breast cancer, low levels of H3K27me3 correlated with additionalprognostic factors such as large tumor size, estrogen receptor-negative and lymph nodepositivestatus, but not with HER-2/Neu status [167]. A similar correlation with high tumorgrade was observed <strong>in</strong> ovarian and pancreatic cancer [167]. Importantly, <strong>in</strong> all these tumortypes, low levels of H3K27me3 correlated with a significantly shorter overall survival time[167]. In contrast with these observations, low levels of H3K27me3 and H3K18ac correlatedwith an improved prognosis <strong>in</strong> patients with esophageal squamous cell carc<strong>in</strong>oma [166,168].In the case of esophageal squamous cell carc<strong>in</strong>oma patients treated with chemo/radiotherapy,a higher expression of H3K27me3 correlated positively with expression of the H3K27methylase EZH2, tumor grade, and worse survival [168]. High levels of H3K27me3 associatedwith advanced cl<strong>in</strong>ical stage and short overall survival have also been reported <strong>in</strong> nasopharyngealcarc<strong>in</strong>oma [169] and hepatocellular carc<strong>in</strong>oma [170].All of the studies reported above provide a first glimpse of the cl<strong>in</strong>ical relevance of the study ofaltered histone modification patterns <strong>in</strong> tumors: they are ma<strong>in</strong>ly if not exclusively correlative,and have been conducted us<strong>in</strong>g methodologies that do not allow a detailed mechanisticanalysis of the molecular consequences of the observed alterations. In some cases, a differenthistone pattern between normal and cancer cell has been reported look<strong>in</strong>g at the level of genepromoter, us<strong>in</strong>g chromat<strong>in</strong> immunoprecipitation-based techniques [171e173]. Interest<strong>in</strong>gly,to our knowledge, little has been done so far to cross these two types of epigenetic analyses(at a more global level and a greater molecular detail) to provide a more ref<strong>in</strong>ed epigeneticprofile of cancer samples; someth<strong>in</strong>g that is urgently needed.69By apply<strong>in</strong>g chromat<strong>in</strong> immunoprecipitation microarrays, <strong>in</strong> a comparative study betweennormal and prostate cancer cells, Kondo and colleagues found that 5% of promoters wereenriched with H3K27me3 <strong>in</strong> cancer cells and that this enrichment corresponded to genesilenc<strong>in</strong>g <strong>in</strong>dependently of DNA methylation [171]. This effect could be reverted by EZH2down-regulation [171]. A subsequent genome-wide analysis of H3K4me3 and H3K27me3 <strong>in</strong>prostate cancer cells and normal epithelial cells confirmed and expanded the observationsreported above [172].As another example of molecular studies, it is worth mention<strong>in</strong>g a genome-wide chromat<strong>in</strong>immunoprecipitation study conducted <strong>in</strong> leukemia patients [173]. The analysis of H3K9me3revealed a specific H3K9me3 alteration pattern <strong>in</strong> leukemia patients which was able todist<strong>in</strong>guish not only normal hematopoietic cells from leukemic blasts, but also acute myeloidleukemia (AML) samples from acute lymphoblastic leukemia (ALL) samples. The analysis ledto the identification of a characteristic signature of AML <strong>in</strong> which a decrease <strong>in</strong> H3K9me3levels, occurr<strong>in</strong>g ma<strong>in</strong>ly at specific promoter regions such as those conta<strong>in</strong><strong>in</strong>g cyclic adenos<strong>in</strong>emonophosphate response elements (CREs), correlated with higher transcriptional activity andmost importantly, predicted survival [173].4.5 MECHANISMS UNDERLYING HISTONE ALTERATIONS INCANCERAn obvious question stemm<strong>in</strong>g from the studies summarized above regards the molecularmechanism(s) lead<strong>in</strong>g to the global alterations <strong>in</strong> the pattern of histone modificationsobserved <strong>in</strong> cancer cells. As an <strong>in</strong>itial level of analysis, perhaps not surpris<strong>in</strong>gly, it was foundthat altered expression levels of enzymes that add, remove, or recognize specific histone


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>CategoriesGenesCategoriesGenesHATsHDACsp300CBPMYST familyHDAC1,HDAC2,HDAC3,HDAC5,HDAC6,HDAC10Sirtun 1,2,3,7HATsAcAcHDACsPPPPK<strong>in</strong>aseK<strong>in</strong>asesAURORA-BJAK2MSK1MST1RSKPI3KsCategoriesGenesCategoriesGenesHMTsKTM1A/Suv39h1KTM2A/MLL1KTM2Z/MLL3SMYD2,SMYD3,SMYD4KTM1C/G9AKTM6B/EZH2NSD1,NSD2,NSD3DOT1LHDMsMeMeHMTsUb E3DubsRNF20USP22USP770PRMTsHDMsPRMT1PRMT4/CARM1PRMT6KDM1AKDM2A,KDM2BKDM4A,B,CKDM5A,B,CKDM6A,KDM6BKDM8DubsUbUbUb E3sFIGURE 4.3Histone-modify<strong>in</strong>g enzymes and cancer. Selection of histone modify<strong>in</strong>g enzymes altered <strong>in</strong> human cancer. This figure is reproduced <strong>in</strong> the color plate section.modifications can be measured <strong>in</strong> cancer cells. At a further level of complexity, <strong>in</strong> some cases,even <strong>in</strong> the presence of normal levels of those factors, an aberrant recruitment of histonemodify<strong>in</strong>genzymes at specific chromat<strong>in</strong> regions could be considered as a determ<strong>in</strong><strong>in</strong>gmolecular event (Figure 4.3).4.5.1 Alteration of the Histone Acetylation Network (HATs, HDACsand Sirtu<strong>in</strong>s)Chromosomal translocations, mutations or simply over-expression <strong>in</strong>volv<strong>in</strong>g several HATs(such as E1A-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> p300 (EP300), cAMP response element-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> (CBP),and enzymes of the MYST family) strongly support the <strong>in</strong>volvement of these enzymes <strong>in</strong>cancer [174e178]. Several <strong>in</strong>activat<strong>in</strong>g mutations <strong>in</strong> p300, correlat<strong>in</strong>g with lack of enzymaticactivity [177], have been detected <strong>in</strong> primary solid tumors and tumor-derived cell l<strong>in</strong>es ofepithelial orig<strong>in</strong>, h<strong>in</strong>t<strong>in</strong>g at a tumor-suppressive role for the enzyme [177,178]. More<strong>in</strong>trigu<strong>in</strong>gly, <strong>in</strong> support of the view that p300 can act both as a tumor suppressor and anoncogene, down-regulation of p300 leads to growth <strong>in</strong>hibition and activation of a senescencecheckpo<strong>in</strong>t <strong>in</strong> human melanocytes [179]. For CBP, several <strong>in</strong>activat<strong>in</strong>g mutations havebeen identified <strong>in</strong> epithelial tumors [180,181]. In AMLs, this enzyme has been foundtranslocated and fused to either the HAT monocytic leukemia z<strong>in</strong>c f<strong>in</strong>ger (MOZ) gene or tothe histone methylase MLL (mixed l<strong>in</strong>eage leukemia) [182e184]. A translocation betweenCBP and a member of the MYST family (MORF), result<strong>in</strong>g <strong>in</strong> a fusion prote<strong>in</strong> with twofunctional HAT doma<strong>in</strong>s, has also been reported <strong>in</strong> some cases of AML [185]. Sequenceordeletion mutations of CBP, impair<strong>in</strong>g its acetyltransferase activity, have been reported both


CHAPTER 4Alterations of Histone Modifications <strong>in</strong> Cancer<strong>in</strong> relapsed acute lymphoblastic leukemia (ALL) and B-cell lymphoma [186,187]. Severalother HATs have also been found as altered <strong>in</strong> cancer. The MOZ gene has been identified asa common retroviral <strong>in</strong>tegration site lead<strong>in</strong>g to myeloid/lymphoid tumors [188], whilehMOF expression appears frequently down-regulated both <strong>in</strong> primary breast carc<strong>in</strong>omas and<strong>in</strong> medulloblastoma [154]. A significant down-regulation of Tip60 expression <strong>in</strong> colon andlung carc<strong>in</strong>omas has been reported [189] as well as a l<strong>in</strong>k between Tip60 down-regulationand disease progression <strong>in</strong> colorectal and gastric cancer [190,191]. Monoallelic loss of theenzyme, with concomitant reduction <strong>in</strong> mRNA levels, has been reported <strong>in</strong> humanlymphomas, head-and-neck and mammary carc<strong>in</strong>omas [192]. As for many other casesdiscussed above, up-regulation of Tip60 has been l<strong>in</strong>ked to promotion of epithelialtumorigenesis, suggest<strong>in</strong>g that the enzyme can have both oncosuppressive and oncogenicproperties [193,194]. A critical po<strong>in</strong>t (valid also for the other cases described below) is thatthere have been few, if any, attempts to correlate these observations with the altered patternsof histone modifications occurr<strong>in</strong>g <strong>in</strong> cancer cells. It is tempt<strong>in</strong>g to speculate that cross<strong>in</strong>gthese two levels of molecular analyses will lead to the def<strong>in</strong>ition of specific molecular lesions<strong>in</strong> histone-modify<strong>in</strong>g enzymes (such as HATs discussed here) that generate def<strong>in</strong>ed alterations<strong>in</strong> the pattern of histone modifications <strong>in</strong> cancer cells.In the case of HDACs, aberrant recruitment of HDAC1, 2 <strong>in</strong> acute promyelocytyc leukemia(APL) represents the first and well-characterized example of the contribution of these familiesof enzymes <strong>in</strong> cancer and particularly <strong>in</strong> hematological malignancies [195e197]. Anotherexample of HDAC-dependent aberrant transcriptional repression can be found <strong>in</strong> the AMLsubtype M2, characterized by the presence of the fusion prote<strong>in</strong> AML1-ETO, derived from the(t8;21) chromosomal translocation [198]. Similarly to the APL fusion prote<strong>in</strong>, AML1-ETOworks as a potent transcription repressor through the <strong>in</strong>teraction with N-CoR and theformation of a complex with N-Cor/S<strong>in</strong>3/HDAC1 [199,200]. HDACs are apparently <strong>in</strong>volved<strong>in</strong> transcriptional repression <strong>in</strong> the case of leukemia due to <strong>in</strong>version of chromosome 16 (8%of AML cases) [201], as well as through the <strong>in</strong>teraction with the over-expressed transcriptionfactor SCL/TAL1 <strong>in</strong> the development of T-cell acute lymphoblastic leukemia (T-ALL) [202].Anelevated expression of HDAC1, HDAC2, and HDAC6 and a possible correlation betweenexpression levels of HDAC2 and histone H4 acetylation with tumor aggressiveness have beenreported for cutaneous T-cell lymphoma patients [203]. Elevated expression of HDACs of classI, except HDAC3, was also reported <strong>in</strong> patients with primary myelofibrosis as compared toother myeloproliferative diseases or normal volunteers [204]. Moreover, over-expression ofHDAC1, HDAC2, and HDAC6 and higher acetylation levels of histone H4 compared tonormal lymphoid tissue have been reported <strong>in</strong> patients with peripheral T-cell lymphoma anddiffuse B large cell lymphoma [205].71Over-expression of s<strong>in</strong>gle HDACs, such as HDAC1, HDAC2, HDAC3, and HDAC6 amongothers, has also been reported <strong>in</strong> solid tumors [206]. Furthermore, reduced expression levels ofHDAC1 <strong>in</strong> gastric cancer, and HDAC5 and HDAC10 <strong>in</strong> lung cancers have been l<strong>in</strong>ked withpoor prognosis [207,208]. The class III HDAC, SIRT1, has been found up-regulated <strong>in</strong> mur<strong>in</strong>elung carc<strong>in</strong>omas, lymphomas, and prostate cancer and <strong>in</strong> human AMLs, glioblastoma,colorectal, prostate, and sk<strong>in</strong> cancer [208e211].4.5.2 Alteration of the Histone Methylation Network (HMTs, PRMTsand HDMs)Similarly to HATs and HDACs, altered expression and/or activity both of HMTs and HDMs,due to genetic mutations or to non-genetic events, has been reported <strong>in</strong> several cancer types[212,213].Exam<strong>in</strong><strong>in</strong>g the various HMTs and HDMs, a differential expression profile of the H3K9me3methyltransferase Suv39h1 (KMT1A) has been found <strong>in</strong> colorectal cancers versus normalcolorectal mucosa [214]. In support of this, double knock-out of Suv39h1 (KMT1A)/Suv39h2


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>(KMT1B) <strong>in</strong> mice compromised genomic stability and led to cancer predisposition andlymphoma development as a consequence of a greatly reduced level of methylated histoneH3K9 [215]. The most strik<strong>in</strong>g example of HMTs <strong>in</strong>volvement <strong>in</strong> cancer is highlighted by themixed l<strong>in</strong>eage leukemia gene MLL1 (KMT2A), encod<strong>in</strong>g an H3K4 HMT. Recurrent chromosomaltranslocations of MLL/KMT2A represent a common and frequent characteristic both <strong>in</strong>human lymphoid and myeloid leukemia [216,217]. More than 50 functional MLL fusionprote<strong>in</strong>s have been found <strong>in</strong> approximately 10% of human acute leukemia, which can bedef<strong>in</strong>ed as AMLs, acute lymphoblastic leukemias (ALLs) or mixed l<strong>in</strong>eage leukemias (MLLs)[216e219]. Furthermore, <strong>in</strong>tragenic mutations of MLL3 (KMT2Z) <strong>in</strong> colorectal cancer havebeen reported [220].72Another H3K4 HMT frequently found up-regulated <strong>in</strong> colorectal and hepatocellular carc<strong>in</strong>omais SMYD3 [221, 222]. An <strong>in</strong>creased expression of this enzyme has also been reported <strong>in</strong> breastcancer <strong>in</strong> which SMYD3 was found to promote carc<strong>in</strong>ogenesis by directly regulat<strong>in</strong>g expressionof the proto-oncogene WNT10B [223]. Similarly, SMYD2 has been found over-expressed <strong>in</strong>hepatocellular carc<strong>in</strong>oma [224], whilst down-regulation of SMYD4 expression has beenreported both <strong>in</strong> medulloblastoma and breast cancer [225,226]. Increased levels of G9A(KMT1C), a H3K9 HMT, has been described <strong>in</strong> leukemia, hepatocellular carc<strong>in</strong>oma, and <strong>in</strong>prostate and lung cancer [227e230]. Another HMT <strong>in</strong>volved <strong>in</strong> cellular transformation andoncogenesis is EZH2 (KMT6B), a H3K27 methytrasferase, that is a member of polycombrepressive complex 2 (PRC2) together with EED, SUZ12, and RbAp48 [231]. Increasedexpression of EZH2 has been found <strong>in</strong> several solid tumors such as prostate, breast, colon, sk<strong>in</strong>,bladder, and lung cancer [231]. Moreover, tissue microarray analysis demonstrated that EZH2prote<strong>in</strong> levels strongly correlated with tumor aggressiveness [232], and recently, a polycombrepression signature, consist<strong>in</strong>g of 14 direct targets of EZH2 repression, has been identified asa prognostic factor for the cl<strong>in</strong>ical outcomes <strong>in</strong> metastatic breast and prostate tumors [233].Additional evidence correlat<strong>in</strong>g high expression of EZH2 with a poor prognosis <strong>in</strong> both breastand bladder cancer have recently been published [234,235]. Interest<strong>in</strong>g data are emerg<strong>in</strong>g onthe role of EZH2, and particularly of EZH2-mediated epigenetic repression of DNA damagerepair, <strong>in</strong> a mechanism that could promote expansion of breast-tumor-<strong>in</strong>itiat<strong>in</strong>g cells [236].Incontrast to the data reported above [231e235], <strong>in</strong>activat<strong>in</strong>g mutation of EZH2, associated witha loss of function, were identified <strong>in</strong> myeloid malignancies [237,238]. Additionally, somaticand heterozygous EZH2 Y641 mutations that lead to a loss of enzymatic activity have beenfound <strong>in</strong> B-cell lymphomas [239,240]. Surpris<strong>in</strong>gly, B-cell lymphoma cell l<strong>in</strong>es and lymphomasamples harbor<strong>in</strong>g heterozygous EZH2 Y641 mutations showed <strong>in</strong>creased levels of H3K27me3[241] . Elegant <strong>in</strong> vitro enzyme assays have shed light on these observations, demonstrat<strong>in</strong>g thatthe Y641 mutation causes a concurrent decrease <strong>in</strong> monomethylation and <strong>in</strong>crease <strong>in</strong> trimethylationactivity of the mutated enzyme relative to its wild-type form [241]. Essentially, all theavailable evidence po<strong>in</strong>ts to an oncogenic role for EZH2 and is consistent with the enhancedlevels of H3K27Me3 found <strong>in</strong> tumor cells over-express<strong>in</strong>g the enzyme.Additional HMTs found altered <strong>in</strong> cancer belong to the NSD family: NSD1 (KMT3B), NSD2,and NSD3 are responsible for methylation of H3K36 and at a less extent of H4K20 [242]. TheNSD1 locus is <strong>in</strong>volved <strong>in</strong> translocations with the nucleopor<strong>in</strong> gene (NUP98) and implicated<strong>in</strong> the pathogenesis of childhood acute myeloid leukemias and myelodysplastic syndromes[243,244]. Moreover, NSD1 expression has been found abrogated by CpG island-promoterhypermethylation both <strong>in</strong> neuroblastoma and glioma cells [245]. The translocation t (4;14)(p16;q32) <strong>in</strong>volv<strong>in</strong>g the NSD2 locus has been reported <strong>in</strong> 20% of multiple myeloma cases[246,247] and a significant over-expression of NSD2 has been measured <strong>in</strong> different tumortypes, correlat<strong>in</strong>g both with tumor aggressiveness and prognosis [248]. Similarly, NSD3translocations with NUP98 <strong>in</strong> leukemia and NSD3 gene amplification <strong>in</strong> breast cancer celll<strong>in</strong>es and primary breast carc<strong>in</strong>oma have been reported [249,250]. A pivotal role of DOT-1, anH3K79 methyltransferase, <strong>in</strong> MLL fusion prote<strong>in</strong>-mediated leukemogenesis has recently beenreported [251,252].


CHAPTER 4Alterations of Histone Modifications <strong>in</strong> CancerAll of the enzymes described above belong to the category of lys<strong>in</strong>e methyltransferases. In thecase of arg<strong>in</strong><strong>in</strong>e methyltransferases, over-expression of CARM1/PRMT4 has been reported <strong>in</strong>breast, colorectal, and hormone-dependent prostate tumors [255e257]. PRMT1 is alsoapparently <strong>in</strong>volved <strong>in</strong> MLL-mediated transformation [257]. Some data regard<strong>in</strong>g the deregulationof expression of both PRMT1 and PRMT6 <strong>in</strong> different cancer types have beenpublished [258].Due to their recent discovery, compared to HMTs, the HDMs have not been extensivelycharacterized as yet; however, several recent results fully support a critical role for them <strong>in</strong>cancer [213].The correlation of KDM1A over-expression with adverse cl<strong>in</strong>ical outcome has been reported <strong>in</strong>bladder, lung, prostate, neuroblastoma, and breast cancer [259e263], whereas low levels ofboth KDM1A and H3K4me2 have been reported <strong>in</strong> hepatocellular carc<strong>in</strong>oma [264]. BothKDM2A and KDM2B have been considered as either tumor suppressors or oncogenesdepend<strong>in</strong>g on the cellular context [213]. Both KDM2A and KDM2B have been found <strong>in</strong>a retroviral-mediated <strong>in</strong>sertional mutagenesis screen<strong>in</strong>g for genes <strong>in</strong>volved <strong>in</strong> the <strong>in</strong>duction oflymphomas <strong>in</strong> rats [265,266]. In support of this, both prote<strong>in</strong>s are <strong>in</strong>volved <strong>in</strong> immortalizationof fibroblasts and oncogene-<strong>in</strong>duced senescence [266,267]. Moreover, KDM2B expressionhas been found significantly <strong>in</strong>creased both <strong>in</strong> B- and T-cell acute lymphoblastic leukemias,AMLs and <strong>in</strong> sem<strong>in</strong>omas cells [268,269]. A relevant role for KDM2B <strong>in</strong> <strong>in</strong>itiation and ma<strong>in</strong>tenanceof AML has recently been described [270]. A significantly decreased expression ofKDM2A and KDM2B has been reported <strong>in</strong> prostate cancer and glioblastoma, respectively[271,272]. Amplification of KDM4C has been reported for esophageal squamous cellcarc<strong>in</strong>omas, breast cancers, medulloblastomas, and metastatic lung sarcomatoid carc<strong>in</strong>omas[273e277]. Over-expression of KDM4A, KDM4B, and KDM4C has been reported <strong>in</strong> prostatecancer [278] and <strong>in</strong> medulloblastoma [279], whilst high expression of KDM4B has also beenreported <strong>in</strong> ER-positive breast cancer [280]. KDM5A has been found over-expressed <strong>in</strong> gastriccancer [281] and has been <strong>in</strong>volved <strong>in</strong> the determ<strong>in</strong>ation of a drug tolerance phenotype <strong>in</strong>a non-small-cell lung carc<strong>in</strong>oma cell system [282]. Increased expression of KDM5B has beenreported for several cancer types [283e286].73A systematic sequenc<strong>in</strong>g of renal carc<strong>in</strong>oma samples revealed the presence of <strong>in</strong>activat<strong>in</strong>gmutations of KDM5C [287]. Functionally, a genome-wide siRNA screen led to identify KDM5Cas one of the E2-dependent regulators of human papilloma virus oncogene expression [288].Inactivat<strong>in</strong>g mutations <strong>in</strong> KDM6A have been reported <strong>in</strong> multiple myeloma, esophagealsquamous cell carc<strong>in</strong>oma and clear cell renal cell carc<strong>in</strong>oma [287,289]. A reduced expression ofKDM6B has been reported <strong>in</strong> lung and liver carc<strong>in</strong>oma, and <strong>in</strong> a subset of lymphomas andleukemias [290]. In contrast, but potentially consistent with a role as tumor suppressor, highexpression of KDM6B has been reported <strong>in</strong> melanocytic nevi [291]. HPV-mediated <strong>in</strong>duction ofthe expression of both KDM6A and KDM6B <strong>in</strong> cervical cancer cells has been reportedsupport<strong>in</strong>g an <strong>in</strong>volvement of both enzymes <strong>in</strong> HPV-mediated oncogenesis [292]. Interest<strong>in</strong>gly,KDM6B expression could also be <strong>in</strong>duced by Epste<strong>in</strong>eBarr virus and appears to be higher <strong>in</strong>Hodgk<strong>in</strong>’s lymphoma [293]. F<strong>in</strong>ally, multiple types of tumors <strong>in</strong>clud<strong>in</strong>g thyroid, adrenal,bladder, uter<strong>in</strong>e, and liver exhibited a higher expression of the H3K36 me2 demethylase KDM8prote<strong>in</strong> <strong>in</strong> comparison to their respective normal tissue controls [294].4.5.3 Alterations of Histone K<strong>in</strong>ases, Histone Ubiquit<strong>in</strong>at<strong>in</strong>g andDeubiquit<strong>in</strong>at<strong>in</strong>g EnzymesA significant amount of data has been generated show<strong>in</strong>g the deregulation of the enzymesresponsible for histone phosphorylation <strong>in</strong> cancer.Aurora B (the primary k<strong>in</strong>ase responsible for H3S10 phosphorylation) has been found overexpressed<strong>in</strong> breast, colorectal, kidney, lung, and prostate cancers [295e298]. JAK2, the k<strong>in</strong>ase


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>that phosphorylates H3Y41 <strong>in</strong> hematopoietic cells, has also been found to be constitutivelyactivated, due to gene mutation and/or rearrangement, <strong>in</strong> several hematological malignancies[299]. Furthermore, gene amplification of JAK2 and KDM4C has been recently found both <strong>in</strong>primary mediast<strong>in</strong>al B-cell lymphoma and Hodgk<strong>in</strong>’s lymphoma [300]. An important role ofMSK1 and RSK2, and consequently of histone H3 phosphorylation, <strong>in</strong> cellular transformationhas been documented [301e303], together with a role of MST1 k<strong>in</strong>ase as a tumor-suppressorgene [304]. Moreover, data <strong>in</strong> support of a possible <strong>in</strong>terdependence between RSK2-H2AX andRSK2-H3 phosphorylation <strong>in</strong> the control of cell transformation have been published [305].The histone ubiquit<strong>in</strong>ation network has not been fully characterized as yet, but a picture isemerg<strong>in</strong>g of its role <strong>in</strong> cancer [306]. The E3 ligase RNF20, responsible for H2B ubiquit<strong>in</strong>ation,has been found commonly silenced via DNA hypermethylation <strong>in</strong> breast cancers [307]. Insupport of this, RNF20-depleted cells showed an <strong>in</strong>creased oncogenic potential aftermeasur<strong>in</strong>g the ability of cells to form tumors once transplanted <strong>in</strong> mice [307]. The deubiquit<strong>in</strong>at<strong>in</strong>genzyme USP22 has been def<strong>in</strong>ed as a putative marker of cancer stem cells [308],and it has also been identified among 11 genes form<strong>in</strong>g a gene signature associated with poorprognosis <strong>in</strong> multiple tumor types [309]. An elevated expression of USP22, correlat<strong>in</strong>g witha poor prognosis, has been reported both for colorectal and breast cancer [310,311], and morerecently <strong>in</strong> melanoma [312]. Similarly, USP7 over-expression has been reported <strong>in</strong> prostate,bladder, colon, liver, and lung cancers [313], although <strong>in</strong> a more recent analysis its expressionwas not found significantly deregulated [312].74Taken together, these data (i.e perta<strong>in</strong><strong>in</strong>g to histone-modify<strong>in</strong>g enzymes and secondaryprote<strong>in</strong>s <strong>in</strong>volved <strong>in</strong> the network) conclusively demonstrate that alterations <strong>in</strong> the histonemodification network occur <strong>in</strong>variably <strong>in</strong> cancer cells and have a complex modality of action.Although an exhaustive molecular description of the events/l<strong>in</strong>ks underly<strong>in</strong>g these alterationsdoes not exist, the data generated to date can expla<strong>in</strong> a large part of the epigenetic alterationsfound <strong>in</strong> cancer cells.4.5.4 Other MechanismsThe abundance and diversity of histone-modify<strong>in</strong>g enzymes, as well as their overlapp<strong>in</strong>g andredundant substrate specificities, clearly exclude a simplistic explanation of the causalcontribution of each enzyme to the altered pattern of histone modifications found <strong>in</strong> cancer.Kurdistani proposed that at the basis of the altered pattern of histone modifications <strong>in</strong> cancerand <strong>in</strong> view of the low levels of histone modifications found <strong>in</strong> several cancer types, there is ananomalous allocation of co-factors as acetyl coenzymeA and S-adenosylmethion<strong>in</strong>e requiredby HATs and HMTs to modify histones, respectively [314]. Thus, metabolic pathways <strong>in</strong>volved<strong>in</strong> cell growth and division are also centrally <strong>in</strong>volved <strong>in</strong> the mechanisms of histone modification.One speculation deriv<strong>in</strong>g from this proposal is that the altered metabolism of cancercells, where there is a high and cont<strong>in</strong>uous need for macromolecular biosynthesis, could leadthe cancer cells to prevalently divert the use of these co-factors from histone modificationpathways to more immediately vital pathways. This diversion of essential cofactors away fromhistone modification enzymes would have a major impact on the global levels of histoneacetylation and methylation. In fact, recent data l<strong>in</strong>k<strong>in</strong>g the alteration of metabolic enzymes(IDH1-2, for example) and deregulated expression of histone methylation tend to support thel<strong>in</strong>k between altered metabolism, histone modification, and cancer [315].4.6 CONCLUSIONSOver the last few years a large number of histone modifications have been identified, <strong>in</strong>formationregard<strong>in</strong>g the role of each type of modification has been generated, the enzymescatalyz<strong>in</strong>g these modifications have been characterized and the elucidation of the molecularmechanisms l<strong>in</strong>k<strong>in</strong>g these modifications to cellular processes has begun.


CHAPTER 4Alterations of Histone Modifications <strong>in</strong> CancerConcurrently, it has become evident that the histone post-translational modifications act <strong>in</strong> anordered and coord<strong>in</strong>ated manner and should be considered <strong>in</strong> the context of an “epigeneticcode”, or better, <strong>in</strong> the context of a “global cross-talk” network between the different histonemodifications. Moreover, <strong>in</strong> agreement with the relevant role of histone modifications <strong>in</strong>several key cellular processes, significant data have been published suggest<strong>in</strong>g that a deregulationof the histone modification pattern is l<strong>in</strong>ked to different human malignancies andparticularly to cancer. In this regard, the most strik<strong>in</strong>g example is the loss of H3K16ac andH4K20me3 which represents a well-recognized cancer histone mark. In addition, several datahave been generated from different cancer types highlight<strong>in</strong>g the correlation between alteredglobal histone modification patterns and cancer aggressiveness and there is now the possibilityto use them as <strong>in</strong>dependent prognostic factors.The reversible nature of epigenetic modifications, <strong>in</strong>clud<strong>in</strong>g all the histone modifications, hasprovided the basis for development of “epigenetic therapies”. So far this has led to the approvalof two DNA methyltransferase <strong>in</strong>hibitors (Vidaza and Decitab<strong>in</strong>e) for myelodysplasticsyndrome and two HDAC <strong>in</strong>hibitors (Vor<strong>in</strong>ostat and Romideps<strong>in</strong>) for cutaneous T-celllymphoma. Large <strong>in</strong>vestments <strong>in</strong> both time and money for identify<strong>in</strong>g <strong>in</strong>hibitors of otherprote<strong>in</strong>s <strong>in</strong>volved <strong>in</strong> regulation of epigenetic modifications, <strong>in</strong>clud<strong>in</strong>g HAT, Sirtu<strong>in</strong>, HMTs, andHDMs, is ongo<strong>in</strong>g.It is now important for both basic and applied science to acquire additional knowledgeregard<strong>in</strong>g the functional relevance of each histone modification and, more importantly,knowledge perta<strong>in</strong><strong>in</strong>g to the <strong>in</strong>terplay between these modifications and the mach<strong>in</strong>ery<strong>in</strong>volved <strong>in</strong> their addition and removal <strong>in</strong> the context of cancer biology. The different patternof histone modifications reported between normal and cancer cells, together with the accumulat<strong>in</strong>gevidence that these differences can be prognostic factors and potential predictors oftherapeutic response, suggest that further research <strong>in</strong> these fields could open the way for betterpersonalized medic<strong>in</strong>e <strong>in</strong> both epigenetic and non-epigenetic therapies. Future studies<strong>in</strong>tended to <strong>in</strong>crease our knowledge of epigenetic modifications, histone modifications <strong>in</strong>particular, could have a strik<strong>in</strong>g impact on the relevance of epigenetic events <strong>in</strong> cancer biologyand on the design of more efficient strategies for epigenetic therapies <strong>in</strong> the treatment ofcancer. In this regard, of significant relevance is the advent and use of new technologies <strong>in</strong> theepigenomic field. The application of techniques such as chromat<strong>in</strong> immunoprecipitationfollowed by modern high-density microarrays, next-generation sequenc<strong>in</strong>g that permitsprofil<strong>in</strong>g of large sample series and the accurate determ<strong>in</strong>ation of the location of differenthistone modifications at global level are expected to have a major impact <strong>in</strong> the field.75Moreover, the identification of new methodologies that would permit the technologiesdescribed above to be performed on paraff<strong>in</strong>-embedded tissue samples (PAT-ChIP-seq) [316]would have an enormous impact. Analysis of historical cancer tissue samples would permit thedeterm<strong>in</strong>ation of the location and type of different histone modifications on a global scale.Detailed analysis on samples where cl<strong>in</strong>ical outcome can be associated with an “epigeneticsignature” would represent a real breakthrough <strong>in</strong> the identification of epigenetic biomarkersfor different pathological conditions.From the cl<strong>in</strong>ical perspective, one can envisage a future where a histone modification signaturewill exist for each type of cancer and that this signature will be correlated with prognosis andmore importantly, with the choice of best possible treatment. Last, but by no means least, onecould expect to use these histone maps to monitor the efficacy of epigenetic drug treatment atthe molecular level.References[1] Alberts B, Johnson A, Lewis J, Raff M, Roberts K, Walter P. Chromosomal DNA and Its Packag<strong>in</strong>g <strong>in</strong> theChromat<strong>in</strong> Fiber. Mol Biol cell 4 th edition 2002.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>76[2] Kouzarides T. Chromat<strong>in</strong> modifications and their function. Cell 2007;128:693e705.[3] Murr R. Interplay between different epigenetic modifications and mechanisms. Adv Genet 2010;70:101e41.[4] Kornerber RD. Chromat<strong>in</strong> structure: A repeat<strong>in</strong>g unit of histones and DNA. <strong>Science</strong> 1974;184:868e71.[5] Oudet P, Gross-Bellard M, Chambon P. Electron microscopic and biochemical evidence that chromat<strong>in</strong>structure is a repeat<strong>in</strong>g unit. Cell 1975;4:281e300.[6] Luger K, Mader AW, Richmond RK, Sargent DF, Richmond TJ. Crystal structure of the nucleosome core particleat 2.8 A resolution. Nature 1997;389:251e60.[7] Kornberg RD, Lorch Y. Twenty-five years of the nucleosome, fundamental particle of the eukaryote chromosome.Cell 1999;98:285e94.[8] Rob<strong>in</strong>son PJ, Fairall L, Huynh VA, Rhodes D. EM measurements def<strong>in</strong>e the dimensions of the ‘‘30-nm’’chromat<strong>in</strong> fiber: evidence for a compact, <strong>in</strong>terdigitated structure. Proc Natl Acad Sci USA 2006;103:6506e11.[9] Van Holde KE. Histone modifications. In: Rich A, editor. Chromat<strong>in</strong>, Spr<strong>in</strong>ger Series <strong>in</strong> Molecular Biology. NewYork: Spr<strong>in</strong>ger; 1988. p. 111e48.[10] Arents G, Moudrianakis EN. The histone fold: a ubiquitous architectural motif utilized <strong>in</strong> DNA compactionand prote<strong>in</strong> dimerization. Proc Natl Acad Sci USA 1995;92:11170e4.[11] Peterson CL, Laniel MA. Histones and histone modifications. Curr Biol 2004;14:R546e51.[12] Paul B, Henikoff T, Henikoff S. Histone variants d ancient wrap artists of the epigenomeS. Henikoff, K.Ahmad, Assembly of variant histones <strong>in</strong>to chromat<strong>in</strong>. Annu Rev Cell Dev Biol 2005;21:133e53.[13] Cosgrove MS, Boeke JD, Wolberger C. Regulated nucleosome mobility and the histone code. Nat Struct MolBiol 2004;11:1037e104.[14] Goll MG, Bestor TH. Histone modification and replacement <strong>in</strong> chromat<strong>in</strong> activation. Genes Dev2002;16:1739e42.[15] Grant PA. A tale of histone modifications. Genome Biol 2001;2. REVIEWS0003.[16] Shahbazian MD, Grunste<strong>in</strong> M. Functions of site-specific histone acetylation and deacetylation. Annu RevBiochem 2007;76:75e100.[17] Struhl K. Histone acetylation and transcriptional regulatory mechanisms. Genes Dev 1998;12:599e606.[18] Yang XJ, Seto E. HATs and HDACs: from structure, function and regulation to novel strategies for therapy andprevention. Oncogene 2007;26:5310e8.[19] Lee KK, Workman JL. Histone acetyltransferase complexes: One size doesn’t fit all. Nat Rev Mol Cell Biol2007;8:284e95.[20] Parthun MR. Hat1: the emerg<strong>in</strong>g cellular roles of a type B histone acetyltransferase. Oncogene2007;26:5319e28.[21] Yang XJ, Seto E. The Rpd3/Hda1 family of lys<strong>in</strong>e deacetylases: from bacteria and yeast to mice and men. NatRev Mol Cell Biol 2008;9:206e18.[22] Thiagal<strong>in</strong>gam S, Cheng KH, Lee HJ, M<strong>in</strong>eva N, Thiagal<strong>in</strong>gam A, Ponte JF. Histone deacetylases: unique players<strong>in</strong> shap<strong>in</strong>g the epigenetic histone code. Ann NY Acad Sci 2003;983:84e100.[23] Mai A. The therapeutic uses of chromat<strong>in</strong>-modify<strong>in</strong>g agents. Expert Op<strong>in</strong> Ther Targets 2007;11:835e51.[24] F<strong>in</strong>kel T, Deng CX, Mostoslavsky R. Recent progress <strong>in</strong> the biology and physiology of sirtu<strong>in</strong>s. Nature2009;460:587e91.[25] Michan S, S<strong>in</strong>clair D. Sirtu<strong>in</strong>s <strong>in</strong> mammals: <strong>in</strong>sights <strong>in</strong>to their biological function. Biochem J2007;404:1e13.[26] Verd<strong>in</strong> E, Dequiedt F, Kasler HG. Class II histone deacetylases: versatile regulators. Trends Genet2003;5:286e93.[27] Gao L, Cueto MA, Asselbergs F, Atadja P. Clon<strong>in</strong>g and functional characterization of HDAC11, a novel memberof the human histone deacetylase family. J Biol Chem 2002;277:25748e55.[28] Tjeertes JV, Miller KM, Jackson SP. Screen for DNA-damage-responsive histone modifications identifiesH3K9Ac and H3K56Ac <strong>in</strong> human cells. EMBO J 2009;28:1878e89.[29] Wang Z, Zang C, Rosenfeld JA, Schones DE, Barski A, Cuddapah S, et al. Comb<strong>in</strong>atorial patterns of histoneacetylations and methylations <strong>in</strong> the human genome. Nat Genet 2008;40:897e903.[30] Wang Z, Zang C, Cui K, Schones DE, Barski A, Peng W, et al. Genome-wide mapp<strong>in</strong>g of HATs and HDACsreveals dist<strong>in</strong>ct functions <strong>in</strong> active and <strong>in</strong>active genes. Cell 2009;138:1019e31.[31] Keogh MC, Kurdistani SK, Morris SA, Ahn SH, Podolny V, Coll<strong>in</strong>s RS, et al. Cotranscriptional Set2 Methylationof Histone H3 Lys<strong>in</strong>e 36 Recruits a Repressive Rpd3 Complex. Cell 2005;123:593e605.[32] Carrozza MJ, Li B, Florens L, Suganuma T, Swanson SK, Lee KK, et al. Histone H3 Methylation by Set2 DirectsDeacetylation of Cod<strong>in</strong>g Regions by Rpd3S to Suppress Spurious Intragenic Transcription. Cell2005;123:581e92.


CHAPTER 4Alterations of Histone Modifications <strong>in</strong> Cancer[33] Li B, Gogol M, Carey M, Pattenden SG, Seidel C, Workman JL. Infrequently transcribed long genes depend onthe Set2/Rpd3S pathway for accurate transcription. Genes Dev 2007;21:1422e30.[34] Shogren-Knaak M, Ishii H, Sun JM, Paz<strong>in</strong> MJ, Davie JR, Peterson CL. Histone H4-K16 acetylation controlschromat<strong>in</strong> structure and prote<strong>in</strong> <strong>in</strong>teractions. <strong>Science</strong> 2006;311:844e7.[35] Cimbora DM, Schubeler D, Reik A, Hamilton J, Francastel C, Epner EM, et al. Long-distance control of orig<strong>in</strong>choice and replication tim<strong>in</strong>g <strong>in</strong> the human beta-glob<strong>in</strong> locus are <strong>in</strong>dependent of the locus control region.Mol Cell Biol 2000;20:5581e91.[36] Doyon Y, Cayrou C, Ullah M, Landry AJ, Cote V, Selleck W, et al. ING tumor suppressor prote<strong>in</strong>s are criticalregulators of chromat<strong>in</strong> acetylation required for genome expression and perpetuation. Mol Cell2006;21:51e64.[37] Bedford MT, Clarke SG. Prote<strong>in</strong> arg<strong>in</strong><strong>in</strong>e methylation <strong>in</strong> mammals: who, what, and why. Mol Cell 2009;33:1e13.[38] Mart<strong>in</strong> C, Zhang Y. The diverse functions of histone lys<strong>in</strong>e methylation. Nat Rev Mol Cell Biol2005;6:838e49.[39] Shilatifard A. Chromat<strong>in</strong> modifications by methylation and ubiquit<strong>in</strong>ation: implications <strong>in</strong> the regulation ofgene expression. Annu Rev Biochem 2006;75:243e69.[40] Ng SS, Yue WW, Oppermann U, Klose RJ. Dynamic prote<strong>in</strong> methylation <strong>in</strong> chromat<strong>in</strong> biology. Cell Mol LifeSci 2009;66:407e22.[41] Zhang Y, Re<strong>in</strong>berg D. Transcription regulation by histone methylation: <strong>in</strong>terplay between different covalentmodifications of the core histone tails. Genes Dev 2001;15:2343e60.[42] Rice JC, Briggs SD, Ueberheide B, Barber CM, Shabanowitz J, Hunt DF, et al. Histone methyltransferases directdifferent degrees of methylation to def<strong>in</strong>e dist<strong>in</strong>ct chromat<strong>in</strong> doma<strong>in</strong>s. Mol Cell 2003;12:1591e8.[43] Keppler BR, Archer TK. Chromat<strong>in</strong>-modify<strong>in</strong>g enzymes as therapeutic targets-Part 1. Expert Op<strong>in</strong> Ther Targets2008;12:1301e12.[44] Wolf SS. The prote<strong>in</strong> arg<strong>in</strong><strong>in</strong>e methyltransferase family: an update about function, new perspectives and thephysiological role <strong>in</strong> humans. Cell Mol Life Sci 2009;66:2109e21.[45] Allis CD, Berger SL, Cote J, Dent S, Jenuwien T, Kouzarides T, et al. New nomenclature for chromat<strong>in</strong>modify<strong>in</strong>genzymes. Cell 2007;131:633e6.[46] Feng Q, Wang H, Ng HH, Erdjument-Bromage H, Tempst P, Struhl K, et al. Methylation of H3-lys<strong>in</strong>e 79 ismediated by a new family of HMTases without a SET doma<strong>in</strong>. Curr Biol 2002;12:1052e8.[47] Fumasoni I, Meani N, Rambaldi D, Scafetta G, Alcalay M, Ciccarelli FD. Family expansion and gene rearrangementscontributed to the functional specialization of PRDM genes <strong>in</strong> vertebrates. BMC Evol Biol2007;4(7):187.[48] Kim KC, Geng L, Huang S. Inactivation of a histone methyltransferase by mutations <strong>in</strong> human cancers. CancerRes 2003;63:7619e23.[49] Funabiki T, Kreider BL, Ihle JN. The carboxyl doma<strong>in</strong> of z<strong>in</strong>c f<strong>in</strong>gers of the Evi-1 myeloid transform<strong>in</strong>g geneb<strong>in</strong>ds a consensus sequence of GAAGATGAG. Oncogene 1994;9:1575e158.[50] Cuthbert GL, Daujat S, Snowden AW, Erdjument-Bromage H, Hagiwara T, Yamada M, et al. Histone deim<strong>in</strong>ationantagonizes arg<strong>in</strong><strong>in</strong>e methylation. Cell 2004;118:545e53.[51] Wang Y, Wysocka J, Sayegh J, Lee YH, Perl<strong>in</strong> JR, Leonelli L, et al. <strong>Human</strong> PAD4 regulates histone arg<strong>in</strong><strong>in</strong>emethylation levels via demethylim<strong>in</strong>ation. <strong>Science</strong> 2004;306:279e83.[52] Trojer P, Re<strong>in</strong>berg D. Histone lys<strong>in</strong>e demethylases and their impact on epigenetics. Cell 2006;125:213e7.[53] Klose RJ, Zhang Y. Regulation of histone methylation by demethylim<strong>in</strong>ation and demethylation. Nat Rev MolCell Biol 2007;8:307e18.[54] Mosammaparast N, Shi Y. Reversal of histone methylation: biochemical and molecular mechanisms of histonedemethylases. Annu Rev Biochem 2010;79:155e79.[55] Shi Y, Lan F, Matson C, Mulligan P, Whetst<strong>in</strong>e JR, Cole PA, et al. Histone demethylation mediated by thenuclear am<strong>in</strong>e oxidase homolog LSD1. Cell 2004;119:941e53.[56] Chang B, Chen Y, Zhao Y, Bruick RK. JMJD6 is a histone arg<strong>in</strong><strong>in</strong>e demethylase. <strong>Science</strong> 2007;318:444e7.[57] Metzger E, Wissmann M, Y<strong>in</strong> N, Müller JM, Schneider R, Peters AH, et al. LSD1 demethylates repressive histonemarks to promote androgen-receptor-dependent transcription. Nature 2005;437:436e9.[58] Yun M, Wu J, Workman JL, Li B. Readers of histone modifications. Cell Res 2011;21:564e78.[59] Wysocka J, Swigut T, Xiao H, Milne TA, Kwon SY, Landry J, et al. A PHD f<strong>in</strong>ger of NURF couples histone H3lys<strong>in</strong>e 4 trimethylation with chromat<strong>in</strong> remodell<strong>in</strong>g. Nature 2006;442:86e90.[60] Bannister AJ, Zegerman P, Partridge JF, Miska EA, Thomas JO, Allshire RC, et al. Selective recognition ofmethylated lys<strong>in</strong>e 9 on histone H3 by the HP1 chromo doma<strong>in</strong>. Nature 2001;410:120e4.[61] Barski A, Cuddapah S, Cui K, Roh TY, Schones DE, Wang Z, et al. High-resolution profil<strong>in</strong>g of histonemethylations <strong>in</strong> the human genome. Cell 2007;129:823e37.77


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>78[62] Bernste<strong>in</strong> BE, Mikkelsen TS, Xie X, Kamal M, Huebert DJ, Cuff J, et al. A bivalent chromat<strong>in</strong> structure marks keydevelopmental genes <strong>in</strong> embryonic stem cells. Cell 2006;125:315e26.[63] Oki M, Aihara H, Ito T. Role of histone phosphorylation <strong>in</strong> chromat<strong>in</strong> dynamics and its implications <strong>in</strong>diseases. Subcell Biochem 2007;41:319e36.[64] Cheung P, Allis CD, Sassone-Corsi P. Signal<strong>in</strong>g to chromat<strong>in</strong> through histone modifications. Cell2000;103:263e71.[65] Dawson MA, Bannister AJ, Göttgens B, Foster SD, Bartke T, Green AR, et al. JAK2 phosphorylates histoneH3Y41 and excludes HP1alpha from chromat<strong>in</strong>. Nature 2009;461:819e22.[66] Ito T. Role of histone modification <strong>in</strong> chromat<strong>in</strong> dynamics. J Biochem 2007;141:609e14.[67] Macdonald N, Welburn JP, Noble ME, Nguyen A, Yaffe MB, Clynes D, et al. Molecular basis for the recognitionof phosphorylated and phosphoacetylated histone h3 by 14-3-3. Mol Cell 2005;20:199e211.[68] W<strong>in</strong>ter S, Simboeck E, Fischle W, Zupkovitz G, Dohnal I, Mechtler K, et al. 14-3-3 prote<strong>in</strong>s recognize a histonecode at histone H3 and are required for transcriptional activation. EMBO Journal 2008;27:88e99.[69] Foster ER, Downs JA. Histone H2A phosphorylation <strong>in</strong> DNA double-strand break repair. FEBS J2005;272:3231e40.[70] Cheung WL, Ajiro K, Samejima K, Kloc M, Cheung P, Mizzen CA, et al. Apoptotic phosphorylation of histoneH2B is mediated by mammalian sterile twenty k<strong>in</strong>ase. Cell 2003;113:507e17.[71] Nowak SJ, Corces VG. Phosphorylation of histone H3: A balanc<strong>in</strong>g act between chromosome condensationand transcriptional activation. Trends Genet 2004;20:214e20.[72] Sugiyama K, Sugiura K, Hara T, Sugimoto K, Shima H, Honda K, et al. Aurora-B associated prote<strong>in</strong> phosphatasesas negative regulators of k<strong>in</strong>ase activation. Oncogene 2002;21:3103e11.[73] Goto H, Yasui Y, Nigg EA, Inagaki M. Aurora-B phosphorylates histone H3 at ser<strong>in</strong>e28 with regard to themitotic chromosome condensation. Genes Cells 2002;7:11e7.[74] Dai J, Sultan S, Taylor SS, Higg<strong>in</strong>s JM. The k<strong>in</strong>ase hasp<strong>in</strong> is required for mitotic histone H3 Thr 3 phosphorylationand normal metaphase chromosome alignment. Genes Dev 2005;19:472e88.[75] Preuss U, Landsberg G, Scheidtmann KH. Novel mitosis-specific phosphorylation of histone H3 at Thr11mediated by Dlk/ZIP k<strong>in</strong>ase. Nucleic Acids Res 2003;31:878e85.[76] Shimada M, Niida H, Z<strong>in</strong>eldeen DH, Tagami H, Tanaka M, Saito H, et al. Chk1 is a histone H3 threon<strong>in</strong>e 11k<strong>in</strong>ase that regulates DNA damage-<strong>in</strong>duced transcriptional repression. Cell 2008;132:221e32.[77] Lau AT, Lee SY, Xu YM, Zheng D, Cho YY, Zhu F, et al. Phosphorylation of Histone H2B Ser<strong>in</strong>e 32 Is L<strong>in</strong>ked toCell Transformation. J Biol Chem 2011;286:26628e37.[78] Zhu F, Zykova TA, Peng C, Zhang J, Cho YY, Zheng D, et al. Phosphorylation of H2AX at Ser139 and a newphosphorylation site Ser16 by RSK2 decreases H2AX ubiquit<strong>in</strong>ation and <strong>in</strong>hibits cell transformation. CancerRes 2011;71:393e403.[79] Chowdhury D, Keogh MC, Ishii H, Peterson CL, Buratowski S, Lieberman J. g-H2AX dephosphorylation byprote<strong>in</strong> phosphatase 2A facilitates DNA double-strand break repair. Mol Cell 2005;20:801e9.[80] Chowdhury D, Xu X, Zhong X, Ahmed F, Zhong J, Liao J, et al. A PP4-phosphatase complex dephosphorylatesg-H2AX generated dur<strong>in</strong>g DNA replication. Mol Cell 2008;31:33e46.[81] Shimada M, Haruta M, Niida H, Sawamoto K, Nakanishi M. Prote<strong>in</strong> phosphatase 1g is responsible fordephosphorylation of histone H3 at Thr 11 after DNA damage. EMBO Rep 2010;11:883e9.[82] Pickart CM, Edd<strong>in</strong>s MJ. Ubiquit<strong>in</strong>: Structures, functions, mechanisms. Biochim Biophy Acta2004;1695:55e72.[83] Hershko A, Ciechanover A. The ubiquit<strong>in</strong> system. Annu Rev Biochem 1998;67:425e79.[84] Zhang Y. Transcriptional regulation by histone ubiquit<strong>in</strong>ation and deubiquit<strong>in</strong>ation. Genes Dev2003;17:2733e40.[85] Tanny JC, Erdjument-Bromage H, Tempst P, Allis CD. Ubiquitylation of histone H2B controls RNApolymerase II transcription elongation <strong>in</strong>dependently of histone H3 methylation. Genes Dev2007;21:835e47.[86] Flem<strong>in</strong>g AB, Kao CF, Hillyer C, Pikaart M, Osley MA. H2B Ubiquitylation Plays a Role <strong>in</strong> NucleosomeDynamics dur<strong>in</strong>g Transcription Elongation. Mol Cell 2008;31:57e66.[87] Shukla A, Bhaumik SR. H2B-K123 ubiquit<strong>in</strong>ation stimulates RNAPII elongation <strong>in</strong>dependent of H3-K4methylation. B.B.R.C 2007;59:214e20.[88] Weake VM, Workman JL. Histone Ubiquit<strong>in</strong>ation: Trigger<strong>in</strong>g Gene Activity. Mol Cell 2008;29:653e63.[89] Osley MA. Regulation of histone H2A and H2B ubiquitylation. Brief Funct Genomic Proteomic 2006;5:179e18.[90] Fierz B, Chatterjee C, McG<strong>in</strong>ty RK, Bar-Dagan M, Raleigh DP, Muir TW. Histone H2B ubiquitylation disruptslocal and higher-order chromat<strong>in</strong> compaction. Nat Chem Biol 2011;7:113e9.


CHAPTER 4Alterations of Histone Modifications <strong>in</strong> Cancer[91] Koken MH, Reynolds P, Jaspers-Dekker I, Prakash L, Prakash S, Bootsma D, et al. Structural and functionalconservation of two human homologs of the yeast DNA repair gene RAD6. Proc Natl Acad Sci USA1991;88:8865e9.[92] Roest HP, van Klaveren J, de Wit J, van Gurp CG, Koken MH, Vermey M, et al. Inactivation of the HR6Bubiquit<strong>in</strong>-conjugat<strong>in</strong>g DNA repair enzyme n mice causes male sterility associated with chromat<strong>in</strong> modification.Cell 1996;86:799e810.[93] Kim J, Hake SB, Roeder RG. The human homolog of yeast BRE1 functions as a transcriptional coactivatorthrough direct activator <strong>in</strong>teractions. Mol Cell 2005;20:759e70.[94] Zhu B, Zheng Y, Pham AD, Mandal SS, Erdjument-Bromage H, Tempst P, et al. Monoubiquit<strong>in</strong>ation ofhuman histone H2B: the factors <strong>in</strong>volved and their roles <strong>in</strong> HOX gene regulation. Mol Cell 2005;20:601e11.[95] Cao R, Tsukada Y, Zhang Y. Role of Bmi-1 and R<strong>in</strong>g1A <strong>in</strong> H2A ubiquitylation and Hox gene silenc<strong>in</strong>g. MolCell 2005;20:845e54.[96] Zhou W, Zhu P, Wang J, Pascual G, Ohgi KA, Lozach J, et al. Histone H2A monoubiquit<strong>in</strong>ation repressestranscription by <strong>in</strong>hibit<strong>in</strong>g RNA polymerase II transcriptional elongation. Mol Cell 2008;29:69e80.[97] Berg<strong>in</strong>k S, Salomons FA, Hoogstraten D, Groothuis TA, de Waard H, Wu J, et al. DNA damage triggersnucleotide excision repair-dependent monoubiquitylation of histone H2A. Genes Dev 2006;20:1343e52.[98] Mailand N, Bekker-Jensen S, Faustrup H, Melander F, Bartek J, Lukas C, et al. RNF8 ubiquitylates histones atDNA double-strand breaks and promotes assembly of repair prote<strong>in</strong>s. Cell 2007;131:887e900.[99] Zhang XY, Varthi M, Sykes SM, Phillips C, Warzecha C, Zhu W, et al. The putative cancer stem cell markerUSP22 Is a subunit of the human SAGA complex required for activated transcription and cell-cycleprogression. Mol Cell 2008;29:102e11.[100] Zhao Y, Lang G, Ito S, Bonnet J, Metzger E, Sawatsubashi S, et al. A TFTC/ STAGA module mediates histoneH2A and H2B deubiquit<strong>in</strong>ation, coactivates nuclear receptors, and counteracts heterochromat<strong>in</strong> silenc<strong>in</strong>g.Mol Cell 2008;29:92e101.[101] Zhang XY, Pfeiffer HK, Thorne AW, McMahon SB. USP22, an hSAGA subunit and potential cancer stemcell marker, reverses the polycomb-catalyzed ubiquitylation of histone H2A. Cell Cycle 2008;7(1):522e4.[102] Zhu P, Zhou W, Wang J, Puc J, Ohgi KA, Erdjument-Bromage H, et al. A histone H2A deubiquit<strong>in</strong>ase complexcoord<strong>in</strong>at<strong>in</strong>g histone acetylation and H1 dissociation <strong>in</strong> transcriptional regulation. Mol Cell2007;27:609e21.[103] Nicassio F, Corrado N, Vissers JH, Areces LB, Berg<strong>in</strong>k S, Marteijn JA, et al. <strong>Human</strong> USP3 is a chromat<strong>in</strong>modifier required for S phase progression and genome stability. Curr Biol 2007;17:1972e7.[104] Van der Knaap JA, Kumar BR, Moshk<strong>in</strong> YM, Langenberg K, Krijgsveld J, Heck AJ, et al. GMPsynthetase stimulates histone H2B deubiquitylation by the epigenetic silencer USP7. Mol Cell2005;17:695e707.[105] Feng L, Wang J, Chen J. The Lys 63-specific deubiquit<strong>in</strong>at<strong>in</strong>g enzyme BRCC36 is regulated by two scaffoldprote<strong>in</strong>s localiz<strong>in</strong>g <strong>in</strong> different subcellular compartments. J Biol Chem 2010;285:30982e8.[106] Wang GG, Allis CD, Chi P. Chromat<strong>in</strong> remodel<strong>in</strong>g and cancer, Part I: Covalent histone modifications. TrendsMol Med 2007;13:363e72.[107] Bannister AJ, Kouzarides T. Regulation of chromat<strong>in</strong> by histone modifications. Cell Res 2011;221:381e95.[108] Taverna SD, Li H, Ruthenburg AJ, Allis CD, Patel DJ. How chromat<strong>in</strong>-b<strong>in</strong>d<strong>in</strong>g modules <strong>in</strong>terpret histonemodifications: lessons from professional pocket pickers. Nat Struct Mol Biol 2007;11:1025e40.[109] Vermeulen M, Eberl HC, Matarese F, Marks H, Denissov S, Butter F, et al. Quantitative <strong>in</strong>teraction proteomicsand genome-wide profil<strong>in</strong>g of epigenetic histone marks and their readers. Cell 2010;142:967e80.[110] Bartke T, Vermeulen M, Xhemalce B, Robson SC, Mann M, Kouzarides T. Nucleosome-<strong>in</strong>teract<strong>in</strong>g prote<strong>in</strong>sregulated by DNA and histone methylation. Cell 2010;43:470e84.[111] Mujtaba S, Zeng L, Zhou MM. Structure and acetyl-lys<strong>in</strong>e recognition of the bromodoma<strong>in</strong>. Oncogene2007;26:5521e7.[112] Maurer-Stroh S, Dickens NJ, Hughes-Davies L, Kouzarides T, Eisenhaber F, Pont<strong>in</strong>g CP. The Tudor doma<strong>in</strong>‘Royal Family’: Tudor, plant Agenet, Chromo, PWWP and MBT doma<strong>in</strong>s. Trends Biochem Sci 2003;28:69e74.[113] Champagne KS, Kutateladze TG. Structural <strong>in</strong>sight <strong>in</strong>to histone recognition by the ING PHD f<strong>in</strong>gers. CurrDrug Targets 2009;10:432e41.[114] Stucki M, Clapperton JA, Mohammad D, Yaffe MB, Smerdon SJ, Jackson SP. MDC1 directly b<strong>in</strong>dsphosphorylated histone H2AX to regulate cellular responses to DNA doublestrand breaks. Cell2005;123:1213e26.[115] Sims RJ, Chen CF, Santos-Rosa H, Kouzarides T, Patel SS, Re<strong>in</strong>berg D. <strong>Human</strong> but not yeast CHD1 b<strong>in</strong>dsdirectly and selectively to histone H3 methylated at lys<strong>in</strong>e 4 via its tandem chromodoma<strong>in</strong>s. J Biol Chem2005;280:41789e92.79


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>80[116] Huang Y, Fang J, Bedford MT, Zhang Y, Xu RM. Recognition of histone H3 lys<strong>in</strong>e-4 methylation by the doubleTudor doma<strong>in</strong> of JMJD2A. <strong>Science</strong> 2006;312:748e51.[117] Trojer P, Li G, Sims 3rd RJ, Vaquero A, Kalakonda N, Boccuni P, et al. L3MBTL1, a histonemethylationdependentchromat<strong>in</strong> lock. Cell 2007;129:915e28.[118] Zegerman P, Canas B, Papp<strong>in</strong> D, Kouzarides T. Histone H3 lys<strong>in</strong>e 4 methylation disrupts b<strong>in</strong>d<strong>in</strong>g ofnucleosome remodel<strong>in</strong>g and deacetylase (NuRD) repressor complex. J Biol Chem 2002;277:11621e4.[119] Adams-Cioaba MA, M<strong>in</strong> J. Structure and function of histone methylation b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s. Biochem Cell Biol2009;87:93e105.[120] Huis<strong>in</strong>ga KL, Brower-Toland B, Elg<strong>in</strong> SCR. The contradictory def<strong>in</strong>itions of heterochromat<strong>in</strong>: Transcriptionand silenc<strong>in</strong>g. Chromosoma 2006;115:110e22.[121] Peters AH, Kubicek S, Mechtler K, O’Sullivan RJ, Derijck AA, Perez-Burgos L, et al. Partition<strong>in</strong>g and plasticityof repressive histone methylation states <strong>in</strong> mammalian chromat<strong>in</strong>. Mol Cell 2003;12:1577e89.[122] Schotta G, Lachner M, Sarma K, Ebert A, Sengupta R, Reuter G, et al. A silenc<strong>in</strong>g pathway to <strong>in</strong>duce H3-K9and H4-K20 trimethylation at constitutive heterochromat<strong>in</strong>. Genes Dev 2004;18:1251e62.[123] Sawan C, Herceg Z. Histone modification and cancer. Adv Genet 2010;70:57e85.[124] Turner BM. Def<strong>in</strong><strong>in</strong>g an epigenetic code. Nat Cell Biol 2007;9:2e6.[125] Hazzal<strong>in</strong> CA, Mahadevan LC. Dynamic acetylation of all lys<strong>in</strong>e 4-methylated histone H3 <strong>in</strong> the mousenucleus: analysis at c-fos and c-jun. PLoS Biol 2005;3:e393.[126] Kohlmaier A, Savarese F, Lachner M, Martens J, Jenuwe<strong>in</strong> T, Wutz A. A chromosomal memory triggered byXist regulates histone methylation <strong>in</strong> X <strong>in</strong>activation. PLoS Biol Jul 2004;2(7):e171.[127] Lee JS, Smith E, Shilatifard A. The language of histone crosstalk. Cell 2010;142:682e5.[128] Latham JA, Dent SY. Cross-regulation of histone modifications. Nat Struct Mol Biol 2007;14:1017e24.[129] Shukla A, Chaurasia P, Bhaumik SR. Histone methylation and ubiquit<strong>in</strong>ation with their cross-talk and roles<strong>in</strong> gene expression and stability. Cell Mol Life Sci 2009;66:1419e33.[130] Sun ZW, Allis CD. Ubiquit<strong>in</strong>ation of histone H2B regulates H3 methylation and gene silenc<strong>in</strong>g <strong>in</strong> yeast.Nature 2002;418:104e8.[131] Dover J, Schneider J, Tawiah-Boateng MA, Wood A, Dean K, Johnston M, et al. Methylation of histone H3 byCOMPASS requires ubiquit<strong>in</strong>ation of histone H2B by Rad6. J Biol Chem 2002;277:28368e71.[132] Lee JS, Shukla A, Schneider J, Swanson SK, Washburn MP, Florens L, et al. Histone crosstalk between H2Bmonoubiquit<strong>in</strong>ation and H3 methylation mediated by COMPASS. Cell 2007;131:1084e96.[133] Kim J, Guermah M, McG<strong>in</strong>ty RK, Lee JS, Tang Z, Milne TA, et al. RAD6-Mediated transcription-coupled H2Bubiquitylation directly stimulates H3K4 methylation <strong>in</strong> human cells. Cell 2009;137:459e71.[134] Fischle W, Tseng BS, Dormann HL, Ueberheide BM, Garcia BA, Shabanowitz J, et al. Regulation of HP1-chromat<strong>in</strong> b<strong>in</strong>d<strong>in</strong>g by histone H3 methylation and phosphorylation. Nature 2005;438:1116e22.[135] Nelson CJ, Santos-Rosa H, Kouzarides T. Prol<strong>in</strong>e isomerization of histone H3 regulates lys<strong>in</strong>e methylationand gene expression. Cell 2006;126:905e16.[136] Clements A, Poux AN, Lo WS, Pillus L, Berger SL, Marmorste<strong>in</strong> R. Structural basis for histone and phosphohistoneb<strong>in</strong>d<strong>in</strong>g by the GCN5 histone acetyltransferase. Mol Cell 2003;12:461e73.[137] Cedar H, Bergman Y. L<strong>in</strong>k<strong>in</strong>g DNA methylation and histone modification: patterns and paradigms. NatRevGenet 2009;10:295e304.[138] Bartke T, Vermeulen M, Xhemalce B, Robson SC, Mann M, Kouzarides T. Nucleosome-<strong>in</strong>teract<strong>in</strong>g prote<strong>in</strong>sregulated by DNA and histone methylation. Cell 2010;143:470e84.[139] Santos-Rosa H, Caldas C. Chromat<strong>in</strong> modifier enzymes, the histone code and cancer. Eur J Cancer2005;41:2381e402.[140] Strahl BD, Allis CD. The language of covalent histone modifications. Nature 2000;403:41e5.[141] Jenuwe<strong>in</strong> T, Allis CD. Translat<strong>in</strong>g the histone code. <strong>Science</strong> 2001;293:1074e80.[142] Gardner KE, Allis CD, Strahl BD. Operat<strong>in</strong>g on chromat<strong>in</strong>, a colorful language where context matters. J MolBiol 2011;409:36e46.[143] Henikoff S, Shilatifard A. Histone modification: cause or cog? Trends Genet 2011;10:389e96.[144] Fraga MF, Ballestar E, Villar-Garea A, Boix-Chornet M, Espada J, Schotta G, et al. Loss of acetylation at Lys16and trimethylation at Lys20 of histone H4 is a common hallmark of human cancer. Nat Genet2005;37:391e400.[145] Seligson DB, Horvath S, Shi T, Yu H, Tze S, Grunste<strong>in</strong> M, et al. Global histone modification patterns predictrisk of prostate cancer recurrence. Nature 2005;435:1262e6.[146] Bagnyukova TV, Tryndyak VP, Montgomery B, Churchwell MI, Karpf AR, James SR, et al. Genetic andepigenetic changes <strong>in</strong> rat preneoplastic liver tissue <strong>in</strong>duced by 2-acetylam<strong>in</strong>ofluorene. Carc<strong>in</strong>ogenesis2008;29:638e46.


CHAPTER 4Alterations of Histone Modifications <strong>in</strong> Cancer[147] Kovalchuk O, Tryndyak VP, Montgomery B, Boyko A, Kutanzi K, Zemp F, et al. Estrogen-<strong>in</strong>duced rat breastcarc<strong>in</strong>ogenesis is characterized by alterations <strong>in</strong> DNA methylation, histone modifications and aberrantmicroRNA expression. Cell Cycle 2007;6:2010e8.[148] Van Den Broeck A, Brambilla E, Moro-Sibilot D, Lantuejoul S, Brambilla C, Eym<strong>in</strong> B, et al. Loss of histoneH4K20 trimethylation occurs <strong>in</strong> preneoplasia and <strong>in</strong>fluences prognosis of non-small cell lung cancer. Cl<strong>in</strong>Cancer Res 2008;14:7237e45.[149] Zhen L, Gui-lan L, P<strong>in</strong>g Y, J<strong>in</strong> H, Ya-li W. The expression of H3K9Ac, H3K14Ac, and H4K20TriMe <strong>in</strong> epithelialovarian tumors and the cl<strong>in</strong>ical significance. Int J Gynecol Cancer 2010;20:82e6.[150] Elsheikh SE, Green AR, Rakha EA, Powe DG, Ahmed RA, Coll<strong>in</strong>s HM, et al. Global histone modifications <strong>in</strong>breast cancer correlate with tumor phenotypes, prognostic factors, and patient outcome. Cancer Res2009;69:3802e9.[151] Li F, Ye B, Hong L, Xu H, Fishbe<strong>in</strong> MC. Epigenetic Modifications of Histone H4 <strong>in</strong> Lung Neuroendocr<strong>in</strong>eTumors. Appl Immunohistochem Mol Morphol 2011;5:389e94.[152] Schneider AC, Heukamp LC, Rogenhofer S, Fechner G, Bastian PJ, von Ruecker A, et al. Global histoneH4K20 trimethylation predicts cancer-specific survival <strong>in</strong> patients with muscle<strong>in</strong>vasive bladder cancer BJUInt; 2011;8(pt2):E290e6.[153] Pogribny IP, Ross SA, Tryndyak VP, Pogribna M, Poirier LA, Karp<strong>in</strong>ets TV. Histone H3 lys<strong>in</strong>e 9 and H4 lys<strong>in</strong>e20 trimethylation and the expression of Suv4-20h2 and Suv-39h1 histone methyltransferases <strong>in</strong> hepatocarc<strong>in</strong>ogenesis<strong>in</strong>duced by methyl deficiency <strong>in</strong> rats. Carc<strong>in</strong>ogenesis 2006;27:1180e6.[154] Pfister S, Rea S, Taipale M, Mendrzyk F, Straub B, Ittrich C, et al. The histone acetyltransferase hMOF isfrequently downregulated <strong>in</strong> primary breast carc<strong>in</strong>oma and medulloblastoma and constitutes a biomarker forcl<strong>in</strong>ical outcome <strong>in</strong> medulloblastoma. Int J Cancer 2008;122:1207e13.[155] Ashktorab H, Belgrave K, Hosse<strong>in</strong>khah F, Brim H, Nouraie M, Takkikto, et al. Global histone H4 acetylationand HDAC2 expression <strong>in</strong> colon adenoma and carc<strong>in</strong>oma. Dig Dis Sci 2009;54:2109e1.[156] Suzuki J, Chen YY, Scott GK, Devries S, Ch<strong>in</strong> K, Benz CC, et al. Prote<strong>in</strong> acetylation and histone deacetylaseexpression associated with malignant breast cancer progression Cl<strong>in</strong> Cancer Res 2009;15:3163e71.[157] Cuomo A, Moretti S, M<strong>in</strong>ucci S, Bonaldi T. SILAC-based proteomic analysis to dissect the "histone modificationsignature" of human breast cancer cells. Am<strong>in</strong>o Acids 2011;41:387e99.[158] Ell<strong>in</strong>ger J, Kahl P, von der Gathen J, Rogenhofer S, Heukamp LC, Gütgemann I, et al. Global levels of histonemodifications predict prostate cancer recurrence. Prostate 2010;70:61e9.[159] Seligson DB, Horvath S, McBrian MA, Mah V, Yu H, Tze S, et al. Global levels of histone modifications predictprognosis <strong>in</strong> different cancers. Am J Pathol 2009;4:1619e28.[160] Mosashvilli D, Kahl P, Mertens C, Holzapfel S, Rogenhofer S, Hauser S, et al. Global histone acetylationlevels: prognostic relevance <strong>in</strong> patients with renal cell carc<strong>in</strong>oma. Cancer Sci 2010;101:2664e9.[161] Manuyakorn A, Paulus R, Farrell J, Dawson NA, Tze S, Cheung-Lau G, et al. Cellular histone modificationpatterns predict prognosis and treatment response <strong>in</strong> resectable pancreatic adenocarc<strong>in</strong>oma: results fromRTOG 9704. J Cl<strong>in</strong> Oncol 2010;28:1358e65.[162] Altucci L, M<strong>in</strong>ucci S. Epigenetic therapies <strong>in</strong> haematological malignancies: search<strong>in</strong>g for true targets. Eur JCancer 2009;45:1137e45.[163] Barlési F, Giaccone G, Gallegos-Ruiz MI, Loundou A, Span SW, Lefesvre P, et al. Global histone modificationspredict prognosis of resected non small-cell lung cancer. J Cl<strong>in</strong> Oncol 2007;25:4358e64.[164] Tzao C, Tung HJ, J<strong>in</strong> JS, Sun GH, Hsu HS, Chen BH, et al. Prognostic significance of global histone modifications<strong>in</strong> resected squamous cell carc<strong>in</strong>oma of the esophagus. Mod Pathol 2009;22:252e60.[165] Liu BL, Cheng JX, Zhang X, Wang R, Zhang W, L<strong>in</strong> H, et al. Global histone modification patterns as prognosticmarkers to classify glioma patients. Cancer Epidemiol Biomarkers Prev 2010;19:2888e96.[166] Das C, Lucia MS, Hansen KC, Tyler JK. CBP/p300-mediated acetylation of histone H3 on lys<strong>in</strong>e 56. Nature2009;459:113e7.[167] Wei Y, Xia W, Zhang Z, Liu J, Wang H, Adsay NV, et al. Loss of trimethylation at lys<strong>in</strong>e 27 of histoneH3 is a predictor of poor outcome <strong>in</strong> breast, ovarian, and pancreatic cancers. Mol Carc<strong>in</strong>og2008;47:701e6.[168] He LR, Liu MZ, Li BK, Rao HL, Liao YJ, Guan XY, et al. Prognostic impact of H3K27me3 expression onlocoregional progression after chemoradiotherapy <strong>in</strong> esophageal squamous cell carc<strong>in</strong>oma. B M C Cancer2009;22(9):461.[169] Cai MY, Tong ZT, Zhu W, Wen ZZ, Rao HL, Kong LL, et al. H3K27me3 Prote<strong>in</strong> Is a Promis<strong>in</strong>g PredictiveBiomarker of Patients’ Survival and Chemoradioresistance <strong>in</strong> <strong>Human</strong> Nasopharyngeal Carc<strong>in</strong>oma. Mol Med2011. doi: 10.2119/molmed.2011.00054. [Epub ahead of pr<strong>in</strong>t].[170] Cai MY, Hou JH, Rao HL, Luo RZ, Li M, Pei XQ, et al. High expression of H3K27me3 <strong>in</strong> human hepatocellularcarc<strong>in</strong>omas correlates closely with vascular <strong>in</strong>vasion and predicts worse prognosis <strong>in</strong> patients. MolMed 2011;(1e2):12e20.81


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>82[171] Kondo Y, Shen L, Cheng AS, Ahmed S, Boumber Y, Charo C, et al. Gene silenc<strong>in</strong>g <strong>in</strong> cancer by histone H3lys<strong>in</strong>e 27 trimethylation <strong>in</strong>dependent of promoter DNA methylation. Nat Genet 2008;40:741e50.[172] Ke XS, Qu Y, Rostad K, Li WC, L<strong>in</strong> B, Halvorsen OJ, et al. Genome-wide profil<strong>in</strong>g of histone h3 lys<strong>in</strong>e 4 andlys<strong>in</strong>e 27 trimethylation reveals an epigenetic signature <strong>in</strong> prostate carc<strong>in</strong>ogenesis. PLoS One 2009;4(3):e4687.[173] Muller-Tidow C, Ulrich Kle<strong>in</strong> H, Hascher A, Isken F, Tickenbrock L, Thoennissen N, et al. Profil<strong>in</strong>g of histoneH3 lys<strong>in</strong>e 9 trimethylation levels predicts transcription factor activity and survival <strong>in</strong> acute myeloid leukemia.Blood 2010;18:3564e71.[174] Dekker FJ, Haisma HJ. Histone acetyl transferases as emerg<strong>in</strong>g drug targets. Drug Discov. Today 2009;14:942e8.[175] Avvakumov N, Cote J. The MYST family of histone acetyltransferases and their <strong>in</strong>timate l<strong>in</strong>ks to cancer.Oncogene 2007;26:5395e407.[176] Yang XJ. The diverse superfamily of lys<strong>in</strong>e acetyltransferases and their roles <strong>in</strong> leukemia and other diseases.Nucleic Acids Res 2004;32:959e76.[177] Muraoka M, Konishi M, Kikuchi-Yanoshita R, Tanaka K, Shitara N, Chong JM, et al. p300 gene alterations <strong>in</strong>colorectal and gastric carc<strong>in</strong>omas. Oncogene 1996;12:1565e9.[178] Gayther SA, Batley SJ, L<strong>in</strong>ger L, Bannister A, Thorpe K, Ch<strong>in</strong> SF, et al. Mutations truncat<strong>in</strong>g the EP300acetylase <strong>in</strong> human cancers. Nat Genet 2000;24:300e3.[179] Bandyopadhyay D, Okan NA, Bales E, Nascimento L, Cole PA, Medrano EE. Down-regulation of p300/CBPhistone acetyltransferase activates a senescence checkpo<strong>in</strong>t <strong>in</strong> human melanocytes. Cancer Res2002;62:6231e9.[180] Iyer NG, Ozdag H, Caldas C. p300/CBP and cancer. Oncogene 2004;23:4225e31.[181] Kishimoto M, Kohno T, Okudela K, Otsuka A, Sasaki H, Tanabe C, et al. Mutations and deletions of the CBPgene <strong>in</strong> human lung cancer. Cl<strong>in</strong> Cancer Res 2005;11:512e9.[182] Crowley JA, Wang Y, Rapoport AP, N<strong>in</strong>g Y. Detection of MOZ-CBP fusion <strong>in</strong> acute myeloid leukemia with 8;16 translocation. Leukemia 2005;19:2344e5.[183] Giles RH, Dauwerse JG, Higg<strong>in</strong>s C, Petrij F, Wessels JW, Beverstock G, et al. Detection of CBP rearrangements<strong>in</strong> acute myelogenous leukemia with t (8; 16). Leukemia 1997;11:2087e96.[184] Taki T, Sako M, Tsuchida M, Hayashi Y. The t (11; 16) (q23; p13) translocation <strong>in</strong> myelodysplastic syndromefuses the MLL gene to the CBP gene. Blood 1997;89:3945e50.[185] Mullighan CG, Zhang J, Kasper LH, Lerach S, Payne-Turner D, Phillips LA, et al. CREBBP mutations <strong>in</strong>relapsed acute lymphoblastic leukaemia. Nature 2011;471:235e9.[186] Pasqualucci L, Dom<strong>in</strong>guez-Sola D, Chiarenza A, Fabbri G, Grunn A, Trifonov V, et al. Inactivat<strong>in</strong>g mutationsof acetyltransferase genes <strong>in</strong> B-cell lymphoma. Nature 2011:471,189e471,195.[187] Panagopoulos I, Fioretos T, Isaksson M, Samuelsson U, Billström R, Strömbeck B, et al. Fusion of theMORF and CBP genes <strong>in</strong> acute myeloid leukemia with the t (10;16)(q22;p13). Hum Mol Genet2001;10:395e404.[188] Lund AH, Turner G, Trubetskoy A, Verhoeven E, Wientjens E, Hulsman D, et al. Genome-wide retroviral<strong>in</strong>sertional tagg<strong>in</strong>g of genes <strong>in</strong>volved <strong>in</strong> cancer <strong>in</strong> Cdkn2a-deficient mice. Nat Genet 2002;32:160e5.[189] Leonart ME, Vidal F, Gallardo D, Diaz-Fuertes M, Rojo F, Cuatrecasas M, et al. New p53 related genes <strong>in</strong>human tumors: significant downregulation <strong>in</strong> colon and lung carc<strong>in</strong>omas. Oncol Rep 2006;16:603e8.[190] Sakuraba K, Yasuda T, Sakata M, Kitamura YH, Shirahata A, Goto T, et al. Down-regulation of Tip60 gene asa potential marker for the malignancy of colorectal cancer. Anticancer Res 2009;10:3953e5.[191] Sakuraba K, Yokomizo K, Shirahata A, Goto T, Saito M, Ishibashi K, et al. TIP60 as a potential marker for themalignancy of gastric cancer. Anticancer Res 2011;31:77e9.[192] Gorr<strong>in</strong>i C, Squatrito M, Luise C, Syed N, Perna D, Wark L, et al. Tip60 is a haplo-<strong>in</strong>sufficient tumoursuppressor required for an oncogene-<strong>in</strong>duced DNA damage response. Nature 2007;448:1063e7:[193] Hobbs CA, Wei G, Defeo K, Paul B, Hayes CS, Gilmour SK. Tip60 prote<strong>in</strong> isoforms and altered function <strong>in</strong>sk<strong>in</strong> and tumors that over-express ornith<strong>in</strong>e decarboxylase. Cancer Res 2006;66:8116e812.[194] Shiota M, Yokomizo A, Masubuchi D, Tada Y, Inokuchi J, Eto M, et al. Tip60 promotes prostate cancer cellproliferation by translocation of androgen receptor <strong>in</strong>to the nucleus. Prostate 2010;70:540e54.[195] M<strong>in</strong>ucci S, Nervi C, Lo Coco F, Pelicci PG. Histone deacetylases: A common molecular target for differentiationtreatment of acute myeloid leukemias? Oncogene 2001;20:3110e5.[196] Peterson LF, Zhang DE. The 8; 21 translocation <strong>in</strong> leukemogenesis. Oncogene 2004;23:4255e62.[197] Mercurio C, M<strong>in</strong>ucci S, Pelicci PG. Histone deacetylases and epigenetic therapies of hematological malignancies.Pharmacol Res 2010;62:18e34.[198] Wang J, Hosh<strong>in</strong>o T, Redner RL, Kajigaya S, Liu JM. ETO, fusion partner <strong>in</strong> t (8; 21) acute myeloid leukemia,represses transcription by <strong>in</strong>teraction with the human N-CoR/mS<strong>in</strong>3/HDAC1 complex. Proc Natl Acad SciUSA 1998;95:10860e5.


CHAPTER 4Alterations of Histone Modifications <strong>in</strong> Cancer[199] Lutterbach B, Westendorf JJ, L<strong>in</strong>ggi B, Patten A, Moniwa M, Davie JR, et al. ETO, a target of t (8; 21) <strong>in</strong> acuteleukemia, <strong>in</strong>teracts with the N-CoR and mS<strong>in</strong>3 corepressors. Mol Cell Biol 1998;18:7176e84.[200] Gelmetti V, Zhang J, Fanelli M, M<strong>in</strong>ucci S, Pelicci PG, Lazar MA. Aberrant recruitment of the nuclear receptorcorepressor-histone deacetylase complex by the acute myeloid leukemia fusion partner ETO. Mol Cell Biol1998;18:7185e91.[201] Shigesada K, van de Sluis B, Liu PP. Mechanism of leukemogenesis by the <strong>in</strong>v (16) chimeric gene CBFB/PEBP2B-MHY11. Oncogene 2004;23:4297e307.[202] Ferrando AA, Herblot S, Palomero T, Hansen M, Hoang T, Fox EA, et al. Biallelic transcriptional activation ofoncogenic transcription factors <strong>in</strong> T-cell acute lymphoblastic leukemia. Blood 2004;103:1909e11.[203] Marquard L, Gjerdrum LM, Christensen IJ, Jensen PB, Sehested M, Ralfkiaer E. Prognostic significance of thetherapeutic targets histone deacetylase 1, 2, 6 and acetylated histone H4 <strong>in</strong> cutaneous T-cell lymphoma.Histopathology 2008;53:267e77.[204] Wang JC, Chen C, Dumlao T, Naik S, Chang T, Xiao YY, et al. Enhanced histone deacetylase enzyme activity<strong>in</strong> primary myelofibrosis. Leuk Lymphoma 2008;49:2321e7.[205] Marquard L, Poulsen CB, Gjerdrum LM, de Nully Brown P, Christensen IJ, Jensen PB. Histone deacetylase 1,2, 6 and acetylated histone H4 <strong>in</strong> B- and T-cell lymphomas. Histopathology 2009;54:688e98.[206] Bolden JE, Peart MJ, Johnstone RW. Anticancer activities of histone deacetylase <strong>in</strong>hibitors. Nat Rev DrugDiscov 2006;5:769e84.[207] Osada H, Tatematsu Y, Saito H, Yatabe Y, Mitsudomi T, Takahashi T. Reduced expression of class IIhistone deacetylase genes is associated with poor prognosis <strong>in</strong> lung cancer patients. Int J Cancer2004;112:26e32.[208] Huffman DM, Grizzle WE, Bamman MM, Kim JS, Eltoum IA, Elgavish A, et al. SIRT1 is significantly elevated<strong>in</strong> mouse and human prostate cancer. Cancer Res 2007;67:6612e8.[209] Liu G, Yuan X, Zeng Z, Tunici P, Ng H, Abdulkadir IR, et al. Analysis of gene expression and chemoresistanceof CD133þ cancer stem cells <strong>in</strong> glioblastoma. Mol Cancer 2006;5:67.[210] Ozdag H, Teschendorff AE, Ahmed AA, Hyland SJ, Blenkiron C, Bobrow L, et al. Differential expression ofselected histone modifier genes <strong>in</strong> human solid cancers. B M C Genomics Apr 2006;25(7):90.[211] Biancotto C, Frigè G, M<strong>in</strong>ucci S. Histone modification therapy of cancer. Adv Genet 2010;70:341e86.[212] Albert M, Hel<strong>in</strong> K. Histone methyltransferases <strong>in</strong> cancer. Sem<strong>in</strong>ars <strong>in</strong> Cell & Developmental Biology2010;21:209e20.[213] Varier RA, Timmers HT. Histone lys<strong>in</strong>e methylation and demethylation pathways <strong>in</strong> cancer. Biochimica etBiophysica Acta 2011;1815:75e89.[214] Ka MY, Lee BB, Kim YH, Chang DK, Kyu Park S, Chun HK, et al. Association of the SUV39H1 histonemethyltransferase with the DNA methyltransferase 1 at mRNA expression level <strong>in</strong> primary colorectal cancer.Int J Cancer 2007;121:2e7.[215] Peters AH, O’Carroll D, Scherthan H, Mechtler K, Sauer S, Schöfer C, et al. Loss of the Suv39h histonemethyltransferases impairs mammalian heterochromat<strong>in</strong> and genome stability. Cell 2001;107:323e37.[216] Hess JL. MLL: a histone methyltransferase disrupted <strong>in</strong> leukemia. Trends Mol Med 2004;10:500e7.[217] Krivtsov AV, Armstrong SA. MLL translocations, histone modifications and leukaemia stem-cell development.Nat Rev Cancer 2007;7:823e33.[218] Shih LY, Liang DC, Fu JF, Wu JH, Wang PN, L<strong>in</strong> TL, et al. Characterization of fusion partner genes<strong>in</strong> 114 patients with de novo acute myeloid leukemia and MLL rearrangement. Leukemia 2006;20. 218-2.[219] Slany RK. The molecular biology of mixed l<strong>in</strong>eage leukemia. Haematologica 2009;94:984e93.[220] Sjöblom T, Jones S, Wood LD, Parsons DW, L<strong>in</strong> J, Barber TD, et al. The consensus cod<strong>in</strong>g sequences of humanbreast and colorectal cancers. <strong>Science</strong> 2006;314:268e74.[221] Hamamoto R, Furukawa Y, Morita M, Iimura Y, Silva FP, Li M, et al. SMYD3 encodes a histone methyltransferase<strong>in</strong>volved <strong>in</strong> the proliferation of cancer cells. Nat Cell Biol 2004;6:731e40.[222] Luo XG, Xi T, Guo S, Liu ZP, Wang N, Jiang Y, et al. Effects of SMYD3 over-expression on transformation,serum dependence, and apoptosis sensitivity <strong>in</strong> NIH3T3 cells. IUBMB Life 2009;61:679e84.[223] Hamamoto R, Silva FP, Tsuge M, Nishidate T, Katagiri T, Nakamura Y, et al. Enhanced SMYD3 expression isessential for the growth of breast cancer cells. Cancer Sci 2006;97:113e8.[224] Skawran B, Ste<strong>in</strong>emann D, Weigmann A, Flemm<strong>in</strong>g P, Becker T, Flik J, et al. Gene expression profil<strong>in</strong>g <strong>in</strong> hepatocellularcarc<strong>in</strong>oma: upregulation of genes <strong>in</strong> amplified chromosome regions. Mod Pathol 2008;21:505e16.[225] Northcott PA, Nakahara Y, Wu X, Feuk L, Ellison DW, Croul S, et al. Multiple recurrent genetic eventsconverge on control of histone lys<strong>in</strong>e methylation <strong>in</strong> medulloblastoma. Nat Genet 2009;41:465e72.[226] Hu L, Zhu YT, Qi C, Zhu YJ. Identification of Smyd4 as a potential tumor suppressor gene <strong>in</strong>volved <strong>in</strong> breastcancer development. Cancer Res 2009;69:4067e72.83


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>84[227] Huang J, Dorsey J, Chuikov S, Pérez-Burgos L, Zhang X, Jenuwe<strong>in</strong> T, et al. G9A and GLP methylate lys<strong>in</strong>e 373<strong>in</strong> the tumor suppressor p53. J Biol Chem 2010;285:9636e41.[228] Kondo Y, Shen L, Ahmed S, Boumber Y, Sekido Y, Haddad BR, et al. Downregulation of histone H3 lys<strong>in</strong>e 9methyltransferase G9a <strong>in</strong>duces centrosome disruption and chromosome <strong>in</strong>stability <strong>in</strong> cancer cells. PLoS ONE2008;3: e2037.[229] Kondo Y, Shen L, Suzuki S, Kurokawa T, Masuko K, Tanaka Y, et al. Alterations of DNA methylation andhistone modifications contribute to gene silenc<strong>in</strong>g <strong>in</strong> hepatocellular carc<strong>in</strong>omas. Hepatol Res2007;37:974e83.[230] Watanabe H, Soejima K, Yasuda H, Kawada I, Nakachi I, Yoda S, et al. Deregulation of histone lys<strong>in</strong>emethyltransferases contributes to oncogenic transformation of human bronchoepithelial cells. Cancer CellInt 2008;8:15.[231] Chase A, Cross NC. Aberrations of EZH2 <strong>in</strong> cancer. Cl<strong>in</strong> Cancer Res 2011;17:2613e8.[232] Kleer CG, Cao Q, Varambally S, Shen R, Ota I, Toml<strong>in</strong>s SA, et al. EZH2 is a marker of aggressive breast cancerand promotes neoplastic transformation of breast epithelial cells. Proc Natl Acad Sci USA2003;100:11606e11.[233] Yu J, Yu J, Rhodes DR, Toml<strong>in</strong>s SA, Cao X, Chen G, et al. A polycomb repression signature <strong>in</strong> metastaticprostate cancer predicts cancer outcome. Cancer Res 2007;67:10657e63.[234] Alford SH, Toy K, Merajver SD, Kleer CG. Increased risk for distant metastasis <strong>in</strong> patients with familial earlystagebreast cancer and high EZH2 expression. Breast Cancer Res Treat 2011. 2011 May 26. [Epub ahead ofpr<strong>in</strong>t].[235] Takawa M, Masuda K, Kunizaki M, Daigo Y, Takagi K, Iwai Y, et al. Validation of the histone methyltransferaseEZH2 as a therapeutic target for various types of human cancer and as a prognostic marker. Cancer Sci2011;102:1298e305.[236] Chang CJ, Yang JY, Xia W, Chen CT, Xie X, Chao CH, et al. EZH2 promotes expansion of breast tumor<strong>in</strong>itiat<strong>in</strong>g cells through activation of RAF1-beta-caten<strong>in</strong> signal<strong>in</strong>g. Cancer Cell 2011;19:86e100.[237] Ernst T, Chase AJ, Score J, Hidalgo-Curtis CE, Bryant C, Jones AV, et al. Inactivat<strong>in</strong>g mutations of the histonemethyltransferase gene EZH2 <strong>in</strong> myeloid disorders. Nat Genet 2010;42:722e6.[238] Nikoloski G, Langemeijer SM, Kuiper RP, Knops R, Massop M, Tönnissen ER, et al. Somatic mutations of thehistone methyltransferase gene EZH2 <strong>in</strong> myelodysplastic syndromes. Nat Genet 2010;42:665e7.[239] Makishima H, Jankowska AM, Tiu RV, Szpurka H, Sugimoto Y, Hu Z, et al. Novel homo- and hemizygousmutations <strong>in</strong> EZH2 <strong>in</strong> myeloid malignancies. Leukemia 2010;24:1799e804.[240] Mor<strong>in</strong> RD, Johnson NA, Severson TM, Mungall AJ, An J, Goya R, et al. Somatic mutations alter<strong>in</strong>g EZH2(Tyr641) <strong>in</strong> follicular and diffuse large B-cell lymphomas of germ<strong>in</strong>al-center orig<strong>in</strong>. Nat Genet 2010;42:181e18.[241] Yap DB, Chu J, Berg T, Schapira M, Cheng SW, Moradian A, et al. Somatic mutations at EZH2 Y641 actdom<strong>in</strong>antly through a mechanism of selectively altered PRC2 catalytic activity, to <strong>in</strong>crease H3K27 trimethylation.Blood 2011;117:2451e9.[242] Morishita M, di Luccio E. Cancers and the NSD family of histone lys<strong>in</strong>e methyltransferases. Biochim BiophysActa 2011;1816:158e63.[243] Wang GG, Caim L, Pasillas MP, Kamps MP. NUP98-NSD1 l<strong>in</strong>ks H3K36 methylation to Hox-A gene activationand leukaemogenesis. Nat Cell Biol 2007;9:804e12.[244] Jaju RJ, Fidler C, Haas OA, Strickson AJ, Watk<strong>in</strong>s F, Clark K, et al. A novel gene, NSD1, is fused to NUP98 <strong>in</strong>the t(5;11)(q35;p15.5) <strong>in</strong> de novo childhood acute myeloid leukemia,. Blood 2001;98:1264e12.[245] Berdasco M, Ropero S, Setien F, Fraga MF, Lapunz<strong>in</strong>a P, Losson R, et al. Epigenetic <strong>in</strong>activation of the Sotosovergrowth syndrome gene histone methyltransferase NSD1 <strong>in</strong> human neuroblastoma and glioma. Proc NatlAcad Sci USA 2009;106:21830e5.[246] Mart<strong>in</strong>ez-Garcia E, Popovic R, M<strong>in</strong> DJ, Sweet SM, Thomas PM, Zamdborg L, et al. The MMSET histone methyltransferase switches global histone methylation and alters gene expression <strong>in</strong> t (4;14) multiple myelomacells. Blood 2011;117:211e20.[247] Brito JL, Walker B, Jenner M, Dickens NJ, Brown NJ, Ross FM, et al. MMSET deregulation affects cell cycleprogression and adhesion regulons <strong>in</strong> t (4; 14) myeloma plasma cells. Haematologica 2009;94:78e86.[248] Kassambara A, Kle<strong>in</strong> B, Moreaux J. MMSET is over-expressed <strong>in</strong> cancers: l<strong>in</strong>k with tumor aggressiveness.Biochem. Biophys Res Comm 2009;379:840e5.[249] Rosati R, La Starza R, Veronese A, Avent<strong>in</strong> A, Schwienbacher C, Vallespi T, et al. NUP98 is fused to the NSD3gene <strong>in</strong> acute myeloid leukemia associated with t (8;11)(p11.2;p15). Blood 2002;99:3857e60.[250] Angrand PO, Apiou F, Stewart AF, Dutrillaux B, Losson R, Chambon P. NSD3, a new SET doma<strong>in</strong>-conta<strong>in</strong><strong>in</strong>ggene, maps to 8p12 and is amplified <strong>in</strong> human breast cancer cell l<strong>in</strong>es. Genomics 2001;74:79e88.[251] Bernt KM, Zhu N, S<strong>in</strong>ha AU, Vempati S, Faber J, Krivtsov AV, et al. MLL-Rearranged Leukemia Is Dependenton Aberrant H3K79 Methylation by DOT1L. Cancer Cell 2011;20:66e78.


CHAPTER 4Alterations of Histone Modifications <strong>in</strong> Cancer[252] Nguyen AT, Taranova O, He J, Zhang Y. DOT1L, the H3K79 methyltransferase, is required for MLL-AF9-mediated leukemogenesis. Blood 2011;117:6912e22.[253] Chang MJ, Wu H, Achille NJ, Reisenauer MR, Chou CW, Zeleznik-Le NJ. Histone H3 lys<strong>in</strong>e 79 methyltransferaseDot1 is required for immortalization by MLL oncogenes. Cell 2005;121:167e78.[254] Barry ER, Corry GN, Rasmussen TP. Target<strong>in</strong>g DOT1L action and <strong>in</strong>teractions <strong>in</strong> leukemia: the role of DOT1L<strong>in</strong> transformation and development. Expert Op<strong>in</strong> Ther Targets 2010;14:405e18.[255] Majumder S, Liu Y, Ford 3rd OH, Mohler JL, Whang YE, Involvement of arg<strong>in</strong><strong>in</strong>e methyltransferase. CARM1<strong>in</strong> androgen receptor function and prostate cancer cell viability. Prostate 2006;66:1292e301.[256] Cheung N, Chan LC, Thompson A, Cleary ML, Eric So CW. Prote<strong>in</strong> arg<strong>in</strong><strong>in</strong>emethyltransferase- dependentoncogenesis. Nat Cell Biol 2007;9:1208e15.[257] Pal S, Vishwanath SN, Erdjument-Bromage H, Tempst P, Sif S. <strong>Human</strong> SWI/SNFassociated PRMT5 methylateshistone H3 arg<strong>in</strong><strong>in</strong>e 8 and negatively regulates expression of ST7 and NM23 tumor suppressor genes. MolCell Biol 2004;24:9630e45.[258] Yoshimatsu M, Toyokawa G, Hayami S, Unoki M, Tsunoda T, et al. Dysregulation of PRMT1 and PRMT6,Type I arg<strong>in</strong><strong>in</strong>e methyltransferases, is <strong>in</strong>volved <strong>in</strong> various types of human cancers. Int J Cancer2011;128:562e73.[259] Kahl P, Gullotti L, Heukamp LC, Wolf S, Friedrichs N, Vorreuther R, et al. Androgen receptor coactivatorslys<strong>in</strong>e-specific histone demethylase 1 and four and a half LIM doma<strong>in</strong> prote<strong>in</strong> 2 predict risk of prostate cancerrecurrence. Cancer Res 2006;66:11341e1134.[260] Schulte JH, Lim S, Schramm A, Friedrichs N, Koster J, Versteeg R, et al. Lys<strong>in</strong>e-specific demethylase 1 isstrongly expressed <strong>in</strong> poorly differentiated neuroblastoma: implications for therapy. Cancer Res2009;69:2065e71.[261] Lim S, Janzer A, Becker A, Zimmer A, Schule R, Buettner R, et al. Lys<strong>in</strong>e specific demethylase 1 (LSD1) ishighly expressed <strong>in</strong> ER-negative breast cancers and a biomarker predict<strong>in</strong>g aggressive biology. Carc<strong>in</strong>ogenesis2010;31:512e5.[262] Hayami S, Kelly JD, Cho HS, Yoshimatsu M, Unoki M, Tsunoda T, et al. Over-expression of LSD1contributes to human carc<strong>in</strong>ogenesis through chromat<strong>in</strong> regulation <strong>in</strong> various cancers. Int J Cancer2011;128:574e86.[263] Kauffman EC, Rob<strong>in</strong>son BD, Downes MJ, Powell LG, Lee MM, Scherr DS, et al. Role of androgen receptor andassociated lys<strong>in</strong>e-demethylase coregulators, LSD1 and JMJD2A, <strong>in</strong> localized and advanced human bladdercancer. Mol Carc<strong>in</strong>og 2011. 2011 Mar 11. doi: 10.1002/mc.20758. Epub ahead of pr<strong>in</strong>t.[264] Magerl C, Ell<strong>in</strong>ger J, Braunschweig T, Kremmer E, Koch LK, Höller T, et al. H3K4 dimethylation <strong>in</strong> hepatocellularcarc<strong>in</strong>oma is rare compared with other hepatobiliary and gastro<strong>in</strong>test<strong>in</strong>al carc<strong>in</strong>omas and correlateswith expression of the methylase Ash2 and the demethylase LSD1. Hum Pathol 2010;41:181e9.[265] Suzuki T, M<strong>in</strong>ehata K, Akagi K, Jenk<strong>in</strong>s NA, Copeland NG. Tumor suppressor gene identification us<strong>in</strong>gretroviral <strong>in</strong>sertional mutagenesis <strong>in</strong> Blm-deficient mice. EMBO J 2006;25:3422e31.[266] Pfau R, Tzatsos A, Kampranis SC, Serebrennikova OB, Bear SE, Tsichlis PN. Members of a family of JmjCdoma<strong>in</strong>-conta<strong>in</strong><strong>in</strong>g oncoprote<strong>in</strong>s immortalize embryonic fibroblasts via a JmjC doma<strong>in</strong> dependent process.Proc Natl Acad Sci USA 2008;105:1907e12.[267] Tzatsos A, Pfau R, Kampranis SC, Tsichlis PN. Ndy1/KDM2B immortalizes mouse embryonic fibroblasts byrepress<strong>in</strong>g the Ink4a/Arf locus. Proc Natl Acad Sci USA 2009;106:2641e6.[268] Andersson A, Ritz C, L<strong>in</strong>dgren D, Edén P, Lassen C, Heldrup J, et al. Microarray-based classification ofa consecutive series of 121 childhood acute leukemias: prediction of leukemic and genetic subtype as well asof m<strong>in</strong>imal residual disease status. Leukemia 2007;21:1198e203.[269] Sperger JM, Chen X, Draper JS, Antosiewicz JE, Chon CH, Jones SB, et al. Gene expression patterns <strong>in</strong> humanembryonic stem cells and human pluripotent germ cell tumors. Proc Natl Acad Sci USA 2003;100:13350e5.[270] He J, Nguyen AT, Zhang Y. KDM2b/JHDM1b, an H3K36me2-specific demethylase, is required for <strong>in</strong>itiationand ma<strong>in</strong>tenance of acute myeloid leukemia. Blood 2001;117:3869e80.[271] Frescas D, Guardavaccaro D, Kuchay SM, Kato H, Poleshko A, Basrur V, et al. KDM2A represses transcriptionof centromeric satellite repeats and ma<strong>in</strong>ta<strong>in</strong>s the heterochromatic state. Cell Cycle 2008;7:3539e47.[272] Frescas D, Guardavaccaro D, Bassermann F, Koyama-Nasu R, Pagano M. JHDM1B/FBXL10 is a nucleolarprote<strong>in</strong> that represses transcription of ribosomal RNA genes. Nature 2007;450:309e13.[273] Yang ZQ, Imoto I, Fukuda Y, Pimkhaokham A, Shimada Y, Imamura M, et al. Identification of a novel gene,GASC1, with<strong>in</strong> an amplicon at 9p23-24 frequently detected <strong>in</strong> esophageal cancer cell l<strong>in</strong>es. Cancer Res2000;60:4735e9.[274] V<strong>in</strong>atzer U, Goll<strong>in</strong>ger M, Mullauer L, Raderer M, Chott A, Streubel B. Mucosa associated lymphoid tissuelymphoma: novel translocations <strong>in</strong>clud<strong>in</strong>g rearrangements of ODZ2, JMJD2C, and CNN3. Cl<strong>in</strong> Cancer Res2008;14:6426e31.85


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>86[275] Italiano A, Attias R, Aurias A, Pérot G, Burel-Vandenbos F, Otto J, et al. Molecular cytogenetic characterizationof a metastatic lung sarcomatoid carc<strong>in</strong>oma: 9p23 neocentromere and 9p23-p24 amplification <strong>in</strong>clud<strong>in</strong>gJAK2 and JMJD2C. Cancer Genet Cytogenet 2006;167:122e30.[276] Liu G, Bollig-Fischer A, Kreike B, van de Vijver MJ, Abrams J, Ethier SP, et al. Genomic amplification andoncogenic properties of the GASC1 histone demethylase gene <strong>in</strong> breast cancer. Oncogene 2009;28:4491e500.[277] Ehrbrecht A, Müller U, Wolter M, Hoischen A, Koch A, Radlwimmer B, et al. Comprehensive genomicanalysis of desmoplastic medulloblastomas: identification of novel amplified genes and separate evaluationof the different histological components. J Pathol 2006;208:554e63.[278] Cloos PA, Christensen J, Agger K, Maiolica A, Rappsilber J, Antal T, et al. The putative oncogene GASC1demethylates tri- and dimethylated lys<strong>in</strong>e 9 on histone H3. Nature 2006;442:307e11.[279] Northcott PA, Nakahara Y, Wu X, Feuk L, Ellison DW, Croul S, et al. Multiple recurrent genetic eventsconverge on control of histone lys<strong>in</strong>e methylation <strong>in</strong> medulloblastoma. Nat Gen 2009;41:465e72.[280] Yang J, Jubb AM, Pike L, Buffa FM, Turley H, Baban D, et al. The histone demethylase JMJD2B is regulated byestrogen receptor alpha and hypoxia, and is a key mediator of estrogen <strong>in</strong>duced growth. Cancer Res2010;70:6456e66.[281] Zeng KJ, Ge Z, Wang L, Li Q, Wang N, Bjorkholm M, et al. The histone demethylase RBP2 Is over-expressed <strong>in</strong>gastric cancer and its <strong>in</strong>hibition triggers senescence of cancer cells. Gastroenterology 2010;138:981e92.[282] Sharma SV, Lee DY, Li B, Qu<strong>in</strong>lan MP, Takahashi F, Maheswaran S, et al. A chromat<strong>in</strong>-mediated reversibledrug-tolerant state <strong>in</strong> cancer cell subpopulations. Cell 2010;141:69e80.[283] Xiang Y, Zhu Z, Han G, Ye X, Xu B, Peng Z, et al. JARID1B is a histone H3 lys<strong>in</strong>e 4 demethylase up-regulated<strong>in</strong> prostate cancer. Proc Natl Acad Sci USA 2007;104:19226e31.[284] Lu PJ, Sundquist K, Baeckstrom D, Poulsom R, Hanby A, Meier-Ewert S, et al. A novel gene (PLU-1)conta<strong>in</strong><strong>in</strong>g highly conserved putative DNA/chromat<strong>in</strong> b<strong>in</strong>d<strong>in</strong>g motifs is specifically up-regulated <strong>in</strong> breastcancer. J Biol Chem 1999;274:15633e45.[285] Yamane K, Tateishi K, Klose RJ, Fang J, Fabrizio LA, Erdjument-Bromage H, et al. PLU-1 is an H3K4 demethylase<strong>in</strong>volved <strong>in</strong> transcriptional repression and breast cancer cell proliferation. Mol Cell 2007;25:801e12.[286] Hayami S, Yoshimatsu M, Veerakumarasivam A, Unoki M, Iwai Y, Tsunoda T, et al. Over-expression of theJmjC histone demethylase KDM5B <strong>in</strong> human carc<strong>in</strong>ogenesis: <strong>in</strong>volvement <strong>in</strong> the proliferation of cancer cellsthrough the E2F/RB pathway. Mol Cancer 2010;13:9e59.[287] Dalgliesh GL, Furge K, Greenman C, Chen L, Bignell G, Butler A, et al. Systematic sequenc<strong>in</strong>g of renalcarc<strong>in</strong>oma reveals <strong>in</strong>activation of histone modify<strong>in</strong>g genes. Nature 2010;463:360e3.[288] Smith JA, White EA, Sowa ME, Powell ML, Ott<strong>in</strong>ger M, Harper JW, et al. Genome-wide siRNA screen identifiesSMCX, EP400, and Brd4 as E2-dependent regulators of human papillomavirus oncogene expression. ProcNatl Acad Sci USA 2010;107:3752e7.[289] van Haaften G, Dalgliesh GL, Davies H, Chen L, Bignell G, Greenman C, et al. Somatic mutations of thehistone H3K27 demethylase gene UTX <strong>in</strong> human cancer. Nat Genet 2009;41:521e3.[290] Agger K, Cloos PAC, Rudkjær L, Williams K, Andersen G, Christensen J, et al. The H3K27me3 demethylaseJMJD3 contributes to the activation of the INK4AeARF locus <strong>in</strong> response to oncogene- and stress-<strong>in</strong>ducedsenescence. Genes Dev 2009;23:1171e6.[291] Barradas M, Anderton E, Acosta JC, Li S, Banito A, Rodriguez-Niedenführ M, et al. Histone demethylaseJMJD3 contributes to epigenetic control of INK4a/ARF by oncogenic RAS. Genes Dev 2009;23:1177e82.[292] McLaughl<strong>in</strong>-Drub<strong>in</strong> ME, Crum CP, Münger K. <strong>Human</strong> papillomavirus E7 oncoprote<strong>in</strong> <strong>in</strong>duces KDM6A andKDM6B histone demethylase expression and causes epigenetic reprogramm<strong>in</strong>g. Proc Natl Acad Sci USA2011;2108:2130e5.[293] Anderton JA, Bose S, Vockerodt M, Vrzalikova K, Wei W, Kuo M, et al. The H3K27me3 demethylase, KDM6B,is <strong>in</strong>duced by Epste<strong>in</strong>eBarr virus and over-expressed <strong>in</strong> Hodgk<strong>in</strong>’s Lymphoma. Oncogene 2011;30:2037e43.[294] Hsia DA, Tepper CG, Pochampalli MR, Hsia EY, Izumiya C, Huerta SB, et al. KDM8, a H3K36me2 histonedemethylase that acts <strong>in</strong> the cycl<strong>in</strong> A1 cod<strong>in</strong>g region to regulate cancer cell proliferation. Proc Natl Acad SciUSA 2010;107:9671e6.[295] Vader G, Lens SM. The Aurora k<strong>in</strong>ase family <strong>in</strong> cell division and cancer. Biochim Biophys Acta2008;1786:60e72.[296] Carvajal RD, Tse A, Schwartz GK. Aurora k<strong>in</strong>ases: new targets for cancer therapy. Cl<strong>in</strong> Cancer Res2006;12:6869e75.[297] Tatsuka M, Katayama H, Ota T, Tanaka T, Odashima S, Suzuki F, et al. Mult<strong>in</strong>uclearity and <strong>in</strong>creased ploidycaused by over-expression of the aurora- and Ipl1-like midbody-associated prote<strong>in</strong> mitotic k<strong>in</strong>ase <strong>in</strong> humancancer cells. Cancer Res 1998;58:4811e6.[298] Katayama H, Ota T, Jisaki F, Ueda Y, Tanaka T, Odashima S, et al. Mitotic k<strong>in</strong>ase expression and colorectalcancer progression. J Natl Cancer Inst 1999;91:1160e2.


CHAPTER 4Alterations of Histone Modifications <strong>in</strong> Cancer[299] Lev<strong>in</strong>e RL, Gilliland DG. JAK-2 mutations and their relevance to myeloproliferative disease. Curr Op<strong>in</strong>Hematol 2007;14:43e7.[300] Rui L, Emre NC, Kruhlak MJ, Chung HJ, Steidl C, Slack G, et al. Cooperative epigenetic modulation by canceramplicon genes. Cancer Cell 2010;18:590e605.[301] Odgerel T, Kikuchi J, Wada T, Shimizu R, Kano Y, Furukawa Y. MSK1 activation <strong>in</strong> acute myeloid leukemiacells with FLT3 mutations. Leukemia 2010;24:1087e90.[302] Kim HG, Lee KW, Cho YY, Kang NJ, Oh SM, Bode AM, et al. Mitogen- and stress-activated k<strong>in</strong>ase 1-mediatedhistone H3 phosphorylation is crucial for cell transformation. Cancer Res 2008;68:2538e47.[303] Cho YY, Yao K, Kim HG, Kang BS, Zheng D, Bode AM, et al. Ribosomal S6 k<strong>in</strong>ase 2 is a key regulator <strong>in</strong> tumorpromoter <strong>in</strong>duced cell transformation. Cancer Res 2007;67:8104e12.[304] Song H, Mak KK, Topol L, Yun K, Hu J, Garrett L, et al. Mammalian Mst1 and Mst2 k<strong>in</strong>ases play essential roles<strong>in</strong> organ size control and tumor suppression. Proc Natl Acad Sci USA 2010;107:1431e6.[305] Zhu F, Zykova TA, Peng C, Zhang J, Cho YY, Zheng D, et al. Phosphorylation of H2AX at Ser139 and a newphosphorylation site Ser16 by RSK2 decreases H2AX ubiquit<strong>in</strong>ation and <strong>in</strong>hibits cell transformation. CancerRes 2011;71:393e403.[306] Esp<strong>in</strong>osa JM. Histone H2B ubiquit<strong>in</strong>ation: the cancer connection. Genes Dev 2008;22:2743e9.[307] Shema E, Tirosh I, Aylon Y, Huang J, Ye C, Moskovits N, et al. The histone H2B-specific ubiquit<strong>in</strong> ligaseRNF20/hBRE1 acts as a putative tumor suppressor through selective regulation of gene expression. Genes Dev2008;22:2664e76.[308] Gl<strong>in</strong>sky GV. Genomic models of metastatic cancer: functional analysis of death from- cancer signature genesreveals aneuploid, anoikis-resistant, metastasis-enabl<strong>in</strong>g phenotype with altered cell cycle control and activatedPolycomb Group (PcG) prote<strong>in</strong> chromat<strong>in</strong> silenc<strong>in</strong>g pathway. Cell Cycle 2006;5:1208e16.[309] Gl<strong>in</strong>sky GV, Berezovska O, Gl<strong>in</strong>skii AB. Microarray analysis identifies a death-from cancer signaturepredict<strong>in</strong>g therapy failure <strong>in</strong> patients with multiple types of cancer. J Cl<strong>in</strong> Invest 2005;115:1503e21.[310] Liu YL, Yang YM, Xu H, Dong XS. Aberrant expression of USP22 is associated with liver metastasis and poorprognosis of colorectal cancer. J Surg Oncol 2011;103:283e9.[311] Zhang Y, Yao L, Zhang X, Ji H, Wang L, Sun S, et al. Elevated expression of USP22 <strong>in</strong> correlation with poorprognosis <strong>in</strong> patients with <strong>in</strong>vasive breast cancer. J Cancer Res Cl<strong>in</strong> Oncol 2011;137:1245e53.[312] Luise C, Capra M, Donzelli M, Mazzarol G, Jodice MG, Nuciforo P, et al. An Atlas of Altered Expression ofDeubiquit<strong>in</strong>at<strong>in</strong>g Enzymes <strong>in</strong> <strong>Human</strong> Cancer. PLoS One 2011;6(1):e15891.[313] Hussa<strong>in</strong> S, Zhang Y, Galardy PJ. DUBs and cancer: the role of deubiquit<strong>in</strong>at<strong>in</strong>g enzymes as oncogenes,nononcogenes and tumor suppressors. Cell Cycle 2009;8:1688e97.[314] Kurdistani SK. Histone modifications <strong>in</strong> cancer biology and prognosis. Prog Drug Res 2011;67:91e106.[315] Xu W, Yang H, Liu Y, Yang Y, Wang P, Kim SH, et al. Oncometabolite 2-hydroxyglutarate is a competitive<strong>in</strong>hibitor of a-ketoglutarate-dependent dioxygenases. Cancer Cell 2011;19:17e30.[316] Fanelli M, Amatori S, Barozzi I, Sonc<strong>in</strong>i M, Zuffo Dal, Bucci R, et al. Pathology tissue-chromat<strong>in</strong> immunoprecipitation,coupled with high-throughput sequenc<strong>in</strong>g, allows the epigenetic profil<strong>in</strong>g of patient samples.Proc Natl Acad Sci USA 2010;107:21535e40.87


This page <strong>in</strong>tentionally left blank


CHAPTER 5MicroRNA <strong>in</strong> OncogenesisNiamh Lynam-Lennon, Steven G. Gray, Stephen G. MaherTr<strong>in</strong>ity College Dubl<strong>in</strong>, Tr<strong>in</strong>ity Centre for Health <strong>Science</strong>s, Dubl<strong>in</strong>, IrelandCHAPTER OUTLINE5.1 Introduction 895.2 miRNA Biogenesis 895.3 miRNA-Mediated Regulation ofTargets 915.4 miRNA and Cancer 925.5 OncomiRs 935.5.1 miRNA as TumorSuppressors 935.5.2 miRNA as Oncogenes 945.6 Mechanisms of miRNADeregulation 955.6.1 ChromosomalAberrations 955.6.2 Dysregulation of TranscriptionFactors 965.6.3 Epigenetic Alterations 975.6.4 Altered Process<strong>in</strong>g 985.7 miRNA and TreatmentResistance 995.7.1 Role of miRNA <strong>in</strong>Chemoresistance 995.7.2 Role of miRNA <strong>in</strong>Radioresistance 1005.8 Cl<strong>in</strong>ical Applications 102References 103895.1 INTRODUCTIONIdentified <strong>in</strong> 2001, microRNA (miRNA) compose a large part of a family of small non-cod<strong>in</strong>gRNA, which <strong>in</strong>cludes small nuclear RNA (snRNA), small nucleolar RNA (snoRNA), small<strong>in</strong>terfer<strong>in</strong>g RNA (siRNA), and piwi-<strong>in</strong>teract<strong>in</strong>g RNA (piRNA). With<strong>in</strong> cells, miRNA function asendogenous gene silencers, repress<strong>in</strong>g target mRNA at a translational level. miRNA are highlyevolutionarily conserved, be<strong>in</strong>g present <strong>in</strong> the genomes of animals, plants, and viruses. It isnow considered that miRNA may represent anywhere <strong>in</strong> the region of 1e3% of the entirehuman genome [1,2] and estimates of the number of miRNA targets <strong>in</strong>dicate that they mayplay a role <strong>in</strong> regulat<strong>in</strong>g as many as 30% of mammalian genes [3]. Consequently, miRNA havebeen shown to play central roles <strong>in</strong> developmental tim<strong>in</strong>g, hematopoietic cell differentiation,programmed cell death, and oncogenesis [4e7].5.2 miRNA BIOGENESISmiRNA are synthesized <strong>in</strong> the nucleus as long primary transcripts (pri-miRNA) up to 1000nucleotides (nt) <strong>in</strong> length, which are characterized by imperfect hairp<strong>in</strong> structures. While themajority of transcripts are less than 1 kb, longer primary transcripts have been documented,for example, pri-miR-21 is 3433 nt [8]. For the majority of miRNA, transcription is mediated byRNA polymerase II (Pol II) [9], although a small subset are transcribed by Pol III [10]. The primiRNAhairp<strong>in</strong> structures have a 5 0 -cap and a 3 0 -polyA tail. Most pri-miRNA arise fromT. Tollefsbol (Ed): <strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>. DOI: 10.1016/B978-0-12-388415-2.00005-6Copyright Ó 2012 Elsevier Inc. All rights reserved.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong><strong>in</strong>tergenic spaces, or are <strong>in</strong> antisense orientation to known genes, <strong>in</strong>dicat<strong>in</strong>g <strong>in</strong>dependenttranscription units. Other genes for miRNA are found <strong>in</strong> <strong>in</strong>tronic regions and could betranscribed as part of the primary transcript for the correspond<strong>in</strong>g gene. Many miRNA alsoform genomic clusters, and can be transcribed as a s<strong>in</strong>gle polycistronic transcript [11,12].A double-stranded RNA (dsRNA)-specific endonuclease RNase III, Drosha, <strong>in</strong> conjunctionwith a dsRNA-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> DGCR8, processes the pri-miRNA <strong>in</strong>to hairp<strong>in</strong> RNAs 70e100-ntlong, called precursor miRNA (pre-miRNA). These pre-miRNA molecules are then transportedfrom the nucleus to the cytoplasm, a process mediated by the nuclear transport receptorexport<strong>in</strong>-5 and the nuclear prote<strong>in</strong> Ran-GTP. In the cytoplasm, GTP is hydrolyzed to guanos<strong>in</strong>ediphosphate (GDP), and the pre-miRNA is released from the transport<strong>in</strong>g complex [13,14].Subsequently, the RNAse III Dicer enzyme cooperates with a dsRNA-b<strong>in</strong>d<strong>in</strong>g partner,transactivat<strong>in</strong>g response RNA-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> (TRBP), and the prote<strong>in</strong> activator of prote<strong>in</strong>90FIGURE 5.1miRNA biogenesis and the mechanism of gene silenc<strong>in</strong>g. The miRNA is transcribed by Pol II <strong>in</strong> the nucleus. The result<strong>in</strong>g pri-miRNA, is then cleaved by Droshaand DGCR8, produc<strong>in</strong>g a pre-miRNA molecule, approximately 70e100 nucleotides <strong>in</strong> length. The pre-miRNA is transported to the cytoplasm by Export<strong>in</strong>-5 andRan-GTP. Here, the pre-miRNA undergoes its f<strong>in</strong>al process<strong>in</strong>g step, which, facilitated by TRBP and PACT <strong>in</strong>volves cleavage by Dicer below the stem-loop. Thisproduces a duplex molecule, conta<strong>in</strong><strong>in</strong>g the s<strong>in</strong>gle-stranded mature miRNA molecule and a miRNA* fragment. The miRNA:miRNA* complex is then <strong>in</strong>corporated<strong>in</strong>to the RNA-<strong>in</strong>duced silenc<strong>in</strong>g complex (RISC), which is activated upon unw<strong>in</strong>d<strong>in</strong>g of the miRNA:miRNA* duplex. Preferentially, the miRNA* fragment is degraded,with the mature miRNA molecule guid<strong>in</strong>g the RISC to the target mRNA. miRNA regulate target genes by base pair<strong>in</strong>g to sites of complementarity <strong>in</strong> the3 0 -UTR and cod<strong>in</strong>g sequence of target mRNA. The degree of complementarity between the seed region of the miRNA and the b<strong>in</strong>d<strong>in</strong>g sites <strong>in</strong> the target mRNAdeterm<strong>in</strong>es the mechanism of regulation. If there is sufficient (near perfect) complementarity, regulation is carried out by RNA <strong>in</strong>terference, whereby the RISCcleaves the target mRNA. If there is <strong>in</strong>sufficient complementarity, regulation is carried out by repression of translation and/or degradation. Degradation <strong>in</strong>volvesdeadenylation of target mRNA followed by decapp<strong>in</strong>g and degradation <strong>in</strong> P-bodies. Translationally repressed targets can also be stored <strong>in</strong> P-bodies. This figureis reproduced <strong>in</strong> the color plate section.


CHAPTER 5MicroRNA <strong>in</strong> Oncogenesisk<strong>in</strong>ase R (PACT) to process the pre-miRNA <strong>in</strong>to a mature form, by cutt<strong>in</strong>g the structure at thebase of the stem-loop. The rema<strong>in</strong><strong>in</strong>g dsRNA duplex structure conta<strong>in</strong>s the mature miRNA aswell as a complementary fragment termed miRNA*. The mature miRNA is s<strong>in</strong>gle-stranded andis loaded <strong>in</strong>to an RNA-<strong>in</strong>duced silenc<strong>in</strong>g complex (RISC). It is the <strong>in</strong>teraction of the miRNA/RISC and its target mRNA that results <strong>in</strong> gene regulation (Figure 5.1).5.3 miRNA-MEDIATED REGULATION OF TARGETSWith<strong>in</strong> the RISC, miRNA <strong>in</strong>teract with target genes via base-pair<strong>in</strong>g. The <strong>in</strong>teraction betweena miRNA and its target mRNA is restricted to the 5 0 end of the miRNA. Sequence complementaritybetween nucleotides 2e8, also known as the “seed region”, is vital for target sequencerecognition [15], although exceptions to this rule have been demonstrated [16,17]. Mostcommonly, miRNA b<strong>in</strong>d<strong>in</strong>g sites are present <strong>in</strong> the 3 0 -untranslated region (UTR) of targetmRNAs, usually <strong>in</strong> multiple copies [18]. However, miRNA have also been demonstrated to targetthe 5 0 -UTR and cod<strong>in</strong>g regions of mRNA [19,20]. A study by Tay et al. demonstrated thata network of miRNA can b<strong>in</strong>d to multiple sites with<strong>in</strong> the cod<strong>in</strong>g and 3 0 -UTR of a s<strong>in</strong>gle mRNAtarget, add<strong>in</strong>g to the complexity of miRNA-mediated target regulation [21]. The degree ofcomplementarity between the seed region of the miRNA and the b<strong>in</strong>d<strong>in</strong>g site <strong>in</strong> the target mRNAdeterm<strong>in</strong>es the mechanism by which the miRNA regulates the target [22]. If the miRNA baressufficient sequence complementarity (near perfect) to the target mRNA, then regulation is carriedout by a process called RNA <strong>in</strong>terference, whereby the RISC is directed to cleave the target mRNA[22]. If there is <strong>in</strong>sufficient complementarity, which is generally the case <strong>in</strong> mammals [15],regulation is achieved by repression of translation and/or destabilization of the mRNA [23].The core components of the RISC are the Argonaute (Ago) family of prote<strong>in</strong>s, which play a keyrole <strong>in</strong> its function [24]. All four mammalian Ago prote<strong>in</strong>s (Ago1eAgo4) can direct thetranslational repression of a target mRNA, however, only Ago2 possesses “slicer” activity, and isresponsible for cleav<strong>in</strong>g target mRNA [25]. The exact mechanism(s) of miRNA-mediatedtranslational repression of target genes is still uncerta<strong>in</strong> [26e28]. Several studies have providedevidence that translational repression occurs pre-<strong>in</strong>itiation of translation [27e29]. However,other studies suggest that repression occurs post-<strong>in</strong>itiation of translation [26,30,31]. It was<strong>in</strong>itially thought that miRNA-mediated repression of target genes was predom<strong>in</strong>antly reflectedat the prote<strong>in</strong> level, with no or m<strong>in</strong>imal effect on mRNA levels. However, it has now beendemonstrated that miRNA-mediated repression of target genes is frequently associated withdestabilization of mRNA, although it is not known if this is a secondary effect of translationalrepression. miRNA-mediated degradation of mRNA targets <strong>in</strong>volves deadenylation (removalof the Poly A tail), followed by decapp<strong>in</strong>g and exonucleolytic digestion [32e34]. In addition,process<strong>in</strong>g bodies (P-bodies), cytoplasmic structures <strong>in</strong>volved <strong>in</strong> the storage and degradationof mRNA, are also thought to play a role <strong>in</strong> miRNA regulation [35,36]. miRNA are thought toguide target mRNA and associated RISC prote<strong>in</strong>s to these storage structures, which areenriched for mRNA degradation and translational repression factors [27,36]. The mechanismsdictat<strong>in</strong>g whether a target mRNA follows the degradation or translational repression pathwayare presently unknown. Add<strong>in</strong>g to the complexity of miRNA-mediated regulation are therecent discoveries that under different stress conditions miRNA-<strong>in</strong>duced repression of targetscan be reversed [37] and that miRNA can activate translation of target mRNA [38].91miRNA-mediated regulation appears to be an extremely dynamic process, its complexity is<strong>in</strong>creased by the fact that perfect complimentarity to the target is not required for regulation.This <strong>in</strong>dicates that a s<strong>in</strong>gle miRNA has the potential to regulate multiple target genes. Inaddition, a network of miRNA can function simultaneously to regulate a s<strong>in</strong>gle mRNA. Thisultimately makes <strong>in</strong> silico identification of target genes and the elucidation of miRNA functionmuch more difficult.The seed region, located at positions 2e7 from the 5 0 end of the miRNA, is employed by theRISC as a nucleation signal for recogniz<strong>in</strong>g target mRNA [15,39,40]. On the mRNA the


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>correspond<strong>in</strong>g sites are referred to as “seed sites”. There are a number of str<strong>in</strong>gencies associatedwith target seed site recognition and b<strong>in</strong>d<strong>in</strong>g [41]. A str<strong>in</strong>gent seed site has perfect WatsoneCrick b<strong>in</strong>d<strong>in</strong>g and can be divided <strong>in</strong>to four “seed” types: 8mer, 7mer-m8, 7mer-A1, and 6mer[41]. Each of these types differs depend<strong>in</strong>g on the comb<strong>in</strong>ation of the nucleotide of position 1and pair<strong>in</strong>g at position 8. 8mer has both an aden<strong>in</strong>e at position 1 of the mRNA target site andbase-pair<strong>in</strong>g at position 8. An aden<strong>in</strong>e on the target site correspond<strong>in</strong>g to position 1 ofa miRNA is known to <strong>in</strong>crease efficiency of target recognition [3]. 7mer-A1 has an aden<strong>in</strong>e atposition 1 only, while 7mer-m8 has base-pair<strong>in</strong>g at position 8 only. By contrast, 6mer hasneither an aden<strong>in</strong>e at position 1 nor base-pair<strong>in</strong>g at position 8 [42].In addition to str<strong>in</strong>gent seed recognition, moderately str<strong>in</strong>gent recognition is also possible,as the RISC can tolerate small mismatches or wobble pair<strong>in</strong>g with<strong>in</strong> the seed region. Thethermodynamic stability of a wobble pair<strong>in</strong>g (such as G:U) is comparable to that ofa WatsoneCrick pair<strong>in</strong>g [41,43].WatsoneCrick pair<strong>in</strong>g <strong>in</strong> the 3 0 part of the miRNA molecule is known to enhance the siterecognition efficacy <strong>in</strong> miRNA targets that have seed pair<strong>in</strong>g [42]. The preferable nucleotidenumber of matches <strong>in</strong> the 3 0 part differs between the site that has str<strong>in</strong>gent-seed pair<strong>in</strong>g and theone that has moderate-str<strong>in</strong>gent-seed pair<strong>in</strong>g [41]. Str<strong>in</strong>gent seeds require 3e4 matches <strong>in</strong> thepositions 13e16, whereas moderate-str<strong>in</strong>gent-seeds require 4e5 matches <strong>in</strong> positions 13e19.Sites with this additional 3 0 pair<strong>in</strong>g are called 3-supplementary and 3 0 compensatory sites [44].92It has been extensively demonstrated that the vast majority of miRNA target recognitionsequences are found <strong>in</strong> the 3 0 -UTR of the target gene, even though miRNA-loaded RISC can <strong>in</strong>theory b<strong>in</strong>d any segment of mRNA. Target genes generally have longer 3 0 UTR, whereas certa<strong>in</strong>ubiquitous genes, such as house-keep<strong>in</strong>g genes, tend to have short 3 0 UTR, potentially to avoidbe<strong>in</strong>g regulated by miRNA [45]. Target sites are not evenly distributed with 3 0 UTR. They arelocated near both ends on long 3 0 UTR (generally 2000 nt). For shorter 3 0 UTR, target sites tendto be ~15e20 nt away from the stop codon [42,43].While it is generally considered that functional miRNA sites are preferentially located <strong>in</strong> 3 0UTR, seed sites <strong>in</strong> the cod<strong>in</strong>g sequence and 5 0 UTR regions can also promote mRNA downregulation[46,47]. The basis for preferential miRNA b<strong>in</strong>d<strong>in</strong>g <strong>in</strong> the 3 0 UTR may have a numberof explanations. For example, the RISC may need to compete with other prote<strong>in</strong> complexes,such as ribosomes, b<strong>in</strong>d<strong>in</strong>g to the cod<strong>in</strong>g sequence and translation <strong>in</strong>itiation complexes <strong>in</strong> the5 0 UTR. As such the 3 0 UTR might simply be more accessible for long-term b<strong>in</strong>d<strong>in</strong>g than theother two sites [41,48].5.4 miRNA AND CANCERmiRNA are <strong>in</strong>volved <strong>in</strong> the regulation of important cellular pathways, such as proliferation[49], cell death [50], angiogenesis [51], <strong>in</strong>vasion and metastasis [52], the deregulation ofwhich are all hallmarks of cancer [53]. Thus, it is not surpris<strong>in</strong>g that aberrant miRNAexpression has been demonstrated <strong>in</strong> many different cancers.The l<strong>in</strong>k between miRNA and cancer was first highlighted by Cal<strong>in</strong> and colleagues [54], whenit was discovered that two miRNA, miR-15a and miR-16-1, are located <strong>in</strong> a region on chromosome13 that is deleted <strong>in</strong> over 65% of chronic lymphocytic leukemia (CLL) patients.Despite extensive profil<strong>in</strong>g, no cancer-associated genes had been identified with<strong>in</strong> this region,suggest<strong>in</strong>g that miR-15a and miR-16-1 were the genomic targets of this frequent deletion.Interest<strong>in</strong>gly, this deletion is present <strong>in</strong> almost all cases of <strong>in</strong>dolent CLL, suggest<strong>in</strong>g that downregulationof miR-15a and miR-16-1 is an early event <strong>in</strong> the pathogenesis of this disease.Support<strong>in</strong>g this, a subsequent study identified the proto-oncogene Bcl2, which is overexpressed<strong>in</strong> CLL, as a target of miR-15a and miR-16-1 [55]. This suggests that the downregulationof these two miRNA <strong>in</strong> CLL provides a mechanism for oncogenesis via the <strong>in</strong>creased


CHAPTER 5MicroRNA <strong>in</strong> Oncogenesisexpression of Bcl2. Further support<strong>in</strong>g a tumor suppressor role for miR-15a and miR-16-1,ectopic expression of both miRNA was demonstrated to prevent tumor growth <strong>in</strong> a xenograftmodel of leukemia [56].A role for miRNA <strong>in</strong> cancer was further supported by several studies that performed genomewideprofil<strong>in</strong>g of miRNA expression <strong>in</strong> multiple primary tumors and cell l<strong>in</strong>es. Vol<strong>in</strong>ia andcolleagues [57] profiled miRNA expression <strong>in</strong> over 500 samples of both normal and tumortissue taken from breast, colon, lung, pancreatic, prostate, and stomach. miRNA expression wasdemonstrated to be significantly dysregulated <strong>in</strong> malignant tissue, demonstrat<strong>in</strong>g a commonmiRNA expression profile <strong>in</strong> solid cancer. A study by Lu et al., profiled the expression of 217miRNA <strong>in</strong> 334 samples comprised of normal non-cancer tissue, primary tumors, and tumorcell-l<strong>in</strong>es, from 20 different cancer types. miRNA profiles were demonstrated to accuratelydist<strong>in</strong>guish normal from malignant tissue, separate cancer type, categorize differentiation stateand cluster samples accord<strong>in</strong>g to their embryonic l<strong>in</strong>eage. Importantly, miRNA profiles weredemonstrated to be more accurate at classify<strong>in</strong>g poorly differentiated tumors than mRNA [58],suggest<strong>in</strong>g a potential role as diagnostic biomarkers. Unique miRNA profiles have now beendemonstrated for all cancers studied to date, these <strong>in</strong>clude breast [59], esophageal [60], colorectal[61], lung [62], prostate [63], and gastric [64], support<strong>in</strong>g a role for miRNA <strong>in</strong> thedevelopment of cancer and suggest<strong>in</strong>g their potential use as diagnostic biomarkers.In addition to their role <strong>in</strong> the development of cancer, evidence also suggests a role for miRNA<strong>in</strong> cancer progression. Metastasis, the process by which cancer cells dissem<strong>in</strong>ate from theprimary tumor site and establish secondary tumors at distant sites, is the predom<strong>in</strong>ant cause ofcancer-related deaths. Dysregulation of miRNA has been demonstrated <strong>in</strong> metastasis, withboth pro- and antimetastatic miRNA identified. miR-10b is highly expressed <strong>in</strong> metastaticbreast cancer. Overexpression of miR-10b <strong>in</strong>creases cell motility and <strong>in</strong>vasiveness, effects thatare reversed upon <strong>in</strong>hibition of expression, both <strong>in</strong> vitro [65] and <strong>in</strong> vivo [66], suggest<strong>in</strong>ga prometastatic role for miR-10b. Conversely, the miR-200 family negatively regulatesepithelialemesenchymal transition (EMT), which is thought to facilitate metastasis via<strong>in</strong>creased cell motility. The miR-200 family, which is repressed dur<strong>in</strong>g EMT, was demonstratedto target the E-cadher<strong>in</strong> repressors ZEB1 and ZEB2, result<strong>in</strong>g <strong>in</strong> <strong>in</strong>creased E-cadher<strong>in</strong> expressionand <strong>in</strong>hibition of EMT [67], suggest<strong>in</strong>g an antimetastatic function for this miRNA family.93In addition, specific miRNA profiles have also been demonstrated to predict progression andoutcome <strong>in</strong> a number of cancers. In breast cancer, dysregulation of miR-145 and miR-21 wasassociated with tumor progression, whilst reduced let-7 expression was associated with<strong>in</strong>creased lymph node metastasis and proliferation capacity [59]. A 9-miRNA signature wasassociated with time to progression <strong>in</strong> CLL [68]. In colorectal cancer, miR-21 expression isassociated with tumor stage, <strong>in</strong>vasion, and prognosis [69]. Whilst <strong>in</strong> lung cancer, reduced let-7was <strong>in</strong>dicative of a poor prognosis <strong>in</strong> two <strong>in</strong>dependent studies [62,70]. The demonstrated roleof miRNA <strong>in</strong> cancer progression highlights their potential as both novel therapeutics for thetreatment of cancer and as prognostic markers.5.5 OncomiRsWhilst miRNA profiles <strong>in</strong> cancer are generally tumor-specific, several miRNA are dysregulatedacross multiple cancers, suggest<strong>in</strong>g a common role <strong>in</strong> tumorigenesis. miRNA that have beenshown to be down-regulated <strong>in</strong> cancers, such as miR-15a, miR-16-1, and the let-7 family, havebeen proposed to be tumor suppressors, whilst up-regulated miRNA, such as miR-21 and themiR-17-92 cluster, have been classified as oncogenes.5.5.1 miRNA as Tumor SuppressorsIn addition to their tumor suppressor role <strong>in</strong> CLL, a potential anticancer role for miR-15a andmiR-16-1 has also been demonstrated <strong>in</strong> several other cancers such as lung [71], pituitary [72],


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>and ovarian [73]. In prostate cancer [74], miR-15a and miR-16-1 were demonstrated to bedown-regulated <strong>in</strong> 80% of tumors. Inhibition of miR-15a and miR-16-1 promoted survival,growth, <strong>in</strong>vasiveness, and tumorigenicity <strong>in</strong> untransformed prostate cells, suggest<strong>in</strong>g downregulationof miR-15a and miR-16-1 as an early event <strong>in</strong> the pathogenesis of prostate cancer.Conversely, overexpression of miR-15a and miR-16-1 reduced growth, <strong>in</strong>duced apoptosis andtumor regression <strong>in</strong> a prostate cancer xenograft model. This study identified the oncogenesCCND1 and WNT3A as direct targets, all of which promote cell growth, survival, and<strong>in</strong>vasiveness, suggest<strong>in</strong>g a mechanism for tumor development <strong>in</strong> prostate tumors deficient <strong>in</strong>miR-15a and miR-16-1.The let-7 family, which consists of 12 closely related miRNA, is also a potential tumorsuppressor miRNA that is frequently down-regulated <strong>in</strong> cancer. A study by Takamizawa andcolleagues was the first to identify a potential tumor suppressor role for let-7 [70], demonstrat<strong>in</strong>gthe down-regulation of let-7 <strong>in</strong> lung cancer, with low expression also associated witha shorter postoperative survival time. Overexpression of let-7 significantly <strong>in</strong>hibited growth <strong>in</strong>lung cancer cells, suggest<strong>in</strong>g a functional role for let-7 <strong>in</strong> modulat<strong>in</strong>g tumorigenesis. This issupported by several studies that have demonstrated the antiproliferative effects of let-7 <strong>in</strong> lung[4], thyroid [75], and prostate [76] cancer. Furthermore, let-7 has been demonstrated to <strong>in</strong>hibittumor growth <strong>in</strong> vivo [77,78], further support<strong>in</strong>g a tumor suppressor role. The effects of let-7on cell growth may be expla<strong>in</strong>ed by its negative regulation of several oncogenes, such asRas [5],myc[79], HMGA2 [80], and cell cycle regulators, <strong>in</strong>clud<strong>in</strong>g Cdc25A, CDK6, and Cycl<strong>in</strong>D2 [4]. This suggests that the frequent down-regulation of let-7 seen <strong>in</strong> multiple cancersprovides a mechanism for tumor development and progression via <strong>in</strong>creased expression ofthese gene targets.945.5.2 miRNA as OncogenesmiR-21 was one of the first miRNA identified <strong>in</strong> humans [8]. Overexpression of miR-21 hasbeen demonstrated <strong>in</strong> multiple cancers, such as glioblastoma [50], breast [59], esophageal[81], andCLL[82], suggest<strong>in</strong>g an oncogenic role for this miRNA. Several studies haveprovided evidence for miR-21 as an antiapoptotic factor. In glioblastoma and breast cancercells, knockdown of miR-21 <strong>in</strong>hibited cell growth and <strong>in</strong>duced apoptosis [50,83], suggest<strong>in</strong>gthat overexpression of miR-21 may promote tumor development via the negative regulationof proapoptotic factors. The oncogenic properties of miR-21 are mediated, at least <strong>in</strong> part, viaits negative regulation of several important tumor suppressor genes. Several studies havedemonstrated miR-21 regulation of the tumor suppressor prote<strong>in</strong>s programmed cell death 4(PDCD4) [84e86] phosphatase and tens<strong>in</strong> homolog (PTEN) [87] and tropomyos<strong>in</strong> 1(TMP1) [88]. In addition, miR-21-mediated regulation of all three tumor suppressors isassociated with <strong>in</strong>creased <strong>in</strong>vasion and metastasis, suggest<strong>in</strong>g a role for miR-21 <strong>in</strong> cancerprogression. miR-21 has also been implicated <strong>in</strong> the resistance of cancer cells to variouschemotherapeutics. miR-21 was demonstrated to modulate sensitivity to the chemotherapeuticagent gemcitab<strong>in</strong>e <strong>in</strong> cholangiocarc<strong>in</strong>oma [89], doxorubic<strong>in</strong> <strong>in</strong> bladder cancer [90],and 5-fluorouracil <strong>in</strong> colorectal cancer [91], suggest<strong>in</strong>g an additional role for miR-21 <strong>in</strong>oncogenesis. The role of miR-21 <strong>in</strong> both the development and progression of cancer, <strong>in</strong>addition to the response to anticancer treatment, highlights the potential of miR-21 as a noveltherapeutic target.The polycistronic cluster miR-17-92, is a family of homologous miRNA that are transcribedas a s<strong>in</strong>gle pri-miRNA and then processed to produce seven mature miRNA molecules(miR-17-5p, miR-17-3p, miR-18, miR-19a, miR-20, miR-19b-1, and miR-92-1) [92]. Thecluster is located on chromosome 13, at a region commonly amplified <strong>in</strong> a number ofhematopoietic malignancies and solid tumors [93e95]. The oncogenic potential of miR-17-92was first highlighted <strong>in</strong> a mouse model of B-cell lymphoma [94]. miR-17-92 was demonstratedto accelerate c-myc-<strong>in</strong>duced lymphoma development, produc<strong>in</strong>g tumors with a more


CHAPTER 5MicroRNA <strong>in</strong> Oncogenesisaggressive phenotype. These tumors also demonstrated lower apoptosis, suggest<strong>in</strong>g that therole of miR-17-92 <strong>in</strong> tumor development may be via antiapoptotic mechanisms. This wassupported by the discovery that the proapoptotic factors PTEN and Bim are both direct targetsof miR-17-92 [96]. miR-17-92 is also overexpressed <strong>in</strong> lung cancer where it was demonstratedto enhance cellular proliferation, whilst <strong>in</strong>hibition of miR-17-92 <strong>in</strong>duced apoptosis [93,97],further support<strong>in</strong>g an antiapoptotic role. The negative regulation of the cell cycle regulator p21by miR-17-92 [98] may expla<strong>in</strong> the effects of this polycistron on proliferation. Interest<strong>in</strong>gly,miR-17-92 has been demonstrated to be directly <strong>in</strong>duced by the oncogene c-myc [99], which isfrequently up-regulated <strong>in</strong> cancer, further support<strong>in</strong>g an oncogenic role for miR-19-72.Surpris<strong>in</strong>gly, loss of the genomic region encod<strong>in</strong>g the miR-17-92 cluster has also beenl<strong>in</strong>ked to malignancy <strong>in</strong> hepatocellular cancer [100]. This potential tumor suppressor role ofmiR-17-92 may be expla<strong>in</strong>ed by its negative regulation of the transcription factor E2F1 [99].E2F1 is <strong>in</strong>duced by c-myc and promotes cell cycle progression [101], it also forms a positivefeedback loop by <strong>in</strong>duc<strong>in</strong>g c-myc expression [102]. Thus, the repression of E2F1 by miR-17-92provides a mechanism for <strong>in</strong>hibition of c-myc-mediated growth. The down-regulation of miR-17-92 <strong>in</strong> hepatocellular cancer may therefore provide a mechanism for tumor development.This suggests that miR-17-92 may have dual tumor suppressor and oncogenic roles <strong>in</strong> a tissue/tumor-dependent manner.5.6 MECHANISMS OF miRNA DEREGULATIONSeveral mechanisms are thought to contribute either alone or <strong>in</strong> comb<strong>in</strong>ation to the dysregulationof miRNA <strong>in</strong> human cancer. These <strong>in</strong>clude chromosomal alterations, dysregulatedtranscriptional activation, epigenetic modifications, and alterations <strong>in</strong> biogenesis. However,the exact mechanism(s) direct<strong>in</strong>g the tumor/tissue-specific dysregulation of miRNA <strong>in</strong> carc<strong>in</strong>ogenesisis still unknown.955.6.1 Chromosomal AberrationsFollow<strong>in</strong>g the <strong>in</strong>itial study which demonstrated the deletion of miR-15a and miR-16-1 <strong>in</strong> CLL,Cal<strong>in</strong> and colleagues [6] used a computational approach to identify the genomic location of186 miRNA. Over 50% of the miRNA <strong>in</strong>vestigated were demonstrated to be located at fragileregions, regions that commonly undergo deletion, amplification or translocation events orgenomic regions associated with human cancers, such as viral DNA <strong>in</strong>tegration sites,suggest<strong>in</strong>g a role for altered miRNA <strong>in</strong> the development of cancer. Interest<strong>in</strong>gly, profil<strong>in</strong>g ofseveral of these sites of cancer-associated chromosomal alterations had previously failed toidentify prote<strong>in</strong>-cod<strong>in</strong>g tumor suppressors or oncogenes implicated <strong>in</strong> the development of thedisease, suggest<strong>in</strong>g that genomic alterations of miRNA may be a causal factor <strong>in</strong> tumorigenesis.Several studies have s<strong>in</strong>ce experimentally validated the non-random genomic location ofmiRNA <strong>in</strong> cancer, demonstrat<strong>in</strong>g significant chromosomal alterations at miRNA-encodedregions across multiple cancer types. A study by Zhang et al. [103] used array-basedcomparative genomic hybridization to profile the genomic loci of 283 miRNA <strong>in</strong> over 200samples of breast, ovarian, and melanoma primary tumors and cancer cell l<strong>in</strong>es. A highproportion of miRNA gene copy number alterations were demonstrated <strong>in</strong> all three cancertypes, with both dist<strong>in</strong>ct and common alterations observed. Furthermore, a strong correlationbetween miRNA copy number alterations and miRNA transcript levels was demonstrated both<strong>in</strong> cancer cell l<strong>in</strong>es and a previously published <strong>in</strong>dependent set of breast tumor samples [59],further support<strong>in</strong>g genomic alterations as a mechanism for miRNA dysregulation. Anotherstudy profil<strong>in</strong>g miRNA expression <strong>in</strong> over 200 normal tissues and the NCI-60 panel of cancercell l<strong>in</strong>es, identified potential tumor suppressor and oncogenic miRNA, which are encoded atknown cancer-associated genomic regions [104].


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>Both genomic loss and ga<strong>in</strong> have been demonstrated to result <strong>in</strong> altered expression of miRNA.The region encod<strong>in</strong>g miR-15a and miR-16-1 is deleted <strong>in</strong> more than 50% of B-cell chroniclymphocytic leukemia cases [54]. In contrast, the miR-17-92 cluster is encoded <strong>in</strong> an 800-bpregion of the non-prote<strong>in</strong> cod<strong>in</strong>g gene C13, which is frequently amplified <strong>in</strong> B-cell lymphomaand lung cancer [93,94]. Increased gene copy number has been demonstrated to correlate withoverexpression of the cluster <strong>in</strong> both cancer types, support<strong>in</strong>g chromosomal amplifications asa mechanism for dysregulation of miR-17-92. Chromosomal translocations also contribute tomiRNA dysregulation. A t(8;17) translocation that juxtaposes the oncogene c-myc to chromosome17 result<strong>in</strong>g <strong>in</strong> its overexpression, is associated with an aggressive form of B-cellleukemia [105]. The underly<strong>in</strong>g mechanism of c-myc up-regulation was unknown until it wasdemonstrated that this translocation positions c-myc at the promoter region of miR-142,which is encoded 50 nt from this chromosomal break [6]. This suggests the <strong>in</strong>volvement ofmiR-142 regulatory elements <strong>in</strong> the overexpression of c-myc. A translocation at chromosome12 disrupts the HMGA2 gene, result<strong>in</strong>g <strong>in</strong> the generation of a truncated version of HMGA2,which is overexpressed <strong>in</strong> lung cancer [106]. The up-regulation of HMGA2 has been attributedto loss of let-7 b<strong>in</strong>d<strong>in</strong>g sites with<strong>in</strong> the 3 0 -UTR of HMGA2, caused by the translocation. Indeed,disruption of let-7-mediated regulation of HMGA2 has been demonstrated to promoteoncogenic transformation [80,107], support<strong>in</strong>g chromosomal translocation as a mechanismfor dysregulation of important tumor suppressor miRNA.96Both dist<strong>in</strong>ct and common miRNA copy alterations have been demonstrated <strong>in</strong> oncogenesis,suggest<strong>in</strong>g that genomic alterations of miRNA play a functional role <strong>in</strong> malignancy, rather thanbe<strong>in</strong>g a mere by-product. Whilst the miR-17-92 cluster loci is frequently amplified <strong>in</strong>lymphoma and lung cancer, it has been demonstrated to be deleted <strong>in</strong> ovarian, breast, andmelanoma tumors [103], suggest<strong>in</strong>g tumor-specific copy number alterations. In contrast,overexpression of miR-21, which has been demonstrated <strong>in</strong> multiple malignancies, is thoughtto be due to amplification, suggest<strong>in</strong>g a common mechanism of dysregulation.5.6.2 Dysregulation of Transcription FactorsAs the majority of miRNA are transcribed by Pol II, transcriptional control is an importantmechanism regulat<strong>in</strong>g miRNA expression. Many transcription factors are dysregulated <strong>in</strong>cancer, result<strong>in</strong>g <strong>in</strong> the altered expression of miRNA, which may promote tumorigenesis.The transcription factor c-myc is <strong>in</strong>volved <strong>in</strong> the regulation of approximately 15% of humangenes, regulat<strong>in</strong>g cell death, proliferation, and differentiation [108] via both positive andnegative regulation of gene expression. c-myc is frequently overexpressed <strong>in</strong> human malignancies,with almost all cases of Burkitt’s lymphoma caused by the juxtaposition of c-myc withimmune regulatory elements, which results <strong>in</strong> hyperactivation [109]. A study by O’Donnelland colleagues [99] demonstrated that the oncogenic miR-17-92 miRNA cluster located onchromosome 13 is a transcriptional target of c-myc. c-myc b<strong>in</strong>ds directly to the genomic locusof the miR-17-92 cluster, activat<strong>in</strong>g transcription. Co-expression of c-myc and the miR-17-92polycistron accelerated oncogenesis <strong>in</strong> a mur<strong>in</strong>e model of B-cell lymphoma [94], support<strong>in</strong>gc-myc-mediated activation of miR-17-92 as a mechanism for tumorigenesis. Add<strong>in</strong>g to thecomplexity of c-myc-mediated miRNA deregulation, <strong>in</strong>duction of c-myc has also beendemonstrated to result <strong>in</strong> the widespread repression of miRNA expression <strong>in</strong> B-cell lymphoma[110]. Many of these c-myc-repressed miRNA (<strong>in</strong>clud<strong>in</strong>g miR-15a, miR-16a, miR-34a, let-7a-1,and let-7f-1), have been demonstrated to be down-regulated <strong>in</strong> human cancers and haveknown tumor suppressor activity, suggest<strong>in</strong>g c-myc-mediated repression as a mechanism foroncogenic transformation. Similar to the direct activation of miR-17-92, c-myc was demonstratedto b<strong>in</strong>d directly to the promoters of several of these down-regulated miRNA, suggest<strong>in</strong>gdirect repression of transcription. Additionally, c-myc has been demonstrated to modulatemiRNA expression via <strong>in</strong>direct mechanisms, as <strong>in</strong> the case of the let-7 family, <strong>in</strong> which c-myc<strong>in</strong>hibits biogenesis via activation of the RNA-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> L<strong>in</strong>-28B [111].


CHAPTER 5MicroRNA <strong>in</strong> OncogenesisThe tumor suppressor p53 regulates the expression of a complex network of genes via bothtranscriptional activation and repression. In response to cellular stresses, such as DNA damage,hypoxia, mitotic sp<strong>in</strong>dle damage, and activation of oncogenes, p53 becomes stabilized andorchestrates cell cycle arrest, apoptosis or senescence, thereby ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g genomic <strong>in</strong>tegrity.Consequently, dysfunction of p53 is considered to be an early event <strong>in</strong> tumorigenesis. This ishighlighted by the fact that p53 is mutated <strong>in</strong> over 50% of human cancers [112]. Several studieshave demonstrated a miRNA component to the p53 network, with the miR-34 family, whichconsist of miR-34a, miR-34b, and miR-34c, identified as direct transcriptional targets of p53[113]. p53 b<strong>in</strong>ds to responsive elements <strong>in</strong> the promoter region, <strong>in</strong>duc<strong>in</strong>g expression. Ectopicexpression of all three miR-34 members has been demonstrated to have antiproliferativeeffects, <strong>in</strong>duc<strong>in</strong>g cell cycle arrest and apoptosis. Consistent with the tumor suppressor effects ofmiR-34 is the f<strong>in</strong>d<strong>in</strong>g that miR-34 is down-regulated <strong>in</strong> a number of human cancers. Thus,p53-mediated activation of miR-34 provides a novel mechanism for tumor suppressor activity,and suggests that dysregulation of this pathway may provide a mechanism for oncogenictransformation. Several other p53-regulated miRNA have s<strong>in</strong>ce been identified, these <strong>in</strong>clude,miR-107 [114], miR-145 [115], miR-192, and miR-215 [116]. In addition, p53 may also be<strong>in</strong>volved <strong>in</strong> the regulation of miRNA process<strong>in</strong>g [117], suggest<strong>in</strong>g an additional mechanism formiRNA dysregulation <strong>in</strong> oncogenesis. Interest<strong>in</strong>gly, both p53 and myc have been demonstratedto be direct targets of miRNA [118,119], add<strong>in</strong>g to the complexity of the transcriptionalregulation of miRNA.5.6.3 Epigenetic AlterationsEpigenetic modifications describe heritable and reversible changes <strong>in</strong> chromat<strong>in</strong> that do notalter the orig<strong>in</strong>al DNA sequence. The most frequently studied epigenetic changes <strong>in</strong> cancer aredifferential methylation of DNA and histone modifications. Methylation of CpG islands,which are associated with the promoter regions of genes, results <strong>in</strong> transcriptional silenc<strong>in</strong>g.Aberrant methylation that results <strong>in</strong> silenc<strong>in</strong>g of tumor suppressor genes is thought tocontribute to tumorigenesis. Conversely, a reduction <strong>in</strong> global methylation levels (hypomethylation)is also associated with cancer. Similarly, post-translational modifications ofhistones, such as acetylation, methylation, and phosphorylation play an important role <strong>in</strong> theregulation of gene expression. Consequently, alterations <strong>in</strong> histone modification patterns,often <strong>in</strong> comb<strong>in</strong>ation with dysregulated hypermethylation, are commonly demonstrated <strong>in</strong>carc<strong>in</strong>ogenesis.97Similar to prote<strong>in</strong>-cod<strong>in</strong>g genes, miRNA have also been demonstrated to be subject toepigenetic regulation. Indeed, over 50% of miRNA genes are associated with CpG islands,suggest<strong>in</strong>g the potential for epigenetic modifications [120]. Saito and colleagues [121]demonstrated that treatment of bladder cancer cells with both the demethyltransferase<strong>in</strong>hibitor 5-aza-2 0 -deoxycytid<strong>in</strong>e and the histone deacetylase <strong>in</strong>hibitor 4-phenylbutyric acid,resulted <strong>in</strong> the reactivation of expression of 17 miRNA, suggest<strong>in</strong>g that these miRNA wereunder epigenetic control. One of these miRNA, miR-127, which is located with<strong>in</strong> a CpG island,was demonstrated to be down-regulated <strong>in</strong> tumor tissue and cell l<strong>in</strong>es across multiple cancertypes, when compared to normal tissue, suggest<strong>in</strong>g that epigenetic regulation may providea mechanism for repression of miR-127 <strong>in</strong> tumorigenesis. Interest<strong>in</strong>gly, both methylation andhistone deacteylation were demonstrated to be <strong>in</strong>volved <strong>in</strong> transcriptional repression of miR-127, as <strong>in</strong>duction of expression was only evident follow<strong>in</strong>g both demethylat<strong>in</strong>g and deacetylase<strong>in</strong>hibition treatment. This <strong>in</strong>duction of miR-127 expression was concomitant witha decrease <strong>in</strong> expression of the miR-127-predicted target, the proto-oncogene BCL6, suggest<strong>in</strong>ga tumor suppressor role for miR-127. S<strong>in</strong>ce then, a plethora of studies have demonstratedepigenetic control of a number of miRNA us<strong>in</strong>g several different methods. A study by Scottet al. [122] demonstrated significant alterations <strong>in</strong> miRNA expression <strong>in</strong> breast cancer cellsfollow<strong>in</strong>g histone deacetylase <strong>in</strong>hibition, suggest<strong>in</strong>g that histone modifications alone canregulate miRNA expression. Lujambio and colleagues [123] used a DNA methyltransferase-


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>deficient cell l<strong>in</strong>e model of colon cancer to <strong>in</strong>vestigate epigenetically silenced miRNA,demonstrat<strong>in</strong>g hypermethylation of miR-124a. Silenc<strong>in</strong>g of miR-124a was associated with the<strong>in</strong>creased expression of target gene CDK6, which plays a demonstrated role <strong>in</strong> oncogenesis,support<strong>in</strong>g a tumor suppressor role for miR-124a. Interest<strong>in</strong>gly, hypermethylation of miR-124a was demonstrated across multiple tumor and cancer cell l<strong>in</strong>es <strong>in</strong>clud<strong>in</strong>g breast, lung, andlymphoma, suggest<strong>in</strong>g a common mechanism of dysregulation <strong>in</strong> tumorigenesis. This wassupported <strong>in</strong> acute lymphoblastic leukemia (ALL), where methylation of miR-124a wasdemonstrated <strong>in</strong> 59% of patients, and was significantly associated with a poor prognosis [124].The epigenetic silenc<strong>in</strong>g of miR-124a <strong>in</strong> ALL was associated with <strong>in</strong>creased cell growth, whichwas mediated by the miR-124a target CDK6 [124], suggest<strong>in</strong>g methylation-<strong>in</strong>duced silenc<strong>in</strong>gof tumor suppressor miRNA as a mechanism for tumorigenesis.In contrast, up-regulation of miRNA via hypomethylation has also been demonstrated.Demethylat<strong>in</strong>g treatment of ovarian cancer cell l<strong>in</strong>es <strong>in</strong>duced expression of miR-21, miR-203,and miR-205, all of which were demonstrated to be up-regulated <strong>in</strong> ovarian tumors, whencompared to normal tissue [125]. Given the known oncogenic properties of miR-21 and its upregulation<strong>in</strong> multiple cancers, these data suggest hypomethylation as a potential mechanismfor overexpression of oncogenic miRNA. Whilst epigenetic regulation of miRNA does appearto be a mechanism for dysregulation of miRNA <strong>in</strong> cancer, evidence does suggest that epigeneticcontrol of miRNA expression is a cell-specific effect. For example, whilst demethylation andhistone deacetylase <strong>in</strong>hibition reactivated miR-127 <strong>in</strong> colon, cervical, and lymphoma cell l<strong>in</strong>es[121], expression was not <strong>in</strong>duced <strong>in</strong> MCF-7 breast cancer or CALU-1 lung cancer cell l<strong>in</strong>es.98Taken together these data suggest epigenetic regulation as a major mechanism of alteredmiRNA expression <strong>in</strong> cancer. The additional discovery that several miRNA negatively regulatecomponents of the epigenetic regulation mach<strong>in</strong>ery [126], only adds to the complexity of theepigenetic-miRNA network.5.6.4 Altered Process<strong>in</strong>gpri-miRNA molecules undergo a co-ord<strong>in</strong>ated multistep process to produce a mature functionalmiRNA. Alterations <strong>in</strong> the process<strong>in</strong>g of miRNA that affect either miRNA maturationand/or the <strong>in</strong>teraction of miRNA and gene targets are therefore likely to play a role <strong>in</strong> thedysregulation of miRNA <strong>in</strong> cancer.A potential role for altered miRNA process<strong>in</strong>g <strong>in</strong> cancer was first highlighted by Kumar andcolleagues [127], who demonstrated that knockdown of Drosha, Dicer, and DGCR8 resulted<strong>in</strong> a global decrease <strong>in</strong> miRNA levels, which enhanced proliferation and transformation <strong>in</strong>cancer cells. Several other studies have s<strong>in</strong>ce demonstrated altered expression of miRNAprocess<strong>in</strong>g mach<strong>in</strong>ery <strong>in</strong> multiple cancer types. A study <strong>in</strong> prostate cancer demonstrated theup-regulation of Dicer and several other components of the miRNA process<strong>in</strong>g pathway [128],suggest<strong>in</strong>g a mechanism for the up-regulation of miRNA previously identified <strong>in</strong> prostatetumors [57]. The up-regulation of Dicer was also demonstrated <strong>in</strong> colorectal cancer [129],where <strong>in</strong>creased expression was significantly associated with disease progression and poorsurvival, suggest<strong>in</strong>g a role for altered miRNA process<strong>in</strong>g <strong>in</strong> tumor progression. Conversely,reduced expression of Dicer was demonstrated <strong>in</strong> a subset of non-small-cell lung cancers witha poor prognosis [130]. This down-regulation was significantly associated with the reducedexpression of let-7, suggest<strong>in</strong>g altered process<strong>in</strong>g as a mechanism for let-7 dysregulation.Interest<strong>in</strong>gly, the downregulation of Dicer was not due to methylation of the promoter,suggest<strong>in</strong>g alternative mechanisms of dysregulation. This was also supported <strong>in</strong> ovarian cancerwhere reduced expression of Dicer was associated with advanced tumor stage [131]. The<strong>in</strong>consistencies <strong>in</strong> these studies may suggest that the role of altered miRNA process<strong>in</strong>g <strong>in</strong>cancer is tissue/tumor-specific, and may be dependent on the expression of cofactors <strong>in</strong>volved<strong>in</strong> biogenesis.


CHAPTER 5MicroRNA <strong>in</strong> OncogenesisA common chromosomal amplification <strong>in</strong> cervical cancer was demonstrated to result <strong>in</strong> upregulationof Drosha <strong>in</strong> advanced squamous cell carc<strong>in</strong>oma (SCC) tumors [132]. This overexpressionresulted <strong>in</strong> an altered miRNA expression profile, suggest<strong>in</strong>g that Drosha-mediateddysregulation of miRNA may play a role <strong>in</strong> the development and progression of cervical cancer.This was supported <strong>in</strong> esophageal SCC [133], where overexpression of Drosha was associatedwith poor prognosis and <strong>in</strong>creased cell growth. In neuroblastoma [134], low expression ofDicer and Drosha was associated with wide-scale down-regulation of miRNA <strong>in</strong> advancedtumors with a poor prognosis, support<strong>in</strong>g dysregulation of miRNA process<strong>in</strong>g mach<strong>in</strong>ery asa mechanism for altered miRNA expression. Interest<strong>in</strong>gly, silenc<strong>in</strong>g of Drosha and Dicerpromoted transformation and cell growth, suggest<strong>in</strong>g a mechanism for tumor development.The altered expression of Dicer and Drosha <strong>in</strong> multiple cancers strongly suggests that dysregulationof miRNA process<strong>in</strong>g prote<strong>in</strong>s plays a role <strong>in</strong> cancer development and progressionvia altered expression of miRNA.In addition to altered levels of miRNA process<strong>in</strong>g prote<strong>in</strong>s, alterations <strong>in</strong> post-transcriptionalregulation of miRNA can also contribute to their dysregulation. Dur<strong>in</strong>g biogenesis, Droshacleaves the pri-miRNA to produce a pre-miRNA molecule. By do<strong>in</strong>g so, Drosha also determ<strong>in</strong>esone end of the mature miRNA molecule. It has been demonstrated that variation <strong>in</strong> the site atwhich Drosha cleavage occurs can produce multiple pre-miRNA molecules from a s<strong>in</strong>gle primiRNA[135]. These pre-miRNA isoforms differ slightly <strong>in</strong> their 5 0 sequence, which may alter<strong>in</strong>teraction with potential gene targets. Interest<strong>in</strong>gly, this shift <strong>in</strong> Drosha cleavage was nonrandomand only occurred dur<strong>in</strong>g the biogenesis of specific miRNA, suggest<strong>in</strong>g that thisalteration may be functionally important.In addition, evidence also suggests that miRNA are subject to RNA edit<strong>in</strong>g, such as theconversion of adenos<strong>in</strong>e residues <strong>in</strong>to <strong>in</strong>os<strong>in</strong>e (A-to-I) at specific edit<strong>in</strong>g sites [136]. Approximately16% of pri-miRNAs undergo A-to-I edit<strong>in</strong>g [137], which has been demonstrated to<strong>in</strong>terfere with both biogenesis and function of miRNA. A study by Yang and colleagues [138],demonstrated that A-to-I edit<strong>in</strong>g of pri-miR-142 <strong>in</strong>terfered with the Drosha process<strong>in</strong>g step,result<strong>in</strong>g <strong>in</strong> degradation of the edited pri-miRNA molecule and consequently, reduced expressionof mature miR-142. Similarly, A-to-I edit<strong>in</strong>g of pri-miR-151 <strong>in</strong>hibited the Dicer process<strong>in</strong>gstep, which resulted <strong>in</strong> an accumulation of pre-miR-151 [139], suggest<strong>in</strong>g this posttranscriptionalregulation as a mechanism for altered miRNA expression levels. In addition,A-to-I edit<strong>in</strong>g has also been demonstrated to provide a mechanism for dysregulation of miRNAfunction. Edit<strong>in</strong>g with<strong>in</strong> the miRNA seed region results <strong>in</strong> the generation of novel edited maturemiRNA isoforms, with altered target specificity [140]. Whilst the role this plays <strong>in</strong> cancer rema<strong>in</strong>sto be elucidated, it does suggest that alterations <strong>in</strong> post-transcriptional process<strong>in</strong>g of miRNAmay play a role <strong>in</strong> tumorigenesis via dysregulation of miRNA expression and/or function.995.7 miRNA AND TREATMENT RESISTANCEWhilst the role of miRNA <strong>in</strong> the <strong>in</strong>itiation, progression, and prognosis of cancer is welldocumented, the role of miRNA <strong>in</strong> the response to cancer therapy is less well known. Given therole of miRNA <strong>in</strong> regulat<strong>in</strong>g pathways <strong>in</strong>volved <strong>in</strong> the cellular response to chemotherapyand radiation, such as cell cycle [141], apoptosis [55], survival [142], oxidative stress [143],and DNA repair [144], it is likely that miRNA are <strong>in</strong>volved <strong>in</strong> the tumor response toanticancer therapy.5.7.1 Role of miRNA <strong>in</strong> ChemoresistanceIt is now becom<strong>in</strong>g <strong>in</strong>creas<strong>in</strong>gly clear that miRNAs may also play important roles <strong>in</strong> resistanceto chemotherapeutic drugs. With regard to cisplat<strong>in</strong> resistance, <strong>in</strong> the non-small-cell lungcancer (NSCLC) cell l<strong>in</strong>e A549, miR-181a, miR-181b, and miR-630 have been shown to be<strong>in</strong>volved with cellular responses to cisplat<strong>in</strong> [145,146]. miR-181a enhances cisplat<strong>in</strong>-mediatedcell death, via the <strong>in</strong>duction of apoptosis through Bax oligomerization, mitochondrial


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>transmembrane potential dissipation, and proteolytic maturation of caspase-9 and caspase-3[145]. miR-181b was found to be down-regulated <strong>in</strong> an isogenic model of cisplat<strong>in</strong> resistance<strong>in</strong> A549 cells (A549/CDDP), and overexpression of this miRNA decreased levels of Bcl2,enhanc<strong>in</strong>g sensitivity to cisplat<strong>in</strong>-<strong>in</strong>duced cell death [146]. Of note, miR-181a has also beenfound to be both significantly down-regulated and associated with poor survival <strong>in</strong> primaryNSCLC tissues [147,148], while <strong>in</strong> head and neck SCC cells, miR-181a was found to be downregulated<strong>in</strong> response to cisplat<strong>in</strong> treatment [149].miR-497 has been shown to be downregulated <strong>in</strong> a cisplat<strong>in</strong>-resistant NSCLC cell l<strong>in</strong>e (A549/CDDP). This was associated with <strong>in</strong>creased expression of Bcl-2, and overexpression ofexogenous miR-497 reduced Bcl2 prote<strong>in</strong> levels and sensitized the A549/CDDP cells tocisplat<strong>in</strong> [150].In the study by Kroemer and colleagues miR-630 was found to be up-regulated <strong>in</strong> A549 cells <strong>in</strong>response to cisplat<strong>in</strong>. This miRNA was subsequently demonstrated to attenuate the DNAdamage response (phosphorylation of ATM, histone H2AX, and p53), with concomitant<strong>in</strong>duction of p27(Kip1), decreased cell proliferation, and G0eG1 phase cell cycle arrest asopposed to the late G2eM cell cycle arrest normally mediated by cisplat<strong>in</strong> [145].100ZEB1 is a master regulator of the epithelialemesenchymal transition (EMT). Reports havedemonstrated that ZEB1 is important for this process <strong>in</strong> lung cancer through its regulation ofmany EMT genes, <strong>in</strong>clud<strong>in</strong>g E-cadher<strong>in</strong> [151e153]. Knockdown of ZEB1 results <strong>in</strong> thesuppression of anchorage-<strong>in</strong>dependent cell growth of lung cancer cells [154]. Of note, themiR-200 family has been shown to target ZEB1 [67,155,156], and one of its members, miR-200c, has been shown to be down-regulated <strong>in</strong> NSCLC, result<strong>in</strong>g <strong>in</strong> an aggressive, <strong>in</strong>vasive, andchemoresistant phenotype <strong>in</strong> NSCLC [157]. The loss of miR-200c expression occurs asa consequence of DNA CpG methylation, and re-expression was shown to restore the sensitivityof a drug-resistant cell l<strong>in</strong>e to cisplat<strong>in</strong> and cetuximab [157].One of the well-established mechanisms <strong>in</strong>volv<strong>in</strong>g cisplat<strong>in</strong> resistance concerns the overexpressionof ERCC1. This DNA repair gene is <strong>in</strong>volved <strong>in</strong> the repair of DNA adducts andstalled DNA replication forks, and its expression levels can predict both survival and cisplat<strong>in</strong>basedtherapeutic benefit <strong>in</strong> patients with resected NSCLC [158,159]. In a cohort of patientsfor which ERCC1 expression and response to cisplat<strong>in</strong> therapy was known, Friboulet et al.identified miR-375 as significantly underexpressed <strong>in</strong> ERCC1-positive tumors [160]. Similar tomiR-200c, this miRNA has also been shown to be epigenetically regulated by DNA CpGmethylation [161].It is becom<strong>in</strong>g clear that miRNA may play important roles <strong>in</strong> tumor cell responses tochemotherapeutics, such as cisplat<strong>in</strong>. Many miRNA appear to be epigenetically silenced byDNA CpG methylation, and as such it may be possible to resensitize patients to cisplat<strong>in</strong>-basedchemotherapy through the use of demethylat<strong>in</strong>g agents, such as Vidaza Ò or Decitab<strong>in</strong>e Ò .Indeed reactivation of genes silenced by DNA CpG methylation can result <strong>in</strong> re-sensitivity tocisplat<strong>in</strong> <strong>in</strong> cell l<strong>in</strong>e models [162], and as such this strategy may work for miRNA.5.7.2 Role of miRNA <strong>in</strong> RadioresistanceRadiation has been demonstrated to modulate miRNA expression <strong>in</strong> lung cancer [163],lymphoblastoma [164], colon cancer [165] and glioma [166] cell l<strong>in</strong>es. Moreover, thisradiation-mediated modulation of miRNA has been demonstrated to be dose-dependent[143], suggest<strong>in</strong>g that miRNA play a functional role <strong>in</strong> the cellular response to radiation. Inaddition, altered basal miRNA expression has also been associated with radioresistance [167],suggest<strong>in</strong>g a role for miRNA <strong>in</strong> determ<strong>in</strong><strong>in</strong>g the <strong>in</strong>itial response to radiation.Several studies have identified a direct role for miRNA <strong>in</strong> regulat<strong>in</strong>g pathways <strong>in</strong>volved <strong>in</strong> thecellular response to radiation. Simone and colleagues demonstrated a role for miRNA <strong>in</strong> the


CHAPTER 5MicroRNA <strong>in</strong> Oncogenesisresponse to oxidative stress and DNA damage. Radiation significantly altered the expression of17 miRNA <strong>in</strong> human fibroblast cells [143]. Interest<strong>in</strong>gly, expression of all 17 miRNA were alsomodulated by treatment with the chemotherapeutic etoposide, which mimics the effect ofradiation by <strong>in</strong>duc<strong>in</strong>g double-strand breaks and H 2 O 2 , which <strong>in</strong>duces oxidative stress,suggest<strong>in</strong>g a common role for these miRNA <strong>in</strong> the cellular response to genotoxic stress.Interest<strong>in</strong>gly, the radiation-<strong>in</strong>duced modulation of miRNA expression was <strong>in</strong>hibited by theaddition of cyste<strong>in</strong>e, a free radical scavenger that abrogates the effects of ioniz<strong>in</strong>g radiation(IR), suggest<strong>in</strong>g a role for miRNA <strong>in</strong> the cellular response to oxidative stress. In addition, thestudy by He et al., which identified the p53-mediated regulation of the miR-34 family, highlighteda role for miR-34 <strong>in</strong> the DNA damage response to IR [113], support<strong>in</strong>g a role formiRNA <strong>in</strong> the regulation of pathways <strong>in</strong>volved <strong>in</strong> the radioresponse.Furthermore, miRNA have been demonstrated to play a role <strong>in</strong> modulat<strong>in</strong>g the cellularresponse to radiation. Inhibition of miR-221/222 sensitized glioma cells to radiation, whichwas mediated by abrogation of miR-221/222-mediated regulation of the cycl<strong>in</strong>-dependentk<strong>in</strong>ase <strong>in</strong>hibitor p27(kip1) [168]. p27 is an important negative regulator of cell cycleprogression, specifically the G1 phase arrest [169]. Loss of p27 is associated with a morea aggressive cancer phenotype [170] and reduced survival <strong>in</strong> patients undergo<strong>in</strong>g radiationtherapy and surgery <strong>in</strong> prostate cancer [171]. Reduced expression is associated with a poorresponse to neoadjuvant chemoradiotherapy (CRT) <strong>in</strong> rectal cancer [172]. Thus, alterations <strong>in</strong>miR-221/222 expression may modulate the cellular response to radiation via regulation ofp27. Another study demonstrated the miR-181a-mediated modulation of radiosensitivity <strong>in</strong>glioma cells [173]. miR-181a was down-regulated <strong>in</strong> response to radiation, however, ectopicexpression significantly sensitized cells to the cytotoxic effects of radiation. Overexpression ofmiR-181a also resulted <strong>in</strong> down-regulation of the antiapoptotic Bcl2, <strong>in</strong>dicat<strong>in</strong>g Bcl2 asa potential target of miR-181a. Bcl2 expression is associated with resistance to radiation <strong>in</strong>numerous cancers [174e176]. This suggests that down-regulation of miR-181a <strong>in</strong> glioma cellsfollow<strong>in</strong>g exposure to radiation, provides a mechanism for radioresistance via abrogation ofmiR-181a-mediated regulation of Bcl2. A study by Kato and colleagues, demonstrated the miR-34-mediated modulation of radiosensitivity <strong>in</strong> breast cancer cells, and highlighted a role formiR-34 <strong>in</strong> the <strong>in</strong> vivo response to IR <strong>in</strong> C. elegans [177]. miRNA-mediated modulation ofradiosensitivity has also been demonstrated <strong>in</strong> prostate cancer. miR-521 was significantlydown-regulated <strong>in</strong> response to radiation <strong>in</strong> two prostate cancer cell l<strong>in</strong>es, suggest<strong>in</strong>g a role <strong>in</strong>the radiation response [178]. Modulation of miR-521 expression altered sensitivity to radiation,with overexpression <strong>in</strong>duc<strong>in</strong>g sensitivity, whilst <strong>in</strong>hibition <strong>in</strong>duced resistance. The DNArepair prote<strong>in</strong> CSA and the antioxidant enzyme MnSOD were identified as potential targets ofmiR-521, suggest<strong>in</strong>g a mechanism for modulation of the radioresponse [178]. A study byWeidhaas and colleagues, demonstrated a role for let-7 <strong>in</strong> determ<strong>in</strong><strong>in</strong>g the sensitivity toradiation <strong>in</strong> lung cancer [179]. let-7 has been demonstrated to regulate the oncogene Ras,which is commonly overexpressed <strong>in</strong> cancer and has been shown to be critical for protectionfrom radiation-<strong>in</strong>duced cell death [180]. The authors identified a common radiation-<strong>in</strong>ducedpattern of miRNA expression <strong>in</strong> both normal and tumor lung cells, with seven members of thelet-7 family significantly down-regulated, suggest<strong>in</strong>g a common global miRNA response toradiation. Ectopic expression of let-7a and let-7b sensitized lung cancer cells to radiation whilst<strong>in</strong>hibition <strong>in</strong>duced a radioprotective effect, suggest<strong>in</strong>g a functional role for let-7 <strong>in</strong> theresponse to radiation <strong>in</strong> lung cancer. let-7 overexpression also <strong>in</strong>duced sensitivity to radiation<strong>in</strong> C. elegans, which was <strong>in</strong>dicated to be regulated, at least <strong>in</strong> part, via let-7-mediatedregulation of Ras and other DNA damage response genes such as Rad51, Rad21, Cdc25, andFancd2 [179]. This is supported by Oh et al., who demonstrated that overexpression of let-7asensitized lung cancer cells to radiation, via regulation of Ras [181].101A role for miRNA <strong>in</strong> the <strong>in</strong> vivo radiation response has also been demonstrated. Svoboda et al.identified the up-regulation of miR-137 and miR-125b <strong>in</strong> rectal tumor biopsies 2 weeksafter the <strong>in</strong>itiation of neoadjuvant capecitab<strong>in</strong>e CRT, suggest<strong>in</strong>g a role for these miRNA <strong>in</strong> the


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>tumor response to CRT. Furthermore, <strong>in</strong>creased expression of both miRNA was associated witha poor response to CRT [182]. A study by Wang and colleagues demonstrated significantlyaltered expression of just 12 miRNA <strong>in</strong> resected lung tissue of patients who were resistant andsensitive to adjuvant radiation therapy. Additionally, miR-126, which was up-regulated <strong>in</strong>radiosensitive tumor tissue, was demonstrated to <strong>in</strong>hibit proliferation and <strong>in</strong>duce IR-mediatedapoptosis via the PI3K-AKT pathway <strong>in</strong> vitro. miR-126 also enhanced radiosensitivity <strong>in</strong> vitro[183]. Therefore, the down-regulation of miR-126 <strong>in</strong> tumor tissue may provide a mechanismfor resistance to radiation therapy <strong>in</strong> vivo, via enhanced proliferation and evasion ofIR-<strong>in</strong>duced apoptosis.Taken together these studies strongly suggest a role for altered miRNA expression as a mechanismfor resistance to radiation. Furthermore, they strongly suggest a role for miRNA as bothpredictive biomarkers of response to radiation, and potential novel therapeutics with which toenhance the efficacy of radiation therapy.1025.8 CLINICAL APPLICATIONSmiRNA have been shown to be <strong>in</strong>timately <strong>in</strong>volved <strong>in</strong> the oncogenic process, and as such theymay also be exploited <strong>in</strong> the cl<strong>in</strong>ical sett<strong>in</strong>g for diagnostic, prognostic, and therapeuticapplications. miRNA array profil<strong>in</strong>g has revealed unique, tissue-specific miRNA profiles thatmay be useful <strong>in</strong> the cl<strong>in</strong>ical situation. Previously, gene expression profil<strong>in</strong>g has been used <strong>in</strong>a diagnostic and prognostic capacity, as well as <strong>in</strong> predict<strong>in</strong>g treatment outcome, but theseapproaches have not translated well <strong>in</strong>to a rout<strong>in</strong>e cl<strong>in</strong>ical sett<strong>in</strong>g for numerous reasons. Forexample, most of the techniques require fresh tumor material, or have issues with reproducibility,have complicated bio<strong>in</strong>formatics due to large data sets, and/or are not cost-effective.Presently, <strong>in</strong> an evolv<strong>in</strong>g field, employ<strong>in</strong>g miRNA <strong>in</strong> diagnostics may be of more efficientcl<strong>in</strong>ical utility. The number of miRNA for profil<strong>in</strong>g is far fewer than the number of genes.miRNA are also very short <strong>in</strong> length, and as such are highly stable, hence they are not subject tothe same degradation problems as mRNA. Many <strong>in</strong>vestigators now agree that given the direct<strong>in</strong>volvement of miRNA <strong>in</strong> the regulation of prote<strong>in</strong> expression, miRNA expression profilesmay be superior to gene expression profiles for cl<strong>in</strong>ical applications, s<strong>in</strong>ce only a small numberof mRNA are regulatory molecules [184].miRNA profiles can be used to discrim<strong>in</strong>ate between normal and malignant tissue, <strong>in</strong> cancerssuch as lung [62], colorectal [185], breast, pancreatic [186,187], hepatocellular [188], and CLL[189], among others. Additionally, it is possible to del<strong>in</strong>eate and stratify tumors of the sameorgan of orig<strong>in</strong>, but that have different histologies, for example pulmonary adenocarc<strong>in</strong>omaand squamous cell carc<strong>in</strong>oma [62] and endocr<strong>in</strong>e and ac<strong>in</strong>ar pancreatic tumors [186]. Ina similar fashion, miRNA profiles may also be employed as prognostic <strong>in</strong>dicators for factorssuch as therapeutic outcome and overall survival, <strong>in</strong> cancers such as lung [190], esophageal[191], gastric [192], osteosarcoma [193], and breast [194,195]. Many studies have identifiedgene expression profiles that are predictive of therapeutic benefit, <strong>in</strong> a variety of cancer types,<strong>in</strong>clud<strong>in</strong>g breast [196,197], esophageal [198], and colon [199,200]. For breast cancer, thesegene signatures have led to the development of cl<strong>in</strong>ical diagnostics, such as the Mammapr<strong>in</strong>t[201] and Oncotype DX [202] signature arrays. Perhaps, the future may provide a miRNAbaseddiagnostic for prognostication <strong>in</strong> cancer patients.A significant advantage of miRNA from a cl<strong>in</strong>ical perspective is the recent f<strong>in</strong>d<strong>in</strong>g that they canbe found free <strong>in</strong> circulation and can be purified from serum and plasma [203e205]. Due totheir small size, miRNA are highly stable molecules <strong>in</strong> serum, be<strong>in</strong>g protected from RNases,and may be ideal candidates for the development of relatively non-<strong>in</strong>vasive screen<strong>in</strong>g tests.A company called mirna therapeutics have developed a new type of anticancer miRNA technology,which <strong>in</strong>volves us<strong>in</strong>g chemically modified synthetic miRNA mimics and a liposomalbaseddelivery system to re<strong>in</strong>troduce a down regulated miRNA back <strong>in</strong>to tumors <strong>in</strong> vivo


CHAPTER 5MicroRNA <strong>in</strong> Oncogenesis[206,207]. This strategy may be used to re<strong>in</strong>troduce miRNA that are important for tumor cellresponsiveness to other anticancer therapeutics. It is favorable to a gene therapy approach, asrather than alter<strong>in</strong>g a s<strong>in</strong>gle gene, which will have limited cellular impact, alter<strong>in</strong>g a s<strong>in</strong>glemiRNA will have multiple downstream effects on tumor cells <strong>in</strong> terms of signal<strong>in</strong>g pathwaysand effector molecules. Indeed a similar strategy has been employed <strong>in</strong> the treatment of thehepatitis C virus (HCV). It is known that miR-122 is a liver-specific miRNA essential for HCVreplication [208]. It has been shown that HCV replication <strong>in</strong> liver can be dramatically reducedus<strong>in</strong>g oligonucleotide <strong>in</strong>hibitors of miR-122 [209]. This anti-miR-122 (miravirsen) HCVtreatment strategy is now <strong>in</strong> phase II cl<strong>in</strong>ical trials. A possibility for the future lies <strong>in</strong> miRNAreplacement therapy for cancer.References[1] Sontheimer EJ, Carthew RW. Silence from with<strong>in</strong>: endogenous siRNAs and miRNAs. Cell 2005;122:9e12.[2] Bentwich I, Avniel A, Karov Y, Aharonov R, Gilad S, Barad O, et al. Identification of hundreds of conservedand nonconserved human microRNAs. Nat Genet 2005;37:766e70.[3] Lewis BP, Burge CB, Bartel DP. Conserved seed pair<strong>in</strong>g, often flanked by adenos<strong>in</strong>es, <strong>in</strong>dicates that thousandsof human genes are microRNA targets. Cell 2005;120:15e20.[4] Johnson CD, Esquela-Kerscher A, Stefani G, Byrom M, Kelnar K, Ovcharenko D, et al. The let-7 microRNArepresses cell proliferation pathways <strong>in</strong> human cells. Cancer Res 2007;67:7713e22.[5] Johnson SM, Grosshans H, Sh<strong>in</strong>gara J, Byrom M, Jarvis R, Cheng A, et al. RAS is regulated by the let-7microRNA family. Cell 2005;120:635e47.[6] Cal<strong>in</strong> GA, Sevignani C, Dumitru CD, Hyslop T, Noch E, Yendamuri S, et al. <strong>Human</strong> microRNA genes arefrequently located at fragile sites and genomic regions <strong>in</strong>volved <strong>in</strong> cancers. Proc Natl Acad Sci USA2004;101:2999e3004.[7] Ambros V. The functions of animal microRNAs. Nature 2004;431:350e5.[8] Cai X, Hagedorn CH, Cullen BR. <strong>Human</strong> microRNAs are processed from capped, polyadenylated transcriptsthat can also function as mRNAs. RNA 2004;10:1957e66.[9] Lee Y, Kim M, Han J, Yeom KH, Lee S, Baek SH, et al. MicroRNA genes are transcribed by RNA polymerase II.EMBO J 2004;23:4051e60.[10] Borchert GM, Lanier W, Davidson BL. RNA polymerase III transcribes human microRNAs. Nat Struct Mol Biol2006;13:1097e101.[11] Lau NC, Lim LP, We<strong>in</strong>ste<strong>in</strong> EG, Bartel DP. An abundant class of t<strong>in</strong>y RNAs with probable regulatory roles <strong>in</strong>Caenorhabditis elegans. <strong>Science</strong> 2001;294:858e62.[12] Lee Y, Jeon K, Lee JT, Kim S, Kim VN. MicroRNA maturation: stepwise process<strong>in</strong>g and subcellular localization.EMBO J 2002;21:4663e70.[13] Bohnsack MT, Czapl<strong>in</strong>ski K, Gorlich D. Export<strong>in</strong> 5 is a RanGTP-dependent dsRNA-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> thatmediates nuclear export of pre-miRNAs. RNA 2004;10:185e91.[14] Lund E, Gutt<strong>in</strong>ger S, Calado A, Dahlberg JE, Kutay U. Nuclear export of microRNA precursors. <strong>Science</strong>2004;303:95e8.[15] Lewis BP, Shih IH, Jones-Rhoades MW, Bartel DP, Burge CB. Prediction of mammalian microRNA targets. Cell2003;115:787e98.[16] Vella MC, Choi EY, L<strong>in</strong> SY, Re<strong>in</strong>ert K, Slack FJ. The C. elegans microRNA let-7 b<strong>in</strong>ds to imperfect let-7complementary sites from the l<strong>in</strong>-41 3’UTR. Genes Dev 2004;18:132e7.[17] Didiano D, Hobert O. Perfect seed pair<strong>in</strong>g is not a generally reliable predictor for miRNA-target <strong>in</strong>teractions.Nat Struct Mol Biol 2006;13:849e51.[18] Brennecke J, Stark A, Russell RB, Cohen SM. Pr<strong>in</strong>ciples of microRNA-target recognition. PLoS Biol2005;3:e85.[19] Duursma AM, Kedde M, Schrier M, le Sage C, Agami R. miR-148 targets human DNMT3b prote<strong>in</strong> cod<strong>in</strong>gregion. RNA 2008;14:872e7.[20] Orom UA, Nielsen FC, Lund AH. MicroRNA-10a b<strong>in</strong>ds the 5’UTR of ribosomal prote<strong>in</strong> mRNAs and enhancestheir translation. Mol Cell 2008;30:460e71.[21] Tay Y, Zhang J, Thomson AM, Lim B, Rigoutsos I. MicroRNAs to Nanog, Oct4 and Sox2 cod<strong>in</strong>g regionsmodulate embryonic stem cell differentiation. Nature 2008;455:1124e8.[22] Hutvagner G, Zamore PD. A microRNA <strong>in</strong> a multiple-turnover RNAi enzyme complex. <strong>Science</strong>2002;297:2056e60.103


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>104[23] Valencia-Sanchez MA, Liu J, Hannon GJ, Parker R. Control of translation and mRNA degradation by miRNAsand siRNAs. Genes Dev 2006;20:515e24.[24] Filipowicz W. RNAi: the nuts and bolts of the RISC mach<strong>in</strong>e. Cell 2005;122:17e20.[25] Liu J, Carmell MA, Rivas FV, Marsden CG, Thomson JM, Song JJ, et al. Argonaute2 is the catalytic eng<strong>in</strong>e ofmammalian RNAi. <strong>Science</strong> 2004;305:1437e41.[26] Petersen CP, Bordeleau ME, Pelletier J, Sharp PA. Short RNAs repress translation after <strong>in</strong>itiation <strong>in</strong>mammalian cells. Mol Cell 2006;21:533e42.[27] Pillai RS, Bhattacharyya SN, Artus CG, Zoller T, Cougot N, Basyuk E, et al. Inhibition of translational <strong>in</strong>itiationby Let-7 MicroRNA <strong>in</strong> human cells. <strong>Science</strong> 2005;309:1573e6.[28] Wang B, Love TM, Call ME, Doench JG, Nov<strong>in</strong>a CD. Recapitulation of short RNA-directed translational genesilenc<strong>in</strong>g <strong>in</strong> vitro. Mol Cell 2006;22:553e60.[29] Humphreys DT, Westman BJ, Mart<strong>in</strong> DI, Preiss T. MicroRNAs control translation <strong>in</strong>itiation by <strong>in</strong>hibit<strong>in</strong>geukaryotic <strong>in</strong>itiation factor 4E/cap and poly(A) tail function. Proc Natl Acad Sci USA 2005;102:16961e6.[30] Maroney PA, Yu Y, Fisher J, Nilsen TW. Evidence that microRNAs are associated with translat<strong>in</strong>g messengerRNAs <strong>in</strong> human cells. Nat Struct Mol Biol 2006;13:1102e7.[31] Nottrott S, Simard MJ, Richter JD. <strong>Human</strong> let-7a miRNA blocks prote<strong>in</strong> production on actively translat<strong>in</strong>gpolyribosomes. Nat Struct Mol Biol 2006;13:1108e14.[32] Behm-Ansmant I, Rehw<strong>in</strong>kel J, Doerks T, Stark A, Bork P, Izaurralde E. mRNA degradation by miRNAs and GW182requires both CCR4:NOT deadenylase and DCP1:DCP2 decapp<strong>in</strong>g complexes. Genes Dev 2006;20:1885e98.[33] Giraldez AJ, Mishima Y, Rihel J, Grocock RJ, Van Dongen S, Inoue K, et al. Zebrafish MiR-430 promotesdeadenylation and clearance of maternal mRNAs. <strong>Science</strong> 2006;312:75e9.[34] Wu L, Fan J, Belasco JG. MicroRNAs direct rapid deadenylation of mRNA. Proc Natl Acad Sci USA2006;103:4034e9.[35] Engels BM, Hutvagner G. Pr<strong>in</strong>ciples and effects of microRNA-mediated post-transcriptional gene regulation.Oncogene 2006;25:6163e9.[36] Liu J, Valencia-Sanchez MA, Hannon GJ, Parker R. MicroRNA-dependent localization of targeted mRNAs tomammalian P-bodies. Nat Cell Biol 2005;7:719e23.[37] Bhattacharyya SN, Habermacher R, Mart<strong>in</strong>e U, Closs EI, Filipowicz W. Relief of microRNA-mediated translationalrepression <strong>in</strong> human cells subjected to stress. Cell 2006;125:1111e24.[38] Vasudevan S, Tong Y, Steitz JA. Switch<strong>in</strong>g from repression to activation: microRNAs can up-regulate translation.<strong>Science</strong> 2007;318:1931e4.[39] Stark A, Brennecke J, Russell RB, Cohen SM. Identification of Drosophila MicroRNA targets. PLoS Biol2003;1:e60.[40] Rajewsky N, Socci ND. Computational identification of microRNA targets. Dev Biol 2004;267:529e35.[41] Saito T, Saetrom P. MicroRNAsetarget<strong>in</strong>g and target prediction. N Biotechnol 2010;27:243e9.[42] Grimson A, Farh KK, Johnston WK, Garrett-Engele P, Lim LP, Bartel DP. MicroRNA target<strong>in</strong>g specificity <strong>in</strong>mammals: determ<strong>in</strong>ants beyond seed pair<strong>in</strong>g. Mol Cell 2007;27:91e105.[43] Gaidatzis D, van Nimwegen E, Hausser J, Zavolan M. Inference of miRNA targets us<strong>in</strong>g evolutionaryconservation and pathway analysis. BMC Bio<strong>in</strong>formatics 2007;8:69.[44] Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell 2009;136:215e33.[45] Stark A, Brennecke J, Bushati N, Russell RB, Cohen SM. Animal MicroRNAs confer robustness to geneexpression and have a significant impact on 3’UTR evolution. Cell 2005;123:1133e46.[46] Lytle JR, Yario TA, Steitz JA. Target mRNAs are repressed as efficiently by microRNA-b<strong>in</strong>d<strong>in</strong>g sites <strong>in</strong> the 5’ UTRas <strong>in</strong> the 3’ UTR. Proc Natl Acad Sci USA 2007;104:9667e72.[47] Kloosterman WP, Wienholds E, Kett<strong>in</strong>g RF, Plasterk RH. Substrate requirements for let-7 function <strong>in</strong> thedevelop<strong>in</strong>g zebrafish embryo. Nucleic Acids Res 2004;32:6284e91.[48] Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 2004;116:281e97.[49] Cheng AM, Byrom MW, Shelton J, Ford LP. Antisense <strong>in</strong>hibition of human miRNAs and <strong>in</strong>dications for an<strong>in</strong>volvement of miRNA <strong>in</strong> cell growth and apoptosis. Nucleic Acids Res 2005;33:1290e7.[50] Chan JA, Krichevsky AM, Kosik KS. MicroRNA-21 is an antiapoptotic factor <strong>in</strong> human glioblastoma cells.Cancer Res 2005;65:6029e33.[51] Hua Z, Lv Q, Ye W, Wong CK, Cai G, Gu D, et al. MiRNA-directed regulation of VEGF and other angiogenicfactors under hypoxia. PLoS One 2006;1:e116.[52] Ma L, Teruya-Feldste<strong>in</strong> J, We<strong>in</strong>berg RA. Tumour <strong>in</strong>vasion and metastasis <strong>in</strong>itiated by microRNA-10b <strong>in</strong> breastcancer. Nature 2007;449:682e8.[53] Hanahan D, We<strong>in</strong>berg RA. The hallmarks of cancer. Cell 2000;100:57e70.


CHAPTER 5MicroRNA <strong>in</strong> Oncogenesis[54] Cal<strong>in</strong> GA, Dumitru CD, Shimizu M, Bichi R, Zupo S, Noch E, et al. Frequent deletions and down-regulationof micro- RNA genes miR15 and miR16 at 13q14 <strong>in</strong> chronic lymphocytic leukemia. Proc Natl Acad Sci USA2002;99:15524e9.[55] Cimm<strong>in</strong>o A, Cal<strong>in</strong> GA, Fabbri M, Iorio MV, Ferrac<strong>in</strong> M, Shimizu, et al. miR-15 and miR-16 <strong>in</strong>duce apoptosisby target<strong>in</strong>g BCL2. Proc Natl Acad Sci USA 2005;102:13944e9.[56] Cal<strong>in</strong> GA, Cimm<strong>in</strong>o A, Fabbri M, Ferrac<strong>in</strong> M, Wojcik SE, Shimizu M, et al. MiR-15a and miR-16-1 clusterfunctions <strong>in</strong> human leukemia. Proc Natl Acad Sci USA 2008;105:5166e71.[57] Vol<strong>in</strong>ia S, Cal<strong>in</strong> GA, Liu CG, Ambs S, Cimm<strong>in</strong>o A, Petrocca F. A microRNA expression signature of humansolid tumors def<strong>in</strong>es cancer gene targets. Proc Natl Acad Sci USA 2006;103:2257e61.[58] Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, et al. MicroRNA expression profiles classifyhuman cancers. Nature 2005;435:834e8.[59] Iorio MV, Ferrac<strong>in</strong> M, Liu CG, Veronese A, Spizzo R, Sabbioni, et al. MicroRNA gene expression deregulation<strong>in</strong> human breast cancer. Cancer Res 2005;65:7065e70.[60] Guo Y, Chen Z, Zhang L, Zhou F, Shi S, Feng X, et al. Dist<strong>in</strong>ctive microRNA profiles relat<strong>in</strong>g to patient survival<strong>in</strong> esophageal squamous cell carc<strong>in</strong>oma. Cancer Res 2008;68:26e33.[61] Michael MZ, SM OC, van Holst Pellekaan NG, Young GP, James RJ. Reduced accumulation of specificmicroRNAs <strong>in</strong> colorectal neoplasia. Mol Cancer Res 2003;1:882e91.[62] Yanaihara N, Caplen N, Bowman E, Seike M, Kumamoto K, Yi M, et al. Unique microRNA molecular profiles<strong>in</strong> lung cancer diagnosis and prognosis. Cancer Cell 2006;9:189e98.[63] Ambs S, Prueitt RL, Yi M, Hudson RS, Howe TM, Petrocca F, et al. Genomic profil<strong>in</strong>g of microRNA andmessenger RNA reveals deregulated microRNA expression <strong>in</strong> prostate cancer. Cancer Res 2008;68:6162e70.[64] Guo J, Miao Y, Xiao B, Huan R, Jiang Z, Meng D, et al. Differential expression of microRNA species <strong>in</strong> humangastric cancer versus non-tumorous tissues. J Gastroenterol Hepatol 2009;24:652e7.[65] Ma L. Role of miR-10b <strong>in</strong> breast cancer metastasis. Breast Cancer Res 2010;12:210.[66] Ma L, Re<strong>in</strong>hardt F, Pan E, Soutschek J, Bhat B, Marcusson EG, et al. Therapeutic silenc<strong>in</strong>g of miR-10b <strong>in</strong>hibitsmetastasis <strong>in</strong> a mouse mammary tumor model. Nat Biotechnol 2010;28:341e7.[67] Korpal M, Lee ES, Hu G, Kang Y. The miR-200 family <strong>in</strong>hibits epithelial-mesenchymal transition and cancercell migration by direct target<strong>in</strong>g of E-cadher<strong>in</strong> transcriptional repressors ZEB1 and ZEB2. J Biol Chem2008;283:14910e4.[68] Cal<strong>in</strong> GA, Ferrac<strong>in</strong> M, Cimm<strong>in</strong>o A, Di Leva G, Shimizu M, Wojcik SE, et al. A MicroRNA signature associatedwith prognosis and progression <strong>in</strong> chronic lymphocytic leukemia. N Engl J Med 2005;353:1793e801.[69] Schetter AJ, Leung SY, Sohn JJ, Zanetti KA, Bowman ED, Yanaihara N, et al. MicroRNA expression profilesassociated with prognosis and therapeutic outcome <strong>in</strong> colon adenocarc<strong>in</strong>oma. JAMA 2008;299:425e36.[70] Takamizawa J, Konishi H, Yanagisawa K, Tomida S, Osada H, Endoh H, et al. Reduced expression of the let-7microRNAs <strong>in</strong> human lung cancers <strong>in</strong> association with shortened postoperative survival. Cancer Res2004;64:3753e6.[71] Bandi N, Zb<strong>in</strong>den S, Gugger M, Arnold M, Kocher V, Hasan L, et al. miR-15a and miR-16 are implicated <strong>in</strong> cellcycle regulation <strong>in</strong> a Rb-dependent manner and are frequently deleted or down-regulated <strong>in</strong> non-small celllung cancer. Cancer Res 2009;69:5553e9.[72] Bottoni A, Picc<strong>in</strong> D, Tagliati F, Luch<strong>in</strong> A, Zatelli MC, degli Uberti EC. miR-15a and miR-16-1 down-regulation<strong>in</strong> pituitary adenomas. J Cell Physiol 2005;204:280e5.[73] Bhattacharya R, Nicoloso M, Arvizo R, Wang E, Cortez A, Rossi S, et al. MiR-15a and MiR-16 control Bmi-1expression <strong>in</strong> ovarian cancer. Cancer Res 2009;69:9090e5.[74] Bonci D, Coppola V, Musumeci M, Addario A, Giuffrida R, Memeo L, et al. The miR-15a-miR-16-1 clustercontrols prostate cancer by target<strong>in</strong>g multiple oncogenic activities. Nat Med 2008;14:1271e7.[75] Ricarte-Filho JC, Fuziwara CS, Yamashita AS, Rezende E, da-Silva MJ, Kimura ET. Effects of let-7 microRNA onCell Growth and Differentiation of Papillary Thyroid Cancer. Transl Oncol 2009;2:236e41.[76] Dong Q, Meng P, Wang T, Q<strong>in</strong> W, Wang F, Yuan J, et al. MicroRNA let-7a <strong>in</strong>hibits proliferation of humanprostate cancer cells <strong>in</strong> vitro and <strong>in</strong> vivo by target<strong>in</strong>g E2F2 and CCND2. PLoS One 2010;5:e10147.[77] Esquela-Kerscher A, Trang P, Wigg<strong>in</strong>s JF, Patrawala L, Cheng A, Ford L, et al. The let-7 microRNA reducestumor growth <strong>in</strong> mouse models of lung cancer. Cell Cycle 2008;7:759e64.[78] Trang P, Med<strong>in</strong>a PP, Wigg<strong>in</strong>s JF, Ruff<strong>in</strong>o L, Kelnar K, Omotola M, et al. Regression of mur<strong>in</strong>e lung tumors bythe let-7 microRNA. Oncogene 2010;29:1580e7.[79] Wong TS, Man OY, Tsang CM, Tsao SW, Tsang RK, Chan JY, et al. MicroRNA let-7 suppresses nasopharyngealcarc<strong>in</strong>oma cells proliferation through downregulat<strong>in</strong>g c-Myc expression. J Cancer Res Cl<strong>in</strong> Oncol 2010.[80] Lee YS, Dutta A. The tumor suppressor microRNA let-7 represses the HMGA2 oncogene. Genes Dev2007;21:1025e30.105


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>106[81] Feber A, Xi L, Luketich JD, Pennathur A, Landreneau RJ, Wu M, et al. MicroRNA expression profiles ofesophageal cancer. J Thorac Cardiovasc Surg 2008;135:255e60. discussion 260.[82] Fulci V, Chiaretti S, Goldoni M, Azzal<strong>in</strong> G, Carucci N, Tavolaro S, et al. Quantitative technologies establisha novel microRNA profile of chronic lymphocytic leukemia. Blood 2007;109:4944e51.[83] Si ML, Zhu S, Wu H, Lu Z, Wu F, Mo YY. miR-21-mediated tumor growth. Oncogene 2007;26:2799e803.[84] Asangani IA, Rasheed SA, Nikolova DA, Leupold JH, Colburn NH, Post S, et al. MicroRNA-21 (miR-21) posttranscriptionallydownregulates tumor suppressor Pdcd4 and stimulates <strong>in</strong>vasion, <strong>in</strong>travasation and metastasis<strong>in</strong> colorectal cancer. Oncogene 2008;27:2128e36.[85] Frankel LB, Christoffersen NR, Jacobsen A, L<strong>in</strong>dow M, Krogh A, Lund AH. Programmed cell death 4 (PDCD4)is an important functional target of the microRNA miR-21 <strong>in</strong> breast cancer cells. J Biol Chem2008;283:1026e33.[86] Lu Z, Liu M, Strib<strong>in</strong>skis V, Kl<strong>in</strong>ge CM, Ramos KS, Colburn NH, et al. MicroRNA-21 promotes cell transformationby target<strong>in</strong>g the programmed cell death 4 gene. Oncogene 2008.[87] Meng F, Henson R, Wehbe-Janek H, Ghoshal K, Jacob ST, Patel T. MicroRNA-21 regulates expression of thePTEN tumor suppressor gene <strong>in</strong> human hepatocellular cancer. Gastroenterology 2007;133:647e58.[88] Zhu S, Si ML, Wu H, Mo YY. MicroRNA-21 targets the tumor suppressor gene tropomyos<strong>in</strong> 1 (TPM1). J BiolChem 2007;282:14328e36.[89] Meng F, Henson R, Lang M, Wehbe H, Maheshwari S, Mendell JT, et al. Involvement of human micro-RNA <strong>in</strong>growth and response to chemotherapy <strong>in</strong> human cholangiocarc<strong>in</strong>oma cell l<strong>in</strong>es. Gastroenterology2006;130:2113e29.[90] Tao J, Lu Q, Wu D, Li P, Xu B, Q<strong>in</strong>g W, et al. microRNA-21 modulates cell proliferation and sensitivity todoxorubic<strong>in</strong> <strong>in</strong> bladder cancer cells. Oncol Rep 2011;25:1721e9.[91] Valeri N, Gaspar<strong>in</strong>i P, Braconi C, Paone A, Lovat F, Fabbri M, et al. MicroRNA-21 <strong>in</strong>duces resistance to 5-fluorouracil by down-regulat<strong>in</strong>g human DNA MutS homolog 2 (hMSH2). Proc Natl Acad Sci USA2010;107:21098e103.[92] Tanzer A, Stadler PF. Molecular evolution of a microRNA cluster. J Mol Biol 2004;339:327e35.[93] Hayashita Y, Osada H, Tatematsu Y, Yamada H, Yanagisawa K, Tomida S, et al. A polycistronic microRNAcluster, miR-17-92, is overexpressed <strong>in</strong> human lung cancers and enhances cell proliferation. Cancer Res2005;65:9628e32.[94] He L, Thomson JM, Hemann MT, Hernando-Monge E, Mu D, Goodson S, et al. A microRNA polycistron asa potential human oncogene. Nature 2005;435:828e33.[95] Ota A, Tagawa H, Karnan S, Tsuzuki S, Karpas A, Kira S, et al. Identification and characterization of a novel gene,C13orf25, as a target for 13q31-q32 amplification <strong>in</strong> malignant lymphoma. Cancer Res 2004;64:3087e95.[96] Xiao C, Sr<strong>in</strong>ivasan L, Calado DP, Patterson HC, Zhang B, Wang J, et al. Lymphoproliferative diseaseand autoimmunity <strong>in</strong> mice with <strong>in</strong>creased miR-17-92 expression <strong>in</strong> lymphocytes. Nat Immunol 2008;9:405e14.[97] Matsubara H, Takeuchi T, Nishikawa E, Yanagisawa K, Hayashita Y, Ebi H, et al. Apoptosis <strong>in</strong>duction byantisense oligonucleotides aga<strong>in</strong>st miR-17-5p and miR-20a <strong>in</strong> lung cancers overexpress<strong>in</strong>g miR-17-92.Oncogene 2007;26:6099e105.[98] Wong P, Iwasaki M, Somervaille TC, Ficara F, Carico C, Arnold C, et al. The miR-17-92 microRNA polycistronregulates MLL leukemia stem cell potential by modulat<strong>in</strong>g p21 expression. Cancer Res 2010;70:3833e42.[99] O’Donnell KA, Wentzel EA, Zeller KI, Dang CV, Mendell JT. c-Myc-regulated microRNAs modulate E2F1expression. Nature 2005;435:839e43.[100] L<strong>in</strong> YW, Sheu JC, Liu LY, Chen CH, Lee HS, Huang GT, et al. Loss of heterozygosity at chromosome 13q <strong>in</strong>hepatocellular carc<strong>in</strong>oma: identification of three <strong>in</strong>dependent regions. Eur J Cancer 1999;35:1730e4.[101] Bracken AP, Ciro M, Cocito A, Hel<strong>in</strong> K. E2F target genes: unravel<strong>in</strong>g the biology. Trends Biochem Sci2004;29:409e17.[102] Matsumura I, Tanaka H, Kanakura Y. E2F1 and c-Myc <strong>in</strong> cell growth and death. Cell Cycle 2003;2:333e8.[103] Zhang L, Huang J, Yang N, Greshock J, Megraw MS, Giannakakis A, et al. microRNAs exhibit high frequencygenomic alterations <strong>in</strong> human cancer. Proc Natl Acad Sci USA 2006;103:9136e41.[104] Gaur A, Jewell DA, Liang Y, Ridzon D, Moore JH, Chen C, et al. Characterization of microRNA expressionlevels and their biological correlates <strong>in</strong> human cancer cell l<strong>in</strong>es. Cancer Res 2007;67:2456e68.[105] Gauwerky CE, Huebner K, Isobe M, Nowell PC, Croce CM. Activation of MYC <strong>in</strong> a masked t(8;17) translocationresults <strong>in</strong> an aggressive B-cell leukemia. Proc Natl Acad Sci USA 1989;86:8867e71.[106] Sarhadi VK, Wikman H, Salmenkivi K, Kuosma E, Sioris T, Salo J, et al. Increased expression of high mobilitygroup A prote<strong>in</strong>s <strong>in</strong> lung cancer. J Pathol 2006;209:206e12.[107] Mayr C, Hemann MT, Bartel DP. Disrupt<strong>in</strong>g the pair<strong>in</strong>g between let-7 and Hmga2 enhances oncogenictransformation. <strong>Science</strong> 2007;315:1576e9.


CHAPTER 5MicroRNA <strong>in</strong> Oncogenesis[108] Grandori C, Cowley SM, James LP, Eisenman RN. The Myc/Max/Mad network and the transcriptional controlof cell behavior. Annu Rev Cell Dev Biol 2000;16:653e99.[109] Dalla-Favera R, Bregni M, Erikson J, Patterson D, Gallo RC, Croce CM. <strong>Human</strong> c-myc onc gene is located onthe region of chromosome 8 that is translocated <strong>in</strong> Burkitt lymphoma cells. Proc Natl Acad Sci USA1982;79:7824e7.[110] Chang TC, Yu D, Lee YS, Wentzel EA, Ark<strong>in</strong>g DE, West KM, et al. Widespread microRNA repression by Myccontributes to tumorigenesis. Nat Genet 2008;40:43e50.[111] Chang TC, Zeitels LR, Hwang HW, Chivukula RR, Wentzel EA, Dews M, et al. L<strong>in</strong>-28B transactivation isnecessary for Myc-mediated let-7 repression and proliferation. Proc Natl Acad Sci USA 2009;106:3384e9.[112] Hollste<strong>in</strong> M, Sidransky D, Vogelste<strong>in</strong> B, Harris CC. p53 mutations <strong>in</strong> human cancers. <strong>Science</strong>1991;253:49e53.[113] He L, He X, Lim LP, de Stanch<strong>in</strong>a E, Xuan Z, Liang Y, et al. A microRNA component of the p53 tumoursuppressor network. Nature 2007;447:1130e4.[114] Yamakuchi M, Lotterman CD, Bao C, Hruban RH, Karim B, Mendell JT, et al. P53-<strong>in</strong>duced microRNA-107<strong>in</strong>hibits HIF-1 and tumor angiogenesis. Proc Natl Acad Sci USA 2010;107:6334e9.[115] Sachdeva M, Zhu S, Wu F, Wu H, Walia V, Kumar S, et al. p53 represses c-Myc through <strong>in</strong>duction of the tumorsuppressor miR-145. Proc Natl Acad Sci USA 2009;106:3207e12.[116] Georges SA, Biery MC, Kim SY, Schelter JM, Guo J, Chang AN, et al. Coord<strong>in</strong>ated regulation of cell cycletranscripts by p53-Inducible microRNAs, miR-192 and miR-215. Cancer Res 2008;68:10105e12.[117] Suzuki HI, Yamagata K, Sugimoto K, Iwamoto T, Kato S, Miyazono K. Modulation of microRNA process<strong>in</strong>g byp53. Nature 2009;460:529e33.[118] Sampson VB, Rong NH, Han J, Yang Q, Aris V, Soteropoulos P, et al. MicroRNA let-7a down-regulates MYCand reverts MYC-<strong>in</strong>duced growth <strong>in</strong> Burkitt lymphoma cells. Cancer Res 2007;67:9762e70.[119] Le MT, Teh C, Shyh-Chang N, Xie H, Zhou B, Korzh V, et al. MicroRNA-125b is a novel negative regulator ofp53. Genes Dev 2009;23:862e76.[120] Weber B, Stresemann C, Brueckner B, Lyko F. Methylation of human microRNA genes <strong>in</strong> normal andneoplastic cells. Cell Cycle 2007;6:1001e5.[121] Saito Y, Liang G, Egger G, Friedman JM, Chuang JC, Coetzee GA, et al. Specific activation of microRNA-127with downregulation of the proto-oncogene BCL6 by chromat<strong>in</strong>-modify<strong>in</strong>g drugs <strong>in</strong> human cancer cells.Cancer Cell 2006;9:435e43.[122] Scott GK, Mattie MD, Berger CE, Benz SC, Benz CC. Rapid alteration of microRNA levels by histonedeacetylase <strong>in</strong>hibition. Cancer Res 2006;66:1277e81.[123] Lujambio A, Ropero S, Ballestar E, Fraga MF, Cerrato C, Setien F, et al. Genetic unmask<strong>in</strong>g of an epigeneticallysilenced microRNA <strong>in</strong> human cancer cells. Cancer Res 2007;67:1424e9.[124] Agirre X, Vilas-Zornoza A, Jimenez-Velasco A, Mart<strong>in</strong>-Subero JI, Cordeu L, Garate L, et al. Epigenetic silenc<strong>in</strong>gof the tumor suppressor microRNA Hsa-miR-124a regulates CDK6 expression and confers a poor prognosis<strong>in</strong> acute lymphoblastic leukemia. Cancer Res 2009;69:4443e53.[125] Iorio MV, Visone R, Di Leva G, Donati V, Petrocca F, Casal<strong>in</strong>i P, et al. MicroRNA signatures <strong>in</strong> human ovariancancer. Cancer Res 2007;67:8699e707.[126] Fabbri M, Garzon R, Cimm<strong>in</strong>o A, Liu Z, Zanesi N, Callegari E, et al. MicroRNA-29 family reverts aberrantmethylation <strong>in</strong> lung cancer by target<strong>in</strong>g DNA methyltransferases 3A and 3B. Proc Natl Acad Sci USA2007;104:15805e10.[127] Kumar MS, Lu J, Mercer KL, Golub TR, Jacks T. Impaired microRNA process<strong>in</strong>g enhances cellular transformationand tumorigenesis. Nat Genet 2007;39:673e7.[128] Chiosea S, Jelezcova E, Chandran U, Acquafondata M, McHale T, Sobol RW, et al. Up-regulation of dicer,a component of the MicroRNA mach<strong>in</strong>ery, <strong>in</strong> prostate adenocarc<strong>in</strong>oma. Am J Pathol 2006;169:1812e20.[129] Faber C, Horst D, Hlubek F, Kirchner T. Overexpression of Dicer predicts poor survival <strong>in</strong> colorectal cancer.Eur J Cancer 2011;47:1414e9.[130] Karube Y, Tanaka H, Osada H, Tomida S, Tatematsu Y, Yanagisawa K, et al. Reduced expression of Dicerassociated with poor prognosis <strong>in</strong> lung cancer patients. Cancer Sci 2005;96:111e5.[131] Merritt WM, L<strong>in</strong> YG, Han LY, Kamat AA, Spannuth WA, Schmandt R, et al. Dicer, Drosha, and outcomes <strong>in</strong>patients with ovarian cancer. N Engl J Med 2008;359:2641e50.[132] Muralidhar B, Goldste<strong>in</strong> LD, Ng G, W<strong>in</strong>der DM, Palmer RD, Good<strong>in</strong>g EL, et al. Global microRNA profiles <strong>in</strong>cervical squamous cell carc<strong>in</strong>oma depend on Drosha expression levels. J Pathol 2007;212:368e77.[133] Sugito N, Ishiguro H, Kuwabara Y, Kimura M, Mitsui A, Kurehara H, et al. RNASEN regulates cell proliferationand affects survival <strong>in</strong> esophageal cancer patients. Cl<strong>in</strong> Cancer Res 2006;12:7322e8.[134] L<strong>in</strong> RJ, L<strong>in</strong> YC, Chen J, Kuo HH, Chen YY, Diccianni MB, et al. microRNA signature and expression of Dicerand Drosha can predict prognosis and del<strong>in</strong>eate risk groups <strong>in</strong> neuroblastoma. Cancer Res 2010;70:7841e50.107


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>108[135] Wu H, Ye C, Ramirez D, Manjunath N. Alternative process<strong>in</strong>g of primary microRNA transcripts by Droshagenerates 5’ end variation of mature microRNA. PLoS One 2009;4:e7566.[136] Luciano DJ, Mirsky H, Vendetti NJ, Maas S. RNA edit<strong>in</strong>g of a miRNA precursor. RNA 2004;10:1174e7.[137] Kawahara Y, Megraw M, Kreider E, Iizasa H, Valente L, Hatzigeorgiou AG, et al. Frequency and fate ofmicroRNA edit<strong>in</strong>g <strong>in</strong> human bra<strong>in</strong>. Nucleic Acids Res 2008;36:5270e80.[138] Yang W, Chendrimada TP, Wang Q, Higuchi M, Seeburg PH, Shiekhattar R, et al. Modulation of microRNAprocess<strong>in</strong>g and expression through RNA edit<strong>in</strong>g by ADAR deam<strong>in</strong>ases. Nat Struct Mol Biol 2006;13:13e21.[139] Kawahara Y, Z<strong>in</strong>shteyn B, Chendrimada TP, Shiekhattar R, Nishikura K. RNA edit<strong>in</strong>g of the microRNA-151precursor blocks cleavage by the Dicer-TRBP complex. EMBO Rep 2007;8:763e9.[140] Kawahara Y, Z<strong>in</strong>shteyn B, Sethupathy P, Iizasa H, Hatzigeorgiou AG, Nishikura K. Redirection of silenc<strong>in</strong>gtargets by adenos<strong>in</strong>e-to-<strong>in</strong>os<strong>in</strong>e edit<strong>in</strong>g of miRNAs. <strong>Science</strong> 2007;315:1137e40.[141] L<strong>in</strong>sley PS, Schelter J, Burchard J, Kibukawa M, Mart<strong>in</strong> MM, Bartz SR, et al. Transcripts targeted by themicroRNA-16 family cooperatively regulate cell cycle progression. Mol Cell Biol 2007;27:2240e52.[142] Meng F, Henson R, Wehbe-Janek H, Smith H, Ueno Y, Patel T. The MicroRNA let-7a modulates <strong>in</strong>terleuk<strong>in</strong>-6-dependent STAT-3 survival signal<strong>in</strong>g <strong>in</strong> malignant human cholangiocytes. J Biol Chem 2007;282:8256e64.[143] Simone NL, Soule BP, Ly D, Saleh AD, Savage JE, Degraff W, et al. Ioniz<strong>in</strong>g radiation-<strong>in</strong>duced oxidative stressalters miRNA expression. PLoS One 2009;4:e6377.[144] Crosby ME, Kulshreshtha R, Ivan M, Glazer PM. MicroRNA regulation of DNA repair gene expression <strong>in</strong>hypoxic stress. Cancer Res 2009;69:1221e9.[145] Galluzzi L, Morselli E, Vitale I, Kepp O, Senovilla L, Criollo A, et al. miR-181a and miR-630 regulate cisplat<strong>in</strong><strong>in</strong>ducedcancer cell death. Cancer Res 2010;70:1793e803.[146] Zhu W, Shan X, Wang T, Shu Y, Liu P. miR-181b modulates multidrug resistance by target<strong>in</strong>g BCL2 <strong>in</strong> humancancer cell l<strong>in</strong>es. Int J Cancer 2010;127:2520e9.[147] Gao W, Shen H, Liu L, Xu J, Shu Y. MiR-21 overexpression <strong>in</strong> human primary squamous cell lung carc<strong>in</strong>omais associated with poor patient prognosis. J Cancer Res Cl<strong>in</strong> Oncol 2011;137:557e66.[148] Gao W, Yu Y, Cao H, Shen H, Li X, Pan S, et al. Deregulated expression of miR-21, miR-143 and miR-181a <strong>in</strong>non small cell lung cancer is related to cl<strong>in</strong>icopathologic characteristics or patient prognosis. BiomedPharmacother 2010;64:399e408.[149] Huang Y, Chuang A, Hao H, Talbot C, Sen T, Tr<strong>in</strong>k B, et al. Phospho-DeltaNp63alpha is a key regulator of thecisplat<strong>in</strong>-<strong>in</strong>duced microRNAome <strong>in</strong> cancer cells. Cell Death Differ 2011;18:1220e30.[150] Zhu W, Zhu D, Lu S, Wang T, Wang J, Jiang B, et al. miR-497 modulates multidrug resistance of human cancercell l<strong>in</strong>es by target<strong>in</strong>g BCL2. Med Oncol 2011.[151] Dohadwala M, Yang SC, Luo J, Sharma S, Batra RK, Huang M, et al. Cyclooxygenase-2-dependent regulationof E-cadher<strong>in</strong>: prostagland<strong>in</strong> E(2) <strong>in</strong>duces transcriptional repressors ZEB1 and snail <strong>in</strong> non-small cell lungcancer. Cancer Res 2006;66:5338e45.[152] Gemmill RM, Roche J, Potiron VA, Nasarre P, Mitas M, Coldren CD, et al. ZEB1-responsive genes <strong>in</strong> nonsmallcell lung cancer. Cancer Lett 2011;300:66e78.[153] Witta SE, Gemmill RM, Hirsch FR, Coldren CD, Hedman K, Ravdel L, et al. Restor<strong>in</strong>g E-cadher<strong>in</strong> expression<strong>in</strong>creases sensitivity to epidermal growth factor receptor <strong>in</strong>hibitors <strong>in</strong> lung cancer cell l<strong>in</strong>es. Cancer Res2006;66:944e50.[154] Takeyama Y, Sato M, Horio M, Hase T, Yoshida K, Yokoyama T, et al. Knockdown of ZEB1, a master epithelialto-mesenchymaltransition (EMT) gene, suppresses anchorage-<strong>in</strong>dependent cell growth of lung cancer cells.Cancer Lett 2010;296:216e24.[155] Gregory PA, Bert AG, Paterson EL, Barry SC, Tsyk<strong>in</strong> A, Farshid G, et al. The miR-200 family and miR-205regulate epithelial to mesenchymal transition by target<strong>in</strong>g ZEB1 and SIP1. Nat Cell Biol 2008;10:593e601.[156] Park SM, Gaur AB, Lengyel E, Peter ME. The miR-200 family determ<strong>in</strong>es the epithelial phenotype of cancercells by target<strong>in</strong>g the E-cadher<strong>in</strong> repressors ZEB1 and ZEB2. Genes Dev 2008;22:894e907.[157] Ceppi P, Mudduluru G, Kumarswamy R, Rapa I, Scagliotti GV, Papotti M, et al. Loss of miR-200c expression<strong>in</strong>duces an aggressive, <strong>in</strong>vasive, and chemoresistant phenotype <strong>in</strong> non-small cell lung cancer. Mol Cancer Res2010;8:1207e16.[158] Cobo M, Isla D, Massuti B, Montes A, Sanchez JM, Provencio M, et al. Customiz<strong>in</strong>g cisplat<strong>in</strong> based onquantitative excision repair cross-complement<strong>in</strong>g 1 mRNA expression: a phase III trial <strong>in</strong> non-small-cell lungcancer. J Cl<strong>in</strong> Oncol 2007;25:2747e54.[159] All<strong>in</strong>gham-Hawk<strong>in</strong>s D, Lea A, Lev<strong>in</strong>e S. ERCC1 Expression Analysis to Guide Therapy <strong>in</strong> Non-Small CellLung Cancer. PLoS Curr 2010;2:RRN1202.[160] Friboulet L, Barrios-Gonzales D, Commo F, Olaussen KA, Vagner S, Adam J, et al. Molecular characteristics ofERCC1 negative vs. ERCC1 positive tumors <strong>in</strong> resected NSCLC. Cl<strong>in</strong> Cancer Res 2011.


CHAPTER 5MicroRNA <strong>in</strong> Oncogenesis[161] Mazar J, Deblasio D, Gov<strong>in</strong>darajan SS, Zhang S, Perera RJ. Epigenetic regulation of microRNA-375 and itsrole <strong>in</strong> melanoma development <strong>in</strong> humans. FEBS Lett 2011.[162] Chang X, Monitto CL, Demokan S, Kim MS, Chang SS, Zhong X, et al. Identification of hypermethylatedgenes associated with cisplat<strong>in</strong> resistance <strong>in</strong> human cancers. Cancer Res 2010;70:2870e9.[163] Sh<strong>in</strong> S, Cha HJ, Lee EM, Lee SJ, Seo SK, J<strong>in</strong> HO, et al. Alteration of miRNA profiles by ioniz<strong>in</strong>g radiation <strong>in</strong>A549 human non-small cell lung cancer cells. Int J Oncol 2009;35:81e6.[164] Cha HJ, Sh<strong>in</strong> S, Yoo H, Lee EM, Bae S, Yang KH, et al. Identification of ioniz<strong>in</strong>g radiation-responsivemicroRNAs <strong>in</strong> the IM9 human B lymphoblastic cell l<strong>in</strong>e. Int J Oncol 2009;34:1661e8.[165] Sh<strong>in</strong> S, Cha HJ, Lee EM, Jung JH, Lee SJ, Park IC, et al. MicroRNAs are significantly <strong>in</strong>fluenced by p53 andradiation <strong>in</strong> HCT116 human colon carc<strong>in</strong>oma cells. Int J Oncol 2009;34:1645e52.[166] Chaudhry MA, Sachdeva H, Omarudd<strong>in</strong> RA. Radiation-<strong>in</strong>duced micro-RNA modulation <strong>in</strong> glioblastoma cellsdiffer<strong>in</strong>g <strong>in</strong> dna-repair pathways. DNA Cell Biol 2010;29:553e61.[167] Chaudhry MA, Kreger B, Omarudd<strong>in</strong> RA. Transcriptional modulation of micro-RNA <strong>in</strong> human cells differ<strong>in</strong>g<strong>in</strong> radiation sensitivity. Int J Radiat Biol 2010;86:569e83.[168] Zhang C, Wang G, Kang C, Du Y, Pu P. [Up-regulation of p27(kip1) by miR-221/222 antisense oligonucleotidesenhances the radiosensitivity of U251 glioblastoma]. Zhonghua Yi Xue Yi Chuan Xue Za Zhi2009;26:634e8.[169] Polyak K, Lee MH, Erdjument-Bromage H, Koff A, Roberts JM, Tempst P, et al. Clon<strong>in</strong>g of p27Kip1,a cycl<strong>in</strong>-dependent k<strong>in</strong>ase <strong>in</strong>hibitor and a potential mediator of extracellular antimitogenic signals. Cell1994;78:59e66.[170] Cheville JC, Lloyd RV, Sebo TJ, Cheng L, Erickson L, Bostwick, et al. Expression of p27kip1 <strong>in</strong> prostaticadenocarc<strong>in</strong>oma. Mod Pathol 1998;11:324e8.[171] Cheng L, Lloyd RV, Weaver AL, Pisansky TM, Cheville JC, Ramnani DM, et al. The cell cycle <strong>in</strong>hibitorsp21WAF1 and p27KIP1 are associated with survival <strong>in</strong> patients treated by salvage prostatectomy after radiationtherapy. Cl<strong>in</strong> Cancer Res 2000;6:1896e9.[172] Esposito G, Pucciarelli S, Alaggio R, Giacomelli L, Marchiori E, Iaderosa, et al. P27kip1 expression is associatedwith tumor response to preoperative chemoradiotherapy <strong>in</strong> rectal cancer. Ann Surg Oncol2001;8:311e8.[173] Chen G, Zhu W, Shi D, Lv L, Zhang C, Liu P, et al. MicroRNA-181a sensitizes human malignant gliomaU87MG cells to radiation by target<strong>in</strong>g Bcl-2. Oncol Rep 2010;23:997e1003.[174] Condon LT, Ashman JN, Ell SR, Stafford ND, Greenman J, Cawkwell L. Overexpression of Bcl-2 <strong>in</strong> squamouscell carc<strong>in</strong>oma of the larynx: a marker of radioresistance. Int J Cancer 2002;100:472e5.[175] An J, Cherv<strong>in</strong> AS, Nie A, Ducoff HS, Huang Z. Overcom<strong>in</strong>g the radioresistance of prostate cancer cells witha novel Bcl-2 <strong>in</strong>hibitor. Oncogene 2007;26:652e61.[176] Streffer JR, Rimner A, Rieger J, Naumann U, Rodemann HP, Weller M. BCL-2 family prote<strong>in</strong>s modulateradiosensitivity <strong>in</strong> human malignant glioma cells. J Neurooncol 2002;56:43e9.[177] Kato M, Paranjape T, Muller RU, Nallur S, Gillespie E, Keane K, et al. The mir-34 microRNA is required for theDNA damage response <strong>in</strong> vivo <strong>in</strong> C. elegans and <strong>in</strong> vitro <strong>in</strong> human breast cancer cells. Oncogene2009;28:2419e24.[178] Josson S, Sung SY, Lao K, Chung LW, Johnstone PA. Radiation modulation of microRNA <strong>in</strong> prostate cancercell l<strong>in</strong>es. Prostate 2008;68:1599e606.[179] Weidhaas JB, Babar I, Nallur SM, Trang P, Roush S, Boehm M, et al. MicroRNAs as potential agents to alterresistance to cytotoxic anticancer therapy. Cancer Res 2007;67:11111e6.[180] Weidhaas JB, Eisenmann DM, Holub JM, Nallur SV. A conserved RAS/mitogen-activated prote<strong>in</strong> k<strong>in</strong>asepathway regulates DNA damage-<strong>in</strong>duced cell death postirradiation <strong>in</strong> Radelegans. Cancer Res2006;66:10434e8.[181] Oh JS, Kim JJ, Byun JY, Kim IA. L<strong>in</strong>28-let7 modulates radiosensitivity of human cancer cells with activation ofK-Ras. Int J Radiat Oncol Biol Phys 2010;76:5e8.[182] Svoboda M, Izakovicova Holla L, Sefr R, Vrtkova I, Kocakova I, Tichy B, et al. Micro-RNAs miR125b andmiR137 are frequently upregulated <strong>in</strong> response to capecitab<strong>in</strong>e chemoradiotherapy of rectal cancer. Int JOncol 2008;33:541e7.[183] Wang XC, Du LQ, Tian LL, Wu HL, Jiang XY, Zhang H, et al. Expression and function of miRNA <strong>in</strong> postoperativeradiotherapy sensitive and resistant patients of non-small cell lung cancer. Lung Cancer2011;72:92e9.[184] Cowland JB, Hother C, Gronbaek K. MicroRNAs and cancer. APMIS 2007;115:1090e106.[185] Bandres E, Cubedo E, Agirre X, Malumbres R, Zarate R, Ramirez N, et al. Identification by Real-time PCR of13 mature microRNAs differentially expressed <strong>in</strong> colorectal cancer and non-tumoral tissues. Mol Cancer2006;5:29.109


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>110[186] Roldo C, Missiaglia E, Hagan JP, Falconi M, Capelli P, Bersani S, et al. MicroRNA expression abnormalities <strong>in</strong>pancreatic endocr<strong>in</strong>e and ac<strong>in</strong>ar tumors are associated with dist<strong>in</strong>ctive pathologic features and cl<strong>in</strong>icalbehavior. J Cl<strong>in</strong> Oncol 2006;24:4677e84.[187] Lee EJ, Gusev Y, Jiang J, Nuovo GJ, Lerner MR, Frankel WL, et al. Expression profil<strong>in</strong>g identifies microRNAsignature <strong>in</strong> pancreatic cancer. Int J Cancer 2007;120:1046e54.[188] Murakami Y, Yasuda T, Saigo K, Urashima T, Toyoda H, Okanoue T, et al. Comprehensive analysis ofmicroRNA expression patterns <strong>in</strong> hepatocellular carc<strong>in</strong>oma and non-tumorous tissues. Oncogene2006;25:2537e45.[189] Cal<strong>in</strong> GA, Liu CG, Sevignani C, Ferrac<strong>in</strong> M, Felli N, Dumitru CD, et al. MicroRNA profil<strong>in</strong>g reveals dist<strong>in</strong>ctsignatures <strong>in</strong> B cell chronic lymphocytic leukemias. Proc Natl Acad Sci USA 2004;101:11755e60.[190] Bianchi F, Nicassio F, Marzi M, Belloni E, Dall’olio V, Bernard L, et al. A serum circulat<strong>in</strong>g miRNA diagnostictest to identify asymptomatic high-risk <strong>in</strong>dividuals with early stage lung cancer. EMBO Mol Med2011;3:495e503.[191] Feber A, Xi L, Pennathur A, Good<strong>in</strong>g WE, Bandla S, Wu M, et al. MicroRNA prognostic signature for nodalmetastases and survival <strong>in</strong> esophageal adenocarc<strong>in</strong>oma. Ann Thorac Surg 2011;91:1523e30.[192] Liu R, Zhang C, Hu Z, Li G, Wang C, Yang C, et al. A five-microRNA signature identified from genome-wideserum microRNA expression profil<strong>in</strong>g serves as a f<strong>in</strong>gerpr<strong>in</strong>t for gastric cancer diagnosis. Eur J Cancer2011;47:784e91.[193] Gougelet A, Pissaloux D, Besse A, Perez J, Duc A, Dutour A, et al. Micro-RNA profiles <strong>in</strong> osteosarcoma asa predictive tool for ifosfamide response. Int J Cancer 2011;129:680e90.[194] Janssen EA, Slewa A, Gudlaugsson E, Jonsdottir K, Skaland I, Soiland H, et al. Biologic profil<strong>in</strong>g of lymphnode negative breast cancers by means of microRNA expression. Mod Pathol 2010;23:1567e76.[195] Iorio MV, Casal<strong>in</strong>i P, Tagliabue E, Menard S, Croce CM. MicroRNA profil<strong>in</strong>g as a tool to understand prognosis,therapy response and resistance <strong>in</strong> breast cancer. Eur J Cancer 2008;44:2753e9.[196] van ’t Veer LJ, Dai H, van de Vijver MJ, He YD, Hart AA, Mao M, et al. Gene expression profil<strong>in</strong>g predictscl<strong>in</strong>ical outcome of breast cancer. Nature 2002;415:530e6.[197] Knauer M, Mook S, Rutgers EJ, Bender RA, Hauptmann M, van de Vijver MJ, et al. The predictive value of the70-gene signature for adjuvant chemotherapy <strong>in</strong> early breast cancer. Breast Cancer Res Treat2010;120:655e61.[198] Maher SG, Gillham CM, Duggan SP, Smyth PC, Miller N, Muldoon C, et al. Gene expression analysis ofdiagnostic biopsies predicts pathological response to neoadjuvant chemoradiotherapy of esophageal cancer.Ann Surg 2009;250:729e37.[199] Del Rio M, Mol<strong>in</strong>a F, Bascoul-Mollevi C, Copois V, Bibeau F, Chalbos P, et al. Gene expression signature <strong>in</strong>advanced colorectal cancer patients select drugs and response for the use of leucovor<strong>in</strong>, fluorouracil, andir<strong>in</strong>otecan. J Cl<strong>in</strong> Oncol 2007;25:773e80.[200] Yi JM, Dhir M, Van Neste L, Down<strong>in</strong>g SR, Jeschke J, Glockner SC, et al. Genomic and epigenomic <strong>in</strong>tegrationidentifies a prognostic signature <strong>in</strong> colon cancer. Cl<strong>in</strong> Cancer Res 2011;17:1535e45.[201] Mook S, Van’t Veer LJ, Rutgers EJ, Piccart-Gebhart MJ, Cardoso F. Individualization of therapy us<strong>in</strong>gMammapr<strong>in</strong>t: from development to the MINDACT Trial. Cancer Genomics Proteomics 2007;4:147e55.[202] Cobleigh MA, Tabesh B, Bitterman P, Baker J, Cron<strong>in</strong> M, Liu ML, et al. Tumor gene expression and prognosis<strong>in</strong> breast cancer patients with 10 or more positive lymph nodes. Cl<strong>in</strong> Cancer Res 2005;11:8623e31.[203] Chen X, Ba Y, Ma L, Cai X, Y<strong>in</strong> Y, Wang K, et al. Characterization of microRNAs <strong>in</strong> serum: a novel class ofbiomarkers for diagnosis of cancer and other diseases. Cell Res 2008;18:997e1006.[204] Lawrie CH, Gal S, Dunlop HM, Pushkaran B, Ligg<strong>in</strong>s AP, Pulford K, et al. Detection of elevated levels oftumour-associated microRNAs <strong>in</strong> serum of patients with diffuse large B-cell lymphoma. Br J Haematol2008;141:672e5.[205] Mitchell PS, Park<strong>in</strong> RK, Kroh EM, Fritz BR, Wyman SK, Pogosova-Agadjanyan EL, et al. Circulat<strong>in</strong>g micro-RNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci USA 2008;105:10513e8.[206] Trang P, Wigg<strong>in</strong>s JF, Daige CL, Cho C, Omotola M, Brown D, et al. Systemic delivery of tumor suppressormicroRNA mimics us<strong>in</strong>g a neutral lipid emulsion <strong>in</strong>hibits lung tumors <strong>in</strong> mice. Mol Ther 2011;19:1116e22.[207] Wigg<strong>in</strong>s JF, Ruff<strong>in</strong>o L, Kelnar K, Omotola M, Patrawala L, Brown D, et al. Development of a lung cancertherapeutic based on the tumor suppressor microRNA-34. Cancer Res 2010;70:5923e30.[208] Roberts AP, Lewis AP, Jopl<strong>in</strong>g CL. miR-122 activates hepatitis C virus translation by a specialized mechanismrequir<strong>in</strong>g particular RNA components. Nucleic Acids Res 2011.[209] Young DD, Connelly CM, Grohmann C, Deiters A. Small molecule modifiers of microRNA miR-122 functionfor the treatment of hepatitis C virus <strong>in</strong>fection and hepatocellular carc<strong>in</strong>oma. J Am Chem Soc2010;132:7976e81.


CHAPTER 6Epigenetic Approaches toCancer TherapySabita N. Saldanha 1,2 , Trygve O. Tollefsbol 11 University of Alabama at Birm<strong>in</strong>gham, Birm<strong>in</strong>gham, AL, USA2 Alabama State University, Montgomery, AL, USACHAPTER OUTLINE6.1 Introduction 1116.2 Histone Acetylation 1126.3 Histone Deacetylases 1136.4 Histone Methylation andDemethylation 1146.5 DNA Methylation 1176.6 Acetylation of Non-HistoneProte<strong>in</strong>s 1186.7 Future Directions 122References 1221116.1 INTRODUCTIONCondensation of DNA is achieved by the <strong>in</strong>teraction of basic prote<strong>in</strong>s called histones thatencircle 147 bp of DNA form<strong>in</strong>g a structure called the nucleosome [1,2]. The histones arearranged as dimers of each subunit; H2A, H2B, H3, and H4 <strong>in</strong> the octet [2]. Histone H1 is<strong>in</strong>dependent of the octet but helps tether the nucleosome complex [2]. The octect complexwith the DNA is so arranged that certa<strong>in</strong> am<strong>in</strong>o acid residues of the histones extend out serv<strong>in</strong>gas regulatory substrates for nucleosomal stability [1]. These substrates establish the condensedand decondensed states of the chromat<strong>in</strong> [1]. Condensation of the chromat<strong>in</strong> prevents thetranscriptome mach<strong>in</strong>ery from b<strong>in</strong>d<strong>in</strong>g and consequently <strong>in</strong>hibits gene expression. However,when these projected tails are modified through enzymatic transformations such as acetylation,methylation, phosphorylation, sumoylation, and ubiquit<strong>in</strong>ation, the accessibility ofDNA changes based on the residue modified [3]. Interest<strong>in</strong>gly, current research has emphasizedthe roles of these modifications <strong>in</strong> the transformation process of a normal cell toa tumorigenic phenotype by creat<strong>in</strong>g imbalances <strong>in</strong> net expression of tumor suppressor versusoncogenes or overall genomic imbalances [4]. These covalent modifications are reversible andtherefore can have profound impacts on the cellular phenotype when the activities of theenzymes that mediate these modifications are altered. Intense <strong>in</strong>terest has been directedtoward the mechanistic pathways of these modifications <strong>in</strong> carc<strong>in</strong>ogenesis. However, substratespecificity and residue-specific alterations still need to be ascerta<strong>in</strong>ed.In addition to histone modifications, CpG d<strong>in</strong>ucleotides can be subjected to epigeneticchanges by the methylation of cytos<strong>in</strong>e residues [5,6]. These methylation patterns are heritableand are governed by four isoforms of DNA methyltransferases; DNMT1, DNMT3a, DNMT3b,T. Tollefsbol (Ed): <strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>. DOI: 10.1016/B978-0-12-388415-2.00006-8Copyright Ó 2012 Elsevier Inc. All rights reserved.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>and DNMT3L [6]. Another area of epigenetics that still requires further exploration andcan potentially compound the effects of chromat<strong>in</strong> epigenomics <strong>in</strong> a neoplastic cell is theepigenetic regulation of non-histone prote<strong>in</strong>s. Epigenetic regulations of non-histone prote<strong>in</strong>scan drastically affect pathways with<strong>in</strong> the cell, the cell cyclical controls, and cellular phenotypes.For example, acetylation of key residues of p53 stabilizes the prote<strong>in</strong> and thus the cellcyclical function with which it is associated [7,8]. This chapter discusses the current treatmentsthat are designed to target epigenetic enzymes with the hope of revers<strong>in</strong>g the epigenome ofcancerous cells. Non-histone prote<strong>in</strong> modifications are also important <strong>in</strong> cancerous cells andtherefore the current approaches to therapy aimed at target<strong>in</strong>g non-histone prote<strong>in</strong>s will alsobe discussed.6.2 HISTONE ACETYLATIONPositively charged am<strong>in</strong>o acids such as lys<strong>in</strong>e (K) and arg<strong>in</strong><strong>in</strong>e (R) located at am<strong>in</strong>o (eNH 2 )term<strong>in</strong>al ends of histones are variously modified (Figure 6.1) [3]. Histones are preferentiallymethylated or phosphorylated at arg<strong>in</strong><strong>in</strong>e residues and acetylated at lys<strong>in</strong>e residues [3,9].Acetylation of lys<strong>in</strong>es <strong>in</strong>itiates active gene expression. Acetylation of histone residues not onlyestablishes euchromat<strong>in</strong> states but has crucial roles <strong>in</strong> nucleosome assembly and ma<strong>in</strong>tenanceof chromat<strong>in</strong> states that affect various phases of the cell, <strong>in</strong>clud<strong>in</strong>g DNA repair [9e11].Currently, no mathematical models are available that can determ<strong>in</strong>e the exact pattern ofepigenetic marks which alter sets of genes <strong>in</strong> cancer tissues. Another h<strong>in</strong>drance <strong>in</strong> determ<strong>in</strong><strong>in</strong>gthese marks is that these chemical transformations are dynamic and affect the genome globally112FIGURE 6.1Effects of acetylation and methylation on histone residues. The red circle represents acetyl groups, the yellow circle symbolizesmethylation and the green symbolizes methylation of arg<strong>in</strong><strong>in</strong>e residue. Acetylation of lys<strong>in</strong>e residues is associated with geneexpression whereas methylation-mediated expression is dependent on the residue methylated and the position. Thesereversible processes exerted by epigenetic enzymes HATs, HDACs, and HMTs largely affect genomic stabilities, local geneexpression, and factors govern<strong>in</strong>g cell fate or phenotype. This figure is reproduced <strong>in</strong> the color plate section.


CHAPTER 6Epigenetic Approaches to Cancer Therapyrather than at specific targets. However, certa<strong>in</strong> histone-lys<strong>in</strong>e residues are specifically acetylatedor deacetylated at key positions. Histone acetyltransferases (HATs) are enzymes thatorchestrate the acetylation of histones and are placed <strong>in</strong> three superfamilies based onhomologies to the yeast class of HATs; GNAT (Gcn5-related N-acetyltransferase), MYST (MOZ,Ybf2/Sas3, Sas2 and TIP60), and p300/CBP145 [4]. In a few cancers, dysregulation of HATactivities by mutations <strong>in</strong> the HAT genes or the dysfunction of the gene by translocationsaccount for tumor promotion [4]. In breast cancer cells, NCOA3 (AIB1) is overexpressed and<strong>in</strong> certa<strong>in</strong> leukemias, p300, CBP, and MYST3 (MOZ) translocations are observed [4]. Target<strong>in</strong>gHAT activity can affect the acetylation patterns and possibly control the expression of oncogenesthat are overexpressed.6.3 HISTONE DEACETYLASESThe dynamic equilibrium of chromat<strong>in</strong> architecture is f<strong>in</strong>ely regulated by the activity of HATsand HDACs. In most cancers, HATs are mutated and <strong>in</strong>clude chromosomal translocations ofthe respective HAT, but HDACs are frequently overexpressed [12]. Certa<strong>in</strong> cofactors exhibit<strong>in</strong>tr<strong>in</strong>sic HAT or HDAC activity and <strong>in</strong> most <strong>in</strong>stances the effects are conglomerative with othercomplexes. Therefore aberrant recruitment of HDACs to transcription factors that affect genessuch as oncogenes or tumor suppressor genes or their expression may facilitate a switch fromnormal to abnormal phenotype. A total of 18 HDACs have been identified and are classified<strong>in</strong>to four major classes [4]. The classification of HDACs is based on the homology of thecatalytic site [13]. Class IV HDACs exhibit homologies similar to class I and II HDACs [4]. TheSir2 HDACs have prom<strong>in</strong>ent roles <strong>in</strong> DNA repair and different Sir2s have varied nuclearfunctional roles [14,15]. Chromat<strong>in</strong> organization is a well-studied area; however, the actualroles of HDACs at the gene level and their roles <strong>in</strong> specific cancers still require further elucidation.In vitro analysis of the effects of HDAC <strong>in</strong>hibitors has demonstrated their profoundeffects on <strong>in</strong>hibit<strong>in</strong>g cell proliferation, and <strong>in</strong>duc<strong>in</strong>g cell differentiation and apoptosis [4].HDAC <strong>in</strong>hibitors are considered important tools <strong>in</strong> cancer therapeutics and are currently be<strong>in</strong>gevaluated for their therapeutic efficacies <strong>in</strong> vitro and <strong>in</strong> cl<strong>in</strong>ical trials. HDAC <strong>in</strong>hibitors testedto date fall under four categories, short-cha<strong>in</strong> fatty acids (SCFA), hydroxamic acid derivatives,benzam<strong>in</strong>des, and cyclic tertrapeptides [4] (Table 6.1). Inhibition of HDACs <strong>in</strong>creases theacetylation levels of specific histone residues and <strong>in</strong> some <strong>in</strong>stances <strong>in</strong>creases stabilities of nonhistoneprote<strong>in</strong>s, both of which are essential to gene regulatory functions (Figure 6.2). Theseobservations have been supported by gene expression profil<strong>in</strong>g studies [16]. Of about 1750prote<strong>in</strong>s that have been identified to be acetylated at lys<strong>in</strong>e residues, 200 of these becomemodified <strong>in</strong> the presence of HDAC <strong>in</strong>hibitors and represent a significant number that maycontribute to changes <strong>in</strong> gene expression and probably <strong>in</strong>itiate antitumor activity [16]. Whatneeds to be determ<strong>in</strong>ed however is the substrate specificities for each HDAC and what drugsare specific for each HDAC.113TABLE 6.1 HDAC InhibitorsClass Type Type CompoundIIIShort-cha<strong>in</strong> fatty acid(SCFA)Hydroxamic acidderivativesSodium butyrateValproatePhenylbutyrateVor<strong>in</strong>ostatBel<strong>in</strong>ostat (PXD-101)Panob<strong>in</strong>ostat (LBH-589)Class of HDACInhibitedClass I and II [4]Class II [4]III Benzam<strong>in</strong>des Ent<strong>in</strong>ostat (SNDX-275) Class I [4]IV Cyclic tetrapeptides Romideps<strong>in</strong> (FK-Not determ<strong>in</strong>ed [4]228)Reference


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>114FIGURE 6.2Effects of acetylation on prote<strong>in</strong> functions. Acetylation of prote<strong>in</strong>s affects many different functions, some of which are listed. Thedouble up-arrows <strong>in</strong>dicate <strong>in</strong>crease and the double down-arrows <strong>in</strong>dicate decrease with respect to the particular function.Some of the genes affected by acetylation under specific prote<strong>in</strong> functions are listed [60].6.4 HISTONE METHYLATION AND DEMETHYLATIONHistone acetylation is key to promot<strong>in</strong>g gene expression. Acetylated lys<strong>in</strong>es are alwaysassociated with gene expression but their methylation status contributes to varied geneexpression and primarily depends on the position and form of the methylated lys<strong>in</strong>e residue(Figure 6.1). Lys<strong>in</strong>es exists <strong>in</strong> mono-, di-, and tri-methylated forms and <strong>in</strong> some <strong>in</strong>stances thesame lys<strong>in</strong>e can be acetylated or methylated, for example, K4 and K9 residues of histone H3[17]. However, <strong>in</strong> this <strong>in</strong>stance the acetylated lys<strong>in</strong>e status will not govern the methylatedstatus of the same lys<strong>in</strong>e <strong>in</strong> the histone [17]. In other cases, either acetylation or methylationwill <strong>in</strong>fluence the covalent modified status of the neighbor<strong>in</strong>g lys<strong>in</strong>e residues and thesummation of these effects will determ<strong>in</strong>e the outcome. A commonly found histone pattern <strong>in</strong>many cancers is the loss of H4K16 acetylation and H4K20 tri-methylation [18].Patterns affect the histone residues globally or histones of gene-specific loci can <strong>in</strong>dependently<strong>in</strong>fluence cancer outcomes (Table 6.2). This implies that when tumor suppressor genes aredown-regulated by hypermethylation, oncogenes may be stimulated by acetylation or


CHAPTER 6Epigenetic Approaches to Cancer TherapyTABLE 6.2 Histone Methylation Marks <strong>in</strong> Cancer DevelopmentHistone Residue Change <strong>in</strong> Histone Pattern Type of Cancer ReferenceH3 K16 Loss of acetylation Cancer [18]K20 Loss of trimethylationH3 K9 Hypomethylation Medulloblastoma [19]H3 K4 Global decrease <strong>in</strong> methylation Poor prognosis or <strong>in</strong>creased risk of [20,21]K18 Global decrease <strong>in</strong> acetylation recurrence <strong>in</strong> prostate, breast,K9 Global decrease <strong>in</strong> methylation kidney, lung, and ovarian cancerK27 Global decrease <strong>in</strong> tri-methylationH3 K9 Hypermethylation Silences tumor suppressor genes <strong>in</strong> [22,23]K27 Hypermethylationcolorectal, breast and prostatecancerhypomethylation. For example, hypermethylation of H3K79 promotes leukemogenesis [24].Tumor-specific epigenetic abnormalities can stem from altered modifications of the histoneresidues, and/or altered expression of the enzymes that catalyze the modifications. Thesechanges are driven by mutations or chromosomal rearrangement of genes that code forepigenetic enzymes regardless of their epigenetic modification. As shown <strong>in</strong> Table 6.2,medulloblastoma arises from the hypomethylation of H3K9, and the loss of H3K9 methyltransferase;amplification of demethylases or acetyltransferases could trigger the outcomeobserved. Hyperacetylation of H3K9 could <strong>in</strong>hibit the methylation of its residue.Like DNA, histone lys<strong>in</strong>e residues are methylated by the activity of methyltransferases andutilize S-adenosyl methion<strong>in</strong>e (SAM) <strong>in</strong> catalyz<strong>in</strong>g the transfer of the methyl group to specifichistone residues [6]. The methyltransferases are specific based on the residues they target.Prote<strong>in</strong> lys<strong>in</strong>e methyltransferases (PKMTs) and arg<strong>in</strong><strong>in</strong>e methyltransferases (PRMTs) arespecific for lys<strong>in</strong>e and arg<strong>in</strong><strong>in</strong>e residues respectively, and mediate mono-, di-, and trimethylation.PRMTs primarily catalyze mono- and di-methylation of histone arg<strong>in</strong><strong>in</strong>e residues2, 8, 17, and 26 of H3 and arg<strong>in</strong><strong>in</strong>e residue 3 of H4 [25]. PKMTs have a conserved SET doma<strong>in</strong>that is required for the methyltransferase activity and several of these have been implicated <strong>in</strong>cancers [25]. H3K27 methylation is mediated by a PKMT called EZH2 [26,27], but this enzymeis over-expressed <strong>in</strong> many tumors and appears to have major roles <strong>in</strong> cancer aggressiveness asseen <strong>in</strong> breast and prostate tissues [26,27]. In another case, leukemogenesis is promoted by theaberrant recruitment of H3K79 non-SET doma<strong>in</strong> DOT1L [28]. Most of the tumor-relatedeffects of HMTs are associated with the over-expression, amplification, and translocation of thegenes cod<strong>in</strong>g the enzymes. Some of these <strong>in</strong>clude SMYD3 [29], CARM1 [30], and PRMT1 [31].Drugs that can <strong>in</strong>hibit the activities of these enzymes are currently be<strong>in</strong>g <strong>in</strong>vestigated anda certa<strong>in</strong> few are show<strong>in</strong>g great promise <strong>in</strong> cl<strong>in</strong>ical trials. 3-Deazaneplanoc<strong>in</strong> (DZNep) isa compound that targets HMTs, <strong>in</strong>clud<strong>in</strong>g EZH2, and is <strong>in</strong>strumental <strong>in</strong> <strong>in</strong>duc<strong>in</strong>g globalhypomethylation at several lys<strong>in</strong>e residues and <strong>in</strong>cludes H3K27 and H4K20 [32]. As seen withHDACs, <strong>in</strong> vitro analysis of DZNep treatment of tumor cell l<strong>in</strong>es <strong>in</strong>duced apoptosis throughthe activation of key target genes [32]. A comb<strong>in</strong>ation of a HMT and HDAC <strong>in</strong>hibitors mayprofoundly affect the synergistic <strong>in</strong>duction of apoptosis and has been demonstrated <strong>in</strong> coloncancer cells [33,34].115Like with most chemical compounds, non-specific and <strong>in</strong>direct mechanisms of action maylimit their cl<strong>in</strong>ical applications. Certa<strong>in</strong> generic compounds may <strong>in</strong>hibit HMT activity directlywith low to no specificity (generic analogs of SAM such as S-adenosyl homocyste<strong>in</strong>e (SAH) ands<strong>in</strong>efung<strong>in</strong>) [35]. However, more selective compounds have been identified through currentscreen<strong>in</strong>g methods. A fungal mycotox<strong>in</strong>, chaetoc<strong>in</strong>, is a potent <strong>in</strong>hibitor of H3K9 HMTasesSUV39H1 and G9a (EHMT2) at IC 50 concentrations of 0.8 mM and 2.5 mM, respectively[36,37]. This compound specifically depletes H3K9 di- and tri-methylation levels. However the


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>compound exhibits cytotoxic effects <strong>in</strong>dependent of its <strong>in</strong>hibitory activity. Another non-SAMcompetitive<strong>in</strong>hibitor of HMT G9a, BIX-01294 has been effective at 1.7 mM and the <strong>in</strong>hibitionis selective toward HMTs with SET doma<strong>in</strong>s such as SETDB1 (ESET), and SETD7 (SET7/9)[38,39]. BIX-01294 was found after screen<strong>in</strong>g 12 500 compounds and is specific <strong>in</strong> actiontowards H3K9me2, reduc<strong>in</strong>g the di-methylated levels <strong>in</strong> mammalian cells. Structural analysisof the SET doma<strong>in</strong> reveals that the compound b<strong>in</strong>ds to the H3 substrate-b<strong>in</strong>d<strong>in</strong>g groove [38].Both natural and chemical analogs with similar aff<strong>in</strong>ities to SET doma<strong>in</strong>s can serve as suitable<strong>in</strong>hibitors <strong>in</strong> cancer treatments. Once the mechanistic action is determ<strong>in</strong>ed, the compoundscan be modified to improve concentration efficacies and m<strong>in</strong>imize non-specific or cytotoxiceffects. Similar <strong>in</strong>hibitors towards PRMTs, have been found [40]. However, more robust<strong>in</strong>hibitors are yet to be discovered as the current PRMT <strong>in</strong>hibitors are non-specific and havelow selectivity and activity. Pyrazole-conta<strong>in</strong><strong>in</strong>g CARM1 <strong>in</strong>hibitors with lower but highlypotent IC 50 concentrations have been reported [41]. It is encourag<strong>in</strong>g that such moleculestarget<strong>in</strong>g essential epigenetic enzymes can potentially reverse epigenetic-mediated cancerousphenotypes and that further optimizations and discoveries of effective yet non-cytotoxic drugsneed to be identified for cl<strong>in</strong>ical test<strong>in</strong>g.116Histone demethylases (HDMs) promote hypomethylation of their target residues and the geneoutput is determ<strong>in</strong>ed by the residue, or position demethylated and/or gene-specific target thatis demethylated. Therapeutically, target<strong>in</strong>g HDMs can be of significant importance as oncogenesthat are normally expressed through histone hypomethylation of their promoters can beswitched off by hypermethylation. Certa<strong>in</strong>ly, smaller molecules with effective catalytic<strong>in</strong>hibitory activity toward HDMs are valuable and screen<strong>in</strong>g for such compounds is crucial.HDMs catalyze the removal of methyl groups from lys<strong>in</strong>e residues <strong>in</strong> two ways. First by theam<strong>in</strong>e oxidation reaction which is specific for mono- and di-methylated residues and secondby the hydroxylation of methylated residues creat<strong>in</strong>g an unstable <strong>in</strong>termediate that degrades torelease formaldehyde [42]. The second process is specific towards mono-, di-, and trimethylatedresidues. The first process requires FAD and the second uses alpha-ketoglutarateand iron as cofactors [42]. The only known HDM to date is lys<strong>in</strong>e-specific demethylase 1(LSD1) which mediates its demethylat<strong>in</strong>g action through the am<strong>in</strong>e oxidation process [43].The family of Jumonji (JmjC) doma<strong>in</strong>-conta<strong>in</strong><strong>in</strong>g prote<strong>in</strong>s demethylate by the hydroxylationof the methyl groups and <strong>in</strong>cludes JARID1B (PLU-1) and Jumonji C (JMJC) 4 doma<strong>in</strong>conta<strong>in</strong><strong>in</strong>gprote<strong>in</strong> (JMJD2C) [42]. These HDMs have been implicated <strong>in</strong> tumor progression aswell. LSD1 has varied roles <strong>in</strong> terms of the residues it catalyzes. H3K4 mono- or di-methylatedresidues are demethylated by LSD1 <strong>in</strong> conjunction with corepressor RE1-silenc<strong>in</strong>g transcriptionfactor (CoREST) [44]. However, H3K9 mono- and di- methylated marks are demethylatedby LSD1, <strong>in</strong> which it <strong>in</strong>teracts with androgen receptor (AR) as a coactivator to enhance thedemethylat<strong>in</strong>g function [45]. This enzyme is over-expressed <strong>in</strong> certa<strong>in</strong> cancers and has beenreported to be associated with aggressive prostate cancer and poorly differentiatedneuroblastomas.S<strong>in</strong>ce LSD1 is homologous to monoam<strong>in</strong>e oxidases (MAO) [46], molecules that are effectiveaga<strong>in</strong>st MAO can <strong>in</strong>hibit the activity of LSD1. One such <strong>in</strong>hibitor is the tranylcyprom<strong>in</strong>e.When used, this <strong>in</strong>hibitor <strong>in</strong>creases the di-methylated levels of H3K4 both <strong>in</strong> vitro and <strong>in</strong> vivoand <strong>in</strong>hibits the neuroblastoma tumor growth [47]. LSD1 has been found to be <strong>in</strong>hibited bypolyam<strong>in</strong>e compounds, a few of which have shown remarkable positive outcomes <strong>in</strong> coloncancer cells [47]. These compounds <strong>in</strong>crease the mono- and di-methylated H3K4 levels andreexpresse many silenced genes important <strong>in</strong> colon cancer development. In breast and prostatecancer, the HMD, JARID1B, is overexpressed and demethylates H3K4me3 that <strong>in</strong>duces cellproliferation, <strong>in</strong>hibits tumor suppressor functions, and results <strong>in</strong> AR coactivation [48].JMJD2C, a H3K9me2/3 demethylase, is amplified and overexpressed <strong>in</strong> esophageal squamouscarc<strong>in</strong>oma and target<strong>in</strong>g this enzyme has been proven effective <strong>in</strong> <strong>in</strong>hibit<strong>in</strong>g esophagealsquamous cell growth <strong>in</strong> vitro [49]. JMJD2C <strong>in</strong> conjunction with LSD1 enhances ARdependentgene expression <strong>in</strong> prostate cancer where this enzyme is overexpressed [50].


CHAPTER 6Epigenetic Approaches to Cancer TherapyTarget<strong>in</strong>g the enzyme or cofactor can effectively <strong>in</strong>hibit the activity of HDMs such as JmjC. Forexample, derivatives of nogg<strong>in</strong> (NOG), analog of alpha-ketoglutarate, are effective aga<strong>in</strong>stmembers of the Jumonji-C family of HDMs and a dimethylester of NOG (DMOG) show<strong>in</strong>gcellular activity has been reported [51]. However, these molecules that are be<strong>in</strong>g used as HDM<strong>in</strong>hibitors are effective at much higher concentrations. Further screen<strong>in</strong>g of potential drugswith higher efficacies and potencies at lower concentrations is still required and the knowledgeof the structural configurations of LSD1 and JMJD2C can assist <strong>in</strong> the f<strong>in</strong>d.6.5 DNA METHYLATIONThe architectural configuration of the nucleosome is strictly governed by histones and theircovalent modifications. However, the DNA encompassed by the histone octet dictates cellularfunctions and stability. Apart from the normally associated transcription factors withpromoters, methylation of CpG residues is another important mechanism regulat<strong>in</strong>g geneexpression. Aberrant expression of this predom<strong>in</strong>ant epigenetic modification has beenreported to play significant roles <strong>in</strong> a variety of diseases, <strong>in</strong>clud<strong>in</strong>g cancers. Fortunately, CpGmethylation can be reversed and therefore this heritable change when exposed to demethylat<strong>in</strong>gcompounds or compounds that <strong>in</strong>hibit the catalytic function of the enzyme itself(DNMTs), presents as potential cancer therapeutic tools. DNMTs are required for CpGmethylation and small molecules that can target these enzymes are be<strong>in</strong>g tested <strong>in</strong> vitro as wellas <strong>in</strong> cl<strong>in</strong>ical trials. The very first DNMT <strong>in</strong>hibitor was Vidaza (5-azacytid<strong>in</strong>e) and wasapproved for use by the US Food and Drug Adm<strong>in</strong>istration (FDA) as treatment aga<strong>in</strong>stmyelodysplastic syndrome (MDS) [52,53]. Another molecule, Dacogen (5-aza-2 0 -deoxycytid<strong>in</strong>e,or decitab<strong>in</strong>e) developed by MGI Pharma Inc. (Bloom<strong>in</strong>gton, MN, USA) has alsobeen used to treat MDS [52,53]. These compounds facilitate their action through bothmethylation-dependent and -<strong>in</strong>dependent pathways and <strong>in</strong> some cases direct proteasomaldegradation of the enzyme has been reported [53]. In theory, the use of these <strong>in</strong>hibitors isaimed at revers<strong>in</strong>g the expression of methylation-silenced critical gene expression. Cl<strong>in</strong>icaltrials us<strong>in</strong>g these molecules have shown great potential as therapeutic agents aga<strong>in</strong>st leukemia,<strong>in</strong>clud<strong>in</strong>g MDS, acute myeloid leukemia, chronic myelogenous leukemia, and chronicmyelomonocytic leukemia [53]. So far, the therapeutic improvements are seen aga<strong>in</strong>stleukemias, although this is not the case with solid tumors s<strong>in</strong>ce it is likely that a multitude offactors govern the growth of the mass. Cellular toxicity is also a major concern and the use ofthese molecules triggers cell cycle arrest by their <strong>in</strong>tegration <strong>in</strong>to the DNA molecule itself.Therefore, it is imperative to develop or screen for drugs that have less cytotoxicity and moreefficacy at lower concentrations. Another concern is the relatively low stability of thesecompounds <strong>in</strong> vivo and therefore modifications that enhance their stability are to beconsidered when select<strong>in</strong>g the drug as a chemotherapeutic agent. It is imperative to design anddevelop drugs that are relatively stable, mediate the degradations of DNMTs without <strong>in</strong>corporation<strong>in</strong>to DNA, and facilitate gene expression crucial to cell differentiation and apoptoticpathways.117In mammalian cells, DNMTs exist as four active forms, DNMT1, DNMT3a, DNMT3b, andDNMT3L and either s<strong>in</strong>gly or <strong>in</strong> comb<strong>in</strong>ation catalyze the methylome pattern<strong>in</strong>g crucial togametogenesis, embryogenesis, development, and carc<strong>in</strong>ogenesis [54]. Structural analysis ofthese enzymes shows that the catalytic doma<strong>in</strong> resides <strong>in</strong> the C-term<strong>in</strong>al region of the prote<strong>in</strong>with the N-term<strong>in</strong>al essential for DNA recognition states, hemimethylated versus unmethylated.DNMT3a is ubiquitously expressed and DNMT3b is present at very low levels with theexception of a few tissues [55]. These levels change <strong>in</strong> tumor cells and global hypomethylationand regional hypermethylation of specific genes becomes an apparent pattern and has beenreported to be the case <strong>in</strong> cervical, prostate, and metastatic hepatocellular carc<strong>in</strong>oma [55].Most precl<strong>in</strong>ical studies focus on the hypermethylation of key genes, <strong>in</strong> some cases tumorsuppressor genes and others oncogenes and the correlation of DNMT levels, methylation


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>patterns of the promoters, and gene expression. There appears to be some level of correlationbetween DNMT levels and hypermethylation; however, regression analysis does not seem tosupport this one-to-one correlation <strong>in</strong>dicative of a much more complex regulatory mechanism<strong>in</strong> vivo [55]. Some of the commonly reported hypermethylated genes <strong>in</strong>clude RAR, RASSF1A,CDNK2A, CHD13, APC, p15, and p16 [56]. p15 is used as a marker to determ<strong>in</strong>e leukemiatransformation, and <strong>in</strong> some other tumors the levels of hypermethylation of tumor suppressorgenes determ<strong>in</strong>e the stage of the tumor [57]. Inhibitors of DNMTs are grouped under twocategories, those that <strong>in</strong>terfere with the methylation of cytos<strong>in</strong>e residues by chelat<strong>in</strong>g <strong>in</strong>tothe DNA complex and second, the non-nucleosides that target DNMT activity or stability(Table 6.3).Results from both non-nucleoside and nucleoside DNMT <strong>in</strong>hibitors (DNMTi) <strong>in</strong> precl<strong>in</strong>icaland cl<strong>in</strong>ical sett<strong>in</strong>gs are encourag<strong>in</strong>g but many of these have drawbacks that need to berevamped. Certa<strong>in</strong>ly the need to screen for and design small molecules with potent DNMTiactivity, less cytotoxicity, and improved specificities is very evident and significant progress hasbeen made <strong>in</strong> this direction. Comb<strong>in</strong>ation treatments of two nucleosides and two nonnucleosidesare <strong>in</strong> progress and the data generated from <strong>in</strong> vitro studies have shownremarkable synergistic DNA-hypermethylat<strong>in</strong>g activity. These have been well described <strong>in</strong>a review by Jiang S-W [55].1186.6 ACETYLATION OF NON-HISTONE PROTEINSHistones are the likely targets for reversible modifications and much focus has been onunderstand<strong>in</strong>g the roles of these modifications <strong>in</strong> cellular processes and the enzymes thatcatalyze these chemistries. However, gene products, primarily prote<strong>in</strong>s, are <strong>in</strong> many wayssubjected to similar regulations, of which acetylation and phosphorylation are key modifications.Many cellular processes are governed by the activity of prote<strong>in</strong>s and are <strong>in</strong>volved <strong>in</strong> cellsignal<strong>in</strong>g, transcription, and even prote<strong>in</strong> degradation (Figure 6.2). Reviews on the acetylationof non-histone prote<strong>in</strong>s are limited, yet this prote<strong>in</strong> modification <strong>in</strong> conjunction with histonemodifications is very relevant to cancer epigenomics. Acetylation of prote<strong>in</strong>s can affect manyaspects of prote<strong>in</strong> function as shown <strong>in</strong> Figure 6.2. Especially pert<strong>in</strong>ent are the effects ofacetylation on p53 (tumor suppressor), nuclear factor-kB (NF-kB ) (metastatic gene), andmyelocytomatosis oncogene (c-Myc) (oncogene) and the plausible roles of <strong>in</strong>direct effects ofHAT/HDAC <strong>in</strong>hibitors on these prote<strong>in</strong>s <strong>in</strong> restor<strong>in</strong>g normal cellular phenotypes.Prote<strong>in</strong> p53 is essential to many cell regulatory functions, and <strong>in</strong> particular, is important torescue a cell from DNA damage and ma<strong>in</strong>ta<strong>in</strong> normal cell division [58]. As seen <strong>in</strong> manycancers, p53 is mutated and the prote<strong>in</strong> is dysfunctional, contribut<strong>in</strong>g to a tumorigenicphenotype. Activation of p53 <strong>in</strong> cells requires the phosphorylation of the prote<strong>in</strong> thatpromotes the acetylation of key lys<strong>in</strong>e residues 120, 164, 320, 370, 372, 373, 381, 382, and386 mediated by different acetyltransferases [59,60]. Although controversial, f<strong>in</strong>d<strong>in</strong>gs stronglysupport that acetylation of p53 at the C-term<strong>in</strong>us end favorably enhances the DNA b<strong>in</strong>d<strong>in</strong>gability of the prote<strong>in</strong> to its target genes [60]. K120 and K164 along with the C-term<strong>in</strong>us arerequired for p53 activity, and s<strong>in</strong>gle-site mutational losses can be rescued by the acetylation ofkey functional residues [60]. Acetylation of p53 K120 by specific HATs, such as Tip60 andhMOF, <strong>in</strong>duces genes of the apoptotic pathway [60]. When K382 is acetylated, p53 recruitsCREB b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> (CBP) that further enhances the transcription of genes, suggestive ofcoactivator functions [60]. In all, acetylation of p53 improves the stability and b<strong>in</strong>d<strong>in</strong>g abilityof p53, allow<strong>in</strong>g for recruitment of coactivators to the transcriptional b<strong>in</strong>d<strong>in</strong>g sites <strong>in</strong> thepromoters of genes crucial to cell cycle regulatory functions, such as for p21. Therefore, HDAC<strong>in</strong>hibitors that <strong>in</strong>crease acetylated levels can contribute to a much more stable p53 even whenit is mutated and can enhance its DNA-b<strong>in</strong>d<strong>in</strong>g abilities, which otherwise would be <strong>in</strong>effective.Further studies are necessary to validate the roles of key acetylated residues that improve theDNA-b<strong>in</strong>d<strong>in</strong>g ability of p53 <strong>in</strong> the mutated form.


TABLE 6.3 Nucleoside and Non-Nucleoside DNMT Inhibitors (DNMTi)Type DNMTi Structure Mode of Action Advantages Disadvantages ReferenceNucleoside<strong>in</strong>hibitorsAzacytid<strong>in</strong>e (5-azacytid<strong>in</strong>e; 5-aza-CR)deoxycytid<strong>in</strong>e (5-aza-2 0 -deoxycytid<strong>in</strong>e; 5-aza-CdR),OHOOOOOONONNNNNNH 2Incorporates <strong>in</strong>to RNA Is effective at loweraffect<strong>in</strong>g nuclear and concentration. Acts oncytoplasmic RNA the S-phase of the cellmetabolism and related cycle and therefore isfunctions <strong>in</strong>clud<strong>in</strong>g very effective aga<strong>in</strong>stprote<strong>in</strong> synthesis highly proliferative cellsIncorporates <strong>in</strong>to DNA as5-Aza-dCTP trapp<strong>in</strong>gDNMT and render<strong>in</strong>g itsusceptible toproteasomal degradationInterferes with the<strong>in</strong>termediate of DNMT-DNA covalent reaction;Traps and <strong>in</strong>activatesDNMTsAdvantage similar toazacytid<strong>in</strong>eCytotoxic and when [55]treatment is withdrawnremethylation isobserved. Low stability.Not effective aga<strong>in</strong>st solidtumorsDisadvantage similar toazacytid<strong>in</strong>e[55]Zebular<strong>in</strong>e (1- (b-D-ribofuranosyl)-2(1H)-pyrimid<strong>in</strong>one)HOOHNO NOOH OHForms a covalentcomplex with DNMT andcytid<strong>in</strong>e deam<strong>in</strong>aseLess cytotoxic and canbe given for extendedperiods of time. Higherstability <strong>in</strong> vivo andtherefore can beadm<strong>in</strong>istered orally.Enhances chemo- andradiosensitivity of thetumor cell. Hasangiostatic andantimitogenicpropertiesUnknown [55]Cont<strong>in</strong>uedCHAPTER 6Epigenetic Approaches to Cancer Therapy119


120TABLE 6.3Nucleoside and Non-Nucleoside DNMT Inhibitors (DNMTi)dcont<strong>in</strong>uedType DNMTi Structure Mode of Action Advantages Disadvantages Reference5-Fluoro-2 0 -deoxycytid<strong>in</strong>e (FdCyd,NSC 48006)HOOONNH 2NInhibits cytid<strong>in</strong>edeam<strong>in</strong>ase. Prevents thetransfer of the methylmoeity at the B-elim<strong>in</strong>ation step mediatedby DNMT. Traps DNMT <strong>in</strong>the covalent complex<strong>in</strong>hibit<strong>in</strong>g its functionUnknown Generation of 5-fluorodeoxyurid<strong>in</strong>e asa metabolite <strong>in</strong> vivo whichis potentially toxic[55]<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>OHNonnucleoside<strong>in</strong>hibitorsProca<strong>in</strong>eOONDemethylates CpG-richresidues by b<strong>in</strong>d<strong>in</strong>g tothem prevent<strong>in</strong>g DNMTsfrom b<strong>in</strong>d<strong>in</strong>g. Reducescell viability and causesgrowth <strong>in</strong>hibitionEffective at very highconcentrations (mM)and is cell-type specificPyrrolid<strong>in</strong>e (proca<strong>in</strong>e [55]derivative) highly effective<strong>in</strong> demethylat<strong>in</strong>g and isconsidered a potentialcompound to be pursuedas a DNMTiNH 2Proca<strong>in</strong>amideONHNSpecifically <strong>in</strong>hibits Reactivates tumorDNMT1 activity and suppressor genes.<strong>in</strong>duces hypomethylation. Effectively reactivatesBlocks enzyme catalytic WIF-1 and <strong>in</strong>hibits Wntactivitypathway. Verypromis<strong>in</strong>g <strong>in</strong> thetreatment of lungcancersUnknown [55]H 2 N


Type DNMTi Structure Mode of Action Advantages Disadvantages ReferenceHydralaz<strong>in</strong>eN NH 2NHNSpecifically <strong>in</strong>hibitsDNMT1 activity and<strong>in</strong>duceshypomethylation.Blocks enzymecatalytic activityComb<strong>in</strong>ation studieswith other epigenetic<strong>in</strong>hibitors make it welltolerable and effectiveaga<strong>in</strong>st solid tumors.The efficacy of thiscompound has alsobeen tested <strong>in</strong> cervicalcancers us<strong>in</strong>g oraldoses <strong>in</strong>dicative ofhigher stabilityUnknown [55]RG108ONHNOOOHExact mechanism notfully understood. Direct<strong>in</strong>teraction with DNMTsor b<strong>in</strong>d<strong>in</strong>g to CpG-richsites are plausibleexplanationsStudies have shownthat the satellite DNAare left <strong>in</strong>tact andhypomethylation isobserved at tumorsuppressor genes.Conserv<strong>in</strong>g themethylation patterns ofsatellite DNA ensures thatchromosomal stability isachievedUnknown [55]CHAPTER 6Epigenetic Approaches to Cancer Therapy121


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>NF-kB is an important gene of the immune system and is observed to be important <strong>in</strong> the<strong>in</strong>flammatory process. This gene is also essential to cell survival, differentiation, and proliferationand is reported to be overexpressed <strong>in</strong> many tumors that are aggressive and metastatic[60]. Nuclear activation of NF-kB target genes occur only when the prote<strong>in</strong> is acetylated butotherwise exists as an <strong>in</strong>active complex tethered by Ik-B <strong>in</strong>hibitor [60]. The ubiquit<strong>in</strong>ationmediateddegradation of Ik-B <strong>in</strong>itiated by its phosphorylation, frees the NF-kB that translocatesto the nucleus and b<strong>in</strong>ds to its target sequence. The NF-kB is a complex of p50, p52,p65(RelA), c-Rel, and RelB [60]. In mammals, the p50/p65 heterodimer is the most commonlyfound complex [60]. Modifications of these dimers are essential to many of its downstreamfunctions. Phosphorylation of p65 <strong>in</strong>itiates its acetylation at multiple sites mediated by therecruitment of p300/CBP. NF-kB target gene expression is enhanced greatly when its K221 andK310 residues are acetylated and the full activity of the prote<strong>in</strong> is dependent on this modification[61]. This observation is supported by the fact that SIRT1 deacetylation of p65 K310,HDAC1, and HDAC3 deacetylation of K221 or K310 <strong>in</strong>hibits transcription of its target genes[61]. Acetylation of NF-kB supports many functions and the acetylation of K122 and K123enhances the export of the prote<strong>in</strong> and re-association with IkB form<strong>in</strong>g an <strong>in</strong>active complex[62]. When p50 subunit is acetylated at positions K431, K440, and K441 the prote<strong>in</strong> moleculesb<strong>in</strong>d with a higher aff<strong>in</strong>ity to its target gene sequences [63,64]. Thus the role of acetylation <strong>in</strong>NF-kB is many-fold. S<strong>in</strong>ce this gene is deregulated <strong>in</strong> many diseases <strong>in</strong>clud<strong>in</strong>g cancers a further<strong>in</strong>vestigation <strong>in</strong>to the application of HDACi or HATi <strong>in</strong> regulat<strong>in</strong>g NF-kB functions iswarranted.122Overexpression of c-Myc has been documented <strong>in</strong> many cancers. c-Myc b<strong>in</strong>ds and activatestarget genes as a complex with Max [64]. However, regulatory functions of the complex aresoley through the transcription activation doma<strong>in</strong> (TAD) of c-Myc located at the N-term<strong>in</strong>us[64]. This doma<strong>in</strong> <strong>in</strong>teracts with HATs, such as GCN5 and Tip60, form<strong>in</strong>g coactivatorcomplexes [64]. The c-Myc <strong>in</strong>teracts with p300 via its TAD region that acetylates lys<strong>in</strong>es atseveral positions between the TAD and DNA-b<strong>in</strong>d<strong>in</strong>g regions of the prote<strong>in</strong>, enhanc<strong>in</strong>g Mycturnover [64]. Thus, HAT-specific <strong>in</strong>teractions with c-Myc dictate its stability and turnover <strong>in</strong>mammalian cells, and molecules that target these <strong>in</strong>teractions by the <strong>in</strong>duction of deacetylatedlevels are promis<strong>in</strong>g strategies <strong>in</strong> cancer therapy.6.7 FUTURE DIRECTIONSEpigenetic phenomena affect histone and non-histone prote<strong>in</strong>s and molecular compoundsthat target enzymes <strong>in</strong>fluenc<strong>in</strong>g these roles are important to further develop. Drawbacks ofcompounds such as those with high cytotoxicity, low specificity, and low stability all haveto be considered when select<strong>in</strong>g an antiepigenetic compound promot<strong>in</strong>g antitumor activity.One way of improv<strong>in</strong>g drug design or compound efficacy is by a dual approach which hasbeen proven to be much more efficient as seen with 5 azacytid<strong>in</strong>e and other chemicalcompounds. Further work <strong>in</strong> these areas is therefore warranted. In addition to drug-basedcompounds, natural plant-based products with similar characteristics need to be screenedand tested.References[1] Bernste<strong>in</strong> BE, Humphrey EL, Erlich RL, Schneider R, Bouman P, Liu JS, et al. Methylation of histone H3 Lys 4 <strong>in</strong>cod<strong>in</strong>g regions of active genes. Proc Natl Acad Sci USA 2002;99:8695e700.[2] Wong CA, Recktenwald AJ, Jones ML, Waterman BM, Boll<strong>in</strong>i ML, Dunagan WC. The cost of serious fall-related<strong>in</strong>juries at three Midwestern hospitals. Jt Comm J Qual Patient Saf 2011;37:81e7.[3] Cohen I, Poreba E, Kamieniarz K, Schneider R. Histone modifiers <strong>in</strong> cancer: friends or foes? Genes Cancer2011;2:631e47.[4] Ma X, Ezzeld<strong>in</strong> HH, Diasio RB. Histone deacetylase <strong>in</strong>hibitors: current status and overview of recent cl<strong>in</strong>icaltrials. Drugs 2009;69:1911e34.


CHAPTER 6Epigenetic Approaches to Cancer Therapy[5] Deaton AM, Bird A. CpG islands and the regulation of transcription. Genes Dev 2011;25:1010e22.[6] Li Y, Tollefsbol TO. Impact on DNA methylation <strong>in</strong> cancer prevention and therapy by bioactive dietarycomponents. Curr Med Chem 2010;17:2141e51.[7] Kim JY, Lee KS, Seol JE, Yu K, Chakravarti D, Seo SB. Inhibition of p53 acetylation by INHAT subunit SET/TAF-I{beta} represses p53 activity. Nucleic Acids Res 2011.[8] Seo SK, J<strong>in</strong> HO, Woo SH, Kim YS, An S, Lee JH, et al. Histone deacetylase <strong>in</strong>hibitors sensitize human non-smallcell lung cancer cells to ioniz<strong>in</strong>g radiation through acetyl p53-mediated c-myc down-regulation. J ThoracOncol 2011;6:1313e9.[9] Torres-Padilla ME, Parfitt DE, Kouzarides T, Zernicka-Goetz M. Histone arg<strong>in</strong><strong>in</strong>e methylation regulatespluripotency <strong>in</strong> the early mouse embryo. Nature 2007;445:214e8.[10] Kouzarides T. SnapShot: Histone-modify<strong>in</strong>g enzymes. Cell 2007;128:802.[11] Kouzarides T. Chromat<strong>in</strong> modifications and their function. Cell 2007;128:693e705.[12] Marks PA, Richon VM, Miller T, Kelly WK. Histone deacetylase <strong>in</strong>hibitors. Adv Cancer Res2004;91:137e68.[13] Yang XJ, Seto E. The Rpd3/Hda1 family of lys<strong>in</strong>e deacetylases: from bacteria and yeast to mice and men. NatRev Mol Cell Biol 2008;9:206e18.[14] Horio Y, Hayashi T, Kuno A, Kunimoto R. Cellular and molecular effects of sirtu<strong>in</strong>s <strong>in</strong> health and disease. Cl<strong>in</strong>Sci (Lond) 2011;121:191e203.[15] McGu<strong>in</strong>ness D, McGu<strong>in</strong>ness DH, McCaul JA, Shiels PG. Sirtu<strong>in</strong>s, bioage<strong>in</strong>g, and cancer. J Ag<strong>in</strong>g Res2011:235754.[16] Choudhary C, Kumar C, Gnad F, Nielsen ML, Rehman M, Walther TC, et al. Lys<strong>in</strong>e acetylation targets prote<strong>in</strong>complexes and co-regulates major cellular functions. <strong>Science</strong> 2009;325:834e40.[17] Rice JC, Allis CD. Histone methylation versus histone acetylation: new <strong>in</strong>sights <strong>in</strong>to epigenetic regulation. CurrOp<strong>in</strong> Cell Biol 2001;13:263e73.[18] Varier RA, Timmers HT. Histone lys<strong>in</strong>e methylation and demethylation pathways <strong>in</strong> cancer. Biochim BiophysActa 2011;1815:75e89.[19] L<strong>in</strong>dsey JC, Lusher ME, Anderton JA, Bailey S, Gilbertson RJ, Pearson AD, et al. Identification of tumourspecificepigenetic events <strong>in</strong> medulloblastoma development by hypermethylation profil<strong>in</strong>g. Carc<strong>in</strong>ogenesis2004;25:661e8.[20] Seligson DB, Horvath S, McBrian MA, Mah V, Yu H, Tze S, et al. Global levels of histone modifications predictprognosis <strong>in</strong> different cancers. Am J Pathol 2009;174:1619e28.[21] Seligson DB, Horvath S, Shi T, Yu H, Tze S, Grunste<strong>in</strong> M, et al. Global histone modification patterns predictrisk of prostate cancer recurrence. Nature 2005;435:1262e6.[22] Kondo Y, Shen L, Cheng AS, Ahmed S, Boumber Y, Charo C, et al. Gene silenc<strong>in</strong>g <strong>in</strong> cancer by histone H3lys<strong>in</strong>e 27 trimethylation <strong>in</strong>dependent of promoter DNA methylation. Nat Genet 2008;40:741e50.[23] Kondo Y, Shen L, Issa JP. Critical role of histone methylation <strong>in</strong> tumor suppressor gene silenc<strong>in</strong>g <strong>in</strong> colorectalcancer. Mol Cell Biol 2003;23:206e15.[24] Muller CI, Ruter B, Koeffler HP, Lubbert M. DNA hypermethylation of myeloid cells, a novel therapeutic target<strong>in</strong> MDS and AML. Curr Pharm Biotechnol 2006;7:315e21.[25] Yost JM, Korboukh I, Liu F, Gao C, J<strong>in</strong> J. Targets <strong>in</strong> epigenetics: <strong>in</strong>hibit<strong>in</strong>g the methyl writers of the histonecode. Curr Chem Genomics 2011;5:72e84.[26] Chase A, Cross NC. Aberrations of EZH2 <strong>in</strong> cancer. Cl<strong>in</strong> Cancer Res 2011;17:2613e8.[27] Chang CJ, Yang JY, Xia W, Chen CT, Xie X, Chao CH, et al. EZH2 promotes expansion of breast tumor<strong>in</strong>itiat<strong>in</strong>g cells through activation of RAF1-beta-caten<strong>in</strong> signal<strong>in</strong>g. Cancer Cell 2011;19:86e100.[28] Okada Y, Feng Q, L<strong>in</strong> Y, Jiang Q, Li Y, Coffield VM, et al. hDOT1L l<strong>in</strong>ks histone methylation to leukemogenesis.Cell 2005;121:167e78.[29] Hamamoto R, Furukawa Y, Morita M, Iimura Y, Silva FP, Li M, et al. SMYD3 encodes a histone methyltransferase<strong>in</strong>volved <strong>in</strong> the proliferation of cancer cells. Nat Cell Biol 2004;6:731e40.[30] Majumder S, Liu Y, Ford 3rd OH, Mohler JL, Whang YE. Involvement of arg<strong>in</strong><strong>in</strong>e methyltransferase CARM1 <strong>in</strong>androgen receptor function and prostate cancer cell viability. Prostate 2006;66:1292e301.[31] Cheung N, Chan LC, Thompson A, Cleary ML, So CW. Prote<strong>in</strong> arg<strong>in</strong><strong>in</strong>e-methyltransferase-dependent oncogenesis.Nat Cell Biol 2007;9:1208e15.[32] Tan J, Yang X, Zhuang L, Jiang X, Chen W, Lee PL, et al. Pharmacologic disruption of Polycomb-repressivecomplex 2-mediated gene repression selectively <strong>in</strong>duces apoptosis <strong>in</strong> cancer cells. Genes Dev2007;21:1050e63.[33] Yadav S, S<strong>in</strong>ghal J, S<strong>in</strong>ghal SS, Awasthi S. hSET1: a novel approach for colon cancer therapy. Biochem Pharmacol2009;77:1635e41.123


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>124[34] Lea MA. Recently identified and potential targets for colon cancer treatment. Future Oncol2010;6:993e1002.[35] Saavedra OM, Isakovic L, Llewellyn DB, Zhan L, Bernste<strong>in</strong> N, Claridge S, et al. SAR around (l)-S-adenosyl-lhomocyste<strong>in</strong>e,an <strong>in</strong>hibitor of human DNA methyltransferase (DNMT) enzymes. Bioorg Med Chem Lett2009;19:2747e51.[36] Iwasa E, Hamashima Y, Fujishiro S, Higuchi E, Ito A, Yoshida M, et al. Total synthesis of (þ)-chaetoc<strong>in</strong> and itsanalogues: their histone methyltransferase G9a <strong>in</strong>hibitory activity. J Am Chem Soc 2010;132:4078e9.[37] Gre<strong>in</strong>er D, Bonaldi T, Eskeland R, Roemer E, Imhof A. Identification of a specific <strong>in</strong>hibitor of the histonemethyltransferase SU(VAR)3-9. Nat Chem Biol 2005;1:143e5.[38] Chang Y, Zhang X, Horton JR, Upadhyay AK, Spannhoff A, Liu J, et al. Structural basis for G9a-like prote<strong>in</strong>lys<strong>in</strong>e methyltransferase <strong>in</strong>hibition by BIX-01294. Nat Struct Mol Biol 2009;16:312e7.[39] Kubicek S, O’Sullivan RJ, August EM, Hickey ER, Zhang Q, Teodoro ML, et al. Reversal of H3K9me2 by a smallmolecule<strong>in</strong>hibitor for the G9a histone methyltransferase. Mol Cell 2007;25:473e81.[40] Lakowski TM, Hart P, Ahern CA, Mart<strong>in</strong> NI, Frankel A. Neta-substituted arg<strong>in</strong>yl peptide <strong>in</strong>hibitors of prote<strong>in</strong>arg<strong>in</strong><strong>in</strong>e N-methyltransferases. ACS Chem Biol 2010;5:1053e63.[41] Sack JS, Thieff<strong>in</strong>e S, Bandiera T, Fasol<strong>in</strong>i M, Duke GJ, Jayaraman L, et al. Structural basis for CARM1 <strong>in</strong>hibitionby <strong>in</strong>dole and pyrazole <strong>in</strong>hibitors. Biochem J 2011;436:331e9.[42] Schneider J, Shilatifard A. Histone demethylation by hydroxylation: chemistry <strong>in</strong> action. ACS Chem Biol2006;1:75e81.[43] Shi Y, Lan F, Matson C, Mulligan P, Whetst<strong>in</strong>e JR, Cole PA, et al. Histone demethylation mediated by thenuclear am<strong>in</strong>e oxidase homolog LSD1. Cell 2004;119:941e53.[44] Yang M, Gocke CB, Luo X, Borek D, Tomchick DR, Machius M, et al. Structural basis for CoREST-dependentdemethylation of nucleosomes by the human LSD1 histone demethylase. Mol Cell 2006;23:377e87.[45] Yu V, Fisch T, Long AM, Tang J, Lee JH, Hierl M, et al. High-Throughput TR-FRET Assays for Identify<strong>in</strong>gInhibitors of LSD1 and JMJD2C Histone Lys<strong>in</strong>e Demethylases. J Biomol Screen 2011.[46] Mao B, Zhao G, Lv X, Chen HZ, Xue Z, Yang B, et al. Sirt1 deacetylates c-Myc and promotes c-Myc/Maxassociation. Int J Biochem Cell Biol 2011;43:1573e81.[47] S<strong>in</strong>gh MM, Manton CA, Bhat KP, Tsai WW, Aldape K, Barton MC, et al. Inhibition of LSD1 sensitizes glioblastomacells to histone deacetylase <strong>in</strong>hibitors. Neuro Oncol 2011;13:894e903.[48] Xiang Y, Zhu Z, Han G, Ye X, Xu B, Peng Z, et al. JARID1B is a histone H3 lys<strong>in</strong>e 4 demethylase up-regulated <strong>in</strong>prostate cancer. Proc Natl Acad Sci USA 2007;104:19226e31.[49] Hamada S, Suzuki T, M<strong>in</strong>o K, Koseki K, Oehme F, Flamme I, et al. Design, synthesis, enzyme-<strong>in</strong>hibitoryactivity, and effect on human cancer cells of a novel series of jumonji doma<strong>in</strong>-conta<strong>in</strong><strong>in</strong>g prote<strong>in</strong> 2 histonedemethylase <strong>in</strong>hibitors. J Med Chem 2010;53:5629e38.[50] Wissmann M, Y<strong>in</strong> N, Muller JM, Greschik H, Fodor BD, Jenuwe<strong>in</strong> T, et al. Cooperative demethylation byJMJD2C and LSD1 promotes androgen receptor-dependent gene expression. Nat Cell Biol 2007;9:347e53.[51] Spannhoff A, Hauser AT, He<strong>in</strong>ke R, Sippl W, Jung M. The emerg<strong>in</strong>g therapeutic potential of histone methyltransferaseand demethylase <strong>in</strong>hibitors. ChemMedChem 2009;4:1568e82.[52] Vigna E, Recchia AG, Madeo A, Gentile M, Bossio S, Mazzone C, et al. Epigenetic regulation <strong>in</strong> myelodysplasticsyndromes: implications for therapy. Expert Op<strong>in</strong> Investig Drugs 2011;20:465e93.[53] Ghoshal K, Bai S. DNA methyltransferases as targets for cancer therapy. Drugs Today (Barc) 2007;43:395e422.[54] Liu L, Li Y, Tollefsbol TO. Gene-environment <strong>in</strong>teractions and epigenetic basis of human diseases. Curr IssuesMol Biol 2008;10:25e36.[55] Ren J, S<strong>in</strong>gh BN, Huang Q, Li Z, Gao Y, Mishra P, et al. DNA hypermethylation as a chemotherapy target. CellSignal 2011;23:1082e93.[56] Tsou JA, Hagen JA, Carpenter CL, Laird-Offr<strong>in</strong>ga IA. DNA methylation analysis: a powerful new tool for lungcancer diagnosis. Oncogene 2002;21:5450e61.[57] Abd El-Hamid TM, Mossallam GI, Sherisher MA. The Cl<strong>in</strong>ical Implications of Methylated p15 and p73 Genes<strong>in</strong> Adult Acute Lymphoblastic Leukemia. J Egypt Natl Canc Inst 2010;22:175e84.[58] Benkirane M, Sardet C, Coux O. Lessons from <strong>in</strong>terconnected ubiquitylation and acetylation of p53: th<strong>in</strong>kmetastable networks. Biochem Soc Trans 2010;38:98e103.[59] Das C, Kundu TK. Transcriptional regulation by the acetylation of nonhistone prote<strong>in</strong>s <strong>in</strong> humans e a newtarget for therapeutics. IUBMB Life 2005;57:137e49.[60] Spange S, Wagner T, He<strong>in</strong>zel T, Kramer OH. Acetylation of non-histone prote<strong>in</strong>s modulates cellular signall<strong>in</strong>gat multiple levels. Int J Biochem Cell Biol 2009;41:185e98.[61] Yeung F, Hoberg JE, Ramsey CS, Keller MD, Jones DR, Frye RA, et al. Modulation of NF-kappaB-dependenttranscription and cell survival by the SIRT1 deacetylase. EMBO J 2004;23:2369e80.


CHAPTER 6Epigenetic Approaches to Cancer Therapy[62] Kiernan R, Bres V, Ng RW, Coudart MP, El Messaoudi S, Sardet C, et al. Post-activation turn-off of NF-kappa B-dependent transcription is regulated by acetylation of p65. J Biol Chem 2003;278:2758e66.[63] Deng WG, Wu KK. Regulation of <strong>in</strong>ducible nitric oxide synthase expression by p300 and p50 acetylation.J Immunol 2003;171:6581e8.[64] Deng WG, Zhu Y, Wu KK. Up-regulation of p300 b<strong>in</strong>d<strong>in</strong>g and p50 acetylation <strong>in</strong> tumor necrosis factor-alpha<strong>in</strong>ducedcyclooxygenase-2 promoter activation. J Biol Chem 2003;278:4770e7.125


This page <strong>in</strong>tentionally left blank


CHAPTER 7Epigenomics <strong>in</strong>Neurobehavioral <strong>Disease</strong>sCassandra L. Smith 1 , Kai Huang 21 Boston University, Boston, MA, USA2 Fortiss geme<strong>in</strong>nützige GmbHAn-Institut der TU, Munich, GermanyCHAPTER OUTLINE7.1 Introduction 1277.2 What is the Epigenome? 1287.3 Epigenomic Modulation ofChromat<strong>in</strong> 1297.4 Issues Unique to Neurobehavioral<strong>Disease</strong>s 1327.5 Genetics 1367.6 Environment 1387.7 Genomic Instability 1397.8 TranscriptionalDysregulation 1427.9 RNA Epigenomics 1437.10 Metabolism 1437.11 Nutritional and DrugInterventions 1457.12 Putt<strong>in</strong>g it all Together 146Glossary 146Acknowledgments 147References 1471277.1 INTRODUCTIONThe spectrum of neurobehavioral diseases, recently named bra<strong>in</strong> diseases by the United StatesNational Institute of Mental Health (NIMH) perhaps to counteract stigma associated with thesediseases, <strong>in</strong>cludes rare s<strong>in</strong>gle gene diseases like Hunt<strong>in</strong>gton’s disease (HD) along with commonmultifactorial and multilevel diseases with genetic and environmental factors, such as schizophrenia(SZ), autism spectrum disorder (ASD), and bipolar disease (BD) (formerly manicdepressive disease). Common neurodegenerative diseases associated with ag<strong>in</strong>g are dementia,Alzheimer’s and Park<strong>in</strong>son’s diseases (discussed <strong>in</strong> Chapter 9). Alzheimer’s disease andprion diseases are epigenomic “templat<strong>in</strong>g” diseases that <strong>in</strong>volve the formation of pathogenicprote<strong>in</strong>s [1].Neurodevelopmental disorders such as Rett (RTT) and Fragile X (FRAX) syndromes are discussedbriefly here and <strong>in</strong> Chapter 8. These diseases arise from specific defects <strong>in</strong> epigenomic processesat specific genetic loci. Another class of neurobehavioral diseases is substance abuse. Thesediseases have well-def<strong>in</strong>ed environmental triggers (e.g. alcohol or coca<strong>in</strong>e) that perturbneurobehavioral processes. Here, our emphasis is on SZ, but <strong>in</strong>cluded are observations on BPand ASD because of their shared features. These disorders have many characteristics <strong>in</strong> commonwith rare neurobehavioral disorders with well-def<strong>in</strong>ed genetic causes, but like other common(cancer and cardiovascular) diseases have strong epigenomic and environmental components.T. Tollefsbol (Ed): <strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>. DOI: 10.1016/B978-0-12-388415-2.00007-XCopyright Ó 2012 Elsevier Inc. All rights reserved.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>An issue with neurobehavioral diseases is that the targets are fuzzy. Here, we have attempted tocover issues that are important <strong>in</strong> understand<strong>in</strong>g epigenomic (another fuzzy term) aspects ofthese diseases. Not surpris<strong>in</strong>gly a complete picture is not possible, <strong>in</strong>stead we show where l<strong>in</strong>ksare known to exist. Other l<strong>in</strong>ks may not exist or may not have been studied. A glossary of termsis presented at the end of the chapter to aid the reader.7.2 WHAT IS THE EPIGENOME?Although the term “epigenetics” was first used by Wadd<strong>in</strong>gton <strong>in</strong> the 1940s, this field of sciencehas multiple orig<strong>in</strong>s. As orig<strong>in</strong>ally def<strong>in</strong>ed by Wadd<strong>in</strong>gton, epigenetics is: “All those eventswhich lead to the unfold<strong>in</strong>g of the genetic program for development.” This def<strong>in</strong>ition was nonspecificbecause although Wadd<strong>in</strong>gton did not propose or know of any mechanisms toconnect genetics with development, importantly he l<strong>in</strong>ked these two aspects of biology. Theidea was that development of a multicellular organism <strong>in</strong>volv<strong>in</strong>g differentiation of primordialtotipotent cells <strong>in</strong>to specific cells and tissues was like a ball mov<strong>in</strong>g down a landscape ofbranch<strong>in</strong>g valleys to its f<strong>in</strong>al dest<strong>in</strong>ation (Figure 7.1). Independently, the term epigenetics was<strong>in</strong>voked to expla<strong>in</strong> a variety of “bizarre” phenotypic phenomena <strong>in</strong> different organisms thatcould not be expla<strong>in</strong>ed by simple Mendelian genetics.<strong>Epigenetics</strong> evolved to mean modifications to DNA that affected gene expression but do not<strong>in</strong>volve base changes (aka mutations). The term “epi”, mean<strong>in</strong>g “on top of genetics” (withgenetic ¼ DNA), certa<strong>in</strong>ly encompasses changes to DNA and to DNA packag<strong>in</strong>g by histones.The best-studied epigenetic changes are DNA and histone (i.e. chromat<strong>in</strong>) modifications thatmodulate access of macromolecules that regulate gene expression. In addition, modificationsimpact chromosome localization with<strong>in</strong> the nucleus; hence, impact<strong>in</strong>g potential <strong>in</strong>teractionsbetween chromat<strong>in</strong> regions [2,3].128We prefer the broad term “epigenomics” that not only refers to changes to DNA and histones,but also encompasses other non-l<strong>in</strong>ear aspects of the <strong>in</strong>formation transfer from genotype tophenotype. Some of these changes are well known. For <strong>in</strong>stance, we <strong>in</strong>clude somatic DNA basechanges, and chromosomal localization <strong>in</strong> the epigenomic landscape. At the RNA level,epigenomics changes <strong>in</strong>clude alternative splic<strong>in</strong>g, edit<strong>in</strong>g, capp<strong>in</strong>g, methylation, and othermodifications, polyA tail<strong>in</strong>g and non-cod<strong>in</strong>g RNA (ncRNA) regulation. At the prote<strong>in</strong> level,epigenomics changes <strong>in</strong>clude the many post-translational modifications to prote<strong>in</strong> (e.g.phosphorylation, methylation, acetylation, palmitoylation, ubiqu<strong>in</strong>ation, etc.).FIGURE 7.1Wadd<strong>in</strong>gton’s epigenetic landscape.


CHAPTER 7Epigenomics <strong>in</strong> Neurobehavioral <strong>Disease</strong>sSome RNA and prote<strong>in</strong> modifications have been studied for a long time and may not begenerally thought of as part of the epigenomic landscape. However, with<strong>in</strong> our viewpo<strong>in</strong>t, allthese changes represent non-l<strong>in</strong>ear <strong>in</strong>formation transfer and fall under the umbrella ofepigenomics. Today, many epigenomic processes have been identified, some studied <strong>in</strong> somedetail, but many others rema<strong>in</strong> to be discovered.In brief, the term “epigenomics” describes non-l<strong>in</strong>ear transfer of genotype to phenotype, and isa more global and <strong>in</strong>clusive term than epigenetics. In the future, an important issue will behow to <strong>in</strong>tegrate and understand the multitude of genetic, environmental, and epigenomicfactors that contribute to disease phenotypes. Epigenomic programm<strong>in</strong>g is specific for <strong>in</strong>dividualgenes for cell type, time, environment, and history.The amount of epigenomic variation is greater than genetic variation. Besides the amount ofvariation, a second difficulty <strong>in</strong> this research area is how to collect the <strong>in</strong>formation, and a thirdis how to <strong>in</strong>tegrate and analyze the multilevel data. These issues are similar but greater thanthose faced with sequenc<strong>in</strong>g the human genome. Although, there are a large number of reviewson epigenomic programm<strong>in</strong>g much of the needed data does not exist today. This review willfocus on epigenomic changes to chromat<strong>in</strong>.7.3 EPIGENOMIC MODULATION OF CHROMATINDNA methylation is the most studied and best-understood epigenomic process with manyreviews published yearly. DNA methyl transferases (DNMT) transfers methyl (eCH 3 ) groupsfrom S-adenosyl methion<strong>in</strong>e (SAM) to the 5 carbon atom of cytos<strong>in</strong>e residues with<strong>in</strong> thed<strong>in</strong>ucleotide sequence, 5 0 CpG3 0 (Figure 7.2). The ma<strong>in</strong>tenance DNA methylase, DNMT1, actssoon after DNA replication to copy parental strand methylation to the newly synthesized DNAstrand, thus conserv<strong>in</strong>g the DNA methylation pattern through mitosis. DNMT 3a and 3b arel<strong>in</strong>ked to de novo DNA methylation. Passive demethylation may occur when DNA is replicatedbut parental strand methylation is not transferred to the newly synthesized strand.129FIGURE 7.2Epigenomic programm<strong>in</strong>g of chromat<strong>in</strong>. This figure is reproduced <strong>in</strong> the color plate section.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>Usually, the absence of DNA methylation at a promoter region and the presence of DNAmethylation <strong>in</strong> s<strong>in</strong>gly occurr<strong>in</strong>g gene body 5 0 CpG3 0 sites is associated with gene expression[4,5]. The rate of CpG methylation is determ<strong>in</strong>ed by the availability of DNMT enzymes andtheir aff<strong>in</strong>ity for a given CpG, the number of CpGs and the non-CpG DNA sequence with<strong>in</strong>a site, as well as other factors like the level of the methyl donor, and with DNMT1, whether thedouble-stranded DNA is hemimethylated. Cytos<strong>in</strong>e methylation can mediate silenc<strong>in</strong>g of geneexpression through the b<strong>in</strong>d<strong>in</strong>g actions of prote<strong>in</strong>s such as methylated CpG b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s(MeCP1 and 2), and the methyl CpG b<strong>in</strong>d<strong>in</strong>g doma<strong>in</strong> prote<strong>in</strong>s MBD1, MBD2, MBD3, andMBD4 (e.g. [6]).A recent excit<strong>in</strong>g caveat to DNA methylation studies is the discovery of modification to the 5r<strong>in</strong>g position carbon of cytos<strong>in</strong>e <strong>in</strong> addition to methylation. The modifications <strong>in</strong>clude 5-hydroxymethyl cytos<strong>in</strong>e, 5-formyl cytos<strong>in</strong>e, and 5-carboxyl cytos<strong>in</strong>e created by the reiterativeoxidation by the 10e11 translocation family of dioxygenases, TET1, TET2, and TET3 [7e9]. Inan active demethylation process, the modified cytid<strong>in</strong>es are de-am<strong>in</strong>ated produc<strong>in</strong>ga mismatched base pair (T-G) <strong>in</strong> a d<strong>in</strong>ucleotide sequence 5 0 TpG3 0 /3 0 GpC5 0 . Thym<strong>in</strong>e DNAglycosylase (TDG) removes the mismatched thym<strong>in</strong>e, which is replaced by an unmethylatedcytos<strong>in</strong>e residue dur<strong>in</strong>g base excision repair [10,11]. TDG <strong>in</strong>teracts with DNA methylases,histone acetyl transferases, and transcription factors.Hydroxymethylcytos<strong>in</strong>e distribution varies <strong>in</strong> a tissue and cell-type-specific manner with highlevels <strong>in</strong> mouse and human embryonic stem cells [7,12,13]. Hydroxymethyl cytos<strong>in</strong>e isenriched at promoter regions and with<strong>in</strong> gene bodies but the former does not correlate withgene expression, and the latter is more positively correlated with gene expression than 5-methyl cytos<strong>in</strong>e [14].130These recent discoveries raise many <strong>in</strong>terest<strong>in</strong>g questions besides reveal<strong>in</strong>g an active mechanismfor DNA de-methylation. For <strong>in</strong>stance, most past studies on DNA methylation did notdist<strong>in</strong>guished between these modifications; hence, almost all past studies need to be revisited<strong>in</strong> order to clarify what modifications are present.Histones condense genomic DNA to fit with<strong>in</strong> the nucleus of a cell. Modifications to histonesare l<strong>in</strong>ked to different levels of compaction referred to as “open” and “closed” chromat<strong>in</strong> thatallow or prevent access and expression of genes. Histones can be methylated, phosphorylated,acetylated, and/or ubiqu<strong>in</strong>ated (for a recent review see [15]).The nucleosome is composed of an octamer of histones H2A, H2B, H3, and H4. Most histonemodifications occur <strong>in</strong> free am<strong>in</strong>o term<strong>in</strong>al histone tails rather than <strong>in</strong> the globular coreportions of the nucleosome. The H3 tail has the most sites for modification, 36 residues, withsome sites hav<strong>in</strong>g multiple modifications simultaneously (e.g. mono-, di-, or trimethylations).The potential variation <strong>in</strong> H3 variation is ~2 40 potentially provid<strong>in</strong>g ~10 14variants. For the most part, the “histone code” for any process is not known and certa<strong>in</strong>ly therelationship with the newly discovered cytos<strong>in</strong>e codes.Histone H3 lys<strong>in</strong>e 4 methylation (H3K4 me ) and histone H3 lys<strong>in</strong>e 9 acetylation (H3K9 ac )are generally associated with open chromat<strong>in</strong> and gene expression. Demethylated H3K4and deacetylated H3K9 me are associated with closed chromat<strong>in</strong> and lack of gene expression.Generally, these modification states correlate with modification of DNA <strong>in</strong> promoterregions and with gene expression (Figure 7.2). One mechanism for coord<strong>in</strong>at<strong>in</strong>g DNAand histone modification <strong>in</strong>volves a complex that b<strong>in</strong>ds to methylated CpG sites and<strong>in</strong>cludes the methylated CpG b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> 2 (MeCP2), a transcriptional repressiondoma<strong>in</strong> (TRD) prote<strong>in</strong>, co-repressor SIN3 homolog A (SIN3A), and histone deacetylase(HDAC) [16].Valproic acid (VPA), a HDAC <strong>in</strong>hibitor, is used to treat BP as an alternative to lithium, to treatepilepsy, and less commonly migra<strong>in</strong>es, major depression, and schizophrenia [17e19]. In


CHAPTER 7Epigenomics <strong>in</strong> Neurobehavioral <strong>Disease</strong>sutero, exposure to VPA <strong>in</strong>creased the risk for autism and neural tube defects [20] and likelyother neurobehavioral diseases like SZ and ASD l<strong>in</strong>ked to folate deficiencies (see below).Epigenomic programm<strong>in</strong>g is used to regulate gene expression as a function of parental orig<strong>in</strong>.Genes that are programmed by parental orig<strong>in</strong> are called impr<strong>in</strong>ted genes (http://www.impr<strong>in</strong>t.com; http://www.igc.otago.ac.nz). However, chromat<strong>in</strong> codes and epigenomic codesare dynamic and may change <strong>in</strong> response to environmental and developmental cues.There are approximately 80 genes proven and another 200 thought-to-be impr<strong>in</strong>ted <strong>in</strong> the humangenome. Impr<strong>in</strong>t<strong>in</strong>g appears to be regional, with 20 proven regions, and 50 addition putativeregions. In some regions, differential impr<strong>in</strong>t<strong>in</strong>g occurs. Impr<strong>in</strong>t<strong>in</strong>g defects at specific chromosomalregions is associated with neurodevelopmental disorders (see Chapter 8). ASD, and <strong>in</strong>some cases SZ, is co-morbid with several impr<strong>in</strong>ted diseases <strong>in</strong>clud<strong>in</strong>g PradereWilli/Angelmansyndrome, CHARGE, fragile X disease and Rett (RTT) syndromes (see below) [21,22].In meiosis, most paternal histones <strong>in</strong> sperm are replaced with protam<strong>in</strong>es [23]. Hence,paternal epigenomic programm<strong>in</strong>g of chromat<strong>in</strong> appears to be through DNA modification. Incontrast, maternal chromat<strong>in</strong> modifications to DNA and histone can be preserved throughgamete production.Epigenomic complexities <strong>in</strong>clude other differences between males and females. Overall DNAmethylation patterns are different <strong>in</strong> male and female cells [24]. For <strong>in</strong>stance, Shimabukuroet al. [25] found global methylation differences <strong>in</strong> leukocytes from males and females, andreported a slight decrease overall <strong>in</strong> samples from males but not female SZ patients, especially<strong>in</strong> younger <strong>in</strong>dividuals.Females have an extra layer of epigenomic complexity. One X chromosome is <strong>in</strong>activatedthrough epigenomic programm<strong>in</strong>g early <strong>in</strong> development (before the 32-cell stage)(for reviewsee [26]). This ensures that genes on the X chromosome are expressed at the same level <strong>in</strong>female (XX) and male (XY) cells. Usually <strong>in</strong>activation is random; hence, females are functionalmosaics. The <strong>in</strong>active X chromosome can be reactivated with age, and <strong>in</strong> some <strong>in</strong>dividuals<strong>in</strong>activation is not random.131An excess of chromosome X aneuploids have been detected <strong>in</strong> neurobehavioral disorders forsome time <strong>in</strong>clud<strong>in</strong>g schizophrenic patients [27,28]. Recently, systematic re-sequenc<strong>in</strong>g ofsynaptic genes on chromosome X revealed a higher than expected level of rare damag<strong>in</strong>gvariants <strong>in</strong> SZ and ASD patients [29]. Such mutations may account for the <strong>in</strong>creased severityseen <strong>in</strong> male versus female patients because males have s<strong>in</strong>gle copies of chromosome X. Xchromosome <strong>in</strong>activation (especially if not random, as found for SZ and BP) may account fordisease discordance <strong>in</strong> tw<strong>in</strong>s [30]. Both X and autosomal chromosome number abnormalities<strong>in</strong> neurons are detected <strong>in</strong> patients with different neurobehavioral disorders <strong>in</strong>clud<strong>in</strong>g SZ, BPand ASD (for review see [31]).In 2009, Lister et al. [32] reported on methylated sites across the genome. As expected, lowlevels of methylation were found at CpG islands, promoter methylation level was <strong>in</strong>verselyl<strong>in</strong>ked gene expression. Gene body and <strong>in</strong>tergenic regions had high levels of methylation(>70%) and were called highly methylated doma<strong>in</strong>s (HMDS). Other regions were partiallymethylated (


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>RTT syndrome, an ASD disease, is usually due to mutations <strong>in</strong> the methylated cytos<strong>in</strong>e b<strong>in</strong>d<strong>in</strong>gprote<strong>in</strong>, MECP (an X l<strong>in</strong>ked gene), although some cases are due to known mutations <strong>in</strong> othergenes (for review see [37]). In RTT syndrome, usually a de novo mutation occurs <strong>in</strong> theproband, although some cases are due to de novo mutations <strong>in</strong> the paternally <strong>in</strong>herited allele.RTT syndrome is more common <strong>in</strong> females because a mutation <strong>in</strong> the s<strong>in</strong>gle copy of the genepresent <strong>in</strong> males leads to embryonic lethality. MECP2 prote<strong>in</strong> b<strong>in</strong>ds to methylated DNA and<strong>in</strong>hibits transcription factor b<strong>in</strong>d<strong>in</strong>g; hence <strong>in</strong>fluenc<strong>in</strong>g the expression level of many genes.DNA, RNA, and histone epigenomic changes can be analyzed by the same approaches used <strong>in</strong>the past to <strong>in</strong>vestigate DNA sequence variation. These approaches have been converted tosecond-generation ultra-high-throughput methods for collect<strong>in</strong>g large amounts of data.Hence, it is likely that much of the miss<strong>in</strong>g epigenomic data on DNA, RNA, and histones willbe uncovered <strong>in</strong> the near future. Here, we focus on chromat<strong>in</strong> because these are the bestcharacterizedepigenomic changes corrected to neurobehavioral disease. Other epigenomicchanges are now be<strong>in</strong>g collected.7.4 ISSUES UNIQUE TO NEUROBEHAVIORAL DISEASESThere are unique major issues related to neurobehavioral disorders. For <strong>in</strong>stance, lack offund<strong>in</strong>g at least by the US National Institute of Health (NIMH) prevents progress on thesediseases and discourages <strong>in</strong>novative approaches. Another issue is that almost any positiveobservation on these diseases is accompanied by a contradictory negative f<strong>in</strong>d<strong>in</strong>g. Other issues<strong>in</strong>clude the subjective diagnosis and the unknown cause(s) of the common neurobehavioraldisease.132Many of the severe neurobehavioral diseases have overlapp<strong>in</strong>g symptoms and can be viewed aspo<strong>in</strong>ts on a cont<strong>in</strong>uum of phenotypes that share characteristics. And there are so manychanges l<strong>in</strong>ked to neuropsychiatric diseases that it is difficult to dist<strong>in</strong>guish between cause andconsequence. Our approach has been to view these seem<strong>in</strong>gly disparate observations asw<strong>in</strong>dows <strong>in</strong>to a disrupted fundamental cellular process such as that described below.Neurobehavioral diseases are diagnosed from subjective behavioral report<strong>in</strong>g by afflicted<strong>in</strong>dividuals and tra<strong>in</strong>ed observers because objective criterion is not established. SZ is diagnosedmore frequently <strong>in</strong> males and BP <strong>in</strong> females. Males tend to be diagnosed <strong>in</strong> their earlytwenties for SZ and BP while women are diagnosed more frequently <strong>in</strong> their late twenties.Attempts to standardize subjective criteria reach back <strong>in</strong> time to Kraepel<strong>in</strong> and Bleuler <strong>in</strong> theearly 1900s. Generally, two major categories of neurobehavioral disease are recognized: SZ andBP based on symptom group<strong>in</strong>gs, course, and outcome.Today, the classification of neurobehavioral disorders is based on criteria developed by theAmerican Psychiatric Association and published as “Diagnostic and Statistical Manual ofMental Disorders (DSM)” and the ICD (International Criteria of <strong>Disease</strong>; published by WHO).These classification schemes undergo periodic revision with current versions DMS-IV and ICD-10, respectively. Generally, classification is based on qualitative behavioral characteristicsrather than quantitative objective criteria.Symptoms for SZ are divided <strong>in</strong>to positive, negative, and cognitive (Table 7.1). The mostcommon positive symptom is halluc<strong>in</strong>ation. Halluc<strong>in</strong>ations are usually auditory but can bevisual, tactile, olfactory, or gustatory. Positive symptoms reflect traits added to the personality.Negative symptoms such as depression represent behavioral deficits such as flat or bluntedaffect, alogia (poverty of speech), anhedonia (<strong>in</strong>ability to experience pleasure), and asociality.The negative symptoms contribute to poor quality of life, functional disability, and lack ofmotivation, and have been l<strong>in</strong>ked to folate deficiencies (see below). Cognitive symptoms,<strong>in</strong>clud<strong>in</strong>g deficits <strong>in</strong> work<strong>in</strong>g memory and executive function, are related to the ability tofunction <strong>in</strong> society.


CHAPTER 7Epigenomics <strong>in</strong> Neurobehavioral <strong>Disease</strong>sTABLE 7.1 Positive, Negative, and Executive Symptoms <strong>in</strong> SZI. Positive Symptoms (Reality Distortion)1. Unusual thought2. Delusions3. Grandiosity4. Suspiciousness/persecution5. Halluc<strong>in</strong>atory behaviorII. Negative Symptoms (Poverty Syndrome)1. Emotional withdrawal2. Passive, apathetic social withdrawal3. Lack of spontaneity and flow of conversation4. Poor rapport5. Blunted affect6. Motor retardation7. Disturbance of volitionIII. Disorganized Symptoms (Formal Thought Disorder, Odd Behaviors, Cognition)1. Conceptual disorganization e circumstantial speech, loose, tangential, illogical associations2. Incoherent speech e <strong>in</strong>comprehensive sentences3. Poverty of speech content e fluent speech with little <strong>in</strong>formation content4. Inappropriate affect e bizarre behaviorA SZ diagnosis us<strong>in</strong>g the DSMIV requires three components be present. (1) The occurrence oftwo or more of the follow<strong>in</strong>g symptoms for 1 month or longer: delusions, halluc<strong>in</strong>ations,disorganized speech, grossly disorganized behavior (e.g. dress<strong>in</strong>g <strong>in</strong>appropriately, cry<strong>in</strong>gfrequently) or catatonic behavior, negative symptoms (blunted affect (lack or decl<strong>in</strong>e <strong>in</strong>emotional response), alogia (lack or decl<strong>in</strong>e <strong>in</strong> speech), or avolition (lack or decl<strong>in</strong>e <strong>in</strong>motivation)). Occurrence of halluc<strong>in</strong>ations with a s<strong>in</strong>gle voice <strong>in</strong> a runn<strong>in</strong>g commentary ofpatient’s activity, or two or more voices or voices that are “bizarre” can be used s<strong>in</strong>gly fordiagnosis. (2) Social or occupational dysfunction (work, <strong>in</strong>terpersonal relationship, or selfcare)that is significantly lower s<strong>in</strong>ce the onset of symptoms. (3) Disturbances present fora significant amount of time, i.e. cont<strong>in</strong>uous signs for 6 months, with at least 1 month of activesymptoms or less if symptoms disappear with treatment.133Today, there is movement to develop and implement objective criteria to patient classificationbased on “endophenotypes” or subphenotypes that can be measured by behavioral, physiological,and DNA test<strong>in</strong>g. Endophenotypes are: (1) heritable characteristics that co-segregatewith disease <strong>in</strong> a family; (2) disease state <strong>in</strong>dependent (i.e. present whether disease is active or<strong>in</strong>active); and (3) present <strong>in</strong> a higher than expected frequency <strong>in</strong> families segregat<strong>in</strong>g disease[38e40]. Endophenotypes for SZ are listed <strong>in</strong> Box 7.1. Today, several companies claim todiagnose SZ with ~85% accuracy based on genetic and/or endophenotypic test<strong>in</strong>g. Thisaccuracy level is <strong>in</strong>adequate given the devastat<strong>in</strong>g consequences of receiv<strong>in</strong>g a SZ diagnosis.BP-related mood disorders <strong>in</strong>cludes episodes of severe and mild to moderate mania, andsevere and mild to moderate depression; some episodes may be mixed episodes. In a manicperiod heightened energy, creativity, and euphoria are common, accompanied by hyperactivity,little sleep, and feel<strong>in</strong>gs of be<strong>in</strong>g all-powerful, <strong>in</strong>v<strong>in</strong>cible, and dest<strong>in</strong>ed for greatness.Dur<strong>in</strong>g this period the <strong>in</strong>dividual may spiral out of control and engage <strong>in</strong> reckless selfdamag<strong>in</strong>gdecisions <strong>in</strong>volv<strong>in</strong>g gambl<strong>in</strong>g, other f<strong>in</strong>ancial activity, sexual activity, etc.ASD is a group<strong>in</strong>g of diseases <strong>in</strong> the DMS-IV- text revision (DMS-IV-TR) called pervasivedevelopmental disorder (PDD) [42]. Besides autism, PDD syndromes <strong>in</strong>clude: Aspergersyndrome, Rett syndrome, childhood dis<strong>in</strong>tegrative disorder, and pervasive developmental


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>BOX 7.1 BEHAVIORAL ENDOPHENOTYPES IN SZ [41]Prepulse <strong>in</strong>hibitionP50 suppressionAntisaccadeCont<strong>in</strong>uous performanceLetterenumber spanVerbal learn<strong>in</strong>gAbstractionFace memorySpatial process<strong>in</strong>gSensorimotor dexterityEmotion recognitiondisorder not otherwise specified. The three behaviors used for diagnosis of ASD <strong>in</strong>clude defects<strong>in</strong> (1) social <strong>in</strong>teractions, (2) communication and imag<strong>in</strong>ative play, and (3) <strong>in</strong>terests andactivities. The age of onset is


CHAPTER 7Epigenomics <strong>in</strong> Neurobehavioral <strong>Disease</strong>strauma, subarachnoid hemorrhage, cerebral tumors, cerebral <strong>in</strong>fection (e.g. syphilis, HIV, etc.),demyel<strong>in</strong>at<strong>in</strong>g diseases, and Wilson’s disease (where excess copper accumulates <strong>in</strong> the bra<strong>in</strong>and other tissues) are l<strong>in</strong>ked to SZ. Traumatic head <strong>in</strong>jury <strong>in</strong>creases risk for SZ by 2e5-fold,although disease may emerge up to 20 years later. Vitam<strong>in</strong> B12 (cobalam<strong>in</strong>) deficiencyand thyroid disease are associated usually with depressive disorder but also with SZ (seebelow). Other rare genetic diseases l<strong>in</strong>ked to secondary SZ are HD, Friedreich’s ataxia(a tr<strong>in</strong>ucleotide repeat disease, see below), and PradereWilli syndrome. Psychosis is l<strong>in</strong>ked toof stimulants like amphetam<strong>in</strong>e, phencyclid<strong>in</strong>e (PCP), lysergic acid diethylam<strong>in</strong>e (LSD), or 3-4-methylenedioxymethamphetam<strong>in</strong>e (ecstasy) (see below).An estimated 5e15% of SZ patients have underly<strong>in</strong>g undiagnosed illness [47]. Whenconventional medical test<strong>in</strong>g of <strong>in</strong>dividuals suspected or given a psychiatric diagnosis is notdone, medical conditions <strong>in</strong> these patients go untreated. The push for rout<strong>in</strong>e health assessmentand care for patients with neurobehavioral disorders is <strong>in</strong>creas<strong>in</strong>g (for <strong>in</strong>stance, see [48]).Medical test<strong>in</strong>g is critical for help<strong>in</strong>g patients with underly<strong>in</strong>g pathologies that impactsymptoms, or that impair the quality of life, and for research purposes where disease andsymptoms need to be clearly def<strong>in</strong>ed.SZ is a systemic disease l<strong>in</strong>ked to loss of taste and/or smell [49e52], palate and/teethabnormalities [53e55], m<strong>in</strong>or dysmorphic features [56,57], and <strong>in</strong>creased risk for metabolicsyndrome <strong>in</strong>clud<strong>in</strong>g diabetes [58]. BP has not been l<strong>in</strong>ked to such systemic abnormalities,whereas immunological and gut abnormalities have been detected <strong>in</strong> some ASD patients.The possible outcomes of SZ are varied (Figure 7.3). Some disease presentations are episodicand others have spontaneous remission. These observations cannot be accounted for bygenetics alone. Few studies have rigorously exam<strong>in</strong>ed the environmental factors that can leadto remission, although recently consensus standards for remission and recovery are be<strong>in</strong>gdeveloped to facilitate treatment and research [59].Besides the paucity of systematic long-term studies on neurobehavioral patients and few, if any,genetic studies have attempted to group observations <strong>in</strong>to primary and secondary SZs. This is135FIGURE 7.3Outcome of schizophrenia diagnosis (adapted from http://www.science.org.au). This figure is reproduced <strong>in</strong> the color platesection.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>unfortunate because patients without severe bra<strong>in</strong> damage may present opportunities forreversal of disease, perhaps through epigenomic manipulation. Clearly, the development ofnew and effective treatment modalities will require further dissection of neurobehavioraldisease types.7.5 GENETICSGenetic dissection of rare s<strong>in</strong>gle gene diseases with high penetrance is effective, providedfamilies and funds are available. Penetrance describes the level of phenotypic expression ofa genetic trait <strong>in</strong> <strong>in</strong>dividuals with a causal mutation. For <strong>in</strong>stance, HD, with 8000patients and >7000 controls. Most meta-analyses have not been this successful, nor been thisextensive.The causes of disappo<strong>in</strong>t<strong>in</strong>g genetic results can be true false positives from small sample sizesand <strong>in</strong>adequate statistical criteria; true heterogeneity <strong>in</strong> disease and/or <strong>in</strong>adequate gene test<strong>in</strong>g.In the past, gene SNP test<strong>in</strong>g focused on known “functional” SNPs with detected phenotypicconsequences. However, any variation <strong>in</strong> a gene is likely to have some phenotypic consequence.Perhaps the application of second-generation ultra-high-throughput sequenc<strong>in</strong>g forcomplete genome and/or complete gene sequenc<strong>in</strong>g will clarify gene association results.Genome side association studies (GWAS) studies analyze SNPs across the genome both with<strong>in</strong>and outside of genes. These studies are generally used to look for l<strong>in</strong>kage disequilibriumbetween genes, i.e. haplotypes and phenotype. A surprise of GWAS studies was the detection ofa slight elevation <strong>in</strong> the occurrence of CNVs <strong>in</strong> SZ, BP, and ASD (for review see [64]). The CNVs<strong>in</strong>clude small and large regions of the genome, and some are common across the multipledisorders (e.g. [66]). De novo mutations are detected as CNVs [67] and with<strong>in</strong> exons [68], andsupport the notion proposed by us <strong>in</strong> 2003 [69] that genome <strong>in</strong>stability is a characteristic of SZ(see below). The absence of strong genetic l<strong>in</strong>ks <strong>in</strong> SZ is similar to f<strong>in</strong>d<strong>in</strong>gs on other commonillnesses like cardiovascular disease and cancer.The results of these genetic studies have led to several generalities. The risk for develop<strong>in</strong>g SZ,BP, or ASD is generally proportional to the degree of genetic relationship with an afflicted<strong>in</strong>dividual. These diseases have high heritability but low penetrance. Low penetrance isattributed to several factors <strong>in</strong>clud<strong>in</strong>g: (a) epistasis (<strong>in</strong>teraction between genes, i.e. polygenic


CHAPTER 7Epigenomics <strong>in</strong> Neurobehavioral <strong>Disease</strong>sdisease); (2) de novo confound<strong>in</strong>g germl<strong>in</strong>e or somatic mutations; and (c) epigenomicprogramm<strong>in</strong>g changes <strong>in</strong>clud<strong>in</strong>g geneeenvironment <strong>in</strong>teractions.Epistasis is commonly <strong>in</strong>voked to expla<strong>in</strong> the large number of genes l<strong>in</strong>ked to neurobehavioralillnesses. There are many possible types of epistatic <strong>in</strong>teractions with different outcomes.Interact<strong>in</strong>g genes may code for prote<strong>in</strong>s or non-cod<strong>in</strong>g RNAs (ncRNAs) with<strong>in</strong> the same,a redundant, or alternative pathway. Other mutations affect<strong>in</strong>g epistasis may decrease or<strong>in</strong>crease prote<strong>in</strong> activity and affect which pathway step is rate-limit<strong>in</strong>g. Mutation may lead toa loss, ga<strong>in</strong>, or change of function, each with different consequences.Despite the spectrum of possible epistatic affect, epistasis does not account for some geneticobservation on neurobehavioral illness. For <strong>in</strong>stance, epistasis cannot expla<strong>in</strong> the discordancerates of disease <strong>in</strong> monozygotic tw<strong>in</strong>s. Discordance <strong>in</strong> monozygotic tw<strong>in</strong>s has traditionallybeen used to dist<strong>in</strong>guish between genetic and environmental components of disease especiallywhen tw<strong>in</strong>s are raised apart. However, although monozygotic tw<strong>in</strong>s share the highestpercentage of their genome, their genomes are not identical because of somatic and epigenomicchanges.Monozygotic tw<strong>in</strong>s that develop from the same fertilized egg are discordant for neurobehavioraldisease at levels that are significantly below 100%, i.e. arguably ~50% (with studiesrang<strong>in</strong>g from ~35% to ~80% for SZ and BP, and ~65% for ASD [70]). Variation <strong>in</strong> concordancerates may be due to several factors <strong>in</strong>clud<strong>in</strong>g differences <strong>in</strong> ascerta<strong>in</strong>ment of disease and/ortw<strong>in</strong>ship. More recently studies have used standardized methods for disease diagnosis andquantitative, rather than qualitative, assessment of tw<strong>in</strong>ship. However, conclusions fromquantitative assessment results can vary also (e.g. [71]) and concordant illness <strong>in</strong> monozygotictw<strong>in</strong>s may not have the same presentation and course.Odds Ratio13701 2 3 4 56 7 8 9 10Paternal ageW<strong>in</strong>terPlace/time of birthUrbanInfluenzaRespiratoryInfection RubellaPoliovirusCNSFam<strong>in</strong>eBereavementPrenatalFloodUnwantednessMaternal deprRh <strong>in</strong>compatibilityHypoxiaObstetricCNS damageLow birth weightPre-eclampsiaFamily historyDOI: 10.1371/Journal.pmed.0020212.g001FIGURE 7.4Odds ratio for schizophrenia as a function of genetic and environmental risk factors (adapted from [73]). This figure is reproduced <strong>in</strong> the color plate section.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>The prevalence of SZ varies from 0.8e2.0% <strong>in</strong> different countries [72]. Some patients comefrom families where related <strong>in</strong>dividuals have the same or a related neurobehavioral disorder(or an associated endophenotype); others have, arguably, no family history (aka spontaneousforms of illness). Despite the high heritability (Figure 7.4) of SZ, prelim<strong>in</strong>ary model<strong>in</strong>gexperiments <strong>in</strong>dicate the rate of spontaneous forms of illness is responsible for ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>gthe level of the disease <strong>in</strong> the population over many generations. This result underscores theimportance of prevention through environmental <strong>in</strong>terventions.7.6 ENVIRONMENTEnvironmental factors l<strong>in</strong>ked to SZ and BP occur early <strong>in</strong> development (for recent reviews see[74,75]) whereas the age of disease onset for SZ and BP is typically young adulthood (seeabove). Early life environmental impacts must persist over many years <strong>in</strong> order to impactdisease phenotype (see Figures 7.4 and 7.5). Figure 7.4 provides the odds ratio of well-studiedfactors for SZ. Figure 7.5 provides developmental time w<strong>in</strong>dows where environmentalcomponents appear to impact subsequent disease development. Environmental factors early<strong>in</strong> life are l<strong>in</strong>ked to other common diseases. Early life periods have uniquely high levels ofDNA replication and epigenomic programm<strong>in</strong>g.138Studies go<strong>in</strong>g back to 1958 l<strong>in</strong>k elder fathers as a risk factor for SZ <strong>in</strong> progeny with a 3-fold<strong>in</strong>crease associated with fathers >50 years of age (for review see [76]). The <strong>in</strong>crease is l<strong>in</strong>earwith paternal age. Similar observations are found for BP77 and ASD78. In fact, paternal age isgenerally l<strong>in</strong>ked to negative neurobehavioral outcomes [79]. Increased paternal age is associatedwith <strong>in</strong>creases <strong>in</strong> other diseases such as Down syndrome; neural tube defects (seediscussion of folate below); congenital cataracts; and reduction defects of the upper limb[80,81], with the greatest affect seen <strong>in</strong> autosomal dom<strong>in</strong>ant mutations [82e84]. All paternalage affects are likely DNA-mediated and due to the high levels of DNA replication and theassociated epigenomic programm<strong>in</strong>g (see below).Undernutrition (general caloric or prote<strong>in</strong> deficiency) and malnutrition (deficiencies <strong>in</strong>specific elements, e.g. folic acid, z<strong>in</strong>c, copper, etc.) occur worldwide and are the most commondiseases of childhood and prenatal life. Moderate to severe undernutrition occurr<strong>in</strong>g prior to 2years of age is associated with persistent behavioral and cognitive deficits that resist nutritionalrehabilitation [85]. Pregnant mothers exposed to fam<strong>in</strong>e or malnourished (especially forfolate), notably <strong>in</strong> the second trimester (e.g. [86]), have an <strong>in</strong>creased risk for children with SZ(Figure 7.4). Prenatal nutrition is l<strong>in</strong>ked to problems <strong>in</strong> dopam<strong>in</strong>e-mediated behaviors anddopam<strong>in</strong>e receptor b<strong>in</strong>d<strong>in</strong>g <strong>in</strong> adults [87].Perhaps some environmental factors shown <strong>in</strong> Figures 7.4 and 7.5 and l<strong>in</strong>ked to disease arebystander markers for poor nutrition. The immune response to an <strong>in</strong>fection agent, ora response to oxidative conditions imposes additional nutritional requirement on a cell andorganism (see below). Measles (rubella <strong>in</strong>fections), associated with severe immune responses,early <strong>in</strong> pregnancy <strong>in</strong>creases the risk for SZ. Other <strong>in</strong>fectious agents l<strong>in</strong>ked to SZ <strong>in</strong>clude<strong>in</strong>fluenza, herpes simplex virus type 2 (HSV-2), Toxoplasma gondii, as well as elevated elevationof <strong>in</strong>flammatory markers l<strong>in</strong>k <strong>in</strong>terleuk<strong>in</strong>-8 (IL-8) early <strong>in</strong> pregnancy, and tumor necrosisfactor (TNF) alpha at birth (for review see [88]). Fam<strong>in</strong>e, <strong>in</strong>fection, hypoxia, and bra<strong>in</strong> damagemight be classified as secondary SZs (see above). The simultaneous occurrence of multiple riskfactors (genetic and/or environmental) may be necessary when <strong>in</strong>dividual risk factors havemild effects.Nutrition may underlie some maternal and rear<strong>in</strong>g environment stress factors. Stress may leadto poor nutrition, as well as <strong>in</strong>creases <strong>in</strong> cytok<strong>in</strong>es. Chronic “defeat stress” is l<strong>in</strong>ked todecreased levels of HDAC-5 <strong>in</strong> the nucleus accubens, long-lasted decreases <strong>in</strong> bra<strong>in</strong>-derivedneurotrophic factor (BDNF) transcription, and aberrant epigenomic changes [74]. Althoughearly life factors appear to be paradoxical, all have the ability to <strong>in</strong>fluence mutation rates and


CHAPTER 7Epigenomics <strong>in</strong> Neurobehavioral <strong>Disease</strong>sFIGURE 7.5Developmental periods affected by environmental factors l<strong>in</strong>ked to SZ, BP, and ASD that impact the epigenome (adapted from [74]).139epigenomic programm<strong>in</strong>g <strong>in</strong> germl<strong>in</strong>e and somatic cells, and be impacted by nutrition (seebelow).Generally, nutrition is an under-considered factor for phenotype (and <strong>in</strong> medic<strong>in</strong>e). Fewstudies have exam<strong>in</strong>ed the affect of nutrition on other risk factors for disease. Further, theimportance of nutrition <strong>in</strong> prevent<strong>in</strong>g or reduc<strong>in</strong>g symptoms <strong>in</strong> neurobehavioral disorders isunder-appreciated and poorly translated to the cl<strong>in</strong>ic although evidence that nutrition isa critical component for bra<strong>in</strong> development and function is well documented [89].Cannabis and drugs are clear risk factors for SZ and other neurobehavioral disorders. Evidencesuggests the cannabis risk is limited to <strong>in</strong>dividuals with the VAL158 allele of the enzymecatechol-O-methyl transferase (COMT) [74]. Tetrahydrocannab<strong>in</strong>ol (THC), the active <strong>in</strong>gredientof cannabis, <strong>in</strong>duces expression of HDAC3 [90]. Abuse of stimulants like coca<strong>in</strong>e,amphetam<strong>in</strong>e, and phenycyclid<strong>in</strong>e are l<strong>in</strong>ked to neurobehavioral disorders (positive andcognitive deficits), and long-term effects of drug use lead to bra<strong>in</strong> changes similar to those seen<strong>in</strong> neurobehavioral disorders, and a sensitization to low dose exposures.7.7 GENOMIC INSTABILITYAt the molecular level, severe neurobehavioral disorders most resemble cancer. Both disordersare l<strong>in</strong>ked to DNA <strong>in</strong>stability (i.e. replication and repair) and aberrant chromat<strong>in</strong> modificationsthroughout the genome. These processes are closely l<strong>in</strong>ked at both the macromolecularlevel and at the metabolic level (see below).The first neurobehavioral diseases l<strong>in</strong>ked to genomic <strong>in</strong>stability were the tr<strong>in</strong>ucleotide repeatsdiseases (for recent review see [91]; Table 7.3). The disease mutations <strong>in</strong>volved <strong>in</strong>crease(“expansions”) <strong>in</strong> tr<strong>in</strong>ucleotide repeat<strong>in</strong>g sequences, usually (CCG) n or (CAG) n located


140TABLE 7.3 Tr<strong>in</strong>ucleotide Repeat <strong>Disease</strong>s<strong>Disease</strong> 1 Sequence Gene 2 Parent of Orig<strong>in</strong> 3 Repeat Length (n) <strong>Disease</strong> SomaticInstabilityNormalPermutation<strong>Disease</strong>s with Cod<strong>in</strong>g Tr<strong>in</strong>ucleotide RepeatsDRLPA CAG ATN1(exon5) P 6-35 35-48 49-88 YesHD CAG HTT (exon1) P 6-29 29-37 38-180 YesOPMD CGN PABPN1 (exon 1) P & M 10 12-17 >11 None is tissuetested (hypothala mus)SCA1 CAG ATXN1 (exon 8) P 6-39 40 41-83 YesSCA2 CAG ATXN2 (exon 1) P 460SCA17 CAG TBP (exon3) P>M 25-42 43-48 45-77 YESSMBA CAG AR (exon 1) P 13-31 32-39 40<strong>Disease</strong>s with Non-Cod<strong>in</strong>g Tr<strong>in</strong>ucleotides RepeatsDM1 CTG DMPK (3’ UTR) M 5-37 37-50 >50DM2 CCTG CNBP (<strong>in</strong>tron 1) Unknown 200FRDA GAA FXN (<strong>in</strong>tron 1) Recessive 5-30 31-100 70-1,000 YesFXS CGG FMR1 (5’ UTR) M 6-50 55-200 >200HDL2 CTG JPH# (exon2A) M 6-27 29-35 36-57SCA8 CTG ATXN*OS (3’ UTR) M 15-34 34-89 89-250SCA10 CATTCT ATXN10 (<strong>in</strong>tron 9) M & P but smaller 10-299 29-400 400-4,500 Yeschanges with MSCA12 CAG PPP2R2B (5’UTR) M&Pbut more unstable with P7-28 28-56 66-78 None found<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>


CHAPTER 7Epigenomics <strong>in</strong> Neurobehavioral <strong>Disease</strong>soutside or with<strong>in</strong> cod<strong>in</strong>g regions, respectively. Expansions outside of cod<strong>in</strong>g regions areusually larger than those with<strong>in</strong> cod<strong>in</strong>g regions, most likely because the latter are constra<strong>in</strong>edby prote<strong>in</strong> function.The first described and best studied repeat disease is FRAX disease e the most frequent cause ofhereditary mental retardation (for review see [92]). FRAX disease, caused by a CGG tr<strong>in</strong>ucleotiderepeat <strong>in</strong> the 3 0 promoter region of the FMR1 gene, is the most common genetic causeof autism, and is associated with SZ. <strong>Disease</strong> occurs when the repeat number becomes greaterthan 230 but can range up to 2000 copies. Between 200 and 230 repeats, disease occurrence isassociated with hypermethylation of the promoter DNA region. FRAX disease is diagnosed bydetection of the FRAXA site, cytologically, on metaphase chromosomes at Xp27 from cellsgrown <strong>in</strong> culture medium deficient <strong>in</strong> folate (see below). The site appears as broken chromosomalregions on metaphase chromosomes. Usually, the chromosome is not broken butthe DNA with<strong>in</strong> the region of site is unreplicated.HD is an example of a tr<strong>in</strong>ucleotide repeat disease with a (CAG) n expansion with<strong>in</strong> a cod<strong>in</strong>gregion of a specific gene. CAG repeat<strong>in</strong>g sequences with<strong>in</strong> exons usually code for the am<strong>in</strong>oacid glutam<strong>in</strong>e.SZ is associated with specific fragile sites <strong>in</strong> some families [28,69]. In addition, a higher thanexpected frequency of genes or regions l<strong>in</strong>ked to SZ co-localize with fragile site regions. Inaddition, the distribution of repeat sizes <strong>in</strong> multiple genes are skewed towards larger but notexpanded size <strong>in</strong> SZ [69,93].Fragile sites are unusual heritable elements. The sites must be <strong>in</strong>duced <strong>in</strong> grow<strong>in</strong>g cells underconditions that <strong>in</strong>terfere with DNA replication. When <strong>in</strong>duced, the sites appear only <strong>in</strong> a smallnumber of chromosomes, i.e. 5% and


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>An <strong>in</strong>creas<strong>in</strong>g number of genetic and epigenetic changes <strong>in</strong> lymphocytes are detected <strong>in</strong> SZ, BP,and ASD, as more laboratories use lymphocytes as sent<strong>in</strong>els of disease. For <strong>in</strong>stance, Uranovaet al. [98] found ultrastructure abnormalities that <strong>in</strong>cluded an <strong>in</strong>creased frequency <strong>in</strong> largeactivated lymphocytes and small mitochondria <strong>in</strong> samples from schizophrenic patients.Cytogenetic studies on fragile site <strong>in</strong>duction have revealed an <strong>in</strong>creased sensitivity to folatedeprivation, here-to-fore unknown fragile sites <strong>in</strong> psychiatric, SZ, and BP patients [99e102].Aberrant DNA damage signal<strong>in</strong>g response reflected as abnormal histone phosphorylationlevels, with unchanged repair rates were detected by Catts et al. [103]. Decondensed chromat<strong>in</strong>changes [104], lower levels of DNA methylation overall and <strong>in</strong> male schizophrenic patients[25] an <strong>in</strong>creased expression of high mobility group A prote<strong>in</strong> 1A (HMGA1), were welldocumented<strong>in</strong> transcription regulator <strong>in</strong>teract<strong>in</strong>g with chromat<strong>in</strong> [105].Several groups are exam<strong>in</strong><strong>in</strong>g the utility of us<strong>in</strong>g whole gene expression <strong>in</strong> blood cells fordiagnos<strong>in</strong>g SZ (for review see [106]). Changes <strong>in</strong> gene expression of specific genes l<strong>in</strong>ked to SZl<strong>in</strong>ked to have been detected, e.g. RELN receptors [107,108]. These cells are also used forfunctional studies. James et al. [109] reported abnormalities <strong>in</strong> the response of lymphoblastoidcells from autistic patient to oxidative stress metabolites (see below). The results are excit<strong>in</strong>gbecause they may provide quantitative diagnostic tests, as well as means of study<strong>in</strong>g epigenomicchanges relevant to bra<strong>in</strong> disease <strong>in</strong> a readily available tissue.1427.8 TRANSCRIPTIONAL DYSREGULATIONRecent studies from several groups, <strong>in</strong>clud<strong>in</strong>g us, have l<strong>in</strong>ked aberrant chromat<strong>in</strong> changes andgene expression to different neurobehavioral diseases. Costa and colleagues have focused onthe GABAergic neurons (see above). Our own work focused on develop<strong>in</strong>g a multi-levelunderstand<strong>in</strong>g of dopam<strong>in</strong>e metabolism <strong>in</strong> the synaptic cleft.We analyzed samples from the prefrontal cortex of <strong>in</strong>dividuals with SZ and BP versus unaffectedcontrols. In a series of manuscripts (e.g. [110e112]), we reported on: the promotermethylation status; transcript level and presence of SNPs of genes important <strong>in</strong> dopam<strong>in</strong>emetabolism <strong>in</strong>clud<strong>in</strong>g membrane-bound catechol-O-methyl transferase (MB-COMT) andRELN previously found to be hypermethylated <strong>in</strong> SZ [113]; and the dopam<strong>in</strong>e receptors(DRD) 1 and 2.Our results revealed that patient samples had significantly lower levels of MB-COMT promotermethylation than controls, particularly <strong>in</strong> the left frontal lobe, and that the level of methylation<strong>in</strong>versely correlated with mRNA expression [111]. The VAL158MET polymorphismwith<strong>in</strong> the MB-COMT gene is correlated with prote<strong>in</strong> activity, with the VAL allele codes fora hyperactive prote<strong>in</strong> that leads to excess dopam<strong>in</strong>e degradation [114]. The val<strong>in</strong>e (VAL) allelewas present <strong>in</strong> more SZ patients, and the VAL/VAL genotype more common <strong>in</strong> both SZ and BPpatients than <strong>in</strong> well controls, although other studies report mixed results. Most <strong>in</strong>dividualswith MB-COMT promoter hypomethylation and the VAL/VAL genotype were <strong>in</strong> the patientgroups (SZ and BD), while most cases with methylated MB-COMT promoters and MET/METgenotypes were <strong>in</strong> the control group.In the same samples, hypomethylation of the MB-COMT and DRD2 promoters almost alwayscorrelated. This implies that synaptic dopam<strong>in</strong>e deficiency result<strong>in</strong>g from overexpression ofMB-COMT was usually compensated for by DRD2 overexpression. However, DRD2 hypomethylationwas significantly lower <strong>in</strong> the patient samples than <strong>in</strong> controls, possibly reflect<strong>in</strong>ga reduced ability to compensate for low levels of dopam<strong>in</strong>e <strong>in</strong> the synaptic cleft. Correlationswere detected with RELN expression and promoter methylation status [111,115].Understand<strong>in</strong>g even a relatively simple function like dopam<strong>in</strong>e metabolism requires multileveldata on DNA sequences, chromat<strong>in</strong> modifications, RNA expression and structure, prote<strong>in</strong>expression and structure, metabolite levels, and phenotype. As discussed above, our own


CHAPTER 7Epigenomics <strong>in</strong> Neurobehavioral <strong>Disease</strong>sstudies have begun to dissect multilevel dopam<strong>in</strong>e metabolism regulation. Integration andunderstand<strong>in</strong>g of any process will require complex mathematical model<strong>in</strong>g of function such asthose pioneered by Reed and colleagues [116] for folate, and by Voit and colleagues [117] fordopam<strong>in</strong>e metabolism.7.9 RNA EPIGENOMICSOther epigenomic modulations known to occur <strong>in</strong> the bra<strong>in</strong> and associated with neurobehavioraldisorders are at the RNA and prote<strong>in</strong> level. Of the known modifications, the mostadvanced studies are focused on alternative splic<strong>in</strong>g. The BDNF gene, l<strong>in</strong>ked to dopam<strong>in</strong>emetabolism (see above) and many neurobehavioral diseases, is a strik<strong>in</strong>g example of theamount of variation that is likely present <strong>in</strong> genes expressed <strong>in</strong> the bra<strong>in</strong>. The BDNF gene has11 exons, n<strong>in</strong>e promoters, >30 splice variants, and antisense regulation [118]. Some variation<strong>in</strong> BDNF RNA process<strong>in</strong>g is likely l<strong>in</strong>ked to neurobehavioral diseases. Disrupted <strong>in</strong> schizophrenia1(DISC1) mRNA splice variants are up-regulated and associated with risk polymorphisms<strong>in</strong> SZ [119]. Splice variations are detected <strong>in</strong> prote<strong>in</strong> regulator of G prote<strong>in</strong>signal<strong>in</strong>g 4 (RFS4) <strong>in</strong> SZ but not BP [120].RNA edit<strong>in</strong>g and alternative splic<strong>in</strong>g changes are l<strong>in</strong>ked to the seroton<strong>in</strong> 2C receptor <strong>in</strong> SZ[121]. Other RNA edit<strong>in</strong>g will be uncovered given that the adenos<strong>in</strong>e deam<strong>in</strong>ases (ADARs) arema<strong>in</strong>ly expressed <strong>in</strong> the nervous system (for review see [122]). microRNA (miRNA) dysregulationoccurs <strong>in</strong> SZ, BP, and ASD (for reviews see [123,124]). Although the function of mostnon-cod<strong>in</strong>g RNAs (ncRNAs) is not known yet, their l<strong>in</strong>k to chromat<strong>in</strong> modification, genomic<strong>in</strong>stability impr<strong>in</strong>t<strong>in</strong>g, X-chromosome, and gene expression argues for their importance <strong>in</strong>a number of processes l<strong>in</strong>ked to disease [125]. Today we are see<strong>in</strong>g the beg<strong>in</strong>n<strong>in</strong>g of tantaliz<strong>in</strong>glevel of here-to-fore unknown RNA complexity.7.10 METABOLISMA key <strong>in</strong>tracellular metabolic hub important for genomic stability and epigenomic programm<strong>in</strong>gis the crossroads (Figure 7.6) of the folateemethion<strong>in</strong>eetransulfurationedopam<strong>in</strong>e(FMTD hub) pathways (for review see [28]). The FMTD hub directly l<strong>in</strong>ks the synthesis of DNAand RNA precursor and metabolites used for energy transduction (i.e. ATP and GTP) tometabolites important for epigenomic programm<strong>in</strong>g, cellular response to oxidative stress, anddopam<strong>in</strong>e metabolism.143DNA and RNA precursor syntheses require folate as a methyl donor. Specifically, the formationof TTP from UTP by thymidylate synthase requires folate. Further, two steps <strong>in</strong> the de novoformation of <strong>in</strong>os<strong>in</strong>e monophosphate (IMP) utilizes folate as a methyl donor. IMP isa precursor for the synthesis all other pur<strong>in</strong>es. Given all the data that po<strong>in</strong>t towards abnormalities<strong>in</strong> the FMTD hub it is not surpris<strong>in</strong>g that homeostatic pur<strong>in</strong>e imbalances are present<strong>in</strong> naive first-episode SZ patients [126].SAM is produced <strong>in</strong> the methion<strong>in</strong>e cycle from exogenous methion<strong>in</strong>e and ATP. SAM is themajor <strong>in</strong>tracellular methyl donor that acts as a cofactor for over 100 methyl transferases,<strong>in</strong>clud<strong>in</strong>g enzymes that participate <strong>in</strong> epigenomic regulation through modification of DNAand RNA, prote<strong>in</strong>s, <strong>in</strong>clud<strong>in</strong>g histones, and <strong>in</strong> other catabolic and anabolic reactions such asthose that degrade dopam<strong>in</strong>e and nicot<strong>in</strong>e. Clearly, <strong>in</strong>terference <strong>in</strong> SAM production will havea major impact on a cell.In the methion<strong>in</strong>e cycle, demethylated SAM is broken down <strong>in</strong>to the key metabolitess-adenosyl homocyste<strong>in</strong>e (SAH) and homocyste<strong>in</strong>e (HCY). HCY is re-methylated by theenzyme beta<strong>in</strong>e homocyste<strong>in</strong>e methyl transferase (BHMT) or the enzyme methion<strong>in</strong>e synthase(MS), to reform methion<strong>in</strong>e. The enzyme BHMT transfers a methyl group from beta<strong>in</strong>e toHCY. Beta<strong>in</strong>e or its precursor chol<strong>in</strong>e is obta<strong>in</strong>ed from the diet. Chol<strong>in</strong>e can also be obta<strong>in</strong>ed


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>144FIGURE 7.6The FMTD hub: Metabolic l<strong>in</strong>k between DNA and RNA synthesis, energy production, epigenomic programm<strong>in</strong>g, oxidative stress responses, <strong>in</strong>flammation, anddopam<strong>in</strong>e metabolism (not all metabolic steps are shown). This figure is reproduced <strong>in</strong> the color plate section.from degradation of the cell membrane. Besides be<strong>in</strong>g used to reform methion<strong>in</strong>e, beta<strong>in</strong>e isused for the synthesis of phospholipids.The MS enzyme is a cobalam<strong>in</strong> (vitam<strong>in</strong> B12)-dependent enzyme sensitive to oxidativeconditions, transfers a methyl groups from a folate derivative, 5-methyl tetrahydrofolateformed <strong>in</strong> the folate cycle. Curiously, demethylated (MET313HCY) dopam<strong>in</strong>e receptor D4(DRD4) competes with HCY as a methyl acceptor from MS. Methylated DRD4 acts as a methyltransferase by methylat<strong>in</strong>g membrane phospholipids and caus<strong>in</strong>g a change <strong>in</strong> membranepermeability. Our recent unpublished studies revealed that the DRD4 promoter is hypomethylatedand the gene overexpressed <strong>in</strong> bra<strong>in</strong> samples from SZ and BP patients.HCY is the <strong>in</strong>put metabolite <strong>in</strong>to the transulfuration pathway. The transulfuration pathwaynot only synthesizes glutathione (GSH), the primary <strong>in</strong>tracellular antioxidant, but alsothe am<strong>in</strong>o acid cyste<strong>in</strong>e <strong>in</strong> mammals. Two rate-limit<strong>in</strong>g enzyme steps require anotheressential vitam<strong>in</strong>, B6, <strong>in</strong> the transulfuration cycle. Inhibition of MS activity by oxidativestress directs HCY towards the transulfuration pathway and away from the production ofSAM.Methion<strong>in</strong>e re-cycl<strong>in</strong>g through the methion<strong>in</strong>e cycle depends on the am<strong>in</strong>o acid’s availability,and metabolic need. Reed and colleagues have pioneered mathematical model<strong>in</strong>g of this hub(e.g. [127e130]) and applied this model to understand<strong>in</strong>g disease [128,131,132] like SNPsl<strong>in</strong>ked to autism [133], cellular processes like DNA methylation [129], and toxic arsenicexposures [134]. These k<strong>in</strong>ds of efforts are needed to understand the multitude of factorsl<strong>in</strong>ked to neuropsychiatric disease.In summary, the FMTD hub depends on five nutrients (methion<strong>in</strong>e, vitam<strong>in</strong>s B9, B12, and B6,and beta<strong>in</strong>e (or chol<strong>in</strong>e)) that must be obta<strong>in</strong>ed from the diet, or from the <strong>in</strong>test<strong>in</strong>al


CHAPTER 7Epigenomics <strong>in</strong> Neurobehavioral <strong>Disease</strong>smicrobiome. Multiple evidence po<strong>in</strong>ts towards abnormalities <strong>in</strong> this pathway <strong>in</strong> neurobehavioraldisorders.Dopam<strong>in</strong>e metabolism is l<strong>in</strong>ked to these pathways <strong>in</strong> three manners: (1) degradation ofdopam<strong>in</strong>e by COMT; (2) methylation regulation of genes l<strong>in</strong>ked to dopam<strong>in</strong>e metabolism(research by others and us, see above); and (3) DRD4 methylation by the enzyme MS. Multiplegenetic variants with<strong>in</strong> the folate cycle contribute to negative symptoms <strong>in</strong> SZ with low serumfolate [135]. SNPs with<strong>in</strong> this hub are l<strong>in</strong>ked to SZ, BP, and ASD patients (see above and[109,136,137]), and even to parents of autistic patients [133]. However, deficits <strong>in</strong> the FMTDhub are l<strong>in</strong>ked to many diseases. Hence, it is not clear whether a particular abnormality isl<strong>in</strong>ked to or affected by disease occurrence or presentation. This dist<strong>in</strong>ction may not beimportant. It is likely that normalization of the FMTD hub dynamics <strong>in</strong> patients will havepositive outcomes.Clearly, nutrition has the potential to impact a large number of cellular processes that requireenergy, for <strong>in</strong>stance, DNA replication, RNA synthesis, epigenomic programm<strong>in</strong>g, and thereduction of oxidative stress. Likewise, processes that require DNA replication and/or RNAsynthesis, or cause oxidative stress like ATP generation <strong>in</strong> the mitochondria (the primarysource of oxidative stress) or <strong>in</strong>flammation, will generally impact multiple cellular processesl<strong>in</strong>ked to this metabolic hub <strong>in</strong>clud<strong>in</strong>g epigenomics programm<strong>in</strong>g.7.11 NUTRITIONAL AND DRUG INTERVENTIONSNutritional and drug <strong>in</strong>terventions used to treat neurobehavioral disorders impact theepigenome. In many cases, the epigenomic l<strong>in</strong>k was detected after disease efficacy wasdiscovered. In reality, most, if not all, nutrients and drugs impact the epigenome. Theimportant issue is to dist<strong>in</strong>guish between primary and secondary affects.A number of required and <strong>in</strong>termediate metabolites <strong>in</strong> the FMTD hub impact neurobehavioralbehaviors. That folic acid is useful for treat<strong>in</strong>g refractory major depression and SZ has beenknown for some time, while SAM and methion<strong>in</strong>e are known to <strong>in</strong>crease psychotic symptoms(see [112] for a review).145Drugs used to treat neurobehavioral disorders impact the epigenome directly. Haloperidol, anolder anti-psychotic drug used to treat schizophrenia, alters DNA methylation <strong>in</strong> a tissue- and<strong>in</strong> a sex-specific manner <strong>in</strong> rats [138]. Clozap<strong>in</strong>e and sulpiride (but not haloperidol orolanzap<strong>in</strong>e) reduced promoter methylation of the GAD 67 and RELN [139] previously shownto be epigenomically remodeled <strong>in</strong> a methion<strong>in</strong>e model of schizophrenia [140]. These effectswere enhanced by VPA. VPA, an <strong>in</strong>hibitory of HDAC, is commonly used to treat BP <strong>in</strong> place oflithium (see above). Fluoxet<strong>in</strong>e, a new generation selective seroton<strong>in</strong>-reuptake <strong>in</strong>hibitor usedto treat depression, <strong>in</strong>creased MeCP2, MBD1, and HDAC2 <strong>in</strong> rat GABAergic neurons aftera 10-day treatment regimen [141]. These changes were accompanied by a decrease <strong>in</strong> acetylationat histone 3 lys<strong>in</strong>e 9 (H3K9) and histone 3 lys<strong>in</strong>e 14 (K3K14). Similar results were foundfollow<strong>in</strong>g adm<strong>in</strong>istration of coca<strong>in</strong>e (an <strong>in</strong>hibitor of the seroton<strong>in</strong>, dopam<strong>in</strong>e, and norep<strong>in</strong>ephr<strong>in</strong>etransporters) but not of nortriptyl<strong>in</strong>e (an <strong>in</strong>hibitor of the seroton<strong>in</strong> and dopam<strong>in</strong>etransporters). These results suggest that some drugs that target the seroton<strong>in</strong>ergic system mayexert their effects through epigenetic mechanisms.Chronic, but not acute, coca<strong>in</strong>e dos<strong>in</strong>g <strong>in</strong>duces histone H3 hyperacetylation at the BDNF andCdk5 promoters that is accompanied by DNA demethylation [142] _ENREF_112. Sodiumbutyrate, a non-specific HDAC <strong>in</strong>hibitor, augments the coca<strong>in</strong>e-<strong>in</strong>duced histone modifications<strong>in</strong> striatum and the coca<strong>in</strong>e-<strong>in</strong>duced association of phosphoacetylated histone H3 with<strong>in</strong> thecFos gene promoter.The list of drugs used for the treatment or <strong>in</strong>duction of disease that impact the epigenome isgrow<strong>in</strong>g. In fact, some modes of action may be mediated through changes <strong>in</strong> the epigenome


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>rather than through, for <strong>in</strong>stance, direct b<strong>in</strong>d<strong>in</strong>g of a dopam<strong>in</strong>e, seroton<strong>in</strong>, or nicot<strong>in</strong>amidereceptor. Today we have tools that allow the exploration for novel <strong>in</strong>teractions that can beimportant for understand<strong>in</strong>g complex diseases.7.12 PUTTING IT ALL TOGETHERResearch and treatment of severe neurobehavioral diseases has a history of many failed claimsof success or ways to succeed. The severity of illness argues for optimism but not overoptimism.Recent disappo<strong>in</strong>tment <strong>in</strong> the outcome of genetic approaches for common diseasesis an example of this phenomenon. Unfortunately, such (over)enthusiasm impairs research onneurobehavioral diseases more severely than other diseases because of the unique deficit <strong>in</strong>research funds.Today, there is an <strong>in</strong>creased <strong>in</strong>terest <strong>in</strong> epigenomics and environmental components ofneuropsychiatric disease. The development of new high-throughput techniques, andcomputational analytical tools, for example <strong>in</strong> system biology and complexity science, hasrenewed <strong>in</strong>terest <strong>in</strong> <strong>in</strong>tegrat<strong>in</strong>g complex, multilevel disease models.Our own view is that the seem<strong>in</strong>gly disparate observations on these diseases provide w<strong>in</strong>dowsonto different and/or overlapp<strong>in</strong>g disease processes that lead to similar behavioral symptoms.Further, although the bra<strong>in</strong> is the major disease target, metabolic and nutritional factors areclearly l<strong>in</strong>ked to these diseases and need to be considered along with other risk factors.Nutritional <strong>in</strong>terventions provide excit<strong>in</strong>g possibilities but s<strong>in</strong>gle supplements as drugs cannotbe considered <strong>in</strong> isolation of their impact of multiple metabolic processes.GLOSSARY146Note: See tables for additional gene names and abbreviationsADAR adenos<strong>in</strong>e deam<strong>in</strong>ase RNAASD autism spectrum disorderBDNF bra<strong>in</strong>-derived neurotrophic factorBHMT beta<strong>in</strong>ehomocyste<strong>in</strong>e methyl transferaseBP bipolar diseaseCNV copy number variantsCOMT catechol-O-methyl transferaseDISC1 Some variation <strong>in</strong> BDNF RNA process<strong>in</strong>g is likely l<strong>in</strong>ked to neurobehavioral diseases. Disrupted <strong>in</strong> schizophrenia1DNMT DNA methyl transferaseDRD dopam<strong>in</strong>e receptorDSM Diagnostic and Statistical Manual of Mental DisordersFRAX Fragile XFMTD folate-methion<strong>in</strong>e-transulfuration-dopam<strong>in</strong>e hubGAD glutamate decarboxylaseGSH glutathioneGWAS genome-wide association studiesH2A histone 2AH2b histone 2BH3 histone 3H4 histone 4H3K4 me histone 3 lys<strong>in</strong>e residue 4 methylatedHDACehistone deacetylaseH3K9 ac histone H3 lys<strong>in</strong>e 9 acetylationH3K9 me histone H3 lys<strong>in</strong>e 9 methylationHCY homocyste<strong>in</strong>eHD Hunt<strong>in</strong>gton diseaseHDAC histone deacetylaseHMD highly methylated doma<strong>in</strong>sICD International Criteria of <strong>Disease</strong>IL-8 <strong>in</strong>terleuk<strong>in</strong>-8IMP <strong>in</strong>os<strong>in</strong>e monophosphateMB-COMT membrane-bound catechol-O-methyl transferase (COMT)


CHAPTER 7Epigenomics <strong>in</strong> Neurobehavioral <strong>Disease</strong>sMBD methylated CpG d<strong>in</strong>g<strong>in</strong>g doma<strong>in</strong>MECP methylated CpG b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>MS methion<strong>in</strong>e synthasemS<strong>in</strong>3A co-repressor SIN3 homolog A, transcription regulatorncRNA non-cod<strong>in</strong>g RNANIMH National Institute of Mental Health (United States)PDD pervasive developmental disordersPMD partially methylated doma<strong>in</strong>sRFS4 regulator of G prote<strong>in</strong> signal<strong>in</strong>g 4RELN reel<strong>in</strong>RTT Rett syndromeSAH S-adenosyl homocyste<strong>in</strong>eSAM S-adenosyl methion<strong>in</strong>eSIN3 SIN3 homolog A, transcription regulatorSZ schizophreniaTDG thym<strong>in</strong>e DNA clygosylaseTET ten eleven-translocation family of dioxygensesTHC tetrahydrocannab<strong>in</strong>olTNF tumor necrosis factorTRD transcriptional repression doma<strong>in</strong>VIPR2 vasoactive <strong>in</strong>test<strong>in</strong>al peptide receptorVPA valproic acidAcknowledgmentsThe authors would like to thank Shiva S<strong>in</strong>gh, Andrew Bolton, and Myron Lewis for comments and encouragement.References[1] Jucker M, Walker LC. Pathogenic prote<strong>in</strong> seed<strong>in</strong>g <strong>in</strong> alzheimer disease and other neurodegenerative disorders.Ann Neurol 2011;70:532e40.[2] Schneider R, Grosschedl R. Dynamics and <strong>in</strong>terplay of nuclear architecture, genome organization, and geneexpression. Genes Dev 2007;21:3027e43.[3] Misteli T, Soutoglou E. The emerg<strong>in</strong>g role of nuclear architecture <strong>in</strong> DNA repair and genome ma<strong>in</strong>tenance.Nat Rev Mol Cell Biol 2009;10:243e54.[4] Deaton AM, Bird A. CpG islands and the regulation of transcription. Genes Dev 2011;25:1010e22.[5] Ball MP, Li JB, Gao Y, Lee JH, LeProust EM, Park IH, et al. Targeted and genome-scale strategies reveal genebodymethylation signatures <strong>in</strong> human cells. Nat Biotechnol 2009;27:361e8.[6] Chatagnon A, Perriaud L, Nazaret N, Croze S, Benhattar J, Lachuer J, et al. Preferential b<strong>in</strong>d<strong>in</strong>g of the methyl-CpG b<strong>in</strong>d<strong>in</strong>g doma<strong>in</strong> prote<strong>in</strong> 2 at methylated transcriptional start site regions. <strong>Epigenetics</strong> 2011;6.[7] Tahiliani M, Koh KP, Shen Y, Pastor WA, Bandukwala H, Brudno Y, et al. Conversion of 5-methylcytos<strong>in</strong>e to5-hydroxymethylcytos<strong>in</strong>e <strong>in</strong> mammalian DNA by MLL partner TET1. <strong>Science</strong> 2009;324:930e5.[8] Ito S, Shen L, Dai Q, Wu SC, Coll<strong>in</strong>s LB, Swenberg JA, et al. Tet prote<strong>in</strong>s can convert 5-methylcytos<strong>in</strong>e to5-formylcytos<strong>in</strong>e and 5-carboxylcytos<strong>in</strong>e. <strong>Science</strong> 2011;333:1300e3.[9] He YF, Li BZ, Li Z, Liu P, Wang Y, Tang Q, et al. Tet-mediated formation of 5-carboxylcytos<strong>in</strong>e and its excisionby TDG <strong>in</strong> mammalian DNA. <strong>Science</strong> 2011;333:1303e7.[10] Cortell<strong>in</strong>o S, Xu J, Sannai M, Moore R, Caretti E, Cigliano A, et al. Thym<strong>in</strong>e DNA glycosylase is essential foractive DNA demethylation by l<strong>in</strong>ked deam<strong>in</strong>ation-base excision repair. Cell 2011;146:67e79.[11] Li YQ, Zhou PZ, Zheng XD, Walsh CP, Xu GL. Association of Dnmt3a and thym<strong>in</strong>e DNA glycosylase l<strong>in</strong>ksDNA methylation with base-excision repair. Nucleic Acids Res 2007;35:390e400.[12] Kriaucionis S, He<strong>in</strong>tz N. The nuclear DNA base 5-hydroxymethylcytos<strong>in</strong>e is present <strong>in</strong> Purk<strong>in</strong>je neurons andthe bra<strong>in</strong>. <strong>Science</strong> 2009;324:929e30.[13] Li W, Liu M. Distribution of 5-hydroxymethylcytos<strong>in</strong>e <strong>in</strong> different human tissues. J Nucleic Acids2011;2011:870726.[14] J<strong>in</strong> SG, Wu X, Li AX, Pfeifer GP. Genomic mapp<strong>in</strong>g of 5-hydroxymethylcytos<strong>in</strong>e <strong>in</strong> the human bra<strong>in</strong>. NucleicAcids Res 2011;39:5015e24.[15] Bannister AJ, Kouzarides T. Regulation of chromat<strong>in</strong> by histone modifications. Cell Res 2011;21:381e95.[16] Nan X, Ng HH, Johnson CA, Laherty CD, Turner BM, Eisenman RN, et al. Transcriptional repression by themethyl-CpG-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> MeCP2 <strong>in</strong>volves a histone deacetylase complex. Nature 1998;393:386e9.147


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>148[17] Muskiet FA, Kemperman RF. Folate and long-cha<strong>in</strong> polyunsaturated fatty acids <strong>in</strong> psychiatric disease. J NutrBiochem 2006;17:717e27.[18] Rosenberg G. The mechanisms of action of valproate <strong>in</strong> neuropsychiatric disorders: can we see the forest forthe trees? Cell Mol Life Sci 2007;64:2090e103.[19] Guidotti A, Auta J, Chen Y, Davis JM, Dong E, Gav<strong>in</strong> DP, et al. Epigenetic GABAergic targets <strong>in</strong> schizophreniaand bipolar disorder. Neuropharmacology 2011;60:1007e16.[20] Ornoy A. Valproic acid <strong>in</strong> pregnancy: how much are we endanger<strong>in</strong>g the embryo and fetus? Reprod Toxicol2009;28:1e10.[21] Grafodatskaya D, Chung B, Szatmari P, Weksberg R. Autism spectrum disorders and epigenetics. J Am AcadChild Adolesc Psychiatry 2010;49:794e809.[22] Robertson KD, Wolffe AP. DNA methylation <strong>in</strong> health and disease. Nat Rev Genet 2000;1:11e9.[23] Z<strong>in</strong>i A, Gabriel MS, Zhang X. The histone to protam<strong>in</strong>e ratio <strong>in</strong> human spermatozoa: comparative study ofwhole and processed semen. Fertil Steril 2007;87:217e9.[24] Fuke C, Shimabukuro M, Petronis A, Sugimoto J, Oda T, Miura K, et al. Age related changes <strong>in</strong> 5-methylcytos<strong>in</strong>econtent <strong>in</strong> human peripheral leukocytes and placentas: an HPLC-based study. Ann Hum Genet2004;68:196e204.[25] Shimabukuro M, Sasaki T, Imamura A, Tsujita T, Fuke C, Umekage T, et al. Global hypomethylation ofperipheral leukocyte DNA <strong>in</strong> male patients with schizophrenia: a potential l<strong>in</strong>k between epigenetics andschizophrenia. J Psychiatr Res 2007;41:1042e6.[26] Wutz A. Gene silenc<strong>in</strong>g <strong>in</strong> X-chromosome <strong>in</strong>activation: advances <strong>in</strong> understand<strong>in</strong>g facultative heterochromat<strong>in</strong>formation. Nat Rev Genet 2011;12:542e53.[27] van Rijn S, Swaab H, Aleman A, Kahn RS. X Chromosomal effects on social cognitive process<strong>in</strong>g and emotionregulation: A study with Kl<strong>in</strong>efelter men (47, XXY). Schizophr Res 2006;84:194e203.[28] Smith CL, Bolton A, Nguyen G. Genomic and epigenomic <strong>in</strong>stability, fragile sites, schizophrenia and autism.Curr Genomics 2011;11:447e69.[29] Piton A, Gauthier J, Hamdan FF, Lafreniere RG, Yang Y, Henrion E, et al. Systematic resequenc<strong>in</strong>g ofX-chromosome synaptic genes <strong>in</strong> autism spectrum disorder and schizophrenia. Mol Psychiatry2011;16:867e80.[30] Rosa A, Picchioni MM, Kalid<strong>in</strong>di S, Loat CS, Knight J, Toulopoulou T, et al. Differential methylation of theX-chromosome is a possible source of discordance for bipolar disorder female monozygotic tw<strong>in</strong>s. Am J MedGenet B Neuropsychiatr Genet 2008;147B:459e62.[31] Iourov IY, Vorsanova SG, Yurov YB. Somatic genome variations <strong>in</strong> health and disease. Curr Genomics2010;11:387e96.[32] Lister R, Ecker JR. F<strong>in</strong>d<strong>in</strong>g the fifth base: genome-wide sequenc<strong>in</strong>g of cytos<strong>in</strong>e methylation. Genome Res2009;19:959e66.[33] Schroeder DI, Lott P, Korf I, LaSalle JM. Large-scale methylation doma<strong>in</strong>s mark a functional subset ofneuronally expressed genes. Genome Res 2011;21:1583e91.[34] Costa E, Dong E, Grayson DR, Guidotti A, Ruzicka W, Veldic M. Review<strong>in</strong>g the role of DNA (cytos<strong>in</strong>e-5)methyltransferase overexpression <strong>in</strong> the cortical GABAergic dysfunction associated with psychosis vulnerability.<strong>Epigenetics</strong> 2007;2:29e36.[35] Veldic M, Caruncho HJ, Liu WS, Davis J, Satta R, Grayson DR, et al. DNA-methyltransferase 1 mRNA isselectively overexpressed <strong>in</strong> telencephalic GABAergic <strong>in</strong>terneurons of schizophrenia bra<strong>in</strong>s. Proc Natl Acad SciUSA 2004;101:348e53.[36] Feng J, Zhou Y, Campbell SL, Le T, Li E, Sweatt JD, et al. Dnmt1 and Dnmt3a ma<strong>in</strong>ta<strong>in</strong> DNA methylation andregulate synaptic function <strong>in</strong> adult forebra<strong>in</strong> neurons. Nat Neurosci 2010;13:423e30.[37] Moretti P, Zoghbi HY. MeCP2 dysfunction <strong>in</strong> Rett syndrome and related disorders. Curr Op<strong>in</strong> Genet Dev2006;16:276e81.[38] Gershon ES, Gold<strong>in</strong> LR. Cl<strong>in</strong>ical methods <strong>in</strong> psychiatric genetics. I. Robustness of genetic marker <strong>in</strong>vestigativestrategies. Acta Psychiatr Scand 1986;74:113e8.[39] Gottesman II , Gould TD. The endophenotype concept <strong>in</strong> psychiatry: etymology and strategic <strong>in</strong>tentions. AmJ Psychiatry 2003;160:636e45.[40] Leboyer M, Bellivier F, Nosten-Bertrand M, Jouvent R, Pauls D, Mallet J. Psychiatric genetics: search forphenotypes. Trends Neurosci 1998;21:102e5.[41] Greenwood TA, Lazzeroni LC, Murray SS, Cadenhead KS, Calk<strong>in</strong>s ME, Dobie DJ, et al. Analysis of 94candidate genes and 12 endophenotypes for schizophrenia from the Consortium on the Genetics ofSchizophrenia. Am J Psychiatry 2011;168:930e46.[42] Mann<strong>in</strong>g-Courtney P, Brown J, Molloy CA, Re<strong>in</strong>hold J, Murray D, Sorensen-Burnworth R, et al. Diagnosis andtreatment of autism spectrum disorders. Curr Probl Pediatr Adolesc Health Care 2003;33:283e304.


CHAPTER 7Epigenomics <strong>in</strong> Neurobehavioral <strong>Disease</strong>s[43] Cohly HH, Panja A. Immunological f<strong>in</strong>d<strong>in</strong>gs <strong>in</strong> autism. Int Rev Neurobiol 2005;71:317e41.[44] Melnyk S, Fuchs GJ, Schulz E, Lopez M, Kahler SG, Fussell JJ, et al. Metabolic Imbalance Associated withMethylation Dysregulation and Oxidative Damage <strong>in</strong> Children with Autism. J Autism Dev Disord 2011.[45] Templer DI, Cappelletty GG, Kauffman I. Schizophrenia and multiple sclerosis. Distribution <strong>in</strong> Italy. Br JPsychiatry 1988;153:389e90.[46] Sachdev PS, Keshavan MS. Secondary Schizophrenia. Cambridge, UK: Cambridge University Press; 2010.[47] Lewis SW. Secondary Schizophrenia. In: Hirsh SR, We<strong>in</strong>berger DR, editors. Schizophrenia. Oxfod, UK:Blackwell <strong>Science</strong>; 1995. p. 324e40.[48] Montejo AL. The need for rout<strong>in</strong>e physical health care <strong>in</strong> schizophrenia. Eur Psychiatry 2010;25(Suppl. 2):S3e5.[49] Rupp CI. Olfactory function and schizophrenia: an update. Curr Op<strong>in</strong> Psychiatry 2010;23:97e102.[50] Stevenson RJ, Langdon R, McGuire J. Olfactory halluc<strong>in</strong>ations <strong>in</strong> schizophrenia and schizoaffective disorder:a phenomenological survey. Psychiatry Res 2011;185:321e7.[51] Javitt DC. Sensory process<strong>in</strong>g <strong>in</strong> schizophrenia: neither simple nor <strong>in</strong>tact. Schizophr Bull 2009;35:1059e64.[52] Turetsky BI, Hahn CG, Arnold SE, Moberg PJ. Olfactory receptor neuron dysfunction <strong>in</strong> schizophrenia.Neuropsychopharmacology 2009;34:767e74.[53] Velasco-Ortega E, Monsalve-Guil L, Velasco-Ponferrada C, Medel-Soteras R, Segura-Egea JJ. Temporomandibulardisorders among schizophrenic patients. A case-control study. Med Oral Patol Oral Cir Bucal2005;10:315e22.[54] Kirkpatrick B, Hack GD, Higg<strong>in</strong>bottom E, Hoffacker D, Fernandez-Egea E. Palate and dentition <strong>in</strong> schizophrenia.Schizophr Res 2007;91:187e91.[55] Moraes N, Moraes E, Cunha Marques HH. Dental anomalies <strong>in</strong> mental patients. Bull Pan Am Health Organ1975;9:325e8.[56] Xu T, Chan RC, Compton MT. M<strong>in</strong>or physical anomalies <strong>in</strong> patients with schizophrenia, unaffected firstdegreerelatives, and healthy controls: a meta-analysis. PLoS One 2011;6:e24129.[57] We<strong>in</strong>berg SM, Jenk<strong>in</strong>s EA, Marazita ML, Maher BS. M<strong>in</strong>or physical anomalies <strong>in</strong> schizophrenia: a metaanalysis.Schizophr Res 2007;89:72e85.[58] Meyer JM, Stahl SM. The metabolic syndrome and schizophrenia. Acta Psychiatr Scand 2009;119:4e14.[59] Emsley R, Chiliza B, Asmal L, Lehloenya K. The concepts of remission and recovery <strong>in</strong> schizophrenia. CurrOp<strong>in</strong> Psychiatry 2011;24:114e21.[60] Group, T. H. s. D. C. R. A novel gene conta<strong>in</strong><strong>in</strong>g a tr<strong>in</strong>ucleotide repeat that is expanded and unstable onHunt<strong>in</strong>gton’s disease chromosomes. Cell 1993;72:971e83.[61] van der Burg JM, Bjorkqvist M, Brund<strong>in</strong> P. Beyond the bra<strong>in</strong>: widespread pathology <strong>in</strong> Hunt<strong>in</strong>gton’s disease.Lancet Neurol 2009;8:765e74.[62] Djousse L, Knowlton B, Hayden MR, Almqvist EW, Br<strong>in</strong>kman RR, Ross CA, et al. Evidence for a modifier ofonset age <strong>in</strong> Hunt<strong>in</strong>gton disease l<strong>in</strong>ked to the HD gene <strong>in</strong> 4p16. Neurogenetics 2004;5:109e14.[63] Andrew SE, Hayden MR. Orig<strong>in</strong>s and evolution of Hunt<strong>in</strong>gton disease chromosomes. Neurodegeneration1995;4:239e44.[64] Gejman PV, Sanders AR, Kendler KS. Genetics of schizophrenia: new f<strong>in</strong>d<strong>in</strong>gs and challenges. Annu RevGenomics Hum Genet 2011;12:121e44.[65] Ald<strong>in</strong>ger KA, Plummer JT, Qiu S, Levitt P. SnapShot: Genetics of Autism. Neuron 2011;72. 418e418 e1.[66] Brunetti-Pierri N, Berg JS, Scaglia F, Belmont J, Bac<strong>in</strong>o CA, Sahoo T, et al. Recurrent reciprocal 1q21.1 deletionsand duplications associated with microcephaly or macrocephaly and developmental and behavioralabnormalities. Nat Genet 2008;40:1466e71.[67] Rees E, Moskv<strong>in</strong>a V, Owen MJ, O’Donovan MC, Kirov G. De Novo Rates and Selection of Schizophrenia-Associated Copy Number Variants. Biol Psychiatry 2011.[68] Xu B, Roos JL, Dexheimer P, Boone B, Plummer B, Levy S, et al. Exome sequenc<strong>in</strong>g supports a de novomutational paradigm for schizophrenia. Nat Genet 2011;43:864e8.[69] Nguyen GH, Bouchard J, Boselli MG, Tolstoi LG, Keith L, Baldw<strong>in</strong> C, et al. DNA stability and schizophrenia <strong>in</strong>tw<strong>in</strong>s. Am J Med Genet B Neuropsychiatr Genet 2003;120B:1e10.[70] Le Couteur A, Bailey A, Goode S, Pickles A, Robertson S, Gottesman I, et al. A broader phenotype of autism:the cl<strong>in</strong>ical spectrum <strong>in</strong> tw<strong>in</strong>s. J Child Psychol Psychiatry 1996;37:785e801.[71] St Clair DM, St Clair JB, Swa<strong>in</strong>son CP, Bamforth F, Mach<strong>in</strong> GA. Tw<strong>in</strong> zygosity test<strong>in</strong>g for medical purposes.Am J Med Genet 1998;77:412e4.[72] McGrath JJ. Variations <strong>in</strong> the <strong>in</strong>cidence of schizophrenia: data versus dogma. Schizophr Bull 2006;32:195e7.[73] Sullivan PF. The genetics of schizophrenia. PLoS Med 2005;2:e212.149


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>150[74] Rutten BP, Mill J. Epigenetic mediation of environmental <strong>in</strong>fluences <strong>in</strong> major psychotic disorders. SchizophrBull 2009;35:1045e56.[75] Brown AS. The environment and susceptibility to schizophrenia. Prog Neurobiol 2011;93:23e58.[76] Malasp<strong>in</strong>a D, Perr<strong>in</strong> M, Kle<strong>in</strong>haus KR, Opler M, Harlap S. Growth and schizophrenia: aetiology, epigemiologyand epigenetics. Novartis Foundation Symposium 2008;289:196e203.[77] Frans EM, Sand<strong>in</strong> S, Reichenberg A, Lichtenste<strong>in</strong> P, Langstrom N, Hultman CM. Advanc<strong>in</strong>g paternal age andbipolar disorder. Arch Gen Psychiatry 2008;65:1034e40.[78] Reichenberg A, Gross R, Weiser M, Bresnahan M, Silverman J, Harlap S, et al. Advanc<strong>in</strong>g paternal age andautism. Arch Gen Psychiatry 2006;63:1026e32.[79] Saha S, Barnett AG, Foldi C, Burne TH, Eyles DW, Buka SL, et al. Advanced paternal age is associated withimpaired neurocognitive outcomes dur<strong>in</strong>g <strong>in</strong>fancy and childhood. PLoS Med 2009;6:e40.[80] Fisch H, Hyun G, Golden R, Hensle TW, Olsson CA, Liberson GL. The <strong>in</strong>fluence of paternal age on downsyndrome. J Urol 2003;169:2275e8.[81] McIntosh GC, Olshan AF, Baird PA. Paternal age and the risk of birth defects <strong>in</strong> offspr<strong>in</strong>g. Epidemiology1995;6:282e8.[82] Risch N, Reich EW, Wishnick MM, McCarthy JG. Spontaneous mutation and parental age <strong>in</strong> humans. Am JHum Genet 1987;41:218e48.[83] Glaser RL, Jabs EW. Dear old dad. Sci Ag<strong>in</strong>g Knowledge Environ 2004;2004. re1.[84] Tiemann-Boege I, Navidi W, Grewal R, Cohn D, Eskenazi B, Wyrobek AJ, et al. The observed human spermmutation frequency cannot expla<strong>in</strong> the achondroplasia paternal age effect. Proc Natl Acad Sci USA2002;99:14952e7.[85] Galler JR, Shumsky JS, Morgane PJ. Malnutrition and bra<strong>in</strong> development. In: Walker WA, Watk<strong>in</strong>s JB, editors.Nutrition <strong>in</strong> Pediatrics: Basic <strong>Science</strong> and Cl<strong>in</strong>ical Application Second edit. Neuilly-sur-Se<strong>in</strong>e, France: J. B.Decker Europe, Inc; 1996. p. 196e212.[86] Brown AS, Susser ES. Prenatal nutritional deficiency and risk of adult schizophrenia. Schizophr Bull2008;34:1054e63.[87] Palmer AA, Brown AS, Keegan D, Siska LD, Susser E, Rotrosen J, et al. Prenatal prote<strong>in</strong> deprivation altersdopam<strong>in</strong>e-mediated behaviors and dopam<strong>in</strong>ergic and glutamatergic receptor b<strong>in</strong>d<strong>in</strong>g. Bra<strong>in</strong> Res2008;1237:62e74.[88] Brown AS, Derkits EJ. Prenatal <strong>in</strong>fection and schizophrenia: a review of epidemiologic and translationalstudies. Am J Psychiatry 2010;167:261e80.[89] Hawk<strong>in</strong>s DR, Paul<strong>in</strong>g L. Orthomolecular psychiatry: treatment of schizophrenia. San Francisco: W. H.Freeman; 1973.[90] Khare M, Taylor AH, Konje JC, Bell SC. Delta9-tetrahydrocannab<strong>in</strong>ol <strong>in</strong>hibits cytotrophoblast cell proliferationand modulates gene transcription. Mol Hum Reprod 2006;12:321e33.[91] McMurray CT. Mechanisms of tr<strong>in</strong>ucleotide repeat <strong>in</strong>stability dur<strong>in</strong>g human development. Nat Rev Genet2010;11:786e99.[92] Santoro MR, Bray SM, Warren ST. Molecular Mechanisms of Fragile X Syndrome: A Twenty-Year Perspective.Annu Rev Pathol 2011.[93] Culjkovic B, Stojkovic O, Savic D, Zamurovic N, Nesic M, Major T, et al. Comparison of the number of triplets<strong>in</strong> SCA1, MJD/SCA3, HD, SBMA, DRPLA, MD, FRAXA and FRDA genes <strong>in</strong> schizophrenic patients anda healthy population. Am J Med Genet 2000;96:884e7.[94] Kam<strong>in</strong>sky ZA, Tang T, Wang SC, Ptak C, Oh GH, Wong AH, et al. DNA methylation profiles <strong>in</strong> monozygoticand dizygotic tw<strong>in</strong>s. Nat Genet 2009;41:240e5.[95] S<strong>in</strong>gh SM, Murphy B, O’Reilly R. Epigenetic contributors to the discordance of monozygotic tw<strong>in</strong>s. Cl<strong>in</strong>Genet 2002;62:97e103.[96] S<strong>in</strong>gh SM, O’Reilly R. (Epi)genomics and neurodevelopment <strong>in</strong> schizophrenia: monozygotic tw<strong>in</strong>s discordantfor schizophrenia augment the search for disease-related (epi)genomic alterations. Genome2009;52:8e19.[97] Gottesman I, Bertelsen A. Confirm<strong>in</strong>g unexpressed genotypes for schizophrenia. Risks <strong>in</strong> the offspr<strong>in</strong>g ofFischer’s Danish identical and fraternal discordant tw<strong>in</strong>s. Arch Gen Psychiatry 1989;46:867e72.[98] Uranova NA, V. V, Vikhreva OV, Zim<strong>in</strong>a IS, Kolomeets NS, Orlovskaya DD. The role of oligodendrocytepathology <strong>in</strong> schizophrenia. Int J Neuropsychopharmacol 2007;10:537e45.[99] Tastemir D, Demirhan O, Sertdemir Y. Chromosomal fragile site expression <strong>in</strong> Turkish psychiatric patients.Psychiatry Res 2006;144:197e203.[100] Demirhan O, Tastemir D. Chromosome aberrations <strong>in</strong> a schizophrenia population. Schizophr Res 2003;65:1e7.[101] Demirhan O, Tastemir D, Sertdemir Y. Chromosomal fragile sites <strong>in</strong> schizophrenic patients. Genetika2006;42:985e92.


CHAPTER 7Epigenomics <strong>in</strong> Neurobehavioral <strong>Disease</strong>s[102] Demirhan O, Tastemir D, Sertdemir Y. The expression of folate sensitive fragile sites <strong>in</strong> patients with bipolardisorder. Yonsei Med J 2009;50:137e41.[103] Catts VS, Catts SV, Jablensky A, Chandler D, Weickert CS, Lav<strong>in</strong> MF. Evidence of aberrant DNA damageresponse signall<strong>in</strong>g but normal rates of DNA repair <strong>in</strong> divid<strong>in</strong>g lymphoblasts from patients with schizophrenia.World J Biol Psychiatry 2011.[104] Klouk<strong>in</strong>a-Pantazidou I, Havaki S, Chrysanthou-Piterou M, Kontaxakis VP, Papadimitriou GN, Issidorides MR.Chromat<strong>in</strong> alterations <strong>in</strong> leukocytes of first-episode schizophrenic patients. Ultrastruct Pathol 2010;34:106e16.[105] Morikawa T, Manabe T, Ito Y, Yamada S, Yoshimi A, Nagai T, et al. The expression of HMGA1a is <strong>in</strong>creased <strong>in</strong>lymphoblastoid cell l<strong>in</strong>es from schizophrenia patients. Neurochem Int 2010;56:736e9.[106] Woelk CH, S<strong>in</strong>ghania A, Perez-Santiago J, Glatt SJ, Tsuang MT. The utility of gene expression <strong>in</strong> blood cells fordiagnos<strong>in</strong>g neuropsychiatric disorders. Int Rev Neurobiol 2011;101:41e63.[107] Suzuki K, Nakamura K, Iwata Y, Sek<strong>in</strong>e Y, Kawai M, Sugihara G, et al. Decreased expression of reel<strong>in</strong> receptorVLDLR <strong>in</strong> peripheral lymphocytes of drug-naive schizophrenic patients. Schizophr Res 2008;98:148e56.[108] Glatt SJ, Everall IP, Kremen WS, Corbeil J, Sasik R, Khanlou N, et al. Comparative gene expression analysis ofblood and bra<strong>in</strong> provides concurrent validation of SELENBP1 up-regulation <strong>in</strong> schizophrenia. Proc Natl AcadSci USA 2005;102:15533e8.[109] James SJ, Rose S, Melnyk S, Jernigan S, Blossom S, Pavliv O, et al. Cellular and mitochondrial glutathioneredox imbalance <strong>in</strong> lymphoblastoid cells derived from children with autism. FASEB J 2009;23:2374e83.[110] Abdolmaleky HM, Cheng KH, Russo A, Smith CL, Faraone SV, Wilcox M, et al. Hypermethylation of the reel<strong>in</strong>(RELN) promoter <strong>in</strong> the bra<strong>in</strong> of schizophrenic patients: a prelim<strong>in</strong>ary report. Am J Med Genet B NeuropsychiatrGenet 2005;134B:60e6.[111] Abdolmaleky HM, Cheng KH, Faraone SV, Wilcox M, Glatt SJ, Gao F, et al. Hypomethylation of MB-COMTpromoter is a major risk factor for schizophrenia and bipolar disorder. Hum Mol Genet 2006;15:3132e45.[112] Abdolmaleky HM, Zhou JR, Thiagal<strong>in</strong>gam S, Smith CL. Epigenetic and pharmacoepigenomic studies of majorpsychoses and potentials for therapeutics. Pharmacogenomics 2008;9:1809e23.[113] Guidotti A, Auta J, Davis JM, Di-Giorgi-Gerev<strong>in</strong>i V, Dwivedi Y, Grayson DR, et al. Decrease <strong>in</strong> reel<strong>in</strong> andglutamic acid decarboxylase67 (GAD67) expression <strong>in</strong> schizophrenia and bipolar disorder: a postmortembra<strong>in</strong> study. Arch Gen Psychiatry 2000;57:1061e9.[114] Lachman HM, Papolos DF, Saito T, Yu YM, Szumlanski CL, We<strong>in</strong>shilboum RM. <strong>Human</strong> catechol-Omethyltransferasepharmacogenetics: description of a functional polymorphism and its potential applicationto neuropsychiatric disorders. Pharmacogenetics 1996;6:243e50.[115] Mart<strong>in</strong>owich K, Hattori D, Wu H, Fouse S, He F, Hu Y, et al. DNA methylation-related chromat<strong>in</strong> remodel<strong>in</strong>g<strong>in</strong> activity-dependent BDNF gene regulation. <strong>Science</strong> 2003;302:890e3.[116] Best JA, Nijhout HF, Reed MC. Homeostatic mechanisms <strong>in</strong> dopam<strong>in</strong>e synthesis and release: a mathematicalmodel. Theor Biol Med Model 2009;6:21.[117] Qi Z, Miller GW, Voit EO. Computational systems analysis of dopam<strong>in</strong>e metabolism. PLoS One 2008;3:e2444.[118] Pruunsild P, Kazantseva A, Aid T, Palm K, Timmusk T. Dissect<strong>in</strong>g the human BDNF locus: bidirectionaltranscription, complex splic<strong>in</strong>g, and multiple promoters. Genomics 2007;90:397e406.[119] Nakata K, Lipska BK, Hyde TM, Ye T, Newburn EN, Morita Y, et al. DISC1 splice variants are upregulated <strong>in</strong>schizophrenia and associated with risk polymorphisms. Proc Natl Acad Sci USA 2009;106:15873e8.[120] D<strong>in</strong>g L, Hegde AN. Expression of RGS4 splice variants <strong>in</strong> dorsolateral prefrontal cortex of schizophrenic andbipolar disorder patients. Biol Psychiatry 2009;65:541e5.[121] Dracheva S, Elhakem SL, Marcus SM, Siever LJ, McGurk SR, Haroutunian V. RNA edit<strong>in</strong>g and alternativesplic<strong>in</strong>g of human seroton<strong>in</strong> 2C receptor <strong>in</strong> schizophrenia. J Neurochem 2003;87:1402e12.[122] Hogg M, Paro S, Keegan LP, O’Connell MA. RNA edit<strong>in</strong>g by mammalian ADARs. Adv Genet 2011;73:87e120.[123] Miller BH, Wahlestedt C. MicroRNA dysregulation <strong>in</strong> psychiatric disease. Bra<strong>in</strong> Res 2010;1338:89e99.[124] Xu B, Karayiorgou M, Gogos JA. MicroRNAs <strong>in</strong> psychiatric and neurodevelopmental disorders. Bra<strong>in</strong> Res2010;1338:78e88.[125] Wap<strong>in</strong>ski O, Chang HY. Long noncod<strong>in</strong>g RNAs and human disease. Trends Cell Biol 2011;21:354e61.[126] Yao JK, Dougherty Jr GG, Reddy RD, Keshavan MS, Montrose DM, Matson WR, et al. Homeostatic imbalanceof pur<strong>in</strong>e catabolism <strong>in</strong> first-episode neuroleptic-naive patients with schizophrenia. PLoS One 2010;5:e9508.[127] Nijhout HF, Reed MC, Ulrich CM. Mathematical models of folate-mediated one-carbon metabolism. VitamHorm 2008;79:45e82.[128] Nijhout HF, Gregory JF, Fitzpatrick C, Cho E, Lamers KY, Ulrich CM, et al. A mathematical model gives <strong>in</strong>sights<strong>in</strong>to the effects of vitam<strong>in</strong> B-6 deficiency on 1-carbon and glutathione metabolism. J Nutr 2009;139:784e91.[129] Ulrich CM, Neuhouser M, Liu AY, Boynton A, Gregory 3rd JF, Shane B, et al. Mathematical model<strong>in</strong>g of folatemetabolism: predicted effects of genetic polymorphisms on mechanisms and biomarkers relevant to carc<strong>in</strong>ogenesis.Cancer Epidemiol Biomarkers Prev 2008;17:1822e31.151


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>152[130] Reed MC, Nijhout HF, Neuhouser ML, Gregory 3rd JF, Shane B, James SJ, et al. A mathematical model gives<strong>in</strong>sights <strong>in</strong>to nutritional and genetic aspects of folate-mediated one-carbon metabolism. J Nutr2006;136:2653e61.[131] Ulrich CM, Reed MC, Nijhout HF. Model<strong>in</strong>g folate, one-carbon metabolism, and DNA methylation. Nutr Rev2008;66(Suppl. 1):S27e30.[132] Boyles AL, Billups AV, Deak KL, Siegel DG, Mehltretter L, Slifer SH, et al. Neural tube defects and folatepathway genes: family-based association tests of gene-gene and gene-environment <strong>in</strong>teractions. EnvironHealth Perspect 2006;114:1547e52.[133] James SJ, Melnyk S, Jernigan S, Hubanks A, Rose S, Gaylor DW. Abnormal Transmethylation/transsulfurationMetabolism and DNA Hypomethylation Among Parents of Children with Autism. J Autism Dev Disord2008;38:1976.[134] Lawley SD, C<strong>in</strong>derella M, Hall MN, Gamble MV, Nijhout HF, Reed MC. Mathematical model <strong>in</strong>sights <strong>in</strong>toarsenic detoxification. Theor Biol Med Model 2011;8:31.[135] Roffman JL, Brohawn DG, Nitenson AZ, Mackl<strong>in</strong> EA, Smoller JW, Goff DC. Genetic Variation Throughout theFolate Metabolic Pathway Influences Negative Symptom Severity <strong>in</strong> Schizophrenia. Schizophr Bull 2011.[136] LaSalle JM. A genomic po<strong>in</strong>t-of-view on environmental factors <strong>in</strong>fluenc<strong>in</strong>g the human bra<strong>in</strong> methylome.<strong>Epigenetics</strong> 2011;6:862e9.[137] Gilbody S, Lewis S, Lightfoot T. Methylenetetrahydrofolate reductase (MTHFR) genetic polymorphisms andpsychiatric disorders: a HuGE review. Am J Epidemiol 2007;165:1e13.[138] Shimabukuro M, J<strong>in</strong>no Y, Fuke C, Okazaki Y. Haloperidol treatment <strong>in</strong>duces tissue- and sex-specific changes<strong>in</strong> DNA methylation: a control study us<strong>in</strong>g rats. Behav Bra<strong>in</strong> Funct 2006;2:37.[139] Dong E, Nelson M, Grayson DR, Costa E, Guidotti A. Clozap<strong>in</strong>e and sulpiride but not haloperidol orolanzap<strong>in</strong>e activate bra<strong>in</strong> DNA demethylation. Proc Natl Acad Sci USA 2008;105:13614e9.[140] Dong E, Agis-Balboa RC, Simon<strong>in</strong>i MV, Grayson DR, Costa E, Guidotti A. Reel<strong>in</strong> and glutamic acid decarboxylase67promoter remodel<strong>in</strong>g <strong>in</strong> an epigenetic methion<strong>in</strong>e-<strong>in</strong>duced mouse model of schizophrenia. ProcNatl Acad Sci USA 2005;102:12578e83.[141] Cassel S, Carouge D, Gensburger C, Anglard P, Burgun C, Dietrich JB, et al. Fluoxet<strong>in</strong>e and coca<strong>in</strong>e <strong>in</strong>duce theepigenetic factors MeCP2 and MBD1 <strong>in</strong> adult rat bra<strong>in</strong>. Mol Pharmacol 2006;70:487e92.[142] Kumar A, Choi KH, Renthal W, Tsankova NM, Theobald DE, Truong HT, et al. Chromat<strong>in</strong> remodel<strong>in</strong>g is a keymechanism underly<strong>in</strong>g coca<strong>in</strong>e-<strong>in</strong>duced plasticity <strong>in</strong> striatum. Neuron 2005;48:303e14.


CHAPTER 8Emerg<strong>in</strong>g Role of<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong>NeurodevelopmentalDisordersK. Naga Mohan 1,2 , J. Richard Chaillet 21 Centre for <strong>Human</strong> Genetics, Bangalore-560100, India; Department of Biological<strong>Science</strong>s, Hyderabad, India2 University of Pittsburgh School of Medic<strong>in</strong>e, Pittsburgh, PA, USACHAPTER OUTLINE8.1 Introduction: FewNeurodevelopmentalDisorders have EpigeneticDefects 1538.2 Components of the EpigeneticMach<strong>in</strong>ery 1548.3 Neurodevelopmental Disordersdue to Defects <strong>in</strong> EpigeneticMach<strong>in</strong>ery 1568.3.1 DNMT1 Levels and NeuronalDevelopment 1578.3.2 Schizophrenia and BipolarDisorders 1578.3.3 ES Cell Models 1588.3.4 HSAN1 1588.3.5 ICF Syndrome 1598.3.6 Rett Syndrome 1598.3.7 ATR-X Syndrome 1628.3.8 Cornelia de LangeSyndrome 1628.3.9 MeCP2-ATRX-Cohes<strong>in</strong> GeneRegulation 1648.3.10 Rubenste<strong>in</strong>eTaybiSyndrome 1658.3.11 Coff<strong>in</strong>eLowrySyndrome 1658.3.12 Kabuki Syndrome 1668.4 Neurodevelopmental Disordersdue to Aberrant EpigeneticPatterns 1668.4.1 Fragile X SpectrumDisorders 1668.4.2 Impr<strong>in</strong>t<strong>in</strong>g Disorders 1688.5 Maternal Duplications versusPaternal Duplications <strong>in</strong> PWS/ASRegion 1698.5.1 Possibilities ofEpimutations 1708.6 Conclusions 170References 1701538.1 INTRODUCTION: FEW NEURODEVELOPMENTAL DISORDERSHAVE EPIGENETIC DEFECTSConsider<strong>in</strong>g the complexity of the processes of epigenetic <strong>in</strong>heritance and epigenetic regulationof transcription, the list of neurodevelopmental disorders associated with mutations <strong>in</strong> genesT. Tollefsbol (Ed): <strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>. DOI: 10.1016/B978-0-12-388415-2.00008-1Copyright Ó 2012 Elsevier Inc. All rights reserved.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>encod<strong>in</strong>g epigenetic regulators is remarkably short. This is particularly noteworthy because of thenumerous prote<strong>in</strong>s <strong>in</strong>volved and the likely profound <strong>in</strong>volvement of epigenetic processes <strong>in</strong>mammalian development. Why are there so few neurodevelopmental disorders with epigeneticetiologies? The most likely explanation is that epigenetic regulatory processes act broadly acrossthe genome, target many genes, function throughout gametogenesis and embryogenesis, andfunction <strong>in</strong> all cells. The ubiquitous role of epigenetic processes likely means humans carry<strong>in</strong>gloss-of-function mutations <strong>in</strong> genes encod<strong>in</strong>g epigenetic mach<strong>in</strong>ery will not be viable due toadverse effects on cell function and organismal development. Because of this, we might expectthat viable outcomes of mutations <strong>in</strong> genes govern<strong>in</strong>g epigenetic processes would be rare, andfound only when the mutations affect a lesser number of target genes, are conf<strong>in</strong>ed to specificdevelopmental stages, or affect only a small percentage of cells. As will be evident <strong>in</strong> the follow<strong>in</strong>gdescription of relevant neurodevelopmental syndromes with aberrant epigenetic modifications,the effects of the causative mutations are usually muted (partial loss-of-function mutations)through a variety of different genetic and/or epigenetic processes. The two general categories ofepigenetic defects we describe here are either mutations <strong>in</strong>volv<strong>in</strong>g components of the epigeneticmach<strong>in</strong>ery or deal with aberrant epigenetic patterns due to genetic defects.1548.2 COMPONENTS OF THE EPIGENETIC MACHINERYThe epigenetic mach<strong>in</strong>ery regulat<strong>in</strong>g genes and controll<strong>in</strong>g cell and organismal physiology iscomplex. It establishes and ma<strong>in</strong>ta<strong>in</strong>s the large variety of chromat<strong>in</strong> states that coat the entiregenome and change <strong>in</strong> programmed ways dur<strong>in</strong>g development and cellular differentiation.As mentioned above, the two ma<strong>in</strong> targets of covalent modifications <strong>in</strong> mammalian genomesare DNA and the core histones. Two types of DNA modifications have been described so far,methylation and hydroxylmethylation of the C-5 position of cytos<strong>in</strong>e residues [1]. Of these twomodifications, the molecular and developmental events whereby cytos<strong>in</strong>e methylation isestablished and ma<strong>in</strong>ta<strong>in</strong>ed are better understood. DNA methylation <strong>in</strong> mammals is localized tocytos<strong>in</strong>e bases <strong>in</strong> the context of CpG d<strong>in</strong>ucleotides. Typically, <strong>in</strong> non-divid<strong>in</strong>g cells or <strong>in</strong> divid<strong>in</strong>gcells outside of S phase of the cell cycle, both cytos<strong>in</strong>e bases of complementary base-paired CpGd<strong>in</strong>ucleotides are methylated (so-called fully methylated DNA). Figure 8.1A showshowfullymethylated DNA can be used as a form of heritable genomic <strong>in</strong>formation. The biochemicalmechanism of <strong>in</strong>heritance of DNA methylation is a well-established two-step process, <strong>in</strong> whichfully methylated DNA replicates to form two short-lived hemimethylated double helices, whichare rapidly acted on by the DNA cytos<strong>in</strong>e methyltransferase 1 (DNMT1) enzyme to regeneratetwo identical fully methylated double helices [2]. The epigenetic <strong>in</strong>formation that is <strong>in</strong>herited istypically <strong>in</strong> the form of patterns of DNA methylation, <strong>in</strong> which a methylation pattern is def<strong>in</strong>edas the positions of methylated CpG d<strong>in</strong>ucleotides among <strong>in</strong>terspersed unmethylated CpGd<strong>in</strong>ucleotides. The density of CpG d<strong>in</strong>ucleotides varies widely <strong>in</strong> the genome, rang<strong>in</strong>g fromahighof5e10 CpG d<strong>in</strong>ucleotides per 100 base-pairs <strong>in</strong> CpG islands to nearly zero CpGs per 100base-pairs <strong>in</strong> <strong>in</strong>tronic or <strong>in</strong>tergenic regions. Methylation that can be <strong>in</strong>herited on CpG islands,and the normal very low degree of CpG methylation on most CpG islands is related to high CpGmethylation <strong>in</strong> the adjacent <strong>in</strong>tronic and <strong>in</strong>tergenic regions [3].The details of the process of <strong>in</strong>herited DNA methylation were established <strong>in</strong> the mouse, and themost enlighten<strong>in</strong>g examples of this process are parental alleles of impr<strong>in</strong>ted genes (Figure 8.1B).In primordial germ cells, unmethylated alleles of impr<strong>in</strong>ted genes become fully methylated asthe cells differentiate <strong>in</strong>to more mature germ cells through a de novo methylation process<strong>in</strong>volv<strong>in</strong>g the DNMT3A cytos<strong>in</strong>e methyltransferase enzyme (Figures 8.1A and 8.1B).The biochemical mechanism to generate fully methylated alleles functions <strong>in</strong> both male andfemale germ cells. For some impr<strong>in</strong>ted genes, the paternal allele becomes fully methylateddur<strong>in</strong>g spermatogenesis, and for the rema<strong>in</strong>der, the maternal allele becomes fully methylateddur<strong>in</strong>g oogenesis. Follow<strong>in</strong>g fertilization, through repetitive rounds of biochemical ma<strong>in</strong>tenancemethylation occurr<strong>in</strong>g co<strong>in</strong>cidentally with every cell cycle, the germ-l<strong>in</strong>e methylated allele


CHAPTER 8Emerg<strong>in</strong>g Role of <strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> Neurodevelopmental Disorders(A)(B)(C)FIGURE 8.1Establish<strong>in</strong>g and ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g DNA methylation <strong>in</strong> mammals. (A) Stepwise progression from unmethylated based-paired CpG d<strong>in</strong>ucleotides (1) to fully methylatedDNA (2), and its ma<strong>in</strong>tenance follow<strong>in</strong>g DNA replication. Transition between (1) and (2) is de novo methylation. Ma<strong>in</strong>tenance requires an obligatehemimethylated <strong>in</strong>termediate (3). (B) De novo methylation of DNA occurs largely <strong>in</strong> the early stages of gametogenesis and ma<strong>in</strong>tenance methylation is primarilya post-fertilization process. (C) Differentially methylated doma<strong>in</strong>s (DMDs) of impr<strong>in</strong>ted genes are created through a process <strong>in</strong> which only one parental alleleundergoes de novo methylation <strong>in</strong> the germ l<strong>in</strong>eage, and follow<strong>in</strong>g its ma<strong>in</strong>tenance <strong>in</strong> the embryo, a difference <strong>in</strong> methylation of the two parental alleles is seen.Snurf/Snrpn is a maternally methylated DMD and Igf2/H19 is a paternally methylated DMD. This figure is reproduced <strong>in</strong> the color plate section.155ma<strong>in</strong>ta<strong>in</strong>s its methylation <strong>in</strong> all somatic cells, whereas the opposite unmethylated parental allelerema<strong>in</strong>s unmethylated. This ma<strong>in</strong>tenance methylation process is an efficient and accuratemechanism of epigenetic <strong>in</strong>heritance us<strong>in</strong>g the DNMT1 cytos<strong>in</strong>e methyltransferase enzyme,which acts immediately follow<strong>in</strong>g DNA replication and ma<strong>in</strong>ta<strong>in</strong>s fully methylated DNApatterns [4]. Dur<strong>in</strong>g this process, hemimethylated DNA is recognized by DNMT1 and convertedto fully methylated DNA. Importantly, this essential aspect of epigenetic <strong>in</strong>heritance functionsafter fertilization and depends on the prior establishment of fully methylated DNA patternsdur<strong>in</strong>g gametogenesis [2]. The parent-specific allelic differences <strong>in</strong> DNA methylation <strong>in</strong>embryonic and adult mammals are found <strong>in</strong> just a few regions of the genome called differentiallymethylated doma<strong>in</strong>s (DMDs). Two examples of DMDs, one with a maternally methylatedallele and the other with a paternally methylated allele, are shown <strong>in</strong> Figure 8.1C.CpG methylation patterns are not <strong>in</strong>ert covalent modifications of genomic DNA. Rather, they are“<strong>in</strong>terpreted” <strong>in</strong> various ways to change genome function, typically by alter<strong>in</strong>g transcriptionefficiency <strong>in</strong> the vic<strong>in</strong>ity of a methylation pattern. A ma<strong>in</strong> mechanism of <strong>in</strong>terpretation is viab<strong>in</strong>d<strong>in</strong>g of methyl-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s (MBPs) that conta<strong>in</strong> methyl-b<strong>in</strong>d<strong>in</strong>g doma<strong>in</strong>s (MBDs).Methyl CpG b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> 2 (MeCP2) is the prom<strong>in</strong>ent example of a MBP that b<strong>in</strong>ds tomethylated DNA, typically <strong>in</strong> promoters of genes (Figure 8.2). The card<strong>in</strong>al consequence of thisb<strong>in</strong>d<strong>in</strong>g is the recruitment of other prote<strong>in</strong> complexes that lead to transcriptional repression [5].In the absence of promoter methylation (e.g. <strong>in</strong> Dnmt-null cell l<strong>in</strong>es), there may be moretranscription, and <strong>in</strong> the absence of MeCP2, there may also be more transcription (Figure 8.2).


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>Normal MeCP2 levels and DNMT1 activity (silenced transcription)Repressor complexMeCP2MeCP2MeCP2Reduced DNMT1 activity (some transcription)Repressor complexMeCP2MeCP2Reduced MeCP2 b<strong>in</strong>d<strong>in</strong>g (some transcription)Repressor complexMeCP2MeCP2FIGURE 8.2Gene repression by DNA methylation occurs via MBD-conta<strong>in</strong><strong>in</strong>g prote<strong>in</strong> b<strong>in</strong>d<strong>in</strong>g and recruitment of a repressor complex (top schematic). Reduced DNMT1activity (middle schematic) or reduced MeCP2 (bottom schematic) lead to promoter activity and some transcription.156DNA <strong>in</strong> the nucleus of cells does not exist alone, but is organized <strong>in</strong>to chromat<strong>in</strong> by a variety ofDNA-<strong>in</strong>teract<strong>in</strong>g prote<strong>in</strong>s. The basic unit of organized chromat<strong>in</strong> is DNA complexed with basichistones <strong>in</strong> the form of nucleosomes. An <strong>in</strong>dividual nucleosome is comprised of a histoneoctomer core with 146 base-pairs of DNA wrapped around the outside. Methylation andacetylation of lys<strong>in</strong>es <strong>in</strong> the N-term<strong>in</strong>al tails are the two ma<strong>in</strong> core histone modifications thataffect gene expression. Histone acetyltransferases (HATs) and methyltransferases (HMTs) areresponsible for these modifications, whereas histone deacetylases (HDACs) remove acetylgroups from the lys<strong>in</strong>es so that the deacetylated lys<strong>in</strong>es are methylated by HMTs [6]. As <strong>in</strong> thecase of DNA methyltransferases, a variety of transcriptional coactivators and corepressors<strong>in</strong>fluence gene expression via local histone modifications <strong>in</strong>volv<strong>in</strong>g any of the three differentclasses of histone-modify<strong>in</strong>g enzymes.Ordered states of chromat<strong>in</strong> are not static, but change <strong>in</strong> a dynamic and highly regulated way.The ma<strong>in</strong> role of the multiple regulated chromat<strong>in</strong> states is the control of gene transcription,either at the level of transcription <strong>in</strong>itiation or elongation. Two types of chromat<strong>in</strong> regulationare relevant to the neurodevelopmental disorders discussed <strong>in</strong> this chapter. The position ofnucleosomes along a stretch of genomic DNA can be adjusted to change transcription efficiency[6]. For example, the nucleosome-remodel<strong>in</strong>g complex SWI/SNF (SWItch defective/Sucrose NonFermentor) can be recruited to sites rich <strong>in</strong> histone acetylation, and mediate thereposition<strong>in</strong>g or clearance of nucleosomes (Figure 8.3A). This clearance occurs <strong>in</strong> genepromoters, and leads to DNA sequence-specific b<strong>in</strong>d<strong>in</strong>g of transcriptional activators and an<strong>in</strong>crease <strong>in</strong> gene transcription. A different chromat<strong>in</strong> organization can be mediated by cohes<strong>in</strong>[7]. In addition to its role <strong>in</strong> pair<strong>in</strong>g of sister chromatids dur<strong>in</strong>g <strong>in</strong>terphase of the cell cycle, atthis and other cell-cycle stages cohes<strong>in</strong> can mediate functional <strong>in</strong>teractions between distantDNA sequence elements, which <strong>in</strong> turn govern gene expression (Figure 8.3B).8.3 NEURODEVELOPMENTAL DISORDERS DUE TO DEFECTS INEPIGENETIC MACHINERYWe will now discuss some neurodevelopmental disorders that are due to defects <strong>in</strong> epigeneticmach<strong>in</strong>ery.


CHAPTER 8Emerg<strong>in</strong>g Role of <strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> Neurodevelopmental Disorders(A)Ac Ac Ac(B)SMC3SMC1ASwi/SnfAc Ac AcNIPBLRAD21Swi/SnfActivatorActivatorInitiation complexFIGURE 8.3Higher-order chromat<strong>in</strong> organization relevant to neurodevelopmental syndromes. (A) Chromat<strong>in</strong> remodel<strong>in</strong>g complexes with ATPase activity such as SWI/SNFare recruited by concentrated acetylated histones, and repositioned nucleosomes, thus lead<strong>in</strong>g to activator b<strong>in</strong>d<strong>in</strong>g and gene transcription. (B) Positionedcohes<strong>in</strong> complexes comprised of structural (SMC3 and SMC1A) and regulatory (NIPBL and RAD21) prote<strong>in</strong>s are thought to be regulated <strong>in</strong> an ONeOFF fashionto control gene expression via long-range <strong>in</strong>teractions between different bound DNA cis-regulatory elements, shown here as activator and <strong>in</strong>itiation complexes.1578.3.1 DNMT1 Levels and Neuronal DevelopmentExperiments <strong>in</strong> mice us<strong>in</strong>g conditional mutant alleles of both Dnmt1 and Dnmt3a suggest thatthese enzymes have different effects <strong>in</strong> different regions of the mouse bra<strong>in</strong>. Loss of DNMT1 <strong>in</strong>postmitotic neurons does not affect DNA methylation or survival of neurons [8]. However, <strong>in</strong>premitotic neurons, absence of DNMT1 results <strong>in</strong> neuronal cell death. Interest<strong>in</strong>gly,a comb<strong>in</strong>ed loss of DNMT1 and DNMT3A results <strong>in</strong> smaller-sized postmitotic hippocampalneurons with hypomethylation and dysregulation of genes <strong>in</strong>volved <strong>in</strong> long-term synapticplasticity and defects <strong>in</strong> learn<strong>in</strong>g and memory [9]. We can conclude from these studies thatproper levels of DNMT1 and probably DNMT3A are clearly important for the developmentand function of the mammalian nervous system, and that the vast majority of mutations <strong>in</strong>these enzymes will lead to embryonic death and therefore the absence of recoverablesyndromes.8.3.2 Schizophrenia and Bipolar DisordersEvidence suggest<strong>in</strong>g the importance of de novo and ma<strong>in</strong>tenance DNA methyltransferaselevels <strong>in</strong> neuronal function and neurological behavior comes from a series of reports. Elevatedlevels of DNMT1 were found <strong>in</strong> GABAergic neurons from layers IeIV of prefrontal cortex ofschizophrenic and bipolar patients [10]. In these cell types, the promoters of GAD 67 and RELNwere hypermethylated and associated with low transcript levels of the two genes, lead<strong>in</strong>g to the


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>suggestion that elevated DNMT1 levels are responsible for their down-regulation. Reducedlevels of GAD 67 have been implicated <strong>in</strong> lower<strong>in</strong>g production of the <strong>in</strong>hibitory transmitterGABA and trophic REELIN prote<strong>in</strong> from the cortical neurons result<strong>in</strong>g <strong>in</strong> reduced dendriticsp<strong>in</strong>es and neuropil hypoplasticity <strong>in</strong> the pyramidal neurons. In turn, reduced dendritic sp<strong>in</strong>eplasticity has been suggested to be responsible for cognitive dysfunctions observed <strong>in</strong>psychotic patients. It is also possible that other genes <strong>in</strong>volved <strong>in</strong> neuronal function are alteredby elevated DNMT1 levels. Recently, it was shown that <strong>in</strong> telencephalic GABAergic neurons,both DNMT1 and DNMT3A are elevated <strong>in</strong> layers I and II of BA10 cortical neurons [11].These f<strong>in</strong>d<strong>in</strong>gs may help <strong>in</strong> understand<strong>in</strong>g the mechanism of de novo hypermethylation andma<strong>in</strong>tenance methylation of GAD 67 and RELN <strong>in</strong> these neurons. Taken together, these resultssuggest that abnormal levels of de novo and ma<strong>in</strong>tenance methyltransferases may at least <strong>in</strong>part expla<strong>in</strong> abnormal neuronal morphology and function <strong>in</strong> psychotic patients.1588.3.3 ES Cell ModelsThe near absence of human syndromes with mutations <strong>in</strong> the molecular mach<strong>in</strong>erycontroll<strong>in</strong>g the establishment and ma<strong>in</strong>tenance of DNA methylation <strong>in</strong>dicates the essentialnature of DNA methylation and the difficulty <strong>in</strong> determ<strong>in</strong><strong>in</strong>g the function of DNA methylation<strong>in</strong> nervous system development through the recognition and study of humancongenital syndromes. Rather, <strong>in</strong>vestigation of the role of DNA methylation <strong>in</strong> developmentof the nervous system requires experimental models of mammalian development such asgenetically modified mice and pluripotent mouse ES cells. Mouse studies are importantbecause of the limited human phenotypes associated with mutations <strong>in</strong> the epigeneticmach<strong>in</strong>ery, mak<strong>in</strong>g the discipl<strong>in</strong>e of human genetics less <strong>in</strong>formative <strong>in</strong> regards to functionof methylation mechanisms and epigenetics <strong>in</strong> general. It is now clear that mutations <strong>in</strong>components of epigenetic processes produce pleiotropic effects, probably lead<strong>in</strong>g mostcommonly to embryonic lethality rather than a syndrome. Broadly pleiotropic effects can bemitigated by either mosaicism or dom<strong>in</strong>ant heterozygous effects. The mouse offers additionaladvantages because of the ability to spatially conf<strong>in</strong>e the mutational effect us<strong>in</strong>gconditional mutant alleles.Overexpression of DNMT1 prote<strong>in</strong> <strong>in</strong> early embryogenesis leads to embryonic death [12],suggest<strong>in</strong>g that the concentration of DNMT1 prote<strong>in</strong> regulates epigenetic features of themouse genome that affect its function, most likely <strong>in</strong> aspects of gene transcription. DNMT1activity can be <strong>in</strong>creased <strong>in</strong> later embryonic development and <strong>in</strong> the adult mouse withoutobvious adverse effects, suggest<strong>in</strong>g that the concentration of DNMT1 prote<strong>in</strong> is criticallyimportant dur<strong>in</strong>g an early period of embryogenesis [13]. Afterwards, above normal activity istolerated by the mouse. Interest<strong>in</strong>gly, overexpression of DNMT1 prote<strong>in</strong> <strong>in</strong> ES cells has noapparent effect on the phenotype of pluripotent ES cells [14]. When these overexpress<strong>in</strong>gcells are subjected to differentiation stimuli however, the result<strong>in</strong>g neurons showedabnormal dendritic arborization, branch<strong>in</strong>g and elevated levels of functional N-methylD-aspartate receptor (NMDAR), a feature also reported <strong>in</strong> some neurological and neurodegenerativedisorders. In contrast to human cortical neurons of schizophrenic patients [10],the promoters of Gad 67 and Reln were not hypermethylated <strong>in</strong> these ES cell-derived mouseneurons. These f<strong>in</strong>d<strong>in</strong>gs suggest that overexpression of DNMT1 may alter the epigeneticcomposition of the ES cell genome, which would <strong>in</strong>volve dysregulation of genes other thanGad 67 and Reln (such as genes encod<strong>in</strong>g components of NMDAR) lead<strong>in</strong>g to abnormalneuronal differentiation.8.3.4 HSAN1Hereditary sensory neuropathy with dementia and hear<strong>in</strong>g loss (HSAN1) is a neurodevelopmentalsyndrome recently reported to be caused by mutations <strong>in</strong> the human DNMT1gene [15]. HSAN1 is one of just a handful of human syndromes <strong>in</strong> which abnormalities <strong>in</strong>genomic methylation are associated with neurodevelopmental disorders. Interest<strong>in</strong>gly, HSAN1


CHAPTER 8Emerg<strong>in</strong>g Role of <strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> Neurodevelopmental Disordersis <strong>in</strong>herited <strong>in</strong> an autosomal dom<strong>in</strong>ant manner, and is due to mutations <strong>in</strong> a region of DNMT1that targets it to nuclear replication foci. The mutations lead to premature degradation ofmutant prote<strong>in</strong>s, reduced enzyme activity, and decreased heterochromat<strong>in</strong> b<strong>in</strong>d<strong>in</strong>g dur<strong>in</strong>g theG2 cell cycle phase result<strong>in</strong>g <strong>in</strong> genome-wide hypomethylation and local hypermethylation.The autosomal dom<strong>in</strong>ant feature is particularly <strong>in</strong>terest<strong>in</strong>g; the wild-type prote<strong>in</strong> presumablyrema<strong>in</strong>s <strong>in</strong> cells, but <strong>in</strong> the presence of the mutant prote<strong>in</strong> is unable to ma<strong>in</strong>ta<strong>in</strong> methylation.To our knowledge, this is the first example of a heterozygous DNMT1 mutation <strong>in</strong> eithermouse or human giv<strong>in</strong>g rise to a mutant developmental phenotype. Possible explanations forthe molecular abnormalities <strong>in</strong> heterozygous HSAN1 <strong>in</strong>dividuals <strong>in</strong>clude haplo<strong>in</strong>sufficiencyand dom<strong>in</strong>ant-negative effects. Because HSAN1 mutations fall with<strong>in</strong> a DNMT1 doma<strong>in</strong>implicated <strong>in</strong> dimerization [15,16], we can speculate that heterodimers between wild-type andmutant DNMT1 prote<strong>in</strong>s readily form and, through a dom<strong>in</strong>ant-negative mechanism, such as<strong>in</strong>stability and impaired heterochromat<strong>in</strong> b<strong>in</strong>d<strong>in</strong>g of the heterodimer, lead to <strong>in</strong>sufficient and/or <strong>in</strong>accurate ma<strong>in</strong>tenance methyltransferase activity.8.3.5 ICF SyndromeDNMT3B mutations are found <strong>in</strong> patients with immunodeficiency-centromeric <strong>in</strong>stabilityfacialanomalies (ICF) syndrome [17]. Facial dysmorphism, mental retardation, recurrent andprolonged respiratory <strong>in</strong>fections, <strong>in</strong>fections of the sk<strong>in</strong> and digestive system, and variableimmune deficiency with a constant decrease of IgA are the most common features observed <strong>in</strong>the ICF syndrome patients. Cytogenetic abnormalities <strong>in</strong> these patients <strong>in</strong>clude chromosomealterations affect<strong>in</strong>g the heterochromatic regions of chromosomes 1, 9, and 16 with despiralization,chromatid and chromosome breaks, somatic pair<strong>in</strong>g, and <strong>in</strong>terchanges betweenhomologous and non-homologous chromosomes. The pericentromeric heterochromat<strong>in</strong> ofthese chromosomes always becomes decondensed and fuses to produce multiradial configurations<strong>in</strong> ICF syndrome. At molecular level, ICF syndrome patients show undermethylationof classical satellite DNA located <strong>in</strong> the pericentromeric regions of chromosomes 1, 9, and 16,and on the distal long arm of the Y chromosome. Alpha satellite DNA is largely unaffected.Studies us<strong>in</strong>g cells from ICF syndrome patients showed extensive hypomethylation, advancedreplication time, nuclease hypersensitivity, and a variable escape from silenc<strong>in</strong>g for genes onthe Y and the <strong>in</strong>active X chromosomes. Global expression profil<strong>in</strong>g of lymphoblastoid celll<strong>in</strong>es from three ICF syndrome patients and five normal controls revealed significant changes<strong>in</strong> expression levels of genes <strong>in</strong>volved <strong>in</strong> immune function, signal transduction, mRNA transcription,development, and neurogenesis. Abnormal epigenetic modifications <strong>in</strong> ICF cells<strong>in</strong>clude loss of methylation at promoter regions of several genes such as LHX2, loss of histoneH3K27 trimethylation (repressive mark), and ga<strong>in</strong> of H3K9 acetylation and H3K4 trimethylation(activation marks). Several of the derepressed genes <strong>in</strong>clude homeobox genes critical forimmune system, bra<strong>in</strong>, and craniofacial development. These genes showed a consistent loss ofb<strong>in</strong>d<strong>in</strong>g of the SUZ12 component of the PRC2 polycomb repression complex. Thesemolecular and cytogenetic f<strong>in</strong>d<strong>in</strong>gs <strong>in</strong> ICF syndrome cells suggests that DNMT3B mutationsare pleiotropic and affect the expression of several genes <strong>in</strong>clud<strong>in</strong>g those related to thedevelopment of the nervous system.1598.3.6 Rett SyndromeA major functional consequence of DNA methylation, whether heritable or not, is its negativeeffect on gene transcription (Figure 8.4). As exemplified by the Rett syndrome, MBD prote<strong>in</strong>splay major roles <strong>in</strong> this process. Rett syndrome is the prototypical neurodevelopmentalsyndrome with a heritable defect <strong>in</strong> the epigenetic mach<strong>in</strong>ery. It is an X-l<strong>in</strong>ked neurologicaldisease caused by genetic defects <strong>in</strong> MeCP2 that affects one girl <strong>in</strong> 10 000e15 000 live births.Girls with Rett syndrome develop normally, but between 6 and 18 months of age, there is theonset of an autistic stage, characterized by loss of cognitive, motor, and social skills. Dur<strong>in</strong>g thisstage, they develop characteristic neurological symptoms, such as stereotypical hand-wr<strong>in</strong>g<strong>in</strong>g


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>(A)Mecp2 -/yMecp2-null malesOR+/-Mecp2Heterozygous femalesSevereSymptoms(~ 5 months)Express MeCP2Loss of symptoms~ 6 months(B)Normal mouseNo symptoms-/yDelete MeCP2 (Mecp2 )Symptoms~ 2 monthsFIGURE 8.4Rett syndrome features require ongo<strong>in</strong>g loss of MeCP2 function. (A) Summary of experimental scheme show<strong>in</strong>g that expression of MeCP2 <strong>in</strong> Mecp2-null adultmale mice or <strong>in</strong> adult female mice heterozygous for an Mecp2-null allele beg<strong>in</strong>n<strong>in</strong>g at approximately 6 months of age will reverse the Rett syndrome-likesymptoms. Scheme is based on [33]. (B) Summary of experimental scheme show<strong>in</strong>g that deletion of the s<strong>in</strong>gle wild-type Mecp2 allele <strong>in</strong> normal adult male miceat approximately 2 months of age leads to Rett syndrome-like symptoms. Scheme is based on [34].160and clapp<strong>in</strong>g, <strong>in</strong>dicative of loss of purposeful motor control. The disease is progressive, andafter a period <strong>in</strong> which the cl<strong>in</strong>ical manifestations appear stable, additional deteriorationoccurs, lead<strong>in</strong>g to severe mental retardation and motor impairments, <strong>in</strong>clud<strong>in</strong>g ataxia, apraxia,and tremors. A few boys with Rett syndrome have been described, and they typically have moresevere disease progression [18]. These observations are consistent with an X-l<strong>in</strong>ked mosaicism<strong>in</strong> females, with normally function<strong>in</strong>g cells (X chromosome with mutant MeCp2 allele is<strong>in</strong>active) <strong>in</strong>termixed with dysfunctional cells (wild-type MeCp2 allele is <strong>in</strong>active), each cellfunction<strong>in</strong>g autonomously with respect to its MeCP2 prote<strong>in</strong> activity. Males are more severelyaffected because they have a s<strong>in</strong>gle, mutant Mecp2 allele. The specific Mecp2 mutation has someeffect on the phenotypic manifestation; mutations <strong>in</strong> the am<strong>in</strong>o-term<strong>in</strong>us of the prote<strong>in</strong>correlated with a more severe cl<strong>in</strong>ical presentation compared with mutations near thecarboxyl-term<strong>in</strong>us [19]. However, it is the pattern of X <strong>in</strong>activation that is most closely associatedwith disease severity; those <strong>in</strong>dividuals whose neurons show <strong>in</strong>activation of X chromosomesconta<strong>in</strong><strong>in</strong>g the mutant MeCp2 allele have milder Rett syndrome phenotypes. Evena few healthy female carriers have been identified because a very skewed pattern of X <strong>in</strong>activationresulted <strong>in</strong> a high percentage of cells with an <strong>in</strong>active mutant MeCp2 allele [20].Skewed X <strong>in</strong>activation is also associated, <strong>in</strong> a poorly understood way, with familial cases of Rettsyndrome. In four out of five families with Rett syndrome, no MECP2 mutations were found[21]. In these familial cases, all the mothers and six out of eight affected girls had a highlyskewed pattern of X <strong>in</strong>activation and it was the paternal X chromosome that was active. Analysisus<strong>in</strong>g polymorphic DNA markers suggested that the two traits, highly skewed X <strong>in</strong>activationand Rett syndrome, are not l<strong>in</strong>ked. Based on these results, Villard et al. [21] proposed thatfamilial Rett syndrome is due to the <strong>in</strong>heritance of two traits: an X-l<strong>in</strong>ked locus abnormallyescap<strong>in</strong>g X <strong>in</strong>activation, and the presence of highly skewed X <strong>in</strong>activation <strong>in</strong> carrier women.The precise molecular mechanism of MeCP2 function has been debated s<strong>in</strong>ce MeCP2 wasidentified as a methyl-cytos<strong>in</strong>e b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>, prior to the realization of MeCp2 mutations <strong>in</strong>Rett syndrome. Studies of mutations <strong>in</strong> Rett syndrome, males with duplications of the Mecp2gene, and mouse models of Rett syndrome have been highly <strong>in</strong>structive <strong>in</strong> advanc<strong>in</strong>g ourknowledge of MeCP2’s function. Mutations <strong>in</strong> MeCp2 lead<strong>in</strong>g to Rett syndrome are found <strong>in</strong>highly conserved regions, and virtually all of these are loss-of-function or partial loss-of-


CHAPTER 8Emerg<strong>in</strong>g Role of <strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> Neurodevelopmental Disordersfunction mutations [22]. This conclusion is consistent with the aforementioned strongcorrelation of Rett syndrome phenotype with skew<strong>in</strong>g of X <strong>in</strong>activation rather than withspecific mutation. Interest<strong>in</strong>gly, males with MeCp2 gene duplications also exhibit neurodevelopmentaland mental retardation that overlaps with phenotypes seen <strong>in</strong> traditional Rettsyndrome [23], <strong>in</strong>dicat<strong>in</strong>g that the proper allelic dose of MeCp2 is important for normalneurodevelopment and nervous system function and that loss or ga<strong>in</strong> of MeCP2 activity lead tosimilar nervous system defects.Observations that both MeCP2-null mice and mice with bra<strong>in</strong>-specific deletions of MeCP2recapitulate a number of key features of Rett syndrome suggest that the site of dosedependentMeCP2 defects is the central nervous system [24]. Furthermore, a mouse modelexpress<strong>in</strong>g MeCP2 under the Tau promoter, which is primarily neuron-specific, rescuedMeCp2-null mice, and transgenic mice overexpress<strong>in</strong>g MeCP2 had phenotypes similar to thehuman MeCP2 duplications [25]. The f<strong>in</strong>d<strong>in</strong>g that MeCP2 is very abundant <strong>in</strong> neuronalnuclei and that MeCP2 deficiency results <strong>in</strong> global changes <strong>in</strong> neuronal chromat<strong>in</strong> structurefurther supports the notion that MeCP2 primarily functions <strong>in</strong> neurons of the CNS and thatRett-syndrome neurodevelopmental abnormalities are a consequence of disturbances <strong>in</strong>MeCP2’s neuronal functions [26]. From these observations, Skene et al. [26] postulated thatMeCP2 may not act as a gene-specific transcriptional repressor <strong>in</strong> neurons, but might <strong>in</strong>steaddampen transcriptional noise genome-wide <strong>in</strong> a DNA methylation-dependent manner. Thisunify<strong>in</strong>g idea may help expla<strong>in</strong> the apparent contradiction that MeCP2 functions as a transcriptionalrepressor <strong>in</strong> complex with the co-repressor S<strong>in</strong>3a and HDACs, and also b<strong>in</strong>ds topromoters of activated genes <strong>in</strong> association with the transcriptional activator CREB1 (cAMPresponse element b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> 1) [27]. Recently, MeCP2 expression <strong>in</strong> astrocytes andmicroglial cells has been described, and loss of glial expression of MeCP2 <strong>in</strong> mouse modelsof Rett syndrome was found to contribute to behavioral phenotypes [28e31]. Collectively,these f<strong>in</strong>d<strong>in</strong>gs suggest that both cell-autonomous neuronal deficits and <strong>in</strong>teractions betweenMeCP2-deficient non-neuronal cells and neurons contribute to the pathologies of Rettsyndrome.161Detailed histomorphological analyses of Rett syndrome bra<strong>in</strong>s revealed abnormal neuronalmorphology but not neuronal death [32]. This observation, together with highly <strong>in</strong>formativemouse studies, has helped to clarify the role of MeCP2 <strong>in</strong> nervous system development andfunction. Guy et al. [33] showed that the neurological symptoms of Rett syndrome can bereversed by reactivat<strong>in</strong>g MeCP2 <strong>in</strong> an adult model where the disease was already established.Specifically, reactivation of MeCP2 expression <strong>in</strong> Mecp2-null male mice beg<strong>in</strong>n<strong>in</strong>g 4 weeksafter birth (before the development of symptoms) prevented their certa<strong>in</strong> death by 11 weeks ofage [33]. In both MeCP2-null males and heterozygous mutant females that developed clearneurological symptoms, the symptoms were reversed upon restoration of MeCP2 expression.Importantly, these studies showed that the developmental absence of MeCP2 does not irreversiblydamage neurons, and they are consistent with the notion that MeCP2 is required tostabilize and ma<strong>in</strong>ta<strong>in</strong> the mature neuronal state. An explanation for this remarkable reversalof phenotype when MeCP2 expression is restored is that DNA methylation patterns are laiddown normally <strong>in</strong> the absence of MeCP2 and restored MeCP2 will distribute accord<strong>in</strong>g to thegenomic patterns of CpG methylation. That is, essential MeCP2 target sites <strong>in</strong> neuronalgenomes are encoded solely by patterns of DNA methylation that are established and ma<strong>in</strong>ta<strong>in</strong>ednormally <strong>in</strong> cells lack<strong>in</strong>g the prote<strong>in</strong>. This work provided a proof of concept thatsymptoms of the disorder may be reversible <strong>in</strong> humans.Recently, it has been shown that switch<strong>in</strong>g off MeCP2 <strong>in</strong> adult mice results <strong>in</strong> the appearance ofbehaviors typically seen <strong>in</strong> Rett syndrome [34]. This f<strong>in</strong>d<strong>in</strong>g argues that early expression of thegene does not protect aga<strong>in</strong>st the development of symptoms if the disease gene is later <strong>in</strong>activated.Thus bra<strong>in</strong> cells must have MeCP2 at all times to function normally, yet loss ofMeCP2 does not cause irreversible cellular damage.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>8.3.7 ATR-X SyndromeMeCP2 <strong>in</strong>teracts with several other prote<strong>in</strong>s [35] <strong>in</strong>clud<strong>in</strong>g other chromat<strong>in</strong> prote<strong>in</strong>s, andmutations <strong>in</strong> some of the genes encod<strong>in</strong>g these prote<strong>in</strong>s are also associated with neurodevelopmentaldisorders. MeCP2 physically <strong>in</strong>teracts with alpha thalassemia/mental retardationsyndrome X-l<strong>in</strong>ked homolog prote<strong>in</strong> (ATRX), which belongs to the sucrosenonferment<strong>in</strong>g 2 (SNF2) family of chromat<strong>in</strong> remodel<strong>in</strong>g prote<strong>in</strong>s. These prote<strong>in</strong>s use energyfrom hydrolysis of ATP to disrupt nucleosome stability. ATR-X syndrome is an X-l<strong>in</strong>keddisorder with mutations <strong>in</strong> the ATRX gene. The syndrome is conf<strong>in</strong>ed to males. Female carriersare <strong>in</strong>tellectually normal and no consistent physical manifestations have been recognized.The syndrome is characterized by severe mental retardation, associated with unique physicalanomalies, vary<strong>in</strong>g degrees of urogenital abnormalities, and alpha thalassemia. Phenotypicabnormalities are not conf<strong>in</strong>ed to the nervous system, as there are non-neurological abnormalities,such as craniofacial defects, skeletal abnormalities, and lung, kidney, and digestiveproblems. Mild cerebral atrophy may be seen, and <strong>in</strong> two cases partial or complete agenesis ofthe corpus callosum was reported [36].162Studies of the pattern of X <strong>in</strong>activation <strong>in</strong> carriers for ATR-X syndrome showed that <strong>in</strong> most ofthem, the abnormal X chromosome is predom<strong>in</strong>antly <strong>in</strong>activated <strong>in</strong> cells from a variety oftissues. ATRX is found <strong>in</strong> the nucleus <strong>in</strong> association with pericentric heterochromat<strong>in</strong> [37].ATRX prote<strong>in</strong> was significantly reduced <strong>in</strong> a group of patients with a wide variety of ATRXmutations, and <strong>in</strong> some patients the prote<strong>in</strong> was virtually undetectable. These f<strong>in</strong>d<strong>in</strong>gs areconsistent with the view that the significant effect of these mutations is a decrease <strong>in</strong> normalcellular ATRX activity. How loss of ATRX activity leads to ATR-X syndrome is not clear. ATRX<strong>in</strong>fluences effects on gene expression through <strong>in</strong>teractions with MeCP2. ATRX mutations <strong>in</strong>humans result <strong>in</strong> aberrant DNA methylation patterns <strong>in</strong> several repetitive elements, <strong>in</strong>clud<strong>in</strong>gribosomal DNA repeats, subtelomeric repeats, and Y-specific satellite repeats [36]. Moreover,the consistent cl<strong>in</strong>ical features of ATR-X syndrome suggest that ATRX regulates expression ofa discrete subset of genes, alpha-glob<strong>in</strong> be<strong>in</strong>g one well-def<strong>in</strong>ed example. This notion issupported by the observation that human ATRX mutations cause alpha-thalassemia by downregulat<strong>in</strong>galpha-glob<strong>in</strong> but not beta-glob<strong>in</strong> expression.The importance of the MeCP2-ATRX <strong>in</strong>teraction <strong>in</strong> neurodevelopment is highlighted <strong>in</strong> thephenotypes of patients harbor<strong>in</strong>g MeCP2 variants that are not Rett syndrome mutations. Inparticular, the A140V mutant MeCP2 prote<strong>in</strong> has been reported <strong>in</strong> hemizygous males ofseveral <strong>in</strong>dependent X-l<strong>in</strong>ked mental retardation (XLMR) families [38]. Because these XLMRmales survive past birth, the A140V mutation is therefore less severe than MeCP2 mutationsassociated with Rett syndrome. The only measurably altered property of the A140V mutant isloss of the ability to <strong>in</strong>teract with ATRX, whereas MeCP2 mutant prote<strong>in</strong>s <strong>in</strong> Rett syndromepatients have additional abnormalities <strong>in</strong> addition to reduced ATRX b<strong>in</strong>d<strong>in</strong>g. Thus, <strong>in</strong>appropriatetarget<strong>in</strong>g of ATRX likely contributes to transcriptional defects, altered neuronalphenotypes, and the neurological abnormalities seen <strong>in</strong> both Rett syndrome patients andMecp2-null mice.8.3.8 Cornelia de Lange SyndromeMeCP2 also <strong>in</strong>teracts with structural components of the cohes<strong>in</strong> complex, and MeCP2, ATRXand cohes<strong>in</strong> co-occupy genomic impr<strong>in</strong>t<strong>in</strong>g control regions <strong>in</strong> cells of the mouse forebra<strong>in</strong>[39]. Because of these <strong>in</strong>teractions, it is not surpris<strong>in</strong>g that mutations <strong>in</strong> genes encod<strong>in</strong>gcohes<strong>in</strong> structural and regulatory prote<strong>in</strong>s are associated with neurodevelopmental abnormalities.Cornelia de Lange syndrome (CdLS) is a dom<strong>in</strong>antly <strong>in</strong>herited disorder witha characteristic facial appearance, malformations of the upper extremities, gastroesophagealdysfunction, growth retardation, and abnormalities <strong>in</strong> cognitive development. Mental retardation<strong>in</strong> CdLS patients, although typically moderate to severe, displays a wide range of


CHAPTER 8Emerg<strong>in</strong>g Role of <strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> Neurodevelopmental Disordersvariability. The majority of CdLS cases are due to mutations <strong>in</strong> the cohes<strong>in</strong> regulatory subunitNIPBL (nipped B-like) or structural subunits SMC1A and SMC3 [7]. Mutations <strong>in</strong> NIPBL, thevertebrate homolog of the yeast Scc2 prote<strong>in</strong> and a regulator of cohes<strong>in</strong> load<strong>in</strong>g andunload<strong>in</strong>g, comprise ~50% of CdLS cases. The predicted outcomes of NIPBL mutations aretruncated or untranslated prote<strong>in</strong>s, suggest<strong>in</strong>g that NIPBL haplo<strong>in</strong>sufficiency results <strong>in</strong> CdLSphenotypes [40]. That haplo<strong>in</strong>sufficiency is a mechanism <strong>in</strong> CdLS is confirmed by the childwith a large deletion of the NIPBL region and severe manifestations of CdLS [41].Mutations <strong>in</strong> genes encod<strong>in</strong>g other components of cohes<strong>in</strong> are associated with CdLS or therelated Roberts and SC phocomelia syndromes (RS/SC). Mutations <strong>in</strong> SMC1A and SMC3,which are structural cohes<strong>in</strong> subunits, contribute to ~5% of CdLS cases and result <strong>in</strong>consistently milder CdLS phenotypes with absence of major anatomical anomalies [42,43].Smc1L1, which encodes the SMC1A cohes<strong>in</strong> subunit, is on the X chromosome and theaffected <strong>in</strong>dividuals are male. This <strong>in</strong>dicates that the mutations are unlikely to be strongloss-of-function alleles, which would likely be lethal. Notably, Smc1L1 escapes X-<strong>in</strong>activation,and of the three female carriers that passed on a Smc1L1 gene mutation, one displayed verymild characteristics consistent with CdLS. This additional cl<strong>in</strong>ical and cytogenetic <strong>in</strong>formationsuggests that human Smc1L1 mutations are probably partial loss-of-function mutations, andthe retention of some function <strong>in</strong> mutant SMC1A prote<strong>in</strong>s is a very likely explanation for theirassociation with viable, yet neurodevelopmentally abnormal, CdLS patients. In this regardanalysis of CdLS-associated mutant SMC1A and SMC3 prote<strong>in</strong>s <strong>in</strong>dicates that they are likely tobe stable prote<strong>in</strong>s <strong>in</strong> vivo [43]. As stable, expressed prote<strong>in</strong>s, it is very likely that they contributeto cohes<strong>in</strong> complexes with altered (partial) function. The Roberts and SC phocomeliasyndromes (RS/SC) are due to mutations <strong>in</strong> the cohes<strong>in</strong> regulator ESCO2 [44]. ESCO2 isa member of a conserved family that is required for the establishment of sister chromatidcohesion dur<strong>in</strong>g S phase and may have acetyltransferase activity (Figure 8.3B).As evidenced by near absence of sister chromatid cohesion defects among CdLS patients, sisterchromatid cohesion seems to be m<strong>in</strong>imally affected or not affected at all by dosage changes ofcohes<strong>in</strong> and its associated prote<strong>in</strong>s. Thus, the cl<strong>in</strong>ical phenotypes of CdLS and RS/SC are morelikely the result of cohes<strong>in</strong>-mediated gene dysregulation dur<strong>in</strong>g embryonic development.Studies <strong>in</strong> Drosophila have shown that the sister chromatid cohesion complex controls geneactivation [45,46]. Such studies suggest a molecular etiology of abnormal phenotypes <strong>in</strong> CdLSpatients, because effects on gene expression dur<strong>in</strong>g human embryogenesis could lead to thedevelopment anomalies seen <strong>in</strong> CdLS patients. The Drosophila Nipped-B gene, an ortholog ofthe budd<strong>in</strong>g yeast gene Scc2, is required for cohes<strong>in</strong> to b<strong>in</strong>d to chromosomes. Nipped-Bregulates the load<strong>in</strong>g of the structural subunits of cohes<strong>in</strong> onto chromosomes. In Drosophila,Nipped-B-mediated cohes<strong>in</strong>-load<strong>in</strong>g is required for long-range activation of two homeoboxprote<strong>in</strong> genes that play critical roles <strong>in</strong> development. Specifically, reduc<strong>in</strong>g cohes<strong>in</strong> concentrationsboosts cut gene expression, whereas reduc<strong>in</strong>g Nipped-B levels dim<strong>in</strong>ishes cut geneexpression <strong>in</strong> the emergent w<strong>in</strong>g marg<strong>in</strong> [45]. The <strong>in</strong>terpretation of these f<strong>in</strong>d<strong>in</strong>gs is thatcohes<strong>in</strong> acts as an <strong>in</strong>sulator that blocks enhancerepromoter communication, similar to themanner <strong>in</strong> which cohes<strong>in</strong> blocks spread<strong>in</strong>g of SIR prote<strong>in</strong> complexes at the HMR locus <strong>in</strong>budd<strong>in</strong>g yeast [47]. How cohes<strong>in</strong> might h<strong>in</strong>der enhancerepromoter communication <strong>in</strong> thismodel is not known. One possibility is similar to that proposed for the gypsy transposon<strong>in</strong>sulator, where the <strong>in</strong>sulator blocks a spread of homeoprote<strong>in</strong> b<strong>in</strong>d<strong>in</strong>g between the enhancerand the promoter [48]. Nipped-B might facilitate gene activation by regulat<strong>in</strong>g the dynamicequilibrium between bound and unbound cohes<strong>in</strong>. Reduced Nipped-B activity, <strong>in</strong> pr<strong>in</strong>ciple,would slow the k<strong>in</strong>etics of cohes<strong>in</strong> removal and therefore block timely gene activation dur<strong>in</strong>gDrosophila development. It is important to note that although effects on sister chromatidcohesion are seen <strong>in</strong> homozygous Nipped-B mutants, they were not observed <strong>in</strong> heterozygousmutants. However significant effects on gene expression were observed <strong>in</strong> heterozygousNipped-B mutants [46]. A schematic of the structure of cohes<strong>in</strong> and how it may control theexpression of genes is shown <strong>in</strong> Figure 8.3B.163


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong><strong>Human</strong> Smc1L1 mutations cause similar developmental problems as NIPBL mutations. An<strong>in</strong>trigu<strong>in</strong>g possibility is that the identified Smc1L1 mutations are not null alleles, but slowdown cohes<strong>in</strong>-b<strong>in</strong>d<strong>in</strong>g dynamics as is proposed above <strong>in</strong> the case of reduced NIPBL levels.Similar effects may also occur <strong>in</strong> RS/SC syndrome patients because there are common features<strong>in</strong> Cornelia de Lange and RS/SC syndrome patients. Nevertheless, the detailed molecularetiology of RS/SC syndromes is probably dist<strong>in</strong>ct from the proposed effects of CdLS mutationson long-range gene activity. For example, key features of RS/SC patients not found <strong>in</strong> CdLSpatients are the presence of chromosomal and mitotic defects. In addition, RS/SC patientsshow effects on cohesion specifically <strong>in</strong> heterochromatic but not euchromatic regions. Inconclusion, the chromosomal and mitotic defects <strong>in</strong> RS/SC syndrome, taken together with thesimilar abnormal developmental phenotypes of Cornelia de Lange syndrome suggest anoverlap <strong>in</strong> the cohes<strong>in</strong>-mediated functions of chromatid cohesion and control of geneexpression.1648.3.9 MeCP2-ATRX-Cohes<strong>in</strong> Gene RegulationThat most human neurodevelopmental disorders are associated with mutations <strong>in</strong> the<strong>in</strong>teract<strong>in</strong>g chromat<strong>in</strong> prote<strong>in</strong>s MeCP2, ATRX, and cohes<strong>in</strong> is <strong>in</strong>terest<strong>in</strong>g and raises the issueof why mutations <strong>in</strong> genes encod<strong>in</strong>g these prote<strong>in</strong>s are compatible with embryonic andpostnatal development, albeit abnormal development? Although we do not know the answerto this question, the modes of <strong>in</strong>heritance of the associated neurodevelopmental disordersprovide some <strong>in</strong>sight <strong>in</strong>to this issue. ATRX is an X-l<strong>in</strong>ked gene and the ATR-X syndrome isfound <strong>in</strong> males carry<strong>in</strong>g mutations that are likely loss-of-function mutations, <strong>in</strong>dicat<strong>in</strong>g thatcomplete loss of ATRX is tolerated and probably required to produce the syndrome. MeCP2 ismore essential for development than ATRX because male carriers have a much more severedisease progression than females, which presumably leads to an early embryonic death.Moreover, females with Rett syndrome are protected from severe early consequences becausethey are X-chromosome mosaics. Likewise, CdLS is <strong>in</strong>herited as an autosomal dom<strong>in</strong>ant traitand more rarely as an X-l<strong>in</strong>ked trait, suggest<strong>in</strong>g that only a partial loss of function is tolerated(reviewed <strong>in</strong> [49]). All <strong>in</strong> all, based on our discussion so far, there are just a handful ofneurodevelopmental disorders due to mutations <strong>in</strong> epigenetic genes, and most of these<strong>in</strong>volve partial loss-of-function of prote<strong>in</strong>s (or loss of just mutant-cell function <strong>in</strong> anX-chromosome mosaic background) associated with higher-order chromat<strong>in</strong> structure andfunction.How might dysregulation of MECP2, ATRX, and cohes<strong>in</strong> <strong>in</strong>teractions lead to neurodevelopmentaldisorders? In <strong>in</strong> vivo and cell culture experiments, us<strong>in</strong>g mice and theirimpr<strong>in</strong>ted genes and DMDs as a model, Kernohan et al. [39] showed that ATRX, cohes<strong>in</strong>, andMeCP2 co-occupy impr<strong>in</strong>t<strong>in</strong>g control regions <strong>in</strong> the mouse forebra<strong>in</strong>. Furthermore, loss ofATRX led to reduced occupancy of cohes<strong>in</strong>, CTCF, and MeCP2 at the Gtl2 and other impr<strong>in</strong>t<strong>in</strong>gDMDs. The fact that ATRX is essential to achieve full occupancy of cohes<strong>in</strong> and CTCF at targetimpr<strong>in</strong>ted doma<strong>in</strong>s potentially implicates ATRX <strong>in</strong> the regulation of higher-order chromat<strong>in</strong>conformation, <strong>in</strong>sulator functions, or monoallelic gene expression. In neurons however, ATRXis not likely to govern all occupancy and effects of MeCP2 because the loss of MeCP2 activity <strong>in</strong>mice and <strong>in</strong> males with MeCP2 mutations leads to more severe effects than loss of ATRXactivity. The <strong>in</strong>vestigators concluded from their studies that ATRX partners with cohes<strong>in</strong>and MeCP2 and contributes to developmental silenc<strong>in</strong>g of impr<strong>in</strong>ted genes <strong>in</strong> the bra<strong>in</strong>.Mechanistically, MeCP2 and cohes<strong>in</strong> function <strong>in</strong> chromosomal loop<strong>in</strong>g, and the presence ofATRX at target genes with cohes<strong>in</strong> and MeCP2 suggests that ATRX may also modulatechromat<strong>in</strong> loop formation by promot<strong>in</strong>g specific long-range <strong>in</strong>teractions. In regards to theATR-X and Rett syndromes, the <strong>in</strong>teraction between ATRX and MeCP2 might also control theexpression of certa<strong>in</strong> MeCP2-bound non-impr<strong>in</strong>ted genes <strong>in</strong>volved <strong>in</strong> cognitive functionthrough their epigenetic regulation.


CHAPTER 8Emerg<strong>in</strong>g Role of <strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> Neurodevelopmental DisordersThe f<strong>in</strong>d<strong>in</strong>g that ATRX is essential to achieve full occupancy of cohes<strong>in</strong> and CTCF at targetimpr<strong>in</strong>ted doma<strong>in</strong>s potentially implicates ATRX <strong>in</strong> the regulation of higher-order chromat<strong>in</strong>conformation, <strong>in</strong>sulator functions, or monoallelic gene regulation. It is possible that CTCF andcohes<strong>in</strong> perform their <strong>in</strong>sulator functions <strong>in</strong> different tissues <strong>in</strong> collaboration with variousmembers of the SNF2 family of chromat<strong>in</strong> remodel<strong>in</strong>g prote<strong>in</strong>s, <strong>in</strong>clud<strong>in</strong>g ATRX. The <strong>in</strong>terplayamong ATRX, MeCP2, CTCF, and cohes<strong>in</strong> prote<strong>in</strong>s at impr<strong>in</strong>t<strong>in</strong>g control regions, and probablyat other sites <strong>in</strong> the neuronal genome, raises the <strong>in</strong>trigu<strong>in</strong>g possibilities that neurodevelopmentalsyndromes may also be associated with mutations <strong>in</strong> the CTCF gene or <strong>in</strong> oneor more of the genes encod<strong>in</strong>g cohes<strong>in</strong> subunits. No human mutations are known <strong>in</strong> CTCFand there are also no mouse knockout models to study <strong>in</strong>activation of Ctcf gene <strong>in</strong> neurodevelopment.Limited <strong>in</strong>formation available based on conditional knockouts of Ctcf ordepletion of CTCF <strong>in</strong> oocytes suggests that CTCF is critical for cell cycle and development.For example, lack of CTCF <strong>in</strong> the female germl<strong>in</strong>e results <strong>in</strong> abnormal meiosis and mitosis,associated with lethality at 4-cell stage of preimplantation development [50]. By extension ofthe studies us<strong>in</strong>g conditional Ctcf alleles, it is possible that CTCF may be essential for survivalof neurons as well. As discussed above, mutations <strong>in</strong> genes encod<strong>in</strong>g cohes<strong>in</strong> structural andregulatory subunits are associated with CdLS, a neurodevelopmental syndrome.8.3.10 Rubenste<strong>in</strong>eTaybi SyndromeThe Rubenste<strong>in</strong>eTaybi syndrome is characterized by a range of phenotypic abnormalities.Many organs are affected, not just the nervous system. It is an autosomal dom<strong>in</strong>ant disordercharacterized primarily by mental retardation. There are also physical abnormalities, <strong>in</strong>clud<strong>in</strong>gpostnatal growth deficiency often followed by excessive weight ga<strong>in</strong> <strong>in</strong> later childhood orpuberty, abnormal craniofacial features and an <strong>in</strong>creased risk of cancer. In approximately 55%of cases, the syndrome is associated with de novo mutations <strong>in</strong> the CREB b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>(CBP). Genetic cause <strong>in</strong> about 42% of the cases is not known. CBP is a histone acetyltransferase(HAT) closely related to the p300 family of prote<strong>in</strong>s, and a transcriptional coactivatorthat is shown to <strong>in</strong>teract with more than 300 transcription factors and the generaltranscription mach<strong>in</strong>ery [51]. CBP and p300 have been shown to regulate hematopoietic stemcells rais<strong>in</strong>g the possibility of their <strong>in</strong>volvement <strong>in</strong> stem cell biology [52]. Consistent with theautosomal <strong>in</strong>heritance of the syndrome, mouse studies showed that haplo<strong>in</strong>sufficiency of CBPresults <strong>in</strong> cognitive dysfunction associated with mature circuit abnormalities [53]. In regard toits role <strong>in</strong> neurological development, knockdown of CBP prote<strong>in</strong> levels <strong>in</strong> utero results <strong>in</strong>significant reduction <strong>in</strong> the efficiency of neurogenesis and gliogenesis from cortical precursorcells [54]. CBP has been shown to b<strong>in</strong>d to neural promoters and mediate histone acetylation,and therefore is thought to be essential for the expression of the neural l<strong>in</strong>eage-specific genesdur<strong>in</strong>g differentiation of neurons. Taken together these results are <strong>in</strong> l<strong>in</strong>e with our earliersuggestion that neurological disorders are only the outcomes of “less severe effects” of mildlyaberrant epigenetic mach<strong>in</strong>ery.1658.3.11 Coff<strong>in</strong>eLowry SyndromeCoff<strong>in</strong>eLowry syndrome is another of the handful of rare neurodevelopmental syndromesl<strong>in</strong>ked to defects <strong>in</strong> chromat<strong>in</strong> remodel<strong>in</strong>g and ma<strong>in</strong>tenance of chromat<strong>in</strong> architecture. It is anX-l<strong>in</strong>ked disorder associated with a broad set of severe systemic abnormalities <strong>in</strong> male patients,<strong>in</strong>clud<strong>in</strong>g neurodevelopmental abnormalities. Phenotypic features of male patients <strong>in</strong>cludegrowth and psychomotor retardation, general hypotonia, and skeletal abnormalities.The mental retardation is usually severe. Facial features <strong>in</strong> affected males are very characteristic,and <strong>in</strong>clude a prom<strong>in</strong>ent forehead, down-slant<strong>in</strong>g palpebral fissures, orbital hypertelorism,thick lips, a thick nasal septum with anteverted nares, and irregular or miss<strong>in</strong>g teeth. Inheterozygous “carrier” females, there is a wide range of milder phenotypes, such as m<strong>in</strong>orfacial coarsen<strong>in</strong>g and obesity.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>At the molecular level, Coff<strong>in</strong>eLowry syndrome is caused by loss-of-function mutations <strong>in</strong>RSK2 (also known as RPS6KA3), which encodes a ser<strong>in</strong>e/threon<strong>in</strong>e prote<strong>in</strong> k<strong>in</strong>ase belong<strong>in</strong>gto the RSK family of k<strong>in</strong>ases (RSK1 to RSK4) <strong>in</strong> humans. Mice lack<strong>in</strong>g RSK2 survive to birth,but have defects <strong>in</strong> osteoblast differentiation result<strong>in</strong>g <strong>in</strong> skeletal abnormalities. They alsoshow impaired spatial learn<strong>in</strong>g and reduced control of exploratory behavior [55]. In uteromouse experiments <strong>in</strong>volv<strong>in</strong>g RSK2 knockdowns <strong>in</strong>dicated that loss of RSK2 results <strong>in</strong>a significant decrease <strong>in</strong> neurogenesis, an <strong>in</strong>crease <strong>in</strong> the radial precursor cells, defectivedopam<strong>in</strong>ergic receptor function, impaired learn<strong>in</strong>g and coord<strong>in</strong>ation [56]. Coff<strong>in</strong>eLowry is anepigenetic disease because RSK2 normally <strong>in</strong>fluences chromat<strong>in</strong> structure through twodifferent mechanisms: direct phosphorylation of histones and by <strong>in</strong>teract<strong>in</strong>g with CBP,a histone acetyltransferase. Even though phosphorylation of H3 <strong>in</strong> response to epidermalgrowth factor is defective <strong>in</strong> cell l<strong>in</strong>es derived from patients with Coff<strong>in</strong>eLowry syndrome,whether dysregulation of transcription through this mechanism contributes to cognitiveimpairment <strong>in</strong> patients with Coff<strong>in</strong>eLowry syndrome is not clear. A possible reason beh<strong>in</strong>dsurvival of Rsk -/- mice with develop<strong>in</strong>g skeletal and neurological abnormalities is that RSK2plays an important role <strong>in</strong> osteogenesis and neurogenesis, whereas <strong>in</strong> development of othertissues, other members of the RSK family play an important role.1668.3.12 Kabuki SyndromeKabuki syndrome is characterized by mental retardation, dist<strong>in</strong>ctive facial appearance,developmental delay, short stature, and urogenital tract anomalies. A recent study identifiedmutations <strong>in</strong> the autosomal MLL2 gene <strong>in</strong> approximately half of Kabuki syndrome patients[57,58]. Most of these cases are due to dom<strong>in</strong>ant de novo MLL2 mutations. Mutations werefound throughout the gene and <strong>in</strong>cluded nonsense mutations, splice-site mutations, smalldeletions or <strong>in</strong>sertions and missense mutations. MLL2 encodes a large 5262-residue prote<strong>in</strong>that is part of the SET family of prote<strong>in</strong>s, of which Trithorax, the Drosophila homolog of MLL, isthe best characterized. The SET doma<strong>in</strong> of MLL2 confers strong histone 3 lys<strong>in</strong>e 4 methyltransferaseactivity and is important <strong>in</strong> the epigenetic control of active chromat<strong>in</strong> states. Mostof the MLL2 variants identified <strong>in</strong> <strong>in</strong>dividuals with Kabuki syndrome are predicted to truncatethe polypeptide cha<strong>in</strong> before translation of the SET doma<strong>in</strong>. The syndrome more likely resultsfrom haplo<strong>in</strong>sufficiency than a ga<strong>in</strong> of function because the few pathogenic missense variantsare located <strong>in</strong> region of MLL2 that encode C-term<strong>in</strong>al doma<strong>in</strong>s. Therefore, a human syndromewith surviv<strong>in</strong>g affected <strong>in</strong>dividuals with MLL2 mutations is probably only evident throughpartial loss of function.8.4 NEURODEVELOPMENTAL DISORDERS DUE TO ABERRANTEPIGENETIC PATTERNSWe will now look at some neurodevelopmental disorders that have arisen due to aberrantepigenetic patterns.8.4.1 Fragile X Spectrum DisordersThe FMR1 locus on the X chromosome is associated with three dist<strong>in</strong>ct fragile X spectrumdisorders: fragile X syndrome (FXS), fragile X-associated tremor/ataxia syndrome (FXTAS), andfragile X-associated primary ovarian <strong>in</strong>sufficiency (FXPOI). FXS is the lead<strong>in</strong>g cause of<strong>in</strong>herited cognitive disability, <strong>in</strong> which affected males have mild to severe cognitive disability,macro-orchidism and connective tissue dysplasia due to an expansion of a CGG tr<strong>in</strong>ucleotide(triplet) repeat located <strong>in</strong> the 5 0 untranslated region (UTR) of the FMR1 gene. The repeat lengthis variable <strong>in</strong> the normal human population, rang<strong>in</strong>g from six to 55 repeats [59]. Uponmaternal transmission, a repeat can become unstable, result<strong>in</strong>g <strong>in</strong> an expansion <strong>in</strong> theoffspr<strong>in</strong>g. Expansions <strong>in</strong>to the range of 55e200 repeats generate premutation FMR1 alleles,and expansions above 200 repeats from a premutation carrier female generate full-mutation


CHAPTER 8Emerg<strong>in</strong>g Role of <strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> Neurodevelopmental Disordersalleles. The latter expansions <strong>in</strong>itiate a series of molecular events lead<strong>in</strong>g to transcriptionalsilenc<strong>in</strong>g of the FMR1 gene. The order of events appears to be histone deacetylation and H3-K9methylation, followed by DNA methylation and H3-K4 demethylation [60]. Repeat b<strong>in</strong>d<strong>in</strong>gprote<strong>in</strong>s that attract HDACs, HMTs, DNMTs, and heterochromat<strong>in</strong> prote<strong>in</strong> 1 (HP1) areimplicated <strong>in</strong> this cascade of silenc<strong>in</strong>g events (reviewed <strong>in</strong> [61]. The absence of FMR1 transcriptionand consequentially FMR1 prote<strong>in</strong> (FMRP) <strong>in</strong> males with full-mutation alleles is thecause of FXS, whereas high expression of CGG-conta<strong>in</strong><strong>in</strong>g transcripts from premutation alleles<strong>in</strong>itiates a cascade of events that culm<strong>in</strong>ate <strong>in</strong> central nervous system degeneration and FXTAS,probably via alterations <strong>in</strong> the availability of RNA-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s. Female carriers ofpremutation alleles are at risk of develop<strong>in</strong>g FXPOI [62].Recent studies us<strong>in</strong>g neuronal and non-neuronal cell l<strong>in</strong>es from normal, premutation, and FXS<strong>in</strong>dividuals showed that the transcription of FMR1 gene is quite complex <strong>in</strong> these <strong>in</strong>dividuals.Exon 1 of FMR1 conta<strong>in</strong>s a sense promoter with three transcription start sites (sites 1e3) andan antisense promoter with multiple start sites. In addition there is another antisensepromoter located >10 kb downstream to the sense promoter. The activity of these promotersand usage of the transcription start sites was shown to be dependent on the repeat length. Incell l<strong>in</strong>es with normal repeat length, the distal-most start site <strong>in</strong> the sense promoter (site 3) ismostly used. As the repeat number <strong>in</strong>creases, the proximal sites (sites 1 and 2) are used moreheavily [63]. This promoter usage may expla<strong>in</strong> the observation that <strong>in</strong> female carriers, theFMR1 transcript levels are higher. In these carriers, the antisense transcripts (ASFMR1) fromthe distal promoter also predom<strong>in</strong>ate. However, <strong>in</strong> full-mutation cells, the antisense transcriptis not detected. It has been suggested that both FMR1 and ASFMR1 may contribute to thevariable phenotypes associated with the repeat expansion [64]. On the basis of the observationsof the antisense transcripts <strong>in</strong> FXS and other patients with triplet nucleotide expansions,Kumari and Usd<strong>in</strong> [61] proposed an RNA-based model for formation of heterochromat<strong>in</strong> <strong>in</strong>the full-mutation cases. In this model, the affected gene generates a region of double-strandedRNA (dsRNA) that is a Dicer substrate. Irrespective of the source of dsRNA, the dicer productsload onto the RNA-<strong>in</strong>duced transcriptional silenc<strong>in</strong>g (RITS) complex and target the complex tothe affected gene. RITS complex then facilitates hypoacetylation of histones H3 and H4,methylation of H3-K9, and DNA methylation.167The <strong>in</strong>volvement of epigenetic mechanisms <strong>in</strong> fragile X spectrum disorders extends beyondtranscriptional silenc<strong>in</strong>g of full-mutation FMR1 alleles by DNA methylation. In rare cases ofmales with unmethylated full mutations, histone H3/H4 acetylation and H3 lys<strong>in</strong>e 9methylation patterns were similar to those found <strong>in</strong> FXS cell l<strong>in</strong>es, suggest<strong>in</strong>g that these posttranslationalchanges alone are not sufficient for full-mutation allele <strong>in</strong>activation [65].Rather, it is likely that the acquisition of stable DNA methylation <strong>in</strong> the repeat-expandedpromoter region is required for complete transcription repression. Consistent with thisnotion are recent observations <strong>in</strong> iPS cells from FXS <strong>in</strong>dividuals of the permanence ofembryonically derived methylation on the full-mutation repeats [66]. In another example ofepigenetic regulation of repeat-expanded FMR1 alleles by mechanisms other than DNAmethylation, overexpression of FMR1 mRNA and the accompany<strong>in</strong>g neurodegeneration <strong>in</strong>a Drosophila model of premutation FXTAS can be effectively suppressed by the application ofhistone acetyltransferase <strong>in</strong>hibitors [67].Recently, much has been learned about the normal function of FMRP, which <strong>in</strong> turn hasprovided <strong>in</strong>sight <strong>in</strong>to pathogenesis of FXS, <strong>in</strong> which FMRP is not expressed or expressed atvery low levels. FMRP is a cytoplasmic RNA-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> found <strong>in</strong> synapses <strong>in</strong> CNSneurons, and known to repress translation of mRNAs [68]. One hypothesis on the cause ofFXS postulates that, <strong>in</strong> the absence of FMRP, loss of repression of metabotropic glutamatereceptor (mGluR)-mediated pathways leads to the dist<strong>in</strong>ct cognitive and behavioral featuresof FXS [69]. More specifically, FMRP may be <strong>in</strong>directly <strong>in</strong>volved <strong>in</strong> the temporal and spatialcontrols of translation by controll<strong>in</strong>g microRNA-mediated translational <strong>in</strong>hibition <strong>in</strong>


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>response to mGluR signals [70,71]. This connection between FMRP and mGluR signal<strong>in</strong>g maybe the reason for the mitigation of behavioral abnormalities <strong>in</strong> <strong>in</strong>dividuals with FXS by theadm<strong>in</strong>istration of an <strong>in</strong>hibitor of metabotropic glutamate receptor 5 (mGluR5)-mediatedsignal<strong>in</strong>g [72].8.4.2 Impr<strong>in</strong>t<strong>in</strong>g DisordersGenomic impr<strong>in</strong>t<strong>in</strong>g is an epigenetic process that dist<strong>in</strong>guishes alleles based on their parentalorig<strong>in</strong>. The ma<strong>in</strong> consequences of the impr<strong>in</strong>t<strong>in</strong>g processes are impr<strong>in</strong>ted genes <strong>in</strong> diploidcells of the develop<strong>in</strong>g mammalian organism <strong>in</strong> which one allele is transcribed and theopposite allele is transcriptionally silent (monoallelic expression). There are approximately100 known impr<strong>in</strong>ted genes. There are a number of different ways <strong>in</strong> which genome-wideimpr<strong>in</strong>t<strong>in</strong>g (set of monoallelically expressed genes) can be experimentally disrupted <strong>in</strong> thedevelop<strong>in</strong>g mouse embryo. These methods <strong>in</strong>clude failure to <strong>in</strong>herit the complete maternalset of impr<strong>in</strong>ts by block<strong>in</strong>g their formation <strong>in</strong> Dnmt3L -/- oocytes, absence of paternal set ofimpr<strong>in</strong>ts (parthenogenotes) and absence of maternal set of impr<strong>in</strong>ts (androgenotes). All ofthese methods lead to a comb<strong>in</strong>ation of loss of expression of some impr<strong>in</strong>ted genes andbialleic expression of other impr<strong>in</strong>ted genes and embryonic death. The approximately 100impr<strong>in</strong>ted genes are organized <strong>in</strong>to 16 dist<strong>in</strong>ct clusters, which are distributed amongapproximately one-half of the autosomes. Impr<strong>in</strong>t<strong>in</strong>g can also be lost locally by disrupt<strong>in</strong>gthe genetic or epigenetic <strong>in</strong>tegrity of a s<strong>in</strong>gle impr<strong>in</strong>ted gene or cluster of impr<strong>in</strong>ted genes.This can occur spontaneously or it can be experimentally <strong>in</strong>duced, and is usually associatedwith viable fetal outcomes.168Mutations or epigenetic changes with<strong>in</strong> one cluster of impr<strong>in</strong>ted genes has been particularlyreveal<strong>in</strong>g about the role of some impr<strong>in</strong>ted genes <strong>in</strong> neurodevelopment. The Angelman andPradereWilli syndromes are associated with mutations <strong>in</strong> a cluster of impr<strong>in</strong>ted genes onchromosome 15. Angelman syndrome is a neurodevelopmental disorder characterized by<strong>in</strong>tellectual and developmental delays, sleep disturbance, seizures, jerky movement,frequent laughter or smil<strong>in</strong>g, and usually a happy demeanor. It is due to a loss of expressionof the Ube3A gene, which is normally impr<strong>in</strong>ted and expressed <strong>in</strong> the central nervoussystem from just the maternal allele [73]. This gene encodes for an E6-AP ubiquit<strong>in</strong> ligase.The frequency is approximately 1 <strong>in</strong> 20 000 live births, and it is most commonly due to denovo deletions <strong>in</strong> an upstream impr<strong>in</strong>t<strong>in</strong>g control element for Ube3A expression. Besidesthe behavioral and neurological symptoms, there are also pathognomonic neurophysiologicalf<strong>in</strong>d<strong>in</strong>gs. Angelman syndrome is not a neurodegenerative syndrome, and <strong>in</strong>dividualswith Angelman’s syndrome have a near normal lifespan. PradereWilli syndrome isa neurodevelopmental disorder dist<strong>in</strong>ct from Angelman syndrome that occurs ata frequency of 1/25 000 to 1/10 000 births. IQ is low, but there is much variability. Childrenshow an unusual cognitive profile, and have behavioral problems, primarily hyperphagiawith the development of morbid obesity. These are recognizable, viable syndromes withdist<strong>in</strong>ct cl<strong>in</strong>ical characteristics and neurodevelopmental abnormalities because of thelocalized nature of the mutations or epigenetic abnormality, rather than a completedisruption of genomic impr<strong>in</strong>t<strong>in</strong>g at all DMDs.Separate disruptions with<strong>in</strong> the same cluster of impr<strong>in</strong>ted genes on chromosome 15 lead to thetwo cl<strong>in</strong>ically dist<strong>in</strong>ct syndromes. Important studies of Angelman and PradereWilli syndromepatients with deletions with<strong>in</strong> the chromosome-15 cluster of impr<strong>in</strong>ted genes identifieda region deleted <strong>in</strong> all PradereWilli patients and a separate region deleted <strong>in</strong> all Angelmansyndrome patients with Chromosome-15 deletions. These are designated SRO (shared regionof overlap; [74], reviewed <strong>in</strong> [75]). Studies of the PradereWilli SRO revealed important<strong>in</strong>sights <strong>in</strong>to the molecular and developmental etiology of the syndrome. With<strong>in</strong> the SRO isa differentially methylated doma<strong>in</strong> (DMD). Approximately one-half of PradereWilli syndromecases are due to microdeletions of paternal chromosome 15, with<strong>in</strong> the SNURF/SNRPN cluster,


CHAPTER 8Emerg<strong>in</strong>g Role of <strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> Neurodevelopmental Disordersremov<strong>in</strong>g the paternal allele of the DMD and usually adjacent sequences/genes. Deletion of theunmethylated paternal SNURF/SNRPN DMD allele leads to transcriptional repression of theimpr<strong>in</strong>ted genes controlled by the DMD. Most of the rema<strong>in</strong><strong>in</strong>g PWS cases are associated withmaternal heteroisodisomy and paternal nullisomy of chromosome 15. In these cases, genesregulated by the DMD sequences are transcriptionally repressed because both (maternal)alleles of the DMD are methylated. Importantly, the other DMDs <strong>in</strong> the genome functionnormally. In a similar ve<strong>in</strong>, most cases of RusselleSilver syndrome (not a neurodevelopmentaldisorder) are caused by loss of paternal IGF2/H19 DMD methylation, lead<strong>in</strong>g to a loss of IGF2expression <strong>in</strong> the develop<strong>in</strong>g fetus, which accounts for the markedly reduced weight at birth.Most cases of BeckwitheWeidemann syndrome (another impr<strong>in</strong>t<strong>in</strong>g disorder) are due to denovo epigenetic events <strong>in</strong> which methylation on the maternal KvDMR (KCNQ1 DMD) is neverestablished dur<strong>in</strong>g oogenesis or, if established, not ma<strong>in</strong>ta<strong>in</strong>ed <strong>in</strong> the period immediatelyfollow<strong>in</strong>g fertilization. Aga<strong>in</strong>, as with PWS and RSS, only a s<strong>in</strong>gle DMD functions abnormally,either because of a mutation or because of an abnormality <strong>in</strong> its normal epigenetic <strong>in</strong>tegrity.The developmental etiologies of the two syndromes can be best understood with the realizationthat the ICs for the two syndromes are sites of critical parent-specific epigeneticmodifications. The impr<strong>in</strong>t<strong>in</strong>g process creates a few DMDs <strong>in</strong> the genome (16 <strong>in</strong> the mouseand probably a similar number <strong>in</strong> the human), which are unique landmarks of <strong>in</strong>heritedparent-specific epigenetic <strong>in</strong>formation. For each DMD, one parental allele is methylated,hav<strong>in</strong>g acquired its methylation <strong>in</strong> the parent’s germl<strong>in</strong>e and ma<strong>in</strong>ta<strong>in</strong>ed (<strong>in</strong>herited) thismethylation after fertilization. The opposite parental DMD allele does not acquire methylationdur<strong>in</strong>g gametogenesis, and thus ma<strong>in</strong>ta<strong>in</strong>s the unmethylated epigenetic state follow<strong>in</strong>gfertilization <strong>in</strong> the embryo. How do DMDs appear <strong>in</strong> the genome? DNA methylation is anepigenetic process that can be logically divided <strong>in</strong>to two dist<strong>in</strong>ct purposes, the <strong>in</strong>heritance of<strong>in</strong>formation and the control of transcription. Impr<strong>in</strong>t<strong>in</strong>g is primarily an issue of the <strong>in</strong>heritanceof epigenetic <strong>in</strong>formation and secondarily an issue of regulation of transcription. Thus,certa<strong>in</strong> impr<strong>in</strong>t<strong>in</strong>g disorders are neurodevelopmental disorders associated with disruption ofcritically important <strong>in</strong>herited methylation patterns.169Another reason for the existence of impr<strong>in</strong>t<strong>in</strong>g disorders is that the majority of impr<strong>in</strong>ted genesare expressed either <strong>in</strong> the placenta or <strong>in</strong> the develop<strong>in</strong>g embryonic nervous system, possiblyat later stages of development, result<strong>in</strong>g <strong>in</strong> survivors with the syndrome. The spatial andtemporal expression of impr<strong>in</strong>ted genes is even more constra<strong>in</strong>ed when referr<strong>in</strong>g to geneswith<strong>in</strong> a s<strong>in</strong>gle cluster.8.5 MATERNAL DUPLICATIONS VERSUS PATERNALDUPLICATIONS IN PWS/AS REGIONFamilial analyses of <strong>in</strong>dividuals affected with neuropsychiatric disorders suggest thatwhereas maternal duplications <strong>in</strong> the human 15q11.2e15q13 regions are associated withautism, paternal duplications are associated with milder phenotypes with reduced penetranceand even benign <strong>in</strong> some cases [76]. A recent study shows that maternal duplicationsare more frequently associated with schizophrenia and schizoaffective disorders thanpaternal duplications [77]. Although the precise molecular basis for pathogenicity is notestablished, it is clear from these studies that altered gene dosage is the underly<strong>in</strong>gmechanism. Consistent with this possibility, maternal isodicentric chromosomes significantly<strong>in</strong>crease the risk for autism spectrum disorder (ASD). Expression analysis of cellsfrom patients carry<strong>in</strong>g the maternal duplications suggested <strong>in</strong>volvement of altered epigeneticpatterns with complex changes <strong>in</strong> expression of the genes <strong>in</strong> the 15q11.2e15q13region. This region also <strong>in</strong>cludes three non-impr<strong>in</strong>ted GABA receptors and thereforechanges <strong>in</strong> the copy number/gene dosage <strong>in</strong>volv<strong>in</strong>g GABA receptors may lead to ASDphenotypes. Consistent with this possibility, altered expression patterns of GABRB3,


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>GABRA5, andGABRG3 was observed <strong>in</strong> some of the postmortem cerebral cortex samplesfrom patients with ASD [78]. Taken together, these reports suggest that mechanism ofpathogenesis due to changes <strong>in</strong> the copy number of genes <strong>in</strong> the 15q11.2e15q13 is not yetclear and can be fairly complex.8.5.1 Possibilities of EpimutationsEpimutation can be described as a heritable change <strong>in</strong> gene expression that does not affect theactual base-pair sequence of DNA [79] and should be able to expla<strong>in</strong> the phenotype observed<strong>in</strong> a patient. From the discussion above, epimutations <strong>in</strong>clude both DNA methylation andhistone modification changes. Epimutations, by def<strong>in</strong>ition should <strong>in</strong>volve one of the twoalleles of a candidate gene that are equivalent to loss of function through silenc<strong>in</strong>g of anessential gene or <strong>in</strong>appropriate activation of a gene which is otherwise silent <strong>in</strong> that particulartissue. At present there are only two cases implicat<strong>in</strong>g epigenetic modifications of candidategenes for neuropsychiatric disorders. One of these two examples <strong>in</strong>cludes FMR1 as describedabove and the second one is RELN. A polymorphic form of RELN conta<strong>in</strong><strong>in</strong>g longer GGCrepeats was shown to be preferentially transmitted to ASD patients and was associated witha reduced expression of RELN [80]. The mechanism of silenc<strong>in</strong>g could <strong>in</strong>volve altered epigeneticmodifications as described <strong>in</strong> the case of FMR1. It may be noted however that epimutations(without <strong>in</strong>volv<strong>in</strong>g a change <strong>in</strong> the DNA sequence) of a vast majority of candidategenes for neuropsychiatric disorders are yet to be discovered.1708.6 CONCLUSIONSAll of the many different cell types of mammalian species use aspects of the epigeneticmach<strong>in</strong>ery for regulation of gene expression or control the <strong>in</strong>heritance of epigenetic modificationsof DNA sequences. Very few epigenetic processes, most notably the <strong>in</strong>fluence of MeCP2on neuronal gene expression, are functionally conf<strong>in</strong>ed to the nervous system. Because of this,autosomal recessive or X-l<strong>in</strong>ked mutations <strong>in</strong> genes encod<strong>in</strong>g components of epigeneticprocesses most likely affect the development and function of many organs and consequentlylead to embryonic death <strong>in</strong> humans. Few neurodevelopmental syndromes associated withgenetic mutations <strong>in</strong> a component of an epigenetic process have been recognized. Most<strong>in</strong>dividuals with neurodevelopmental syndromes due to mutations <strong>in</strong> genes controll<strong>in</strong>gepigenetic processes are X-chromosomal mosaics (with recessive X-l<strong>in</strong>ked mutations), haveautosomal dom<strong>in</strong>ant mutations, or have partial loss-of-function mutations.References[1] Tahiliani M, Koh KP, Shen Y, Pastor WA, Bandukwala H, Brudno Y, et al. Conversion of 5-methylcytos<strong>in</strong>e to5-hydroxylmethylcytos<strong>in</strong>e <strong>in</strong> mammalian DNA by MLL partner TET1. <strong>Science</strong> 2009;324:930e5.[2] Re<strong>in</strong>hart B, Chaillet JR, Genomic impr<strong>in</strong>t<strong>in</strong>g: cis-act<strong>in</strong>g sequences and regional control. Int Rev Cytol2005;243:173e213.[3] Ji H, Ehrlich LI, Seita J, Murakami P, Doi A, L<strong>in</strong>dau P, et al. Comprehensive methylome map of l<strong>in</strong>eagecommitment from haematopoietic progenitors. Nature 2010;467:338e42.[4] Bestor TH. The DNA methyltransferases of mammals. Hum Mol Genet 2000;9:2395e402.[5] Nan X, Campoy FJ, Bird A. MeCP2 is a transcriptional repressor with abundant b<strong>in</strong>d<strong>in</strong>g sites <strong>in</strong> genomicchromat<strong>in</strong>. Cell 1997;88:471e81.[6] Suganuma T, Workman JL. Signals and comb<strong>in</strong>atorial functions of histone modifications. Annu Rev Biochem2011;80:473e99.[7] Liu J, Krantz ID. Cohes<strong>in</strong> and human <strong>Disease</strong>. Annu Rev Genom <strong>Human</strong> Genetics 2008;9:303e20.[8] Fan G, Beard C, Chen RZ, Csankovszki G, Sun Y, S<strong>in</strong>iaia M, et al. DNA hypomethylation perturbs the functionand survival of CNS neurons <strong>in</strong> postnatal animals. J Neurosci 2001;21:788e97.[9] Feng J, Zhou Y, Campbell SL, Le T, Li E, Sweatt JD, et al. Dnmt1 and Dnmt13a ma<strong>in</strong>ta<strong>in</strong> DNA methylation andregulate synaptic function <strong>in</strong> adult forebra<strong>in</strong> neurons. Nat Neurosci 2010;13:423e30.


CHAPTER 8Emerg<strong>in</strong>g Role of <strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> Neurodevelopmental Disorders[10] Veldic M, Caruncho HJ, Liu WS, Davis J, Satta R, Grayson DR, et al. DNA-methyltransferase 1 mRNA isselectively overexpressed <strong>in</strong> telencephalic GABAergic <strong>in</strong>terneurons of schizophrenia bra<strong>in</strong>s. Proc Natl Acad SciUSA 2004;101:348e53.[11] Zhubi A, Veldic M, Puri NV, Kadriu B, Caruncho H, Loza I, et al. An upregulation of DNA-methyltransferase 1and 3a expressed <strong>in</strong> telencephalic GABAergic neurons of schizophrenia patients is also detected <strong>in</strong> peripheralblood lymphocytes. Schizophr Res 2009;111:115e22.[12] B<strong>in</strong>iszkiewicz D, Gribnau J, Ramsahoye B, Gaudet F, Eggan K, Humpherys D, et al. Dnmt1 overexpressioncauses genomic hypermethylation, loss of impr<strong>in</strong>t<strong>in</strong>g, and embryonic lethality. Mol Cell Biol2002;22:2124e35.[13] D<strong>in</strong>g F, Chaillet JR. In vivo stabilization of the Dnmt1 (cytos<strong>in</strong>e-5)-methyltransferase prote<strong>in</strong>. Proc Natl AcadSci USA 2002;99:14861e6.[14] D’Aiuto LD, Di Maio R, Mohan KN, M<strong>in</strong>erv<strong>in</strong>i C, Saporiti F, Soreca I, et al. Mouse ES cells overexpress<strong>in</strong>gDNMT1 produce abnormal neurons with upregulated NMDA/NR1 subunit. Differentiation 2011;82:9e17.[15] Kle<strong>in</strong> CJ, Botuyan MV, Wu Y, Ward CJ, Nicholson GA, Hammans S, et al. Mutations <strong>in</strong> DNT1 cause hereditarysensory neuropathy with dementia and hear<strong>in</strong>g loss. Nat Genet 2011;43:595e600.[16] Fell<strong>in</strong>ger K, Rothbauer U, Felle M, Langst G, Leonhardt H. Dimerization of DNA methyltransferase 1 ismediated by its regulatory doma<strong>in</strong>. J Cell Biochem 2009;106:521e8.[17] Xu GL, Bestor TH, Bourc’his D, Hsieh CL, Tommerup N, Bugge M, et al. Chromosome <strong>in</strong>stability and immunodeficiencysyndrome caused by mutations <strong>in</strong> a DNA methyltransferase gene. Nature 1999;402:187e91.[18] Urd<strong>in</strong>guio RG, Sanchez-Mut JV, Esteller M. Epigenetic mechanisms <strong>in</strong> neurological diseases: genes, syndromes,and therapies. Lancet Neurol 2009;8:1056e72.[19] Amir RE, Zoghbi HY. Rett syndrome: Methyl-CpG b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> 2 mutations and phenotype-genotypecorrelations. Am J Med Genet 2000;97:147e52.[20] Ishii T, Makita Y, Ogawa A, Amamiya S, Yamamoto M, Miyamoto A, et al. The role of different X-<strong>in</strong>activationpattern on the variable cl<strong>in</strong>ical phenotype with Rett syndrome. Bra<strong>in</strong> Dev 2001;23:S161e4.[21] Villard L, Levy N, Xiang F, Kpebe A, Labelle V, Chevillard C, et al. Segregation of a totally skewed pattern of Xchromosome <strong>in</strong>activation <strong>in</strong> four familial cases of Rett syndrome without MECP2 mutation: implications forthe disease. J Med Genet 2001;38:435e42.[22] Amir RE, Van den Veyver IB, Wan M, Tran CQ, Francke U, Zoghbi HY. Rett syndrome is caused by mutations <strong>in</strong>X-l<strong>in</strong>ked MWCP2, encod<strong>in</strong>g methyl-CpG-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> 2. Nat Genet 1999;23:185e8.[23] Del Gaudio D, Fang P, Scaglia F, Ward PA, Craigen WJ, Glaze DG, et al. Increased MECP2 gene copynumber as the result of genomic duplication <strong>in</strong> neurodevelopmentally delayed males. Genet Med 2006;8:784e92.[24] Chen RZ, Akbarian S, Tudor M, Jaenisch R. Deficiency of methyl-CpG b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>-2 <strong>in</strong> CNS neuronsresults <strong>in</strong> a Rett-like phenotype <strong>in</strong> mice. Nat Genet 2001;27:327e31.[25] Coll<strong>in</strong>s AL, Levenson JM, Vilaythong AP, Richman R, Armstrong DL, Noebels JL, et al. Mild overexpression ofMeCP2 causes a progressive neurological disorder <strong>in</strong> mice. Hum Molec Genet 2004;13:2679e89.[26] Skene PJ, Ill<strong>in</strong>gworth RS, Webb S, Kerr ARW, James KD, Turner DJ, et al. Neuronal MeCP2 is expressed at nearhistone-octomer levels and globally alters the chromat<strong>in</strong> state. Mol Cell 2010;37:457e68.[27] Chahrour M, Jung SY, Shaw C, Zhou X, Wong STC, Q<strong>in</strong> J, et al. MeCP2, a key contributor to neurologicaldisease, activates and represses transcription. <strong>Science</strong> 2008;320:1224e9.[28] Ballas N, Lioy DT, Grunseich C, Mandel G. Non-cell autonomous <strong>in</strong>fluence of MeCP2-deficient glia onneuronal dendritic morphology. Nat Neurosci 2009;12:311e7.[29] Maezawa I, Swanberg S, Harvey D, LaSalle JM, J<strong>in</strong> LW. Rett syndrome astrocytes are abnormal and spreadMeCP2 deficiency through gap junctions. J Neurosci 2009;29:5051e61.[30] Maezawa I, J<strong>in</strong> LW. Rett syndrome microglia damage dendrites and synapses by the elevated release ofglutamate. J Neurosci 2010;30:5346e56.[31] Lioy DT, Garg SK, Monaghan CE, Raber J, Foust KD, Kaspar BK, et al. A role for glia <strong>in</strong> the progression of Rett’ssyndrome. Nature 2011;475:497e500.[32] Armstrong D, Dunn JK, Antalffy B, Trivedi R. Selective dendritic alterations <strong>in</strong> the cortex of Rett syndrome.J. Neuropathol Exp Neurol 1995;54:195e201.[33] Guy J, Gan J, Selfridge J, Cobb S, Bird A. Reversal of neurological defects <strong>in</strong> a mouse model of Rett syndrome.<strong>Science</strong> 2007;315:1143e7.[34] McGraw CM, Samaco RC, Zoghbi HY. Adult neural function requires MeCP2. <strong>Science</strong> 2011;333:186.[35] Nan X, Hou J, Maclean A, Nasir J, Lafuente MJ, Shu X, et al. Interaction between chromat<strong>in</strong> prote<strong>in</strong>s MECP2and ATRX is disrupted by mutations that cause <strong>in</strong>herited mental retardation. Proc Natl Acad Sci USA2007;104:2709e14.[36] Gibbons RJ, Higgs DR. Molecular-cl<strong>in</strong>ical spectrum of the ATR-X syndrome. Am J Med Genet 2000;97:204e12.171


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>172[37] McDowell TL, Gibbons RJ, Sutherland H, O’Rourke DM, Bickmore WA, Pombo A, et al. Localization ofa putative transcriptional regulator (ATRX) at pericentromeric heterochromat<strong>in</strong> and the short arms of acrocentricchromosomes. Proc Natl Acad Sci USA 1999;96:13983e8.[38] W<strong>in</strong>nepenn<strong>in</strong>ckx B, Errijgers V, Hayez-Delatte F, Reyniers E, Kooy RF. Identification of a family withnonspecific mental retardation (MRX79) with the A140V mutation <strong>in</strong> the MECP2 gene: Is there a need forrout<strong>in</strong>e screen<strong>in</strong>g? Hum Mutat 2002;20:249e52.[39] Kernohan KD, Jiang Y, Tremblay DC, Bonvissuto AC, Eubanks JH, Mann MRW, et al. ATRX partners withcohes<strong>in</strong> and MeCP2 and contributes to developmental silenc<strong>in</strong>g of impr<strong>in</strong>ted genes <strong>in</strong> the bra<strong>in</strong>. Dev Cell2010;18:191e202.[40] Krantz ID, McCallum J, DeScipio C, Kaur M, Gillis LA, Yaeger D, et al. Cornelia de Lange syndrome is caused bymutations <strong>in</strong> NIPBL, the human homolog of Drosophila melanogaster Nipped-B. Nat Genet 2004;36:631e5.[41] Hul<strong>in</strong>sky R, Byrne JLB, Lowchik A, Viskochil DH. Fetus with <strong>in</strong>terstitial del(5)(p13.1p14.2) diagnosed postnatallywith Cornelia de Lange Syndrome. Am J Med Genet 2005;137A:336e8.[42] Musio A, Selicorni A, Focarelli ML, Gervas<strong>in</strong>i C, Milani D, Russo S, et al. X-l<strong>in</strong>ked Cornelia de Lange syndromeow<strong>in</strong>g to SMC1L1 mutations. Nat Genet 2005;38:528e30.[43] Deardorff MA, Kaur M, Yaeger D, Rampuria A, Korolev S, Pie J, et al. Mutations <strong>in</strong> cohes<strong>in</strong> complex membersSMC3 and SMC1A cause a mild variant of Cornelia de Lange syndrome with predom<strong>in</strong>ant mental retardation.Am J Hum Genet 2007;80:485e94.[44] Vega H, Waisfisz Q, Gordillo M, Sakai N, Yanagihara I, Yamada M, et al. Roberts syndrome is caused bymutations <strong>in</strong> ESCO2, a human homolg of yeast ECO1 that is essential for the establishment of sister chromatidcohes<strong>in</strong>. Nat Genet 2005;37:468e70.[45] Roll<strong>in</strong>s RA, Morcillo P, Dorsett D. Nipped-B, a Drosophila homologue of chromosomal adher<strong>in</strong>s, participates<strong>in</strong> activation by remote enhancers <strong>in</strong> the cut and Ultrabithorax genes. Genetics 1999;152:577e93.[46] Roll<strong>in</strong>s RA, Korom M, Aulnr N, Martens A, Dorsett D. Drosophila Nipped-B prote<strong>in</strong> supports sister chromatidcohes<strong>in</strong> and opposes the Stromal<strong>in</strong>/Scc3 cohesion factor to facilitate long-range activation of the cut gene. MolCell Biol 2004;24:3100e11.[47] Chang CR, Wu CS, Hom Y, Gartenberg MR. Target<strong>in</strong>g of cohes<strong>in</strong> by transcriptionally silent chromat<strong>in</strong>. GenesDevelopment 2005;19:3031e42.[48] Dorsett D. Distant liaisons: long range enhancer-promoter <strong>in</strong>teractions <strong>in</strong> Drosophila. Curr Op<strong>in</strong> Genet Dev1999;9:505e14.[49] Dorsett D. Roles of the sister chromatid cohes<strong>in</strong> apparatus <strong>in</strong> gene expression, development, and humansyndromes. Chromosoma 2007;116:1e13.[50] Wan LB, Pan H, Hannenhalli S, Cheng Y, Ma J, Fedoriw A, et al. Maternal depletion of CTCF reveals multiplefunctions dur<strong>in</strong>g oocyte and preimplantation embryo development. Development 2008;135:2729e38.[51] Lee J, Hagerty S, Cormier KA, Kim J, Kung AL, Ferrante RJ, et al. Monoallelic deletion of CBP leads to pericentromericheterochromat<strong>in</strong> condensation through ESET expression and histone H3 (K9) methylation. HumMol Genet 2008;17:1774e82.[52] Rebel VI, Kung AL, Tanner EA, Yang H, Bronson RT, Liv<strong>in</strong>gston DM. Dist<strong>in</strong>ct roles for CREB-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>and p300 <strong>in</strong> hematopoietic stem cell self-renewal. Proc Natl Acad Sci USA 2002;99:14789e94.[53] Josselyn SA. What’s right with my mouse model? New <strong>in</strong>sights <strong>in</strong>to the molecular and cellular basis ofcognition from mouse models of Rub<strong>in</strong>ste<strong>in</strong>-Taybi syndrome. Learn Mem 2005;12:80e3.[54] Wang J, Weaver IC, Gauthier-Fisher A, Wang H, He L, Yeomans J, et al. CBP histone acetyltransferase activityregulates embryonic neural differentiation <strong>in</strong> the normal and Rub<strong>in</strong>ste<strong>in</strong>-Taybi syndrome bra<strong>in</strong>. Dev Cell2010;18:114e25.[55] Poirier R, Jacquot S, Vaillend C, Soutthiphong AA, Libbey M, Davis S, et al. Deletion of the Coff<strong>in</strong>eLowrysyndrome gene Rsk2 <strong>in</strong> mice is associated with impaired spatial learn<strong>in</strong>g and reduced control of exploratorybehavior. Behav Genet 2007;37:31e50.[56] Dugani CB, Paqu<strong>in</strong> A, Kaplan DR, Miller FD. Coff<strong>in</strong>eLowry syndrome: a role for RSK2 <strong>in</strong> mammalianneurogenesis. Dev Biol 2010;347:348e59.[57] Ng SB, Bigham AW, Buck<strong>in</strong>gham KJ, Hannibal MC, McMill<strong>in</strong> MJ, Gildersleeve HI, et al. Exome sequenc<strong>in</strong>gidentifies MLL2 mutations as a cause of Kabuki syndrome. Nat Genet 2010;42:790e3.[58] Li Y, Bogershausen N, Alanay Y, Kiper POS, Plume N, Keupp K, et al. A mutation screen <strong>in</strong> patients with Kabukisyndrome. Hum Genet 2011;130:715e24.[59] Willemsen R, Levenga J, Oostra BA. CGG repeat <strong>in</strong> the FMR1 gene: size matters. Cl<strong>in</strong> Genet 2011;80:214e25.[60] Pietrobono R, Tabolacci E, Zalfa F, Zito I, Terracciano A, Moscato U, et al. Molecular dissection of the eventslead<strong>in</strong>g to <strong>in</strong>activation of the FMR1 gene. Hum Mol Genet 2005;14:267e77.[61] Kumari D, Usd<strong>in</strong> K. Chromat<strong>in</strong> remodel<strong>in</strong>g <strong>in</strong> the noncod<strong>in</strong>g repeat expansion diseases. J Biol Chem2009;284:7413e7.


CHAPTER 8Emerg<strong>in</strong>g Role of <strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> Neurodevelopmental Disorders[62] All<strong>in</strong>gham-Hawk<strong>in</strong>s DJ, Babul-Hirji R, Chitayat D, Holden JJA, Yang KT, Lee C, et al. Fragile X premutation isa significant risk factor for premature ovarian failure: The <strong>in</strong>ternational collaborative POF <strong>in</strong> fragile Xstudydprelim<strong>in</strong>ary data. J Med Genet 1999;83:322e5.[63] Ladd PD, Smith LE, Rabaia NA, Moore JM, Georges SA, Hansen RS, et al. An antisense transcript spann<strong>in</strong>g theCGG repeat region of FMR1 is upregulated <strong>in</strong> premutation carriers but silenced <strong>in</strong> full mutation <strong>in</strong>dividuals.Hum Mol Genet 2007;24:3174e87.[64] Beil<strong>in</strong>a A, Tassone F, Schwartz PH, Sahota P, Hagerman PJ. Redistribution of transcription start sites with<strong>in</strong> theFMR1 promoter region with expansion of the downstream CGG-repeat element. Hum Mol Genet2004;13:543e9.[65] Tabolacci E, Moscato U, Zalfa F, Bagni C, Chiurazzi P, Neri G. Epignetic analysis reveals a euchromaticconfiguration <strong>in</strong> the FMR1 unmethylated full mutations. Eur J Hum Genet 2008;16:1487e98.[66] Urbach A, BarNur O, Daley GQ, Bevenistry N. Differential model<strong>in</strong>g of fragile X syndrome by humanembryonic stem cells and <strong>in</strong>duced pluripotent stem cells. Cell Stem Cell 2010;6:407e11.[67] Todd PK, Oh SY, Krans A, Pandey UB, Di Prospero NA, M<strong>in</strong> K-T, et al. Histone deacetylases suppress CGGrepeat-<strong>in</strong>duced neurodegeneration via transcriptional silenc<strong>in</strong>g <strong>in</strong> models of fragile X tremor ataxia syndrome.PLoS Genet 2010;6:12.e1001240.[68] Zalfa F, Giorgi M, Primerano B, Moro A, D Penta A, et al. The fragile X syndrome prote<strong>in</strong> FMRP associates withBC1 RNA and regulates the translation of specific mRNAs at synapses. Cell 2003;112:317e27.[69] Bear MF, Huber KM, Warren ST. The mGluR theory of fragile X mental retardation. Trends Neurosci2004;27:370e7.[70] J<strong>in</strong> P, Zarnescu DC, Ceman S, Nakamoto M, Mowrey J, Jongens TA, et al. Biochemical and genetic <strong>in</strong>teractionbetween the fragile X mental retardation prote<strong>in</strong> and the microRNA pathway. Nat Neurosci 2004;2:113e7.[71] Muddashetty RS, Nalavadi VC, Gross C, Yao X, X<strong>in</strong>g L, Laur O, et al. Reversible <strong>in</strong>hibition of PSD-95 mRNAtranslation by mir-125a, FMRP phosphorylation, and mGluR signal<strong>in</strong>g. Mol Cell 2011;42:673e88.[72] Jacquemont S, Curie A, des Portes V, Torrioli MG, Berry-Kravis E, Hagerman RJ, et al. Epigenetic modificationof he FMR1 gene <strong>in</strong> fragile X syndrome is associated with differential response to the mGluR antagonistAFQ056. Sci Trans Med 2011;3:1e9.[73] Kish<strong>in</strong>o T, Lalande M, Wagstaff J. UBE3A/E6-AP mutations cause Angelman syndrome. Nat Genet1997;15:70e3.[74] Ohta T, Gray TA, Rogan PK, Buit<strong>in</strong>g K, Gabriel JM, Saitoh S, et al. Impr<strong>in</strong>t<strong>in</strong>g-mutation mechanisms <strong>in</strong>PradereWilli syndrome. Am J Hum Genet 1999;64:397e413.[75] Horsthemke B, Wagstaff J. Mechanisms of impr<strong>in</strong>t<strong>in</strong>g of the PradereWilli/Angelman region. Am J Med Genet2008;146A:2041e52.[76] Cook Jr EH, L<strong>in</strong>dgren V, Leventhal BL, Courchesne R, L<strong>in</strong>coln A, Shulman C, et al. Autism or atypical autism <strong>in</strong>maternally but not paternally derived proximal 15q duplication. Am J Hum Genet 1997;60:928e34.[77] Ingason A, Kirov G, Giegl<strong>in</strong>g I, Hansen T, Isles AR, Jakobsen KD, et al. Maternally derived microduplications at15q11-q13: implication of impr<strong>in</strong>ted genes <strong>in</strong> psychotic illness. Am J Psychiatry 2011;168:408e17.[78] Hogart A, Nagarajan RP, Patzel KA, Yasui DH, Lasalle JM. 15q11 to 15q13 GABAA receptor genes are normallybiallelically expressed <strong>in</strong> bra<strong>in</strong> yet are subject to epigenetic dysregulation <strong>in</strong> autism-spectrum disorders. HumMol Genet 2007;16:691e703.[79] Jablonka E, Lamb MJ. Evolution <strong>in</strong> Four Dimensions: Genetic, Epigenetic, Behavioral, and Symbolic Variation<strong>in</strong> the History of Life. Cambridge: MIT Press; 2005.[80] Persico AM, Levitt P, Pimenta AF. Polymorphic GGC repeat differentially regulates human reel<strong>in</strong> geneexpression levels. J Neural Transm 2006;113:1373e82.173


This page <strong>in</strong>tentionally left blank


CHAPTER 9The <strong>Epigenetics</strong> ofAlzheimer’s <strong>Disease</strong>Fabio CoppedèUniversity of Pisa, Pisa, ItalyCHAPTER OUTLINE9.1 Alzheimer’s <strong>Disease</strong> 1759.2 One-Carbon Metabolism and DNAMethylation <strong>in</strong> Alzheimer’s<strong>Disease</strong> 1779.2.1 An Overview of One-CarbonMetabolism 1789.2.2 One-Carbon Metabolism <strong>in</strong>Alzheimer’s <strong>Disease</strong> 1789.2.3 Studies <strong>in</strong> Cell Cultures andAnimal Models 1809.2.4 Studies <strong>in</strong> <strong>Human</strong>s 1829.2.5 L<strong>in</strong>k<strong>in</strong>g the MethylationPotential to TauPhosphorylation<strong>in</strong> Alzheimer’s <strong>Disease</strong> 1829.3 Histone Tail Modifications andAlzheimer’s <strong>Disease</strong> 1839.3.1 Histone Modifications <strong>in</strong>Alzheimer’s <strong>Disease</strong>Bra<strong>in</strong>s 1849.3.2 HDAC Inhibitors and MemoryFunction <strong>in</strong> AD AnimalModels 1849.4 RNA-Mediated Mechanisms andAlzheimer’s <strong>Disease</strong> 1859.5 Discussion andConclusions 186References 1871759.1 ALZHEIMER’S DISEASEAlzheimer’s disease (AD) is an age-related neurodegenerative disorder and the most commonform of dementia <strong>in</strong> the elderly. The disease is cl<strong>in</strong>ically characterized by a progressiveneurodegeneration <strong>in</strong> selected bra<strong>in</strong> regions, <strong>in</strong>clud<strong>in</strong>g the temporal and parietal lobes andrestricted regions with<strong>in</strong> the frontal cortex and the c<strong>in</strong>gulate gyrus, lead<strong>in</strong>g to memory lossaccompanied by changes <strong>in</strong> behavior and personality severe enough to affect daily life. It wasreported that worldwide 26.6 million <strong>in</strong>dividuals suffered from AD <strong>in</strong> 2006, and this numberwill quadruple by 2050 follow<strong>in</strong>g the global ag<strong>in</strong>g of the world’s population [1]. Therefore, ADis becom<strong>in</strong>g a serious health concern <strong>in</strong> both developed and develop<strong>in</strong>g countries, particularlybecause there is actually no cure for the disease.Affected bra<strong>in</strong> regions <strong>in</strong> AD are characterized by the occurrence of extracellular amyloiddeposits or senile plaques (SP) and by the presence of neurofibrillary tangles (NFT) composedof <strong>in</strong>traneuronal aggregates of hyperphosphorylated tau prote<strong>in</strong> [2]. The primary componentof SP is an approximately 40-residue-long peptide, known as amyloid b (Ab) peptide, result<strong>in</strong>gT. Tollefsbol (Ed): <strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>. DOI: 10.1016/B978-0-12-388415-2.00009-3Copyright Ó 2012 Elsevier Inc. All rights reserved.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>from the proteolytic process<strong>in</strong>g of its precursor, the amyloid precursor prote<strong>in</strong> (APP). APP canbe processed either by a-secretase and g-secretase (a prote<strong>in</strong> complex composed by presenil<strong>in</strong>sand other prote<strong>in</strong>s) produc<strong>in</strong>g non-amyloidogenic peptides, or by b-secretase (b-site APPcleav<strong>in</strong>g enzyme 1, BACE1) and g-secretase-produc<strong>in</strong>g Ab peptides. Therefore the balancebetween different secretase activities is very important <strong>in</strong> the ma<strong>in</strong>tenance of the physiologicallevels of non-amyloidogenic and amyloidogenic fragments. The two major forms of Ab thatare produced by APP process<strong>in</strong>g under normal conditions are 40 and 42 residues <strong>in</strong> length(Ab 40 and Ab 42 , respectively). Ab 42 is the major component of SP. In a normal <strong>in</strong>dividual themajority of Ab produced is of the shorter variety, Ab 40 ; whereas mutations caus<strong>in</strong>g familial ADlead to <strong>in</strong>creased Ab 42 /Ab 40 ratio [3,4].176AD is a complex multifactorial disorder. Rare mutations <strong>in</strong> APP, presenil<strong>in</strong>-1 (PSEN1), andpresenil<strong>in</strong>-2 (PSEN2) genes cause early-onset (< 65 years) familial forms of the diseaseaccount<strong>in</strong>g for less than 1% of the total AD cases [5]. AD-causative mutations lead to alteredAPP production and/or process<strong>in</strong>g and the disease is transmitted <strong>in</strong> families follow<strong>in</strong>ga Mendelian <strong>in</strong>heritance pattern [3,4]. Interest<strong>in</strong>gly, 50% or more of early-onset AD cases arenot expla<strong>in</strong>ed by the known APP, PSEN1, and PSEN2 mutations, suggest<strong>in</strong>g the existence of yetunknown genetic factors [5]. Most of AD (90e95%) are however sporadic forms diagnosed <strong>in</strong>people over 65 years of age and referred to as late-onset (LOAD) forms, likely result<strong>in</strong>g fromthe <strong>in</strong>teraction among genetic, epigenetic, environmental, and stochastic factors superimposedon the physiological decl<strong>in</strong>e of cognitive functions with age [6]. Hundreds of geneshave been <strong>in</strong>vestigated <strong>in</strong> genetic association studies as possible AD susceptibility or modifiergenes, and more-recent genome-wide association studies (GWAS) are reveal<strong>in</strong>g novel polymorphismsthat could account for <strong>in</strong>creased LOAD risk. The ALZGene database (www.alzgene.org) is a cont<strong>in</strong>uously updated database conta<strong>in</strong><strong>in</strong>g meta-analyses of published studies,<strong>in</strong>clud<strong>in</strong>g GWAS, <strong>in</strong> order to provide a list of the genetic variants that most likely affect diseaserisk [7]. Accessed on July 2011, the database conta<strong>in</strong>ed data on 1395 genetic association studiescover<strong>in</strong>g 695 genes and 2973 polymorphisms. In addition, 320 meta-analyses were availablefor those genetic polymorphisms which had been <strong>in</strong>vestigated <strong>in</strong> at least four <strong>in</strong>dependentresearch studies. Accord<strong>in</strong>g to these meta-analyses the top ten genes that most likely contributeto LOAD risk are the follow<strong>in</strong>g: (1) apolipoprote<strong>in</strong> E (APOE ε4 allele, OR ¼ 3.68), (2) bridg<strong>in</strong>g<strong>in</strong>tegrator 1 (BIN1 rs744373, OR ¼ 1.17), (3) cluster<strong>in</strong> (CLU rs11136000, OR ¼ 0.88), (4)ATP-b<strong>in</strong>d<strong>in</strong>g cassette sub-family A member 7 (ABCA7 rs3764650, OR ¼ 1.23), (5) complementreceptor 1 (CR1 rs3818361, OR ¼ 1.17), (6) phosphatidyl<strong>in</strong>ositol b<strong>in</strong>d<strong>in</strong>g clathr<strong>in</strong>assembly prote<strong>in</strong> (PICALM rs3851179, OR ¼ 0.88), (7) membrane-spann<strong>in</strong>g 4-doma<strong>in</strong>s,subfamily A, member 6A (MS4A6A rs610932, OR ¼ 0.90), (8) myeloid cell surface antigenCD33 (CD33 rs3865444, OR ¼ 0.89), (9) membrane-spann<strong>in</strong>g 4-doma<strong>in</strong>s, subfamily A,member 4E (MS4A4E rs670139, OR ¼ 1.08), and (10) CD2-associated prote<strong>in</strong> (CD2APrs9349407, OR ¼ 1.12) (www.alzgeme.org). Additional genes l<strong>in</strong>ked to LOAD risk <strong>in</strong>cludethose <strong>in</strong>volved <strong>in</strong> sort<strong>in</strong>g mechanisms, such as the sortil<strong>in</strong>-related receptor (SORL1, severalpolymorphisms, OR ¼ 1.10e1.20) and its homolog, the sortil<strong>in</strong>-related VPS10 doma<strong>in</strong>conta<strong>in</strong><strong>in</strong>g receptor 1 (SORCS1, rs600879, OR ¼ 1.24), both caus<strong>in</strong>g APP, BACE1 and g-secretases to localize <strong>in</strong> the same compartment that is critical <strong>in</strong> the regulation of Abproduction (www.alzgene.org). Overall, with the exception of the APOE ε4 variant which givesan odds ratio of 3.7, all the other variants only confer a modest LOAD risk with ORs rang<strong>in</strong>gfrom 0.9 to 1.2, suggest<strong>in</strong>g a multifactorial nature of the disease and complex geneegene andgeneeenvironment <strong>in</strong>teractions yet to be clarified [7].Alongside with genetic variants, also several environmental agents have been <strong>in</strong>vestigated aspossible LOAD risk factors, often with conflict<strong>in</strong>g or <strong>in</strong>conclusive results [6]. Among themmetals, pesticides, bra<strong>in</strong> <strong>in</strong>juries, and the result<strong>in</strong>g <strong>in</strong>flammation and oxidative stress, havebeen suggested to <strong>in</strong>crease LOAD risk; by contrast, Mediterranean diet, the consumption offruit, vegetables and fish, dietary antioxidants, regular physical activity, and bra<strong>in</strong> stimulationuntil late <strong>in</strong> life are considered to be among LOAD protective factors [6]. However, despite


CHAPTER 9The <strong>Epigenetics</strong> of Alzheimer’s <strong>Disease</strong>active research <strong>in</strong> the field the etiology of sporadic AD is still uncerta<strong>in</strong>. Interest<strong>in</strong>gly, a recenthypothesis l<strong>in</strong>k<strong>in</strong>g environmental exposure to AD risk suggests that several environmentalfactors could epigenetically modify the expression of AD-related genes, such as APP, PSEN1,and many others [6]. This chapter describes the current evidence of AD-related epigeneticmodifications. <strong>Epigenetics</strong> has also a role <strong>in</strong> ag<strong>in</strong>g, cognitive functions, and several other agerelateddiseases than AD [8,9]. All these po<strong>in</strong>ts are nicely addressed <strong>in</strong> Chapter 26.9.2 ONE-CARBON METABOLISM AND DNA METHYLATION INALZHEIMER’S DISEASEFolates are essential nutrients required for one-carbon biosynthetic and epigenetic processes.They are derived entirely from dietary sources, ma<strong>in</strong>ly from the consumption of green vegetables,fruits, cereals, and meat. After <strong>in</strong>test<strong>in</strong>al absorption, folate metabolism requiresreduction and methylation <strong>in</strong>to the liver to form 5-methyltetrahydrofolate (5-MTHF), release<strong>in</strong>to the blood and cellular uptake; then it can be used for the synthesis of DNA and RNAprecursors or for the conversion of homocyste<strong>in</strong>e (Hcy) to methion<strong>in</strong>e, which is then used toform S-adenosylmethion<strong>in</strong>e (SAM), the ma<strong>in</strong> DNA-methylat<strong>in</strong>g agent. Several enzymes andcofactors, such as vitam<strong>in</strong> B12 and vitam<strong>in</strong> B6, participate <strong>in</strong> one-carbon metabolism(Figure 9.1), and common polymorphisms of genes <strong>in</strong>volved <strong>in</strong> folate uptake and metabolismhave been often associated with <strong>in</strong>creased AD risk, though results are still conflict<strong>in</strong>g or<strong>in</strong>conclusive for most of them [10]. Folic acid is the synthetic form added to foods and found<strong>in</strong> dietary supplements, it is converted to a natural biological form of the vitam<strong>in</strong> as it passesthrough the <strong>in</strong>test<strong>in</strong>al wall, with enzymatic reduction and methylation result<strong>in</strong>g <strong>in</strong> thecirculat<strong>in</strong>g form of the vitam<strong>in</strong>, 5-methylTHF [10]. Given its role <strong>in</strong> the DNA methylationpathway, the contribution of folate metabolism (also referred to as one-carbon metabolism) toepigenetic modifications of AD-related genes is currently <strong>in</strong>vestigated by several researchgroups (see below).177GSH(Cys-Gly-Glu)CysCH 3 CH 3| |-----CGCGCGCG----Methylated DNACystathion<strong>in</strong>e B6SAHMTHFRCBS5-MTHFUnmethylatedHcySAMDNAMTRRMTRB12MetDNMTsMAT -----CGCGCGCG----5,10-methyleneTHF10-formylTHFdUMPTYMSPur<strong>in</strong>esMTHFDGARTTHFRFC1ReducedfolatesdTMPDHFDNA syhthesisFIGURE 9.1Overview of the folate metabolic pathway, adapted from Coppedè et al. [86]: Metabolites: Cys, cyste<strong>in</strong>e; dTMP, deoxythymid<strong>in</strong>emonophosphate; dUMP, deoxyurid<strong>in</strong>e monophosphate; DHF, dihydrofolate; 10-formyl-THF, 10-formyl-tetrahydrofolate; GSH,glutathione; Hcy, homocyste<strong>in</strong>e; Met, methion<strong>in</strong>e; 5-MTHF, 5- methyltetrahydrofolate; 5,10-MTHF, 5,10-methylentetrahydrofolate; SAH, S-adenosylhomocyste<strong>in</strong>e; SAM, S-adenosylmethion<strong>in</strong>e; THF, tetrahydrofolate. Enzymes: CBS,cystathion<strong>in</strong>e b-synthase; DNMTs, DNA methyltransferases; GART, phosphoribosylglyc<strong>in</strong>amide transformylase; MAT,methion<strong>in</strong>e adenosyltransferase; MTHFD, methylenetetrahydrofolate dehydrogenase; MTHFR, methylenetetrahydrofolatereductase; MTR, methion<strong>in</strong>e synthase; MTRR, methion<strong>in</strong>e synthase reductase; RFC1, reduced folate carrier; TYMS, thymidylatesynthase. Cofactors: B6, vitam<strong>in</strong> B6; B12, vitam<strong>in</strong> B12.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>1789.2.1 An Overview of One-Carbon MetabolismAs illustrated <strong>in</strong> Figure 9.1 cellular folates can be used either for DNA methylation processes orfor the synthesis of nucleic acid precursors (Figure 9.1). Folates are highly hydrophilicmolecules that do not cross biological membranes by diffusion alone, so it is not surpris<strong>in</strong>gthat sophisticated membrane transport systems have evolved for facilitat<strong>in</strong>g their uptake bymammalian cells and tissues, the most ubiquitous and best characterized be<strong>in</strong>g the reducedfolate carrier (RFC1). Methylenetetrahydrofolate reductase (MTHFR) is the first enzyme <strong>in</strong> theDNA methylation pathway and reduces 5,10-methylentetrahydrofolate (5,10-MTHF) to 5-methylTHF. Subsequently, methion<strong>in</strong>e synthase (MTR) transfers a methyl group from 5-methylTHF to homocyste<strong>in</strong>e form<strong>in</strong>g methion<strong>in</strong>e and tetrahydrofolate (THF). Methion<strong>in</strong>e isthen converted to SAM <strong>in</strong> a reaction catalyzed by methion<strong>in</strong>e adenosyltransferase (MAT). Mostof the SAM generated is used <strong>in</strong> transmethylation reactions, whereby SAM is converted to S-adenosylhomocyste<strong>in</strong>e (SAH) by transferr<strong>in</strong>g the methyl group to diverse biological acceptors,<strong>in</strong>clud<strong>in</strong>g prote<strong>in</strong>s and DNA. Vitam<strong>in</strong> B12 (or cobalam<strong>in</strong>) is a cofactor of MTR, and methion<strong>in</strong>esynthase reductase (MTRR) is required for the ma<strong>in</strong>tenance of MTR <strong>in</strong> its active state. Ifnot converted <strong>in</strong>to methion<strong>in</strong>e, Hcy can be condensed with ser<strong>in</strong>e to form cystathion<strong>in</strong>e <strong>in</strong>a reaction catalyzed by cystathion<strong>in</strong>e b-synthase (CBS), which requires vitam<strong>in</strong> B6 asa cofactor. Cystathion<strong>in</strong>e can be then utilized to form the antioxidant compound glutathione(GSH). Another important function of folates is <strong>in</strong> the de novo synthesis of DNA and RNAprecursors, required dur<strong>in</strong>g nucleic acid synthesis and for DNA repair processes. Therefore,depend<strong>in</strong>g on cellular demands 5,10-MTHF can be used for the synthesis of SAM or for thesynthesis of nucleic acids precursors. Thymidylate synthase (TYMS) converts deoxyurid<strong>in</strong>emonophosphate (dUMP) and 5,10-MTHF to deoxythym<strong>in</strong>e monophospate (dTMP) anddihydrofolate (DHF) <strong>in</strong> the de novo synthesis of pyrimid<strong>in</strong>es (Figure 9.1). Methylenetetrahydrofolatedehydrogenase (MTHFD1) consists of three activities: 5,10-MTHF dehydrogenase,5,10-methenyltetrahydrofolate cyclohydrolase, and 10-formyltetrahydrofolatesynthetase, respectively, and catalyzes three sequential reactions <strong>in</strong> the <strong>in</strong>terconversion of THFderivatives. Once generated by the 10-formyltetrahydrofolate synthetase activity, 10-formyltetrahydrofolate can be utilized for the production of pur<strong>in</strong>es by the phosphoribosylglyc<strong>in</strong>eamidetransformylase (GART) enzyme. Overall, the folate metabolic pathway<strong>in</strong>volves several enzymes and is tightly regulated by <strong>in</strong>tracellular levels of metabolites andcofactors [10].9.2.2 One-Carbon Metabolism <strong>in</strong> Alzheimer’s <strong>Disease</strong>The author recently reviewed the literature deal<strong>in</strong>g with one-carbon metabolism <strong>in</strong> AD, andmost of the published studies agree that AD <strong>in</strong>dividuals are characterized by decreasedfolate values, as well as <strong>in</strong>creased plasma homocyste<strong>in</strong>e levels (hyperhomocyste<strong>in</strong>emia)[10]. The hypothesis that Hcy is a risk factor for AD was <strong>in</strong>itially prompted by the observationthat patients with histologically confirmed AD had higher plasma levels of Hcy, thanage-matched controls. Although most evidence accumulated so far implicates hyperhomocyste<strong>in</strong>emiaas an AD risk factor, there are also conflict<strong>in</strong>g results [11]. For example,a recent meta-analysis of relevant studies suggests that <strong>in</strong>dividuals with AD and vasculardementia have higher plasma Hcy levels than controls, but a causal relationship betweenhyperhomocyste<strong>in</strong>emia and the risk of develop<strong>in</strong>g dementia was not observed [12].Therefore, the role of hyperhomocyste<strong>in</strong>emia as a risk factor for dementia is stillcontroversial.Alongside data concern<strong>in</strong>g plasma Hcy values, there is also some “although controversial”<strong>in</strong>dication that Hcy levels are <strong>in</strong>creased <strong>in</strong> the cerebrosp<strong>in</strong>al fluid (CSF) of AD patients withrespect to controls [13e15]. A recent study <strong>in</strong>clud<strong>in</strong>g 70 AD patients, 33 patients with anothertype of dementia, and 30 age-matched control subjects, revealed that folates <strong>in</strong> CSF weresignificantly different between groups, but not Hcy. In addition, the average folate <strong>in</strong> CSF was


CHAPTER 9The <strong>Epigenetics</strong> of Alzheimer’s <strong>Disease</strong>lower <strong>in</strong> AD patients compared with controls, and <strong>in</strong> AD CSF there was a significant <strong>in</strong>versecorrelation between Hcy and folate, support<strong>in</strong>g the hypothesis of a possible role of folate <strong>in</strong> theonset or worsen<strong>in</strong>g of AD [16].DNA methylation is closely dependent on the DNA methylation potential, which is referred toas the ratio between SAM and SAH levels. There is an <strong>in</strong>dication of reduced DNA methylationpotential <strong>in</strong> AD bra<strong>in</strong>s [17], and it has been suggested that <strong>in</strong>creased SAH concentrations <strong>in</strong> thebra<strong>in</strong>s of AD patients might <strong>in</strong>hibit DNA methyltransferases (DNMTs) [18]. A recent studyshowed that Hcy plasma levels <strong>in</strong> the highest quartile were more frequent <strong>in</strong> AD patients than<strong>in</strong> controls. In addition, AD patients had significantly lower CSF levels of the methyl groupdonor SAM. Accord<strong>in</strong>gly, the SAM/SAH ratio, i.e. the methylation capacity, was significantlylower <strong>in</strong> the CSF of the AD patients. Further, explorative analysis of all subjects showed thatCSF SAM levels were lower <strong>in</strong> carriers of the APOE ε4 allele compared with non-carriers [19].Studies <strong>in</strong> AD animal models also revealed altered SAM levels <strong>in</strong> the bra<strong>in</strong>. However, theanalysis of SAM and SAH levels <strong>in</strong> the ag<strong>in</strong>g bra<strong>in</strong> of APP/PS1 Alzheimer mice dur<strong>in</strong>g ag<strong>in</strong>g,revealed that SAM levels decreased <strong>in</strong> the bra<strong>in</strong>s of wild-type mice, whereas SAH levelsdim<strong>in</strong>ished <strong>in</strong> both wild-type and APP/PS1 mice. In contrast to wild-type mice, SAM levels <strong>in</strong>APP/PS1 mice are not decreased dur<strong>in</strong>g ag<strong>in</strong>g, probably related to less demand due toneurodegeneration [20].DNMTs are the key enzymes for DNA methylation and catalyze the transfer of a methyl groupfrom SAM to cytos<strong>in</strong>e, thus form<strong>in</strong>g 5-methyl-cytos<strong>in</strong>e and SAH (Figure 9.2). There are multiplefamilies of DNMTs <strong>in</strong> mammals. DNMT1 is primarily <strong>in</strong>volved <strong>in</strong> the ma<strong>in</strong>tenance of DNA179FIGURE 9.2The reaction catalyzed by DNA methyltransferases (DNMTs). DNMTs are the key enzymes for DNA methylation and catalyze thetransfer of a methyl group from SAM to cytos<strong>in</strong>e, thus form<strong>in</strong>g 5-methyl-cytos<strong>in</strong>e and SAH. Methylation of CpG sequencesmight <strong>in</strong>duce chromat<strong>in</strong> conformational modifications and <strong>in</strong>hibit the access of the transcriptional mach<strong>in</strong>ery to gene promoterregions, thus alter<strong>in</strong>g gene expression levels. Therefore, promoter rmethylation of CpG islands is commonly associated withgene silenc<strong>in</strong>g and promoter demethylation with gene expression, though several exceptions to this rule are known.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>methylation patterns dur<strong>in</strong>g development and cell division, whereas DNMT3a and DNMT3b arethe de novo methyltransferases and establish DNA methylation patterns dur<strong>in</strong>g early development.DNMT3L <strong>in</strong>duces de novo DNA methylation by recruitment or activation of DNMT3a,whilst DNMT2 is primarily <strong>in</strong>volved <strong>in</strong> the methylation of transfer RNA molecules [21,22].Follow<strong>in</strong>g the observation of impaired one-carbon metabolism and methylation potential <strong>in</strong>AD, some <strong>in</strong>vestigators raised the possibility of a l<strong>in</strong>k between DNA methylation and AD risk,and several studies are now available (Table 9.1), ma<strong>in</strong>ly from animal disease models andneuronal cell cultures, suggest<strong>in</strong>g that dietary factors, but also other environmental factors, canlead to epigenetic modifications and deregulated expression of key AD genes [23]. Little is,however, still known <strong>in</strong> humans.9.2.3 Studies <strong>in</strong> Cell Cultures and Animal ModelsOne of the most excit<strong>in</strong>g hypothesis l<strong>in</strong>k<strong>in</strong>g one-carbon metabolism to AD risk suggests thatimpaired folate/Hcy metabolism and subsequent reduction of SAM levels might result <strong>in</strong>epigenetic modifications of the promoters of AD-related genes lead<strong>in</strong>g to <strong>in</strong>creased Ab peptideproduction [24,25]. DNA methylation represents one of the most important epigeneticTABLE 9.1 Evidence of DNA Methylation <strong>in</strong> AD and AD Model Systems180Experimental Model Observation References<strong>Human</strong> neuroblastomaSK-N-SH or SK-N-BEcellsBV-2 mouse microglialcellsMur<strong>in</strong>e cerebralendothelial cellsRodentsRodents and monkeysPost-mortem humanbra<strong>in</strong>sBlood DNAFolate and vitam<strong>in</strong> B12 deprivation <strong>in</strong>duced epigeneticmodifications <strong>in</strong> the promoter of PSEN1, result<strong>in</strong>g <strong>in</strong> upregulationof gene expression and <strong>in</strong>creased production ofpresenil<strong>in</strong> 1, BACE1, and APPB vitam<strong>in</strong> deficiency <strong>in</strong>duced decreased de novo DNAmethylation activitySAM adm<strong>in</strong>istration resulted <strong>in</strong> down-regulation of PSEN1expressionSAM adm<strong>in</strong>istration was able to modulate the expression ofseven out of 588 genes of the nervous systemSAH adm<strong>in</strong>istration <strong>in</strong>creased the production of Ab peptide likelythrough <strong>in</strong>duction of hypomethylation of APP and PSEN1 genepromotersAb reduces global DNA demethylation whilst <strong>in</strong>creas<strong>in</strong>g DNAmethylation of the gene encod<strong>in</strong>g neprilys<strong>in</strong>B vitam<strong>in</strong> deprivation altered the methylation potential and<strong>in</strong>duced hypomethylation of the promoter of PSEN1, result<strong>in</strong>g <strong>in</strong><strong>in</strong>creased gene expressionB vitam<strong>in</strong> deficiency <strong>in</strong>duced decreased de novo DNAmethylation activityEarly life exposure to Pb resulted <strong>in</strong> <strong>in</strong>hibition of DNAmethyltransferase,hypomethylation of the promoter of APP anddelayed up-regulation of gene expression later <strong>in</strong> lifeThe analysis of DNA methylation <strong>in</strong> selected regions of MAPT,APP, and PSEN1 genes revealed no difference between controlsamples and AD samplesOnly two (SORBS3 and S100A2) out of 50 analyzed loci showedAD-related methylation changes, which appeared to reflect anacceleration of age-related changesAD bra<strong>in</strong>s showed an <strong>in</strong>creased epigenetic drift with unusualmethylation patterns, and <strong>in</strong>ter-<strong>in</strong>dividual variations concern<strong>in</strong>gPSEN1, APOE, MTHFR, and DNMT1 genesAD bra<strong>in</strong>s showed a marked reduction of DNA methylationHigher levels of LINE-1 methylation <strong>in</strong> AD subjectswith respect to controls[25][32][24][35][31][36][27,28][32][37,38][42][43][44][40,41][45]


CHAPTER 9The <strong>Epigenetics</strong> of Alzheimer’s <strong>Disease</strong>processes, alongside histone tail modifications and mechanisms <strong>in</strong>volv<strong>in</strong>g small RNA molecules.Methylation of CpG sequences might <strong>in</strong>duce chromat<strong>in</strong> conformational modificationsand <strong>in</strong>hibit the access of the transcriptional mach<strong>in</strong>ery to gene promoter regions, thus alter<strong>in</strong>ggene expression levels. Therefore, promoter hypermethylation is commonly associated withgene silenc<strong>in</strong>g and promoter demethylation with gene expression, though several exceptionsto this rule have been reported [26].APP and PSEN1 promoter methylation was analyzed <strong>in</strong> human neuroblastoma SK-N-SH or SK-N-BE cell l<strong>in</strong>es under conditions of folate and vitam<strong>in</strong> B12 deprivation from the media. Thestudy revealed demethylation of a CpG site <strong>in</strong> the promoter of PSEN1, with a subsequent<strong>in</strong>creased production of presenil<strong>in</strong>1, BACE1, and APP prote<strong>in</strong>s [25]. By contrast, SAMadm<strong>in</strong>istration to human neuroblastoma SK-N-SH cells resulted <strong>in</strong> down-regulation of PSEN1gene expression and reduced Ab peptide production [24]. Similarly, a comb<strong>in</strong>ation of dietaryfolate, vitam<strong>in</strong> B12, and vitam<strong>in</strong> B6 deprivation (B vitam<strong>in</strong> deprivation) resulted <strong>in</strong> hyperhomocyste<strong>in</strong>emia,<strong>in</strong>creased bra<strong>in</strong> SAH levels, depletion of bra<strong>in</strong> SAM, hypomethylation ofspecific CpG moieties <strong>in</strong> the 5 0 -flank<strong>in</strong>g region of PSEN1 and enhancement of presenil<strong>in</strong> 1 andBACE1 expression and Ab deposition <strong>in</strong> mice [27,28]. Dietary deficiency of folate and vitam<strong>in</strong> E,<strong>in</strong> condition of oxidative stress (the diet conta<strong>in</strong>ed iron as a pro-oxidant), <strong>in</strong>creased presenil<strong>in</strong> 1expression, BACE1 activity, and Ab levels <strong>in</strong> normal adult mice. These <strong>in</strong>creases were particularlyevident <strong>in</strong> mice lack<strong>in</strong>g apolipoprote<strong>in</strong> E. On the contrary, dietary SAM supplementationattenuated these deleterious consequences [29]. A similar experiment was performed <strong>in</strong> miceexpress<strong>in</strong>g the human APOE gene. Mice express<strong>in</strong>g human apolipoprote<strong>in</strong> ε4, apolipoprote<strong>in</strong>ε3, or apolipoprote<strong>in</strong> ε2, were subjected to a diet lack<strong>in</strong>g folate and vitam<strong>in</strong> E, and conta<strong>in</strong><strong>in</strong>giron as a pro-oxidant. The study revealed that presenil<strong>in</strong> 1 and gamma-secretase were overexpressed<strong>in</strong> ε3 mice to the same extent as <strong>in</strong> ε4 mice even under a complete diet, and were notalleviated by SAM supplementation. Ab <strong>in</strong>creased only <strong>in</strong> ε4 mice ma<strong>in</strong>ta<strong>in</strong>ed under thecomplete diet, and was alleviated by SAM supplementation [30]. L<strong>in</strong> and co-workers showedthat SAH <strong>in</strong>creases the production of Ab <strong>in</strong> BV-2 mouse microglial cells possibly by an <strong>in</strong>creasedexpression of APP and <strong>in</strong>duction of hypomethylation of APP and PSEN1 gene promoters [31].181A recent study demonstrated that B vitam<strong>in</strong> deficiency <strong>in</strong>duces DNMT3a and DNMT3bprote<strong>in</strong> down-regulation <strong>in</strong> both neuroblastoma cell cultures and mice bra<strong>in</strong> regions.Moreover, the de novo DNA methylation activity was modulated <strong>in</strong> both neuroblastomacells and mice, and resulted decreased under B vitam<strong>in</strong> deficiency conditions and <strong>in</strong>creasedby SAM supplementation, whilst the activity of a putative DNA demethylase (MBD2) showedan opposite tendency [32]. It is however worth mention<strong>in</strong>g that little is still knownconcern<strong>in</strong>g DNA demethylation <strong>in</strong> the adult mammalian bra<strong>in</strong>. Guo and co-workers [33]have recently demonstrated that the 5-methylcytos<strong>in</strong>e hydroxylase TET1, by convert<strong>in</strong>g 5-methylcytos<strong>in</strong>e to 5-hydroxymethylcytos<strong>in</strong>es (5hmCs), promotes DNA demethylation <strong>in</strong>mammalian cells through a process that requires the DNA base excision repair pathway.Demethylation of 5hmCs is promoted by the AID (activation-<strong>in</strong>duced deam<strong>in</strong>ase)/APOBEC(apolipoprote<strong>in</strong> B mRNA-edit<strong>in</strong>g enzyme complex) family of cytid<strong>in</strong>e deam<strong>in</strong>ases.Furthermore, Tet1 and Apobec1 are <strong>in</strong>volved <strong>in</strong> neuronal activity-<strong>in</strong>duced, region-specific,active DNA demethylation and subsequent gene expression <strong>in</strong> the dentate gyrus of the adultmouse bra<strong>in</strong> <strong>in</strong> vivo [33]. Nutritional B vitam<strong>in</strong> restriction was also used to study the variationof prote<strong>in</strong> expression profile <strong>in</strong> mice bra<strong>in</strong> regions. A group of prote<strong>in</strong>s ma<strong>in</strong>ly <strong>in</strong>volved<strong>in</strong> neuronal plasticity and mitochondrial functions was identified as modulated by onecarbonmetabolism [34]. The same group also analyzed 588 genes of the central nervoussystem <strong>in</strong> SK-N-BE neuroblastoma cells, observ<strong>in</strong>g that only seven genes were modulated bySAM treatment (and therefore by DNA methylation); three were up-regulated and fourdown-regulated [35]. Others observed that the Ab peptide <strong>in</strong>duces global DNA hypomethylation<strong>in</strong> mur<strong>in</strong>e cerebral endothelial cells, but specifically leads to hypermethylationof the gene encod<strong>in</strong>g neprilys<strong>in</strong> (NEP), one of the enzymes responsible for Ab degradation,thus suppress<strong>in</strong>g NEP expression<strong>in</strong>mRNAandprote<strong>in</strong>levels[36].


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>Alongside with the dietary factors (dietary B vitam<strong>in</strong>s), metal exposure has also been suggestedto be able to epigenetically modulate the expression of AD-related genes. Particularly, theanalysis of rodents and monkeys exposed to lead (Pb) dur<strong>in</strong>g embryogenesis and/or earlypostnatal period revealed that early life exposure to this metal could result <strong>in</strong> epigeneticmodifications of AD-related genes, such as APP, and subsequent deregulated expression later <strong>in</strong>life [37,38]. Particularly, it was observed that developmental exposure of rats to lead resulted <strong>in</strong>a delayed overexpression (20 months later) of the amyloid precursor prote<strong>in</strong> and itsamyloidogenic Ab product. Similarly, aged monkeys exposed to lead as <strong>in</strong>fants also responded<strong>in</strong> the same way [37,38]. These data <strong>in</strong>dicate that environmental <strong>in</strong>fluences occurr<strong>in</strong>g dur<strong>in</strong>gbra<strong>in</strong> development predeterm<strong>in</strong>e the expression and regulation of APP later <strong>in</strong> life, potentially<strong>in</strong>fluenc<strong>in</strong>g the course of amyloidogenesis, and suggest<strong>in</strong>g that early life neurogenesis is likelyto be a critical step for environmentally <strong>in</strong>duced epigenetic modifications [39].1829.2.4 Studies <strong>in</strong> <strong>Human</strong>sDespite ev<strong>in</strong>ce of possible epigenetic modifications of AD-related genes obta<strong>in</strong>ed <strong>in</strong> neuronalcell cultures as well as <strong>in</strong> rodents and primates, epigenetic studies <strong>in</strong> AD patients are still scarce,likely depend<strong>in</strong>g on the difficulty to obta<strong>in</strong> human post-mortem bra<strong>in</strong> specimens. Mastroeniand co-workers exam<strong>in</strong>ed global DNA methylation <strong>in</strong> post-mortem bra<strong>in</strong> regions of monozygotictw<strong>in</strong>s discordant for AD, observ<strong>in</strong>g significantly reduced levels of DNA methylation <strong>in</strong>the temporal neocortex of the AD tw<strong>in</strong> [40]. The same group analyzed bra<strong>in</strong> tissues from 20 ADpatients and 20 age-matched controls, and used immunocytochemistry for two markers of DNAmethylation (5-methylcytos<strong>in</strong>e and 5-methylcytid<strong>in</strong>e) and eight methylation ma<strong>in</strong>tenancefactors <strong>in</strong> the entorh<strong>in</strong>al cortex layer II, aga<strong>in</strong> observ<strong>in</strong>g significant decrements <strong>in</strong> AD cases [41].However, Barrach<strong>in</strong>a and Ferrer [42] analyzed DNA methylation <strong>in</strong> selected regions of MAPT(the gene encod<strong>in</strong>g for microtubule-associated tau prote<strong>in</strong>), APP, andPSEN1 <strong>in</strong> post-mortemfrontal cortex and hippocampus of healthy controls and various stages of AD progression,observ<strong>in</strong>g no differences <strong>in</strong> the percentage of CpG methylation <strong>in</strong> any region analyzed [42].Siegmund and co-workers exam<strong>in</strong>ed the DNA methylation status at 50 loci, encompass<strong>in</strong>gprimarily 5 0 CpG islands of genes related to CNS growth and development, <strong>in</strong> the temporalneocortex of 125 subjects rang<strong>in</strong>g <strong>in</strong> age from 17 weeks of gestation to 104 years old, and<strong>in</strong>clud<strong>in</strong>g a cohort of AD patients [43]. Only two loci showed AD-related changes (SORBS3 andS100A2, encod<strong>in</strong>g a cell adhesion molecule and a calcium-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>, respectively), whichappeared to reflect an acceleration of age-related changes [43]. Wang and collaborators [44]analyzed 12 potential AD susceptibility loci <strong>in</strong> peripheral lymphocytes and post-mortem bra<strong>in</strong>samples of LOAD patients and matched controls. From the 12 analyzed CpG-rich regions, onlyone was hypermethylated, and was the only region analyzed outside of a promoter region (DNAmethylation can also occur <strong>in</strong> CpG sites outside the promoter region “gene-body methylation”,and is usually associated with transcriptional activation). Overall, the authors failed to f<strong>in</strong>dsignificant differences <strong>in</strong> DNA methylation between LOAD cases and controls [44]. However,four of the studied genes (PSEN1, APOE, MTHFR, andDNMT1) showed a significant <strong>in</strong>ter<strong>in</strong>dividualepigenetic variability, and LOAD bra<strong>in</strong> samples showed an “age-specific epigeneticdrift” associated with unusual methylation patterns [44]. More recently, Bollati and co-workersanalyzed the methylation pattern of repetitive elements (i.e. Alu, LINE-1, and a-satellite DNA) <strong>in</strong>the DNA obta<strong>in</strong>ed from blood samples of 43 AD patients and 38 matched controls, observ<strong>in</strong>ghigher levels of LINE-1 methylation <strong>in</strong> AD subjects with respect to controls [45].9.2.5 L<strong>in</strong>k<strong>in</strong>g the Methylation Potential to Tau Phosphorylation<strong>in</strong> Alzheimer’s <strong>Disease</strong>The results of prospective cohort studies suggest that <strong>in</strong>creased serum Hcy levels mightpredispose to AD [46e48], and there is also an <strong>in</strong>dication that higher folate <strong>in</strong>take is related tolower AD risk <strong>in</strong> the elderly [48,49]. Folate deficiency fosters a decl<strong>in</strong>e <strong>in</strong> SAM levels, thusdecreas<strong>in</strong>g DNA methylation dur<strong>in</strong>g ag<strong>in</strong>g and AD. Moreover, folate deficiency and the


CHAPTER 9The <strong>Epigenetics</strong> of Alzheimer’s <strong>Disease</strong>resultant SAM depletion lead to <strong>in</strong>creased levels of Hcy [46]. Hcy is a critical branch po<strong>in</strong>tmetabolite that can <strong>in</strong>fluence cellular levels of SAM and SAH, which <strong>in</strong> turn regulate theactivity of methyltransferases dur<strong>in</strong>g DNA methylation and post-translational modification ofprote<strong>in</strong>s [50]. There is an <strong>in</strong>dication that elevated Hcy causes tau hyperphosphorylation, NFTformation, and SP formation via <strong>in</strong>hibition of methyltransferases and reduced methylation ofprote<strong>in</strong> phosphatase 2A (PP2A) [51,52]. Tau phosphorylation is regulated by the equilibriumbetween prote<strong>in</strong> k<strong>in</strong>ases and phosphatases. PP2A is the major prote<strong>in</strong> phosphatase thatdephosphorylates tau. Methylation of the PP2A catalytic subunit can potently activate PP2A.Therefore, demethylation of PP2A result<strong>in</strong>g from <strong>in</strong>hibition of methyltransferases could result<strong>in</strong> <strong>in</strong>creased production of hyperphosphorylated tau prote<strong>in</strong> [51,52]. Moreover, B vitam<strong>in</strong>deficiency was shown to reduce PP2A activity and <strong>in</strong>crease that of glycogen synthase k<strong>in</strong>ase3beta (GSK3beta) one of the most important prote<strong>in</strong> k<strong>in</strong>ase <strong>in</strong>volved <strong>in</strong> tau phosphorylation[53]. In addition, a B vitam<strong>in</strong>-deficient diet <strong>in</strong>creased phosphorylated tau levels <strong>in</strong> APOE ε4but not <strong>in</strong> APOE ε3 mice, and tau phosphorylation was prevented by SAM supplementation[54]. Overall, there is an <strong>in</strong>dication that deficiency <strong>in</strong> one-carbon metabolism, reduced SAMlevels, and SAH-mediated <strong>in</strong>hibition of methyltransferase prote<strong>in</strong>s could also contribute toNFT formation dur<strong>in</strong>g AD pathogenesis.9.3 HISTONE TAIL MODIFICATIONS AND ALZHEIMER’S DISEASEChromat<strong>in</strong> can exist <strong>in</strong> a condensate <strong>in</strong>active state (heterochromat<strong>in</strong>) or <strong>in</strong> a decondensed andtranscriptionally active state (euchromat<strong>in</strong>). Conformational changes <strong>in</strong> histone prote<strong>in</strong>s ormodifications of the way <strong>in</strong> which DNA wraps around the histone octamer <strong>in</strong> nucleosomesmay either alter or facilitate the access of the transcriptional mach<strong>in</strong>ery to the promoter regionof some genes, lead<strong>in</strong>g to gene silenc<strong>in</strong>g or activation, respectively. Histone tail modifications<strong>in</strong>clude acetylation, methylation, phosphorylation, ubiquitylation, sumoylation, and otherpost-translational modifications (Figure 9.3). Histone tail acetylation represents one of themost studied modifications and is associated with chromat<strong>in</strong> relaxation and transcriptionalactivation, while deacethylation is related to a more condensed chromat<strong>in</strong> state and transcriptionalrepression [55]. Acetylation occurs at lys<strong>in</strong>e residues on the am<strong>in</strong>o-term<strong>in</strong>al tails ofthe histones, thereby neutraliz<strong>in</strong>g the positive charge of the histone tails and decreas<strong>in</strong>g theiraff<strong>in</strong>ity for DNA. As a consequence, histone acetylation alters nucleosomal conformation,which can <strong>in</strong>crease the accessibility of transcriptional regulatory prote<strong>in</strong>s to chromat<strong>in</strong>templates [55]. Histone acetyltransferases (HATs) catalyze the acetylation of lys<strong>in</strong>e residues <strong>in</strong>histone tails, whereas histone deacetylation is mediated by histone deacetylases (HDACs).Another frequently studied modification of histone tails is methylation on either lys<strong>in</strong>e orarg<strong>in</strong><strong>in</strong>e residues. Methylation of histone tails can be associated with either condensation orrelaxation of the chromat<strong>in</strong> structure, s<strong>in</strong>ce several sites for methylation are present on eachtail thus allow<strong>in</strong>g several comb<strong>in</strong>ations [56]. A number of lys<strong>in</strong>e methylation sites of H3 andH4 have been well characterized. Methylation at H3K4, H3K36, and H3K79 has been l<strong>in</strong>ked toactively transcribed genes, whereas di- and tri-methylated H3K9, methylated H3K27 andH4K20 are considered repressive marks [57]. Prote<strong>in</strong> lys<strong>in</strong>e methyltransferases (PKMTs) andprote<strong>in</strong> arg<strong>in</strong><strong>in</strong>e methyltransferases (PRMTs) are two of the writers responsible for add<strong>in</strong>g themethyl marks to histones. Moreover, these writers only methylate the residue to a specificmethylation level, which is known as product specificity. For example, lys<strong>in</strong>e residues can bemono-, di-, or trimethylated, while arg<strong>in</strong><strong>in</strong>es can be mono- or dimethylated <strong>in</strong> eithera symmetric (one methyl group on each of the two N term<strong>in</strong>al atoms) or asymmetric (bothmethyl groups on the same N term<strong>in</strong>al atom) manner. Both PKMTs and PRMTs require SAM asthe methyl donor for histone tail methylation reactions [58]. G9a and GLP are the primaryenzymes for mono- and dimethylation at Lys 9 of histone H3 (H3K9me1 and H3K9me2), andexist predom<strong>in</strong>antly as a G9aeGLP heteromeric complex that appears to be a functional H3K9methyltransferase <strong>in</strong> vivo. It was shown that <strong>in</strong> mammals G9a/GLP suppresses transcription by<strong>in</strong>dependently <strong>in</strong>duc<strong>in</strong>g both H3K9 and DNA methylation [59].183


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>FIGURE 9.3Histone tail modifications. The figure showssome examples of histone tail modifications.Acetylation of lys<strong>in</strong>e residues is associated withchromat<strong>in</strong> relaxation and transcriptionalactivation, while deacethylation is related toa more condensed chromat<strong>in</strong> state andtranscriptional repression. DNA methylation canbe associated with either transcriptionalrepression (for example the H3K9 methylation,shown <strong>in</strong> the figure) or with transcriptionalactivation (see the text, for further details on thehistone code).1849.3.1 Histone Modifications <strong>in</strong> Alzheimer’s <strong>Disease</strong> Bra<strong>in</strong>sSeveral authors reported histone modifications <strong>in</strong> AD (Table 9.2). For example Ogawa et al.[60] observed <strong>in</strong>creased phosphorylation of histone 3 (H3) <strong>in</strong> AD hippocampal neurons. An<strong>in</strong>creased phosphorylation of histone H2AX was observed <strong>in</strong> hippocampal astrocytes of AD<strong>in</strong>dividuals [61]. Concern<strong>in</strong>g HDACs, elevated levels of HDAC6 have been observed <strong>in</strong> postmortembra<strong>in</strong> regions of AD subjects, and it was proposed that HDAC6 is a tau-<strong>in</strong>teract<strong>in</strong>gprote<strong>in</strong> and a potential modulator of tau phosphorylation and accumulation [62].9.3.2 HDAC Inhibitors and Memory Function <strong>in</strong> AD Animal ModelsSeveral <strong>in</strong>vestigators have manipulated histone acetylation with HDAC <strong>in</strong>hibitors (HDACi) <strong>in</strong>AD animal models, often observ<strong>in</strong>g prevention of cognitive deficits and memory recovery. Forexample, Francis and co-workers have undertaken precl<strong>in</strong>ical studies <strong>in</strong> the APP/PS1 mousemodel of AD to determ<strong>in</strong>e whether there are differences <strong>in</strong> histone acetylation levels dur<strong>in</strong>gassociative memory formation. After fear-condition<strong>in</strong>g tra<strong>in</strong><strong>in</strong>g, levels of hippocampalacetylated histone 4 (H4) <strong>in</strong> APP/PS1 mice were about 50% lower than <strong>in</strong> wild-type littermates[63]. Interest<strong>in</strong>gly, an acute treatment with the HDACi trichostat<strong>in</strong> A prior to tra<strong>in</strong><strong>in</strong>g rescuedboth acetylated H4 levels and contextual freez<strong>in</strong>g performance to wild-type values [63]. It wasalso observed that <strong>in</strong>hibitors of class 1 histone deacetylases (sodium valproate, sodiumbutyrate, or vor<strong>in</strong>ostat) reverse contextual memory deficits <strong>in</strong> a mouse model of AD thatshowed pronounced contextual memory impairments beg<strong>in</strong>n<strong>in</strong>g at 6 months of age [64].Cluster<strong>in</strong> is a secreted molecular chaperone, also called apolipoprote<strong>in</strong> J, and a significantsusceptibility gene for LOAD (www.alzgene.org). Cluster<strong>in</strong> shares several properties with


CHAPTER 9The <strong>Epigenetics</strong> of Alzheimer’s <strong>Disease</strong>TABLE 9.2 Evidence of Histone Modifications <strong>in</strong> AD and AD Model SystemsExperimental Model Observation Refs.AD hippocampal neurons Increased phosphorylation of histone H3 [60]AD hippocampal astrocytes Increased phosphorylation of histone H2AX [61]Post-mortem human bra<strong>in</strong>s Increased levels of HDAC6, primarily <strong>in</strong>volved <strong>in</strong> [62]cytoplasmic acetylation, and likely <strong>in</strong>volved <strong>in</strong> tauphosphorylation and accumulationAD animal modelsHDAC <strong>in</strong>hibitors improved cognition and memoryfunctions[63e67]apolipoprote<strong>in</strong> E, s<strong>in</strong>ce they b<strong>in</strong>d to Ab peptides and are present <strong>in</strong> neuritic plaques, enhancethe clearance of Ab peptides <strong>in</strong> bra<strong>in</strong>, and are <strong>in</strong>cluded <strong>in</strong> lipid particles and thus regulatecholesterol traffic. It was shown that HDACi, such as valproic acid and vor<strong>in</strong>ostat, stimulatedthe expression of cluster<strong>in</strong> <strong>in</strong> human astrocytes, and might therefore be able to prevent Abaggregation <strong>in</strong> AD [65]. A recent paper by Gov<strong>in</strong>darajan et al. showed that severe amyloidpathology correlates with a pronounced deregulation of histone acetylation <strong>in</strong> the forebra<strong>in</strong> ofAPP/PS1-21 mice, and that a prolonged treatment with the pan-HDAC <strong>in</strong>hibitor sodiumbutyrate improved associative memory <strong>in</strong> APP/PS1-21 mice even when adm<strong>in</strong>istered at a veryadvanced stage of pathology. The recovery of memory function correlated with elevatedhippocampal histone acetylation and <strong>in</strong>creased expression of genes implicated <strong>in</strong> associativelearn<strong>in</strong>g [66]. Others observed that systemic adm<strong>in</strong>istration of the HDAC <strong>in</strong>hibitor 4-phenylbutyrate (PBA) re<strong>in</strong>stated fear learn<strong>in</strong>g <strong>in</strong> the Tg2576 mouse model of AD. Memoryre<strong>in</strong>statement by PBA was observed <strong>in</strong>dependently of the disease stage: both <strong>in</strong> young mice atthe onset of the first symptoms, but also <strong>in</strong> aged mice, when amyloid plaque deposition andmajor synaptic loss has occurred [67]. In this section the author has discussed the most recentf<strong>in</strong>d<strong>in</strong>gs concern<strong>in</strong>g HDACi and memory function <strong>in</strong> AD animal models. Several additionalexamples can be found <strong>in</strong> the literature, overall suggest<strong>in</strong>g that the adm<strong>in</strong>istration of HDACiwas able to improve cognition <strong>in</strong> various animal models of AD.1859.4 RNA-MEDIATED MECHANISMS AND ALZHEIMER’S DISEASEMicroRNAs (miRNAs) are a group of small non-cod<strong>in</strong>g RNAs that b<strong>in</strong>d to the 3 0 untranslatedregion (3 0 -UTR) of target mRNAs and mediate their post-transcriptional regulation lead<strong>in</strong>g toeither degradation or translational <strong>in</strong>hibition (Figure 9.4), depend<strong>in</strong>g on the degree ofsequence complementarity. MiRNA-mediated mechanisms are therefore commonly consideredamong epigenetic regulators, and J. Satoh (68) has recently reviewed all the studiesperformed <strong>in</strong> recent years and report<strong>in</strong>g an aberrant expression of miRNAs <strong>in</strong> AD bra<strong>in</strong>s(Table 9.3). For most of them the target mRNA has not yet been clarified, however miR-107,miR29a/b-1, miR-15a, miR-9, and miR-19b have as a target BACE1 mRNA, and were found tobe down-regulated <strong>in</strong> the temporal cortex of AD patients, likely result<strong>in</strong>g <strong>in</strong> <strong>in</strong>creased BACE1expression and production of Ab peptides [69,70]. Similarly miR-15a, miR-101, let-7i, andmiR-106b, target the APP mRNA and were down-regulated <strong>in</strong> the anterior temporal cortex ofAD patients [70,71]. Additionally, miR-132 was down-regulated <strong>in</strong> cerebellum, hippocampus,and medial frontal gyrus of AD patients [72]. MiR132 targets the ARHGAP32 mRNA encod<strong>in</strong>gfor the rho GTPase activat<strong>in</strong>g prote<strong>in</strong> 32, a member of a family of prote<strong>in</strong>s that have beenimplicated <strong>in</strong> regulat<strong>in</strong>g multiple processes <strong>in</strong> the morphological development of neurons,<strong>in</strong>clud<strong>in</strong>g axonal growth and guidance, dendritic elaboration, and formation of synapses [73].Non-cod<strong>in</strong>g ribonucleic acids (ncRNAs) are transcribed from <strong>in</strong>trons and <strong>in</strong>tergenic regions.They are also implicated <strong>in</strong> epigenetic regulation and might be <strong>in</strong>volved <strong>in</strong> site-specificrecruitment of chromat<strong>in</strong>-modify<strong>in</strong>g complexes [74]. Faghihi et al. [75] identified a conservednon-cod<strong>in</strong>g antisense transcript for BACE1 (BACE1-AS) that regulates BACE1 mRNA and


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>FIGURE 9.4MicroRNAs (miRNAs) mediated gene regulation. miRNAs are a group of small non-cod<strong>in</strong>g RNAs that b<strong>in</strong>d to the 3 0 untranslatedregion (3 0 -UTR) of target mRNAs and mediate their post-transcriptional regulation. A mature s<strong>in</strong>gle-strand miRNA complexedwith prote<strong>in</strong>s forms the RISCemiRNA complex that b<strong>in</strong>ds to the target mRNA lead<strong>in</strong>g to either degradation or translational<strong>in</strong>hibition, depend<strong>in</strong>g on the degree of sequence complementarity.TABLE 9.3 Some Examples of Altered miRNAs <strong>in</strong> AD Bra<strong>in</strong>sExperimental Model Observation Refs.Post-mortem AD bra<strong>in</strong>s Down-regulation of miR-107, miR29a/b-1, miR- [69,70]15a, miR-9, and miR-19b that target BACE1 mRNADown-regulation of miR-15a, miR-101, let-7i, and [70,71]miR-106b that target APP mRNADown-regulation of miR-132 that targetsARHGAP32 mRNA[72]186prote<strong>in</strong> expression <strong>in</strong> vitro and <strong>in</strong> vivo. BACE1-AS concentrations were elevated <strong>in</strong> subjectswith AD and <strong>in</strong> APP transgenic mice [75]. Others observed that a subset of mRNA-like senseantisensetranscript pairs are co-expressed near synapses of the adult mouse forebra<strong>in</strong> [76].Several of these pairs <strong>in</strong>volve mRNAs that have been implicated <strong>in</strong> synaptic functions and <strong>in</strong>AD pathways, <strong>in</strong>clud<strong>in</strong>g BACE1, reticulon 3 (a prote<strong>in</strong> that b<strong>in</strong>ds BACE1 and <strong>in</strong>hibits APPcleavage); APP b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> 2 (which b<strong>in</strong>ds the cytoplasmic tail of APP); rab6 (a prote<strong>in</strong>that is elevated <strong>in</strong> AD cortex); sirtu<strong>in</strong>-3 (a member of a class of prote<strong>in</strong>s thought to beneuroprotective <strong>in</strong> AD); and <strong>in</strong>tegr<strong>in</strong>-l<strong>in</strong>ked k<strong>in</strong>ase (which phosphorylates GSK3beta to<strong>in</strong>hibit its activity) [76].9.5 DISCUSSION AND CONCLUSIONSEpigenetic mechanisms have been l<strong>in</strong>ked to ag<strong>in</strong>g, age-related diseases, and memory formation[8,9,77,78]. Overall, there is consensus <strong>in</strong>dicat<strong>in</strong>g that DNA methylation is dynamicallyregulated <strong>in</strong> the adult central nervous system (CNS) <strong>in</strong> response to experience, and that thiscellular mechanism is a crucial step <strong>in</strong> memory formation, that DNMT activity is necessary formemory, and that DNA methylation may work <strong>in</strong> concert with histone modifications <strong>in</strong>memory formation [78]. The analysis of Dnmt3a and Dnmt3b prote<strong>in</strong>s dur<strong>in</strong>g the developmentof the mouse CNS revealed that whereas Dnmt3b is important for the early phase ofneurogenesis, Dnmt3a likely plays a dual role <strong>in</strong> regulat<strong>in</strong>g neurogenesis prenatally and CNSmaturation and function postnatally. Particularly, Dnmt3b is specifically expressed <strong>in</strong>progenitor cells dur<strong>in</strong>g neurogenesis, suggest<strong>in</strong>g an important role <strong>in</strong> the <strong>in</strong>itial steps ofprogenitor cell differentiation. Dnmt3a is expressed <strong>in</strong> post-mitotic young neurons follow<strong>in</strong>gthe Dnmt3b expression and may be required for the establishment of tissue-specific methylationpatterns of the genome [79,80]. Dnmt1 and Dnmt3a are expressed <strong>in</strong> post-mitoticneurons and required for synaptic plasticity, learn<strong>in</strong>g and memory through their overlapp<strong>in</strong>g


CHAPTER 9The <strong>Epigenetics</strong> of Alzheimer’s <strong>Disease</strong>roles <strong>in</strong> ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g DNA methylation and modulat<strong>in</strong>g neuronal gene expression <strong>in</strong> adultCNS rodent neurons [81]. The study by Siegmund et al. [43] clearly showed that DNMT3a isexpressed <strong>in</strong> human neurons across all ages, and that DNA methylation is dynamicallyregulated <strong>in</strong> the human cerebral cortex throughout the lifespan, and <strong>in</strong>volves differentiatedneurons [43]. The studies performed <strong>in</strong> mice and monkeys exposed to Pb as <strong>in</strong>fants have nicelydemonstrated that early life might be particularly vulnerable to epigenetic modifications thatcould predispose to AD pathogenesis <strong>in</strong> adulthood [37,38]. The same group has recentlyshown that Dnmt1 and Dnmt3a prote<strong>in</strong>s, methyl-CpG b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s, and prote<strong>in</strong>s<strong>in</strong>volved <strong>in</strong> histone acetylation and methylation were decreased <strong>in</strong> the bra<strong>in</strong> of 23-year-oldmonkeys exposed to Pb as <strong>in</strong>fants, with respect to non-exposed monkeys [82]. Even if DNAdemethylation processes are less clear than methylation ones, base excision repair is likely tobe <strong>in</strong>volved [32,78]. The studies by Fuso and colleagues suggest that dietary manipulation <strong>in</strong>rodents is able to methylate/demethylate the promoter of the PSEN1 gene, with subsequentregulation of gene expression [27,28], and that the same dietary manipulation can modulatethe expression of de novo DNMTs [32]. Similarly, all the follow<strong>in</strong>g studies [63e67] <strong>in</strong>dicatethat HDAC <strong>in</strong>hibitors improved cognition and memory functions <strong>in</strong> AD animal models.Although promis<strong>in</strong>g, the limit of all these papers is that most of the experiments have beenperformed <strong>in</strong> disease animal models, and little is still known <strong>in</strong> humans. Severe alterations ofthe one-carbon metabolic pathway <strong>in</strong> humans are associated with disease, for exampleDNMT3B mutations cause a rare chromosome breakage disease characterized by aberrantDNA methylation and called the immunodeficiency, centromeric region <strong>in</strong>stability, and facialanomalies syndrome (ICF syndrome) [83]. Complex <strong>in</strong>teractions between polymorphisms <strong>in</strong>one-carbon metabolic genes and plasma Hcy or serum folate and vitam<strong>in</strong> B12 levels areknown <strong>in</strong> LOAD subjects [84], however we still do not know to what extent dietary “onecarbonnutrients” and their metabolism can modulate and/or reverse AD-related epigeneticmarks <strong>in</strong> the human bra<strong>in</strong>. Therefore, some caution should be taken prior to recommend“one-carbon nutrients” or epigenetic drugs, such as HDACi, as preventative strategies for agerelatedneurodegeneration. Indeed, many issues still need to be clarified before compoundswith epigenetic properties could be effectively used to treat patients with a cognitive decl<strong>in</strong>e,the most important be<strong>in</strong>g the possible short- and long- term side effects. If a diet rich <strong>in</strong>antioxidants and <strong>in</strong> methyl donor compounds, such as fruit and vegetables, is likely to protectaga<strong>in</strong>st the risk of late-onset degenerative diseases, a manipulation of our genome by means ofa widespread usage of HDACi or other epigenetic compounds could have several side-effects <strong>in</strong>different organs and tissues of the body, such as, for example, cancer promotion [10,56,85].Additional studies are therefore required to clarify the <strong>in</strong>terplay between dietary methyl-donorcompounds, polymorphisms of metabolic genes and epigenetic modifications of key ADgenes, as well as the <strong>in</strong>terconnections between DNA methylation/demethylation reactions,histone tail modifications and RNA-mediated mechanisms <strong>in</strong> the regulation of AD-relatedgenes [10]. Only a better understand<strong>in</strong>g of the networks and the <strong>in</strong>terplay of these threeepigenetic mechanisms <strong>in</strong> AD pathophysiology could help the design of preventative strategiesto delay disease onset and/or progression.187References[1] Brookmeyer R, Johnson E, Ziegler-Graham K, Arrighi HM. Forecast<strong>in</strong>g the global burden of Alzheimer’sdisease. Alzheimer’s Dement 2007;307:186e91.[2] Wenk GL. Neuropathologic changes <strong>in</strong> Alzheimer’s disease. J Cl<strong>in</strong> Psychiatry 2003;64:7e10.[3] F<strong>in</strong>deis MA. The role of amyloid beta peptide 42 <strong>in</strong> Alzheimer’s disease. Pharmacol Ther 2007;116:266e86.[4] Bentahir M, Nyabi O, Verhamme J, Tolia A, Horré K, Wiltfang J, et al. Presenil<strong>in</strong> cl<strong>in</strong>ical mutations can affectgamma secretase activity by different mechanisms. J Neurochem 2006;96:732e42.[5] Ertek<strong>in</strong>-Taner N. Genetics of Alzheimer’s disease: a centennial review. Neurol Cl<strong>in</strong> 2007;25:611e7.[6] Migliore L, Coppedè F. Genetics, environmental factors and the emerg<strong>in</strong>g role of epigenetics <strong>in</strong> neurodegenerativediseases. Mutat Res 2009;667:82e97.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>188[7] Bertram L, McQueen MB, Mull<strong>in</strong> K, Blacker D, Tanzi RE. Systematic meta-analyses of Alzheimer disease geneticassociation studies: the AlzGene database. Nat Genet 2007;39:17e23.[8] Rodríguez-Rodero S, Fernández-Morera JL, Fernandez AF, Menéndez-Torre E, Fraga MF. Epigenetic regulation ofag<strong>in</strong>g. Discov Med 2010;10:225e33.[9] Reichenberg A, Mill J, MacCabe JH. <strong>Epigenetics</strong>, genomic mutations and cognitive function. Cogn Neuropsychiatry2009;14:377e90.[10] Coppedè F. One-carbon metabolism and Alzheimer’s disease: focus on epigenetics. Curr Genomics2010;11:246e60.[11] Zhuo JM, Wang H, Praticò D. Is hyperhomocyste<strong>in</strong>emia an Alzheimer’s disease (AD) risk factor, an AD marker,or neither? Trends Pharmacol Sci 2011. In press.[12] Ho RC, Cheung MW, Fu E, W<strong>in</strong> HH, Zaw MH, Ng A, et al. Is high homocyste<strong>in</strong>e level a risk factor for cognitivedecl<strong>in</strong>e <strong>in</strong> elderly? A systematic review, meta-analysis, and meta-regression. Am J Geriatr Psychiatry2011;19:607e17.[13] Selley ML, Close DR, Stern SE. The effect of <strong>in</strong>creased concentrations of homocyste<strong>in</strong>e on the concentration of(E)-4-hydroxy-2-nonenal <strong>in</strong> the plasma and cerebrosp<strong>in</strong>al fluid of patients with Alzheimer’s disease. NeurobiolAg<strong>in</strong>g 2002;23:383e8.[14] Hasegawa T, Ukai W, Jo DG, Xu X, Mattson MP, Nakagawa M, et al. Homocysteic acid <strong>in</strong>duces <strong>in</strong>traneuronalaccumulation of neurotoxic Abeta42: implications for the pathogenesis of Alzheimer’s disease. J Neurosci Res2005;80:869e76.[15] Isobe C, Murata T, Sato C, Terayama Y. Increase of total homocyste<strong>in</strong>e concentration <strong>in</strong> cerebrosp<strong>in</strong>al fluid <strong>in</strong>patients with Alzheimer’s disease and Park<strong>in</strong>son’s disease. Life Sci 2005;77:1836e43.[16] Smach MA, Jacob N, Golmard JL, Charfedd<strong>in</strong>e B, Lammouchi T, Ben Othman L, et al. Folate and homocyste<strong>in</strong>e<strong>in</strong> the cerebrosp<strong>in</strong>al fluid of patients with Alzheimer’s disease or dementia: a case control study. EurNeurol 2011;65:270e8.[17] Morrison LD, Smith DD, Kish SJ. Bra<strong>in</strong> S-adenosylmethion<strong>in</strong>e levels are severely decreased <strong>in</strong> Alzheimer’sdisease. J Neurochem 1996;67:1328e31.[18] Kennedy BP, Bottiglieri T, Arn<strong>in</strong>g E, Ziegler MG, Hansen LA, Masliah E. Elevated S-adenosyhomocyste<strong>in</strong>e <strong>in</strong>Alzheimer bra<strong>in</strong>: <strong>in</strong>fluence on methyltransferases and cognitive function. J Neural Transm2004;111:547e67.[19] L<strong>in</strong>nebank M, Popp J, Smulders Y, Smith D, Semmler A, Farkas M, et al. S-adenosylmethion<strong>in</strong>e is decreased <strong>in</strong>the cerebrosp<strong>in</strong>al fluid of patients with Alzheimer’s disease. Neurodegener Dis 2010;7:373e8.[20] Hooijmans CR, Blom HJ, Oppenraaij-Emmerzaal D, Ritskes-Hoit<strong>in</strong>ga M, Kiliaan AJ. The analysis ofS-adenosylmethion<strong>in</strong>e and S-adenosylhomocyste<strong>in</strong>e levels <strong>in</strong> the ag<strong>in</strong>g bra<strong>in</strong> of APP/PS1 Alzheimer mice.Neurol Sci 2009;30:439e45.[21] Goll MG, Bestor TH. Eukaryotic cytos<strong>in</strong>e methyltransferases. Annu Rev Biochem 2005;74:481e514.[22] Goll MG, Kirpekar F, Maggert KA, Yoder JA, Hsieh CL, Zhang X, et al. Methylation of tRNAAsp by the DNAmethyltransferase homolog Dnmt2. <strong>Science</strong> 2006;311:395e8.[23] Mastroeni D, Grover A, Delvaux E, Whiteside C, Coleman PD, Rogers J. Epigenetic mechanisms <strong>in</strong> Alzheimer’sdisease. Neurobiol Ag<strong>in</strong>g 2011. In Press.[24] Scarpa S, Fuso A, D’Anselmi F, Cavallaro RA. Presenil<strong>in</strong> 1 gene silenc<strong>in</strong>g by S-adenosylmethion<strong>in</strong>e: a treatmentfor Alzheimer disease? FEBS Lett 2003;541:145e8.[25] Fuso A, Sem<strong>in</strong>ara L, Cavallaro RA, D’Anselmi F, Scarpa S. S-adenosylmethion<strong>in</strong>e/homocyste<strong>in</strong>e cycle alterationsmodify DNA methylation status with consequent deregulation of PS1 and BACE and beta-amyloidproduction. Mol Cell Neurosci 2005;28:195e204.[26] Gius D, Cui H, Bradbury CM, Smart DK, Zhao S, Young L, et al. Dist<strong>in</strong>ct effects on gene expression of chemicaland genetic manipulation of the cancer epigenome revealed by a multimodality approach. Cancer Cell2004;6:361e71.[27] Fuso A, Nicolia V, Cavallaro RA, Ricceri L, D’Anselmi F, Coluccia P, et al. B-vitam<strong>in</strong> deprivation <strong>in</strong>duceshyperhomocyste<strong>in</strong>emia and bra<strong>in</strong> S-adenosylhomocyste<strong>in</strong>e, depletes bra<strong>in</strong> S-adenosylmethion<strong>in</strong>e, andenhances PS1 and BACE expression and amyloid-beta deposition <strong>in</strong> mice. Mol Cell Neurosci2008;37:731e46.[28] Fuso A, Nicolia V, Pasqualato A, Fiorenza MT, Cavallaro RA, Scarpa S. Changes <strong>in</strong> Presenil<strong>in</strong> 1 genemethylation pattern <strong>in</strong> diet-<strong>in</strong>duced B vitam<strong>in</strong> deficiency. Neurobiol Ag<strong>in</strong>g 2011;32:187e99.[29] Chan A, Shea TB. Folate deprivation <strong>in</strong>creases presenil<strong>in</strong> expression, gamma-secretase activity, and Abeta levels<strong>in</strong> mur<strong>in</strong>e bra<strong>in</strong>: potentiation by ApoE deficiency and alleviation by dietary S-adenosyl methion<strong>in</strong>e.J Neurochem 2007;102:753e60.[30] Chan A, Tchantchou F, Rogers EJ, Shea TB. Dietary deficiency <strong>in</strong>creases presenil<strong>in</strong> expression, gamma-secretaseactivity, and Abeta levels: potentiation by ApoE genotype and alleviation by S-adenosyl methion<strong>in</strong>e.J Neurochem 2009;110:831e6.


CHAPTER 9The <strong>Epigenetics</strong> of Alzheimer’s <strong>Disease</strong>[31] L<strong>in</strong> HC, Hsieh HM, Chen YH, Hu ML. S-Adenosylhomocyste<strong>in</strong>e <strong>in</strong>creases beta-amyloid formation <strong>in</strong> BV-2microglial cells by <strong>in</strong>creased expressions of beta-amyloid precursor prote<strong>in</strong> and presenil<strong>in</strong> 1 and by hypomethylationof these gene promoters. Neurotoxicology 2009;30:622e7.[32] Fuso A, Nicolia V, Cavallaro RA, Scarpa S. DNA methylase and demethylase activities are modulated by onecarbonmetabolism <strong>in</strong> Alzheimer’s disease models. J Nutr Biochem 2011;22:242e51.[33] Guo JU, Su Y, Zhong C, M<strong>in</strong>g GL, Song H. Hydroxylation of 5-methylcytos<strong>in</strong>e by TET1 promotes active DNAdemethylation <strong>in</strong> the adult bra<strong>in</strong>. Cell 2011;145:423e34.[34] Borro M, Cavallaro RA, Gentile G, Nicolia V, Fuso A, Simmaco M, et al. One-carbon metabolism alterationaffects bra<strong>in</strong> proteome profile <strong>in</strong> a mouse model of Alzheimer’s disease. J Alzheimer’s Dis 2010;22:1257e68.[35] Cavallaro RA, Fuso A, D’Anselmi F, Sem<strong>in</strong>ara L, Scarpa S. The effect of S-adenosylmethion<strong>in</strong>e on CNS geneexpression studied by cDNA microarray analysis. J Alzheimer’s Dis 2006;9:415e9.[36] Chen KL, Wang SS, Yang YY, Yuan RY, Chen RM, Hu CJ. The epigenetic effects of amyloid-beta(1-40) on globalDNA and neprilys<strong>in</strong> genes <strong>in</strong> mur<strong>in</strong>e cerebral endothelial cells. Biochem Biophys Res Commun2008;378:57e61.[37] Basha MR, Wei W, Bakheet SA, Benitez N, Siddiqi HK, Ge YW, et al. The fetal basis of amyloidogenesis:exposure to lead and latent overexpression of amyloid precursor prote<strong>in</strong> and beta-amyloid <strong>in</strong> the ag<strong>in</strong>g bra<strong>in</strong>.J Neurosci 2005;25:823e9.[38] Wu J, Basha MR, Brock B, Cox DP, Cardozo-Pelaez F, McPherson CA, et al. Alzheimer’s disease (AD)-likepathology <strong>in</strong> aged monkeys after <strong>in</strong>fantile exposure to environmental metal lead (Pb): evidence for a developmentalorig<strong>in</strong> and environmental l<strong>in</strong>k for AD. J Neurosci 2008;28:3e9.[39] Lahiri DK, Zawia NH, Greig NH, Sambamurti K, Maloney B. Early-life events may trigger biochemical pathwaysfor Alzheimer’s disease: the “LEARn” model. Biogerontology 2008;9:375e9.[40] Mastroeni D, McKee A, Grover A, Rogers J, Coleman PD. Epigenetic differences <strong>in</strong> cortical neurons from a pairof monozygotic tw<strong>in</strong>s discordant for Alzheimer’s disease. PLoS One 2009;4:e6617.[41] Mastroeni D, Grover A, Delvaux E, Whiteside C, Coleman PD, Rogers J. Epigenetic changes <strong>in</strong> Alzheimer’sdisease: Decrements <strong>in</strong> DNA methylation. Neurobiol Ag<strong>in</strong>g 2010;31:2025e37.[42] Barrach<strong>in</strong>a M, Ferrer I. DNA methylation of Alzheimer disease and tauopathy-related genes <strong>in</strong> postmortembra<strong>in</strong>. J Neuropathol Exp Neurol 2009;68:880e91.[43] Siegmund KD, Connor CM, Campan M, Long TI, Weisenberger DJ, B<strong>in</strong>iszkiewicz D, et al. DNA methylation <strong>in</strong>the human cerebral cortex is dynamically regulated throughout the life span and <strong>in</strong>volves differentiatedneurons. PLoS One 2007;2:e895.[44] Wang SC, Oelze B, Schumacher A. Age-specific epigenetic drift <strong>in</strong> late-onset Alzheimer’s disease. PLoS One2008;3:e2698.[45] Bollati V, Galimberti D, Pergoli L, Dalla Valle E, Barretta F, Cort<strong>in</strong>i F, et al. DNA methylation <strong>in</strong> repetitiveelements and Alzheimer disease. Bra<strong>in</strong> Behav Immun 2011. In press.[46] Seshadri S, Beiser A, Selhub J, Jacques PF, Rosenberg IH, D’Agost<strong>in</strong>o RB, et al. Plasma homocyste<strong>in</strong>e as a riskfactor for dementia and Alzheimer’s disease. N Engl J Med 2002;346:476e83.[47] Luchs<strong>in</strong>ger JA, Tang MX, Shea S, Miller J, Green R, Mayeux R. Plasma homocyste<strong>in</strong>e levels and risk ofAlzheimer disease. Neurology 2004;62:1972e6.[48] Ravaglia G, Forti P, Maioli F, Martelli M, Servadei L, Brunetti N, et al. Homocyste<strong>in</strong>e and folate as risk factorsfor dementia and Alzheimer disease. Am J Cl<strong>in</strong> Nutr 2005;82:636e43.[49] Luchs<strong>in</strong>ger JA, Tang MX, Miller J, Green R, Mayeux R. Relation of higher folate <strong>in</strong>take to lower risk ofAlzheimer disease <strong>in</strong> the elderly. Arch Neurol 2007;64:86e92.[50] Fowler B. Homocyste<strong>in</strong>e: overview of biochemistry, molecular biology, and role <strong>in</strong> disease processes. Sem<strong>in</strong>Vasc Med 2005;5:77e86.[51] Vafai SB, Stock JB. Prote<strong>in</strong> phosphatase 2A methylation: a l<strong>in</strong>k between elevated plasma homocyste<strong>in</strong>e andAlzheimer’s <strong>Disease</strong>. FEBS Lett 2002;518:1e4.[52] Sontag E, Nunbhakdi-Craig V, Sontag JM, Diaz-Arrastia R, Ogris E, Dayal S, et al. Prote<strong>in</strong> phosphatase 2Amethyltransferase l<strong>in</strong>ks homocyste<strong>in</strong>e metabolism with tau and amyloid precursor prote<strong>in</strong> regulation.J Neurosci 2007;27:2751e9.[53] Nicolia V, Fuso A, Cavallaro RA, Di Luzio A, Scarpa S. B vitam<strong>in</strong> deficiency promotes tau phosphorylationthrough regulation of GSK3beta and PP2A. J Alzheimer’s disease 2010;19:895e907.[54] Chan A, Rogers E, Shea TB. Dietary Deficiency <strong>in</strong> Folate and Vitam<strong>in</strong> E Under Conditions of Oxidative StressIncreases Phospho-Tau Levels: Potentiation by ApoE4 and Alleviation by S-Adenosylmethion<strong>in</strong>e. J Alzheimer’sDis 2009;17:483e7.[55] Berger SL. The complex language of chromat<strong>in</strong> regulation dur<strong>in</strong>g transcription. Nature 2007;447:407e12.[56] Chouliaras L, Rutten BP, Kenis G, Peerbooms O, Visser PJ, Verhey F, et al. Epigenetic regulation <strong>in</strong> the pathophysiologyof Alzheimer’s disease. Prog Neurobiol 2010;90:498e510.189


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>190[57] Mart<strong>in</strong> C, Zhang Y. The diverse functions of histone lys<strong>in</strong>e methylation. Nat Rev Mol Cell Biol 2005;6:838e49.[58] Yost JM, Korboukh I, Liu F, Gao C, J<strong>in</strong> J. Targets <strong>in</strong> epigenetics: <strong>in</strong>hibit<strong>in</strong>g the methyl writers of the histonecode. Curr Chem Genomics 2011;5:72e84.[59] Tachibana M, Matsumura Y, Fukuda M, Kimura H, Sh<strong>in</strong>kai Y. G9a/GLP complexes <strong>in</strong>dependently mediateH3K9 and DNA methylation to silence transcription. EMBO J 2008;27:2681e90.[60] Ogawa O, Zhu X, Lee HG, Ra<strong>in</strong>a A, Obrenovich ME, Bowser R, et al. Ectopic localization of phosphorylatedhistone H3 <strong>in</strong> Alzheimer’s disease: a mitotic catastrophe? Acta Neuropathol 2003;105:524e8.[61] Myung NH, Zhu X, Kruman II, Castellani RJ, Petersen RB, Siedlak SL, et al. Evidence of DNA damage <strong>in</strong>Alzheimer disease: phosphorylation of histone H2AX <strong>in</strong> astrocytes. Age (Dordr) 2008;30:209e15.[62] D<strong>in</strong>g H, Dolan PJ, Johnson GV. Histone deacetylase 6 <strong>in</strong>teracts with the microtubule-associated prote<strong>in</strong> tau.J Neurochem 2008;106:2119e30.[63] Francis YI, Fà M, Ashraf H, Zhang H, Staniszewski A, Latchman DS, et al. Dysregulation of histone acetylation<strong>in</strong> the APP/PS1 mouse model of Alzheimer’s disease. J Alzheimer’s Dis 2009;18:131e9.[64] Kilgore M, Miller CA, Fass DM, Hennig KM, Haggarty SJ, Sweatt JD, et al. Inhibitors of class 1 histonedeacetylases reverse contextual memory deficits <strong>in</strong> a mouse model of Alzheimer’s disease. Neuropsychopharmacology2009;35:870e80.[65] Nuut<strong>in</strong>en T, Suuronen T, Kaupp<strong>in</strong>en A, Salm<strong>in</strong>en A. Valproic acid stimulates cluster<strong>in</strong> expression <strong>in</strong> humanastrocytes: Implications for Alzheimer’s disease. Neurosci Lett 2010;475:64e8.[66] Gov<strong>in</strong>darajan N, Agis-Balboa RC, Walter J, Sananbenesi F, Fischer A. Sodium Butyrate Improves MemoryFunction <strong>in</strong> an Alzheimer’s <strong>Disease</strong> Mouse Model When Adm<strong>in</strong>istered at an Advanced Stage of <strong>Disease</strong>Progression. J Alzheimer’s Dis 2011. In Press.[67] Ricobaraza A, Cuadrado-Tejedor M, Marco S, Pérez-Otaño I, García-Osta A. Phenylbutyrate rescues dendriticsp<strong>in</strong>e loss associated with memory deficits <strong>in</strong> a mouse model of Alzheimer disease. Hippocampus 2010. InPress.[68] Satoh J. MicroRNAs and their therapeutic potential for human diseases: aberrant microRNA expression <strong>in</strong>Alzheimer’s disease bra<strong>in</strong>s. J Pharmacol Sci 2010;114:269e75.[69] Wang WX, Rajeev BW, Stromberg AJ, Ren N, Tang G, Huang Q, et al. The expression of microRNA miR-107decreases early <strong>in</strong> Alzheimer’s disease and may accelerate disease progression through regulation of beta-siteamyloid precursor prote<strong>in</strong>-cleav<strong>in</strong>g enzyme 1. J Neurosci 2008;28:1213e23.[70] Hébert SS, Horré K, Nicolaï L, Papadopoulou AS, Mandemakers W, Silahtaroglu AN, et al. Loss of microRNAcluster miR-29a/b-1 <strong>in</strong> sporadic Alzheimer’s disease correlates with <strong>in</strong>creased BACE1/beta-secretase expression.Proc Natl Acad Sci USA 2008;105:6415e20.[71] Hébert SS, Horré K, Nicolaï L, Bergmans B, Papadopoulou AS, Delacourte A, et al. MicroRNA regulation ofAlzheimer’s Amyloid precursor prote<strong>in</strong> expression. Neurobiol Dis 2009;33:422e8.[72] Cogswell JP, Ward J, Taylor IA, Waters M, Shi Y, Cannon B, et al. Identification of miRNA changes <strong>in</strong>Alzheimer’s disease bra<strong>in</strong> and CSF yields putative biomarkers and <strong>in</strong>sights <strong>in</strong>to disease pathways. J Alzheimer’sDis 2008;14:27e41.[73] Nasu-Nishimura Y, Hayashi T, Ohishi T, Okabe T, Ohwada S, Hasegawa Y, et al. Role of the Rho GTPaseactivat<strong>in</strong>gprote<strong>in</strong> RICS <strong>in</strong> neurite outgrowth. Genes Cells 2006;11:607e14.[74] Costa FF. Non-cod<strong>in</strong>g RNAs: Meet thy masters. Bioessays 2010;32:599e608.[75] Faghihi MA, Modarresi F, Khalil AM, Wood DE, Sahagan BG, Morgan TE, et al. Expression of a noncod<strong>in</strong>g RNAis elevated <strong>in</strong> Alzheimer’s disease and drives rapid feed-forward regulation of beta-secretase. Nat Med2008;14:723e30.[76] Smalheiser NR, Lugli G, Torvik VI, Mise N, Ikeda R, Abe K. Natural antisense transcripts are co-expressed withsense mRNAs <strong>in</strong> synaptoneurosomes of adult mouse forebra<strong>in</strong>. Neurosci Res. 2008;62:236e9.[77] Lub<strong>in</strong> FD. Epigenetic gene regulation <strong>in</strong> the adult mammalian bra<strong>in</strong>: Multiple roles <strong>in</strong> memory formation.Neurobiol Learn Mem 2011;96:68e78.[78] Day JJ, Sweatt JD. DNA methylation and Memory Formation. Nat Neurosci 2010;13:1319e23.[79] Feng J, Chang H, Li E, Fan G. Dynamic expression of de novo DNA methyltransferases Dnmt3a and Dnmt3b<strong>in</strong> the central nervous system. J Neurosci Res 2005;79:734e46.[80] Watanabe D, Uchiyama K, Hanaoka K. Transition of mouse de novo methyltransferases expression fromDnmt3b to Dnmt3a dur<strong>in</strong>g neural progenitor cell development. Neuroscience 2006;142:727e37.[81] Feng J, Zhou Y, Campbell SL, Le T, Li E, Sweatt JD, et al. Dnmt1 and Dnmt3a ma<strong>in</strong>ta<strong>in</strong> DNA methylation andregulate synaptic function <strong>in</strong> adult forebra<strong>in</strong> neurons. Nat Neurosci 2010;13:423e30.[82] Bihaqi SW, Huang H, Wu J, Zawia NH. Infant Exposure to Lead (Pb) and Epigenetic Modifications <strong>in</strong> the Ag<strong>in</strong>gPrimate Bra<strong>in</strong>: Implications for Alzheimer’s <strong>Disease</strong>. J Alzheimer’s Dis 2011. In press.[83] Ehrlich M. The ICF syndrome, a DNA methyltransferase 3B deficiency and immunodeficiency disease. Cl<strong>in</strong>Immunol 2003;109:17e28.


CHAPTER 9The <strong>Epigenetics</strong> of Alzheimer’s <strong>Disease</strong>[84] Coppedè F, Tannorella P, Pezz<strong>in</strong>i I, Migheli F, Ricci G, Caldarazzo-Ienco E, et al. Folate, homocyste<strong>in</strong>e, vitam<strong>in</strong>B12 and polymorphisms of genes participat<strong>in</strong>g <strong>in</strong> one-carbon metabolism <strong>in</strong> late onset Alzheimer’s diseasepatients and healthy controls. Antioxid Redox Signal 2011. In press.[85] Mason JB. Unravel<strong>in</strong>g the complex relationship between folate and cancer risk. Biofactors 2011;37:253e60.[86] Coppedè F, Grossi E, Migheli F, Migliore L. Polymorphisms <strong>in</strong> folate-metaboliz<strong>in</strong>g genes, chromosomedamage, and risk of Down syndrome <strong>in</strong> Italian women: identification of key factors us<strong>in</strong>g artificial neuralnetworks. BMC Med Genomics 2010;3:42.191


This page <strong>in</strong>tentionally left blank


CHAPTER 10Epigenetic Modulationof <strong>Human</strong> NeurobiologicalDisordersTakeo Kubota 1 , Kunio Miyake 1 , Takae Hirasawa 1 , Tatsushi Onaka 2 , Hidenori Yamasue 31 University of Yamanashi, Yamanashi, Japan2 Jichi Medical University, Tochigi, Japan3 The University of Tokyo, Tokyo, JapanCHAPTER OUTLINE10.1 Introduction 19310.2 Epigenetic MechanismAssociated with CongenitalNeurobiological Disorders 19510.3 Epigenetic MechanismUnderly<strong>in</strong>g Alteration of theBra<strong>in</strong> Function byEnvironmental Factors 19610.4 Transgenerational EpigeneticInheritance (Non-Mendelian<strong>Disease</strong> Inheritance) 19710.5 Epigenetic Medic<strong>in</strong>e forNeurobiological Disorders 19810.6 Conclusion 199References 20019310.1 INTRODUCTIONNeurons are sensitive to the dosage of genes, and a subset of neurobiological disorders can becaused by either an underexpression or an overexpression of a molecule <strong>in</strong> the neuron.Examples of such neurodevelopmental disorders are Pelizaeus-Merzbacher disease that isassociated with a deletion, mutation, or duplication of the PLP1 gene [1], adult-onsetneuromuscular disease, CharcoteMarieeTooth disease associated with a mutation or duplicationof PMP22 [2], and several types of mental retardation that are associated with deletionor duplication of the neuronal migration factor LIS1 [3,4]. Also, it was recently demonstratedthat mutation or duplication (multiplication) of the a-synucle<strong>in</strong> gene is associated withPark<strong>in</strong>son’s disease [5]. These cl<strong>in</strong>ical f<strong>in</strong>d<strong>in</strong>gs suggest that the bra<strong>in</strong> is extremely sensitive toperturbations <strong>in</strong> gene-regulation, and further <strong>in</strong>dicate that the bra<strong>in</strong> is an organ that requiresa proper control system for gene expression.“Epigenetic” mechanism is one such system of control of gene expression <strong>in</strong> higher vertebrates.The term “epigenetics” was first used by Conrad Wadd<strong>in</strong>gton <strong>in</strong> 1939 to describe “the causal<strong>in</strong>teractions between genes and their products, which br<strong>in</strong>g the phenotype <strong>in</strong>to be<strong>in</strong>g” [6]. Thecurrent def<strong>in</strong>ition is “the study of heritable changes <strong>in</strong> gene expression that occur <strong>in</strong>dependentT. Tollefsbol (Ed): <strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>. DOI: 10.1016/B978-0-12-388415-2.00010-XCopyright Ó 2012 Elsevier Inc. All rights reserved.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>of changes <strong>in</strong> the primary DNA sequence” [7]. Wadd<strong>in</strong>gton’s def<strong>in</strong>ition <strong>in</strong>itially referred to therole of epigenetics <strong>in</strong> embryonic development, <strong>in</strong> which cells develop dist<strong>in</strong>ct identities despitehav<strong>in</strong>g the same genetic <strong>in</strong>formation; however, the def<strong>in</strong>ition of epigenetics has evolved overtime as it is implicated <strong>in</strong> a wide variety of biological processes. The mechanism is essential fornormal development dur<strong>in</strong>g embryogenesis [8] and neural cell differentiation [9] as well asdifferentiation of other types of cells [10]. Therefore, epigenetics, which <strong>in</strong>cludes DNAmethylation, histone modifications, and regulation by microRNAs, may be a key concept tounderstand<strong>in</strong>g of the pathogenesis of neurobiological disorders [11e14].Precise understand<strong>in</strong>g of neurobiological disorders is important, because the number ofchildren with autism and related disorders (autistic spectrum disorder; ASD) have <strong>in</strong>creased30-fold (prevalence from 1/2500 to 1/86) <strong>in</strong> the last 50 years <strong>in</strong> England [15]. Affectedchildren are presently numbered approximately 100 (between 34 and 264) per 10 000 childrenworldwide [15e20]. Autism is one of the most common neurobiological disorders, which ischaracterized by three specific criteria: (1) abnormal reciprocal social <strong>in</strong>teractions: reduced<strong>in</strong>terest <strong>in</strong> peers and difficulty ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g social <strong>in</strong>teraction and failure to use eye gaze andfacial expressions to communicate efficiently, (2) impaired communication: language delays,deficits <strong>in</strong> language comprehension and response to voices, stereotyped or literal use of wordsand phrases, poor pragmatics and lack of prosody, result<strong>in</strong>g <strong>in</strong> monotone or exaggeratedspeech patterns, and (3) repetitive behaviors: motor stereotypies, repetitive use of objects,compulsions and rituals, <strong>in</strong>sistence on sameness, upset to change and unusual or very narrowrestricted <strong>in</strong>terests [21]. However, there have so far not been appropriate biological markers forautism, and thus, diagnosis is made based purely on behavioral criteria, such as DSM-IV99, thediagnostic manual of the American Psychiatric Association, and ICD-10100, the diagnosticmanual of the World Health Organization [22,23].194While various environmental factors are thought to contribute to the pathogenesis of autism[24], recent genetic studies have revealed rare mutations <strong>in</strong> more than 20 genes <strong>in</strong> a subset ofautistic children [25]. Many of the genes encode prote<strong>in</strong>s that are associated with synapticfunction, <strong>in</strong>clud<strong>in</strong>g synaptic scaffold<strong>in</strong>g prote<strong>in</strong>s, receptors, transporters on synapses, andneuronal cell adhesion molecules [25]. These f<strong>in</strong>d<strong>in</strong>gs suggest that autism may be a disorder ofthe synapse [26] (Figure 10.1).However, the <strong>in</strong>crease <strong>in</strong> the <strong>in</strong>cidence of autism cannot be solely attributed to genetic factors,because it is unlikely that mutation rates suddenly <strong>in</strong>creased <strong>in</strong> recent years. Therefore,FIGURE 10.1Location of the molecules <strong>in</strong> the synapse, which are associated with the pathogenesis of autism (references [25] and [45]).This figure is reproduced <strong>in</strong> the color plate section.


CHAPTER 10Epigenetic Modulation of <strong>Human</strong> Neurobiological Disordersenvironmental factors are very likely to be <strong>in</strong>volved <strong>in</strong> this <strong>in</strong>crease. This is partly supported bya study <strong>in</strong> tw<strong>in</strong>s that revealed that environmental factors contribute to the occurrence ofautism [27,28]. Furthermore epigenetic mechanisms are affected by environmental factors[11], and environment-<strong>in</strong>duced epigenetics changed <strong>in</strong> early life can persist through adulthoodand can be transmitted across generations result<strong>in</strong>g <strong>in</strong> abnormal behavior traits <strong>in</strong> theoffspr<strong>in</strong>g [29]. Consideration of the forego<strong>in</strong>g led us to propose a hypothesis that variousenvironmental factors change the epigenetic status of bra<strong>in</strong>-specific genes lead<strong>in</strong>g to alterations<strong>in</strong> the expression of a number of neuronal genes associated with the synapse, result<strong>in</strong>g<strong>in</strong> abnormal bra<strong>in</strong> function (aberrant synaptic function) <strong>in</strong> certa<strong>in</strong> neurobiological disorders(namely autism and ASD).In this chapter, we show various examples of neurobiological disorders associated withepigenetics, environmental factors that affect epigenetic gene regulation, and discuss futuredirections <strong>in</strong> medic<strong>in</strong>e for neurobiological disorders based on recent epigeneticunderstand<strong>in</strong>gs.10.2 EPIGENETIC MECHANISM ASSOCIATED WITH CONGENITALNEUROBIOLOGICAL DISORDERSEpigenetic control of gene expression is an <strong>in</strong>tr<strong>in</strong>sic mechanism for normal bra<strong>in</strong> development[30] and abnormalities <strong>in</strong> the molecules associated with this mechanism are associated withcongenital neurodevelopmental disorders <strong>in</strong>clud<strong>in</strong>g autistic disorders [31e40]. Genomicimpr<strong>in</strong>t<strong>in</strong>g is the epigenetic phenomenon <strong>in</strong>itially discovered <strong>in</strong> human disorders. In animpr<strong>in</strong>ted gene, one out of the two parental alleles is active and the other allele is <strong>in</strong>active dueto epigenetic mechanisms such as DNA methylation. Therefore, mutations <strong>in</strong> the active alleleor deletion of the active allele of the impr<strong>in</strong>ted gene results <strong>in</strong> no expression. This has beenfound <strong>in</strong> autistic disorders, Angelman syndrome, and PradereWilli syndrome [31].S<strong>in</strong>ce there are more genes <strong>in</strong> the two X chromosomes than <strong>in</strong> the X and Y chromosomes,females (XX) have more genes than males (XY). To m<strong>in</strong>imize this sex imbalance, one of the twoX chromosomes <strong>in</strong> females is <strong>in</strong>activated by an epigenetic mechanism [32]. If X-<strong>in</strong>activationdoes not properly occur <strong>in</strong> a female, such a female is believed to die <strong>in</strong> utero and abort. Thishypothesis is supported by the recent f<strong>in</strong>d<strong>in</strong>gs <strong>in</strong> cloned animals produced by somatic nucleartransfer <strong>in</strong> which failure of X-chromosome <strong>in</strong>activation <strong>in</strong>duces embryonic abortion [33,34].Even if one of the X chromosomes is extremely small due to a large term<strong>in</strong>al deletion, mak<strong>in</strong>gthe overdosage effect of X-l<strong>in</strong>ked genes small, the female shows a severe congenital neurodevelopmentaldelay [35], <strong>in</strong>dicat<strong>in</strong>g that proper epigenetic control of gene expression isessential for normal bra<strong>in</strong> development.195DNA methylation and histone modifications are a fundamental step <strong>in</strong> epigenetic gene control.DNA methylation is achieved by the addition of a methyl group (CH 3 ) to CpG d<strong>in</strong>ucleotides <strong>in</strong>a reaction that is mediated by DNA methyltransferases (e.g. DNMT1, 3A, 3B). Defect <strong>in</strong>a methyltransferase DNMT3B activity causes a syndrome characterized by immunodeficiency,centromere <strong>in</strong>stability, facial anomalies, and mild mental retardation (ICF syndrome) [36e38].Methyl-CpG b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s, which b<strong>in</strong>d to the methylated DNA region of genes, are alsoimportant molecules for control of gene expression. Mutations <strong>in</strong> one of the methyl-CpGb<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s, named methyl CpG b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> 2 (MeCP2), cause Rett syndrome, whichis characterized by seizures, ataxic gait, language dysfunction, and autistic behavior [39,40].Thus, it is thought that the mutation <strong>in</strong>MeCP2 results <strong>in</strong> a dysfunctional prote<strong>in</strong> and misb<strong>in</strong>d<strong>in</strong>gof MeCP2 to the methylated regions of genes, which leads to malsuppression of geneexpression <strong>in</strong> the bra<strong>in</strong> lead<strong>in</strong>g to manifestation of Rett syndrome <strong>in</strong>clud<strong>in</strong>g autism. Recentstudies have shown that MeCP2 controls the expression of some neuronal genes, such as bra<strong>in</strong>derivedneurotrophic factor (BDNF), distal-less homeobox 5 (DLX5), <strong>in</strong>sul<strong>in</strong>-like growthfactor b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> 3 (IFGBP3), and protocadher<strong>in</strong>s PCDHB1 and PCDH7 (neuronal cell


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>adhesion molecules) [41e45]. These f<strong>in</strong>d<strong>in</strong>gs suggest that not only mutations [25], but alsoepigenetic dysregulation of genes that encode synaptic molecules is possibly associated withautism (Figure 10.2). In fact, either deficiency of MeCP2 (i.e. Rett syndrome) or excessiveproduction of MeCP2 (duplication of the MECP2 genomic region) can lead to autisticfeatures [46].10.3 EPIGENETIC MECHANISM UNDERLYING ALTERATION OF THEBRAIN FUNCTION BY ENVIRONMENTAL FACTORSIn autism, both environmental factors (e.g. tox<strong>in</strong>s and <strong>in</strong>fections) and genetic factors (e.g.mutations <strong>in</strong> synaptic molecules) have historically been implicated [19,47,48]. However,a biological mechanism that l<strong>in</strong>ks these two groups of factors has not been identified.<strong>Epigenetics</strong> may bridge these two groups of factors contribut<strong>in</strong>g to disease development [11].Besides the <strong>in</strong>tr<strong>in</strong>sic (congenital) epigenetic defects (associated with the diseases describedabove), several l<strong>in</strong>es of evidence suggest that extr<strong>in</strong>sic (environmental) factors, such asmalnutrition, drugs, mental stress, maternal care, and neuronal stimulation, alter theepigenetic status of genes thereby affect<strong>in</strong>g bra<strong>in</strong> function [49e63]. Therefore, it is <strong>in</strong>trigu<strong>in</strong>gto th<strong>in</strong>k that acquired neurodevelopmental disorders, <strong>in</strong>clud<strong>in</strong>g autistic disorders, may bethe result of epigenetic dysregulation caused by environmental factors (Figure 10.3).196The epigenetic mechanism is also likely to be relevant <strong>in</strong> drug addiction. Gene expression <strong>in</strong>the dopam<strong>in</strong>ergic and glutamatergic systems is mediated by an epigenetic mechanism, andcoca<strong>in</strong>e and alcohol alter the epigenetic state. For example, coca<strong>in</strong>e <strong>in</strong>duces eitherhyperacetylation or hypoacetylation of histones H3 and H4 <strong>in</strong> the nucleus accumbens of mice,and alcohol <strong>in</strong>duces hyperacetylation of histones H3 and H4 <strong>in</strong> the frontal cortex and thenucleus accumbens of adolescent rats; such alterations may be associated with permanentbehavioral consequences [64,65].The above studies were ma<strong>in</strong>ly performed <strong>in</strong> animals, and there is little evidence for suchchanges <strong>in</strong> humans. However, the fact that epigenomic differences are greater <strong>in</strong> oldermonozygotic tw<strong>in</strong>s than <strong>in</strong> younger tw<strong>in</strong>s suggests that epigenetic status may be altered dur<strong>in</strong>gag<strong>in</strong>g by environmental factors <strong>in</strong> humans [66].FIGURE 10.2Schematic representation of epigenetic control of the gene expression. Either an excessive amount or deficiency of MeCP2leads to aberrant expression pattern of its target genes, lead<strong>in</strong>g to neurobiological disorders that exhibit autistic features. Yellowcircle: MeCP2, orange circle: prote<strong>in</strong> associated with histone modification, red circle: methyl-residue, green square:chromosomal histone prote<strong>in</strong>, arrow: transcription start site. This figure is reproduced <strong>in</strong> the color plate section.


CHAPTER 10Epigenetic Modulation of <strong>Human</strong> Neurobiological DisordersFIGURE 10.3DNA methylation mechanisms can be affected by environmental factors. This figure is reproduced <strong>in</strong> the color plate section.10.4 TRANSGENERATIONAL EPIGENETIC INHERITANCE(NON-MENDELIAN DISEASE INHERITANCE)In the current understand<strong>in</strong>g of biology, one’s acquired character is not <strong>in</strong>herited <strong>in</strong> the nextgeneration. We call this way of th<strong>in</strong>k<strong>in</strong>g Darw<strong>in</strong>ian <strong>in</strong>heritance. Therefore, we can rest easy,because this leads us to believe that the bad habits obta<strong>in</strong>ed dur<strong>in</strong>g one’s lifetime will not be<strong>in</strong>herited by our progeny. However, recent advances <strong>in</strong> epigenetics have revealed that suchundesirable acquired traits might be transmitted <strong>in</strong>to the next generation.Epigenetic marks, e.g. DNA methylation or histone modifications, allow the transmission ofgene activity states from one cell to its daughter cells. A fundamental question <strong>in</strong> epigenetics iswhether these marks can also be transmitted through the germl<strong>in</strong>e. If so, an aberrant epigeneticmark acquired <strong>in</strong> one generation could be <strong>in</strong>herited by the next generation. In general,epigenetic marks should be erased by demethylat<strong>in</strong>g factors such as the cytid<strong>in</strong>e deam<strong>in</strong>ases(e.g. AID, APOBEC1) [67] and re-established <strong>in</strong> each generation, but there have been reportsthat this erasure is <strong>in</strong>complete at some loci <strong>in</strong> the genome of several model organisms, possiblydue to deficiency of demethylat<strong>in</strong>g factors (e.g. AID) [67]. “Transgenerational epigenetic<strong>in</strong>heritance” refers to the germl<strong>in</strong>e transmission of an epigenetic mark [68,69], which mayprovide direct biological proof for an unexpla<strong>in</strong>ed hypothesis, Lamarckism, which is the ideaof heritability of acquired characteristics.197Transgenerational <strong>in</strong>heritance of epigenetic marks was first demonstrated <strong>in</strong> a specific mousestra<strong>in</strong>. The methylation status at the Ax<strong>in</strong> (Fu) locus <strong>in</strong> mature sperm reflects the methylationstate of the allele <strong>in</strong> the somatic tissue of the animal. This epigenetic status is l<strong>in</strong>ked to theshape of tail of the animals, and it does not undergo epigenetic reprogramm<strong>in</strong>g dur<strong>in</strong>ggametogenesis [70]. This observation was recently confirmed <strong>in</strong> Drosophila, <strong>in</strong> which anaberrant epigenetic mark (defective chromat<strong>in</strong> state) acquired <strong>in</strong> one generation <strong>in</strong>duced byenvironmental stress (e.g. heat shock) was <strong>in</strong>herited by the next generation [71].It has also been demonstrated that an aberrant epigenetic mark acquired <strong>in</strong> one generation bymental stress (maternal separation <strong>in</strong> early life) can be <strong>in</strong>herited by the next generation [29].Chronic maternal separation alters behaviors as well as the profile of DNA methylation <strong>in</strong> thepromoter of several candidate genes <strong>in</strong> both germl<strong>in</strong>e of the separated mice and the bra<strong>in</strong>s ofthe offspr<strong>in</strong>g with altered gene expression (e.g. decreases <strong>in</strong> the expression of the corticotrop<strong>in</strong>releas<strong>in</strong>g factor receptor 2 <strong>in</strong> the amygdala and the hypothalamus) [72]. These f<strong>in</strong>d<strong>in</strong>gs mayprovide the biological evidence for the current social issue that traumatic experiences <strong>in</strong> earlylife are risk factors for the development of behavioral and emotional disorders.Based on the evidences described above, the readers of this chapter might be led to believe thatepigenetics is a scientific field that portends adverse news for society. However, if we couldcreate an environment conducive to good human health, we will be able to sever the


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>deleterious environment-<strong>in</strong>duced epigenetic patterns across the generations. Even if epigeneticmark<strong>in</strong>gs provide a "memory" of past experiences and the mark<strong>in</strong>gs persist across the lifespanof an <strong>in</strong>dividual and then be transmitted to the offspr<strong>in</strong>g via epigenetic <strong>in</strong>heritance, futureepigenetic research can possibly establish restorative methods tak<strong>in</strong>g advantage of thereversibility of stress-<strong>in</strong>duced epigenetic modifications. It can also show us the appropriateenvironment for keep<strong>in</strong>g a healthy physical and mental condition [73,74].10.5 EPIGENETIC MEDICINE FOR NEUROBIOLOGICAL DISORDERSEnvironmental factors via epigenetic mechanisms are not always harmful. Imipram<strong>in</strong>e,a major antidepressant, was recently found to have the effect of restor<strong>in</strong>g a depressive state byalteration of the epigenetic state (<strong>in</strong>creas<strong>in</strong>g histone H3 and H4 acetylation at the Bdnf P3 andP4 promoters and histone H3-K4 dimethylation at Bdnf P3 promoter), lead<strong>in</strong>g to upregulationof Bdnf (bra<strong>in</strong>-derived neurotrophic factor) <strong>in</strong> the hippocampus [53]. It wasrecently shown that other drugs used for mental illness also have an epigenetic restor<strong>in</strong>g effect;these <strong>in</strong>clude valproic acid [51,54], clozap<strong>in</strong>e [55], sulpiride [55], and lithium [56]. Oxytoc<strong>in</strong>,which is a hormone associated with social <strong>in</strong>teractions [57e59], is tested for the treatment forautism and ASD, s<strong>in</strong>ce previous studies have revealed the effects of this neuropeptide onautistic social cognition <strong>in</strong> adults with autism [60,61]. S<strong>in</strong>ce aberrant hypermethylation of thepromoter region of the oxytoc<strong>in</strong> receptor gene is found <strong>in</strong> the DNA from bra<strong>in</strong> and bloodsamples of some autistic children [62], oxytoc<strong>in</strong> as well as other drugs for autism may haveepigenetic restor<strong>in</strong>g effects. Based on these f<strong>in</strong>d<strong>in</strong>gs, chemicals that alter epigenetic geneexpression are candidates of new drugs for a subset of the patients with mental and neurobiologicaldisorders [65].198Recent research has shown that not only drugs, but also general nutrition alters the epigeneticstatus of DNA. The best example is folic acid. Folic acid is the substrate that supplies methylresiduesdur<strong>in</strong>g methylation of cytos<strong>in</strong>e <strong>in</strong> DNA. Therefore, <strong>in</strong> order to ma<strong>in</strong>ta<strong>in</strong> DNAmethylation, proper <strong>in</strong>take of folic acid is essential. In Japan, the number of young womenwho do not take a sufficient amount of folic acid dur<strong>in</strong>g pregnancy is <strong>in</strong>creas<strong>in</strong>g, and this<strong>in</strong>creases the risk of hav<strong>in</strong>g babies with neural tube defects [75]. In rats, <strong>in</strong>appropriate supplyof nutrients from the mother to the fetus also <strong>in</strong>creases the susceptibility of the fetus to developdiabetes mellitus through epigenetic effects [76]. These hypotheses are supported by observationfrom a rat study <strong>in</strong> which prote<strong>in</strong> restriction dur<strong>in</strong>g pregnancy <strong>in</strong>duced a state ofmalnutrition and hyperlipidemia <strong>in</strong> the fetus. However, supplementation of the maternalprote<strong>in</strong>-restricted diet with folic acid dur<strong>in</strong>g pregnancy relieved these abnormalities. The effectwas achieved by an <strong>in</strong>crease <strong>in</strong> DNA methylation of the promoter regions of the PPAR-alphaand glucocorticoid receptor genes <strong>in</strong> the liver by folic acid, which led to proper suppressionof the genes [49]. These f<strong>in</strong>d<strong>in</strong>gs <strong>in</strong>dicate that specific nutrient <strong>in</strong>takes may alter the phenotypeof the offspr<strong>in</strong>g through epigenetic changes. Prote<strong>in</strong> restriction dur<strong>in</strong>g pregnancy also reducedthe expression of Mecp2 gene <strong>in</strong> the liver of the fetus [50]. This observation implies thatmalnutrition dur<strong>in</strong>g pregnancy may contribute to the development of neurobiologicaldisorders, although the effect of malnutrition on Mecp2 has not been <strong>in</strong>vestigated <strong>in</strong> the bra<strong>in</strong>.Royal jelly, known to change the phenotype from a genetically identical female honeybee toa fertile queen, also has epigenetic effects. A recent study revealed that royal jelly has the effectof eras<strong>in</strong>g global DNA methylation, because silenc<strong>in</strong>g the expression of Dnmt3, a DNAmethyltransferase, had a similar effect on the larval development [77].S<strong>in</strong>ce the 1980s, folic acid is empirically used for the treatment of autistic patients, and thesestudies have shown that it is effective only for a subset of the patients [78e80]. However, it hasnot been completely proven that the effect of folic acid on autistic patients is based onepigenetic effects. Therefore, it is important to confirm the genomic regions (genes) whereDNA methylation is altered by treatment with folic acid. Improved methods for the genome-


CHAPTER 10Epigenetic Modulation of <strong>Human</strong> Neurobiological DisordersFIGURE 10.4Pathogenesis of Rett syndrome and putative pathogenesis of autism. This figure is reproduced <strong>in</strong> the color plate section.wide methylation studies [81,82] may allow us to identify folate-responsive genes where DNAmethylation is altered by adm<strong>in</strong>istration of folic acid <strong>in</strong> autistic children. Such gene, if identified,can be a therapeutic marker for folic acid treatment, and the folic acid responsive(treatable) autistic patients can be dist<strong>in</strong>guished from the non-responsive (non-treatable)patients (Figure 10.4).Folic acid-based treatment may be safe, s<strong>in</strong>ce it is a nutrient, and it is expected to have globaleffects, but not on <strong>in</strong>dividual genes. Besides these nutrition-based treatments, several alternativeepigenetic-based treatments are currently developed, which is gene-specific and able torestore the epigenetic status that has been changed by an environmental factor. Examples are<strong>in</strong>hibitors of either DNA methylation or histone deacetylases are bound to pyrrole-imidazole(PI) polyamides, small synthetic molecules that recognize and attach to the m<strong>in</strong>or groove ofDNA, which can be designed for DNA sequences of any genes and can regulate gene transcriptionby b<strong>in</strong>d<strong>in</strong>g to DNA [83]. In fact, molecules that consist of an epigenetic <strong>in</strong>hibitor anda PI polyamide have been delivered to a target gene and have altered its expression [84].199It has recently been discovered that the DNA sequence is not the same <strong>in</strong> each neuron [85],and that epigenetic change underlies the somatic change of the DNA sequence [86]. . This is thephenomenon that is referred to as retrotransposition, <strong>in</strong> which a repetitive LINE-1 element is<strong>in</strong>serted <strong>in</strong>to various genomic regions when it is hypomethylated and can change theexpression of adjacent genes. The retrotransposition is accelerated by deficiency of MeCP2[87]. Interest<strong>in</strong>gly, <strong>in</strong> mice, the retrotransposition is also activated by voluntary exercise(runn<strong>in</strong>g) [88], suggest<strong>in</strong>g that exercise may alter the DNA methylation status <strong>in</strong> neurons.Studies us<strong>in</strong>g a Rett syndrome mouse model (Mecp2 knockout mice) show that environmentalenrichment (e.g. availability of stimulat<strong>in</strong>g toys) dur<strong>in</strong>g early postnatal development produceseffects on neural development and ameliorates the neurological phenotypes associated withRett syndrome [89,90]. This suggests that DNA methylation status may be corrected by anappropriate environmental stimulus, compensat<strong>in</strong>g for the <strong>in</strong>sufficient MeCP2 function.The reversibility of this epigenetic change is supported by a different study <strong>in</strong> which activationof Mecp2 expression after birth leads to relief of neurological symptoms <strong>in</strong> Mecp2 knockoutmice [91].10.6 CONCLUSIONVarious environmental factors potentially rewrite epigenetic codes. However, s<strong>in</strong>ce epigeneticcode is reversible, be<strong>in</strong>g different from genetic codes, it is potentially treatable, and preventable


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>FIGURE 10.5Overview of the DNA methylation changes and environmental factors. Note: This diagram depicts aberrant expression ofnormally suppressed gene by promoter hypermethylation; however, suppression of normally expressed genes by promoterhypermethylation is equally important for pathogenesis of human neurobiological disorders. This figure is reproduced <strong>in</strong> thecolor plate section.when we further understand the mechanism(s) of how environmental factors <strong>in</strong>duce epigeneticchanges (Figure 10.5). In this context, better understand<strong>in</strong>g of epigenetics is nowimportant for development of new therapies for neurobiological disorders.200References[1] Inoue K, Kanai M, Tanabe Y, Kubota T, Kashork CD, Wakui K, Fukushima Y, Lupski JR, Shaffer LG. Prenatal<strong>in</strong>terphase FISH diagnosis of PLP1 duplication associated with Pelizaeus-Merzbacher disease. Prenat Diagn2001;21:1133e6.[2] Onl<strong>in</strong>e Mendelian Inheritance <strong>in</strong> Man (OMIM): #118220 http://www.ncbi.nlm.nih.gov/entrez/[3] Re<strong>in</strong>er O, Carrozzo R, Shen Y, Wehnert M, Faust<strong>in</strong>ella F, Dobyns WB, Caskey CT, Ledbetter DH. Isolation ofa Miller-Dieker lissencephaly gene conta<strong>in</strong><strong>in</strong>g G prote<strong>in</strong> beta-subunit-like repeats. Nature 1993;364:717e21.[4] Bi W, Sapir T, Shchelochkov OA, Zhang F, Withers MA, Hunter JV. Increased LIS1 expression affects human andmouse bra<strong>in</strong> development. Nat Genet 2009;41:168e77.[5] Obi T, Nishioka K, Ross OA, Terada T, Yamazaki K, Sugiura A, Takanashi M, Mizoguchi K, Mori H, Mizuno Y,Hattori N. Cl<strong>in</strong>icopathologic study of a SNCA gene duplication patient with Park<strong>in</strong>son disease and dementia.Neurology 2008;70:238e41.[6] Wadd<strong>in</strong>gton CH. Epigenotype. Endeavour 1942;1:18e20.[7] Sharma S, Kelly TK, Jones PA. <strong>Epigenetics</strong> <strong>in</strong> cancer. Carc<strong>in</strong>ogenesis 2010;31:27e36.[8] Li E, Beard C, Jaenisch R. Role for DNA methylation <strong>in</strong> genomic impr<strong>in</strong>t<strong>in</strong>g. Nature 1993;366:362e5.[9] Takizawa T, Nakashima K, Namihira M, Ochiai W, Uemura A, Yanagisawa M, et al. DNA methylation isa critical cell-<strong>in</strong>tr<strong>in</strong>sic determ<strong>in</strong>ant of astrocyte differentiation <strong>in</strong> the fetal bra<strong>in</strong>. Dev Cell 2001;1:749e58.[10] Sakashita K, Koike K, K<strong>in</strong>oshita T, Shiohara M, Kamijo T, Taniguchi S, Kubota T. Dynamic DNA methylationchange <strong>in</strong> the CpG island region of p15 dur<strong>in</strong>g human myeloid development. J Cl<strong>in</strong> Invest2001;108:1195e204.[11] Qiu J. <strong>Epigenetics</strong>: unf<strong>in</strong>ished symphony. Nature 2006;441:143e5.[12] Abel T, Zuk<strong>in</strong> RS. Epigenetic targets of HDAC <strong>in</strong>hibition <strong>in</strong> neurodegenerative and psychiatric disorders. CurrOp<strong>in</strong> Pharmacol 2008;8:57e64.[13] Urd<strong>in</strong>guio RG, Sanchez-Mut JV, Esteller M. Epigenetic mechanisms <strong>in</strong> neurological diseases: genes, syndromes,and therapies. Lancet Neurol 2009;8:1056e72.[14] Wu H, Tao J, Chen PJ, Shahab A, Ge W, Hart RP, Ruan X, Ruan Y, Sun YE. Genome-wide analysis revealsmethyl-CpG-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> 2-dependent regulation of microRNAs <strong>in</strong> a mouse model of Rett syndrome. ProcNatl Acad Sci USA 2010;107:18161e6.[15] Baird G, Simonoff E, Pickles A, Chandler S, Loucas T, Meldrum D, Charman T. Prevalence of disorders of theautism spectrum <strong>in</strong> a population cohort of children <strong>in</strong> South Thames: the Special Needs and Autism Project(SNAP). Lancet 2006;368:210e5.


CHAPTER 10Epigenetic Modulation of <strong>Human</strong> Neurobiological Disorders[16] Yearg<strong>in</strong>-Allsopp M, Rice C, Karapurkar T, Doernberg N, Boyle C, Murphy C. Prevalence of Autism <strong>in</strong> a USMetropolitan Area. JAMA 2003;289:49e55.[17] Honda H, Shimizu Y, Imai M, Nitto Y. Cumulative <strong>in</strong>cidence of childhood autism: a total population study ofbetter accuracy and precision. Dev Med Child Neurol 2005;47:10e8.[18] Holden C. Autism Now. <strong>Science</strong> 2009;323:565.[19] Fombonne E. Epidemiology of pervasive developmental disorders. Pediatr Res 2009;65:591e8.[20] Kim YS, Leventhal BL, Koh YJ, Fombonne E, Laska E, Lim EC, et al. Prevalence of Autism Spectrum Disorders<strong>in</strong> a Total Population Sample. Am J Psychiatry 2011;168:904e12.[21] Silverman JL, Yang M, Lord C, Crawley JN. Behavioural phenotyp<strong>in</strong>g assays for mouse models of autism. NatRev Nerusosci 2010;490:490e502.[22] American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders. 4th ed. Wash<strong>in</strong>gtonD.C: APA; 1994.[23] World Health Organization. The ICD-10 Classification of Mental and Behavioural Disorders, Geneva,Switzerland; 1992[24] Herbert MR. Contributions of the environment and environmentally vulnerable physiology to autism spectrumdisorders. Curr Op<strong>in</strong> Neurol 2010;23:103e10.[25] Persico AM, Bourgeron T. Search<strong>in</strong>g for ways out of the autism maze: genetic, epigenetic and environmentalclues. Trends Neurosci 2006;29:349e58.[26] Zoghbi HY. Postnatal neurodevelopmental disorders: meet<strong>in</strong>g at the synapse? <strong>Science</strong> 2003;302:826e30.[27] Hoekstra RA, Bartels M, Hudziak JJ, Van Beijsterveldt TC, Boomsma DI. Genetics and environmental covariationbetween autistic traits and behavioral problems. Tw<strong>in</strong> Res Hum Genet 2007;10:853e86.[28] Hallmayer J, Cleveland S, Torres A, Phillips J, Cohen B, Torigoe T, et al. Genetic heritability and sharedenvironmental factors among tw<strong>in</strong> pairs with autism. Arch Gen Psychiatry 2011;68:1095e102.[29] Frankl<strong>in</strong> TB, Russig H, Weiss IC, Gräff J, L<strong>in</strong>der N, Michalon A, Vizi S, Mansuy IM. Epigenetic transmission ofthe impact of early stress across generations. Biol Psychiatry 2010;68:408e15.[30] Guy J, Hendrich B, Holmes M, Mart<strong>in</strong> JE, Bird A. A mouse Mecp2-null mutation causes neurological symptomsthat mimic Rett syndrome. Nat Genet 2001;27:322e6.[31] Kubota T, Das S, Christian SL, Bayl<strong>in</strong> SB, Herman JG, Ledbetter DH. Methylation-specific PCR symplifiesimpr<strong>in</strong>t<strong>in</strong>g analysis. Nat Genet 1997;16:16e7.[32] Kubota T, Nonoyama S, Tonoki H, Masuno M, Imaizumi K, Kojima M, Wakui K, Shimadzu M, Fukushima Y.A new assay for the analysis of X-chromosome <strong>in</strong>activation based on methylation-specific PCR. Hum Genet1999;104:49e55.[33] Xue F, Tian XC, Du F, Kubota C, Taneja M, D<strong>in</strong>nyes A, Dai Y, Lev<strong>in</strong>e H, Pereira LV, Yang X. Aberrant patterns ofX chromosome <strong>in</strong>activation <strong>in</strong> bov<strong>in</strong>e clones. Nat Genet 2002;31:216e20.[34] Nolen LD, Gao S, Han Z, Mann MR, Gie Chung Y, Otte AP, Bartolomei MS, Latham KE. X chromosomereactivation and regulation <strong>in</strong> cloned embryos. Dev Biol 2005;279:525e40.[35] Kubota T, Wakui K, Nakamura T, Ohashi H, Watanabe Y, Yosh<strong>in</strong>o M, et al. Proportion of the cells withfunctional X disomy is associated with the severity of mental retardation <strong>in</strong> mosaic r<strong>in</strong>g X Turner syndromefemales. Cytogenet Genome Res 2002;99:276e84.[36] Okano M, Bell DW, Haber DA, Li E. DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novomethylation and mammalian development. Cell 1999;99:247e57.[37] Shirohzu H, Kubota T, Kumazawa A, Sado T, Chijiwa T, Inagaki K, Suetake I, Tajima S, Wakui K, Miki Y,Hayashi M, Fukushima Y, Sasaki H. Three novel DNMT3B mutations <strong>in</strong> Japanese patients with ICF syndrome.Am J Med Genet 2002;112:31e7.[38] Kubota T, Furuumi H, Kamoda T, Iwasaki N, Tobita N, Fujiwara N, Goto Y, Matsui A, Sasaki H, Kajii T. ICFsyndrome <strong>in</strong> a girl with DNA hypomethylation but without detectable DNMT3B mutation. Am J Med Genet A2004;129:290e3.[39] Amir RE, Van den Veyver IB, Wan M, Tran CQ, Francke U, Zoghbi HY. Rett syndrome is caused by mutations <strong>in</strong>X-l<strong>in</strong>ked MECP2, encod<strong>in</strong>g methyl-CpG-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> 2. Nat Genet 1999;23:185e8.[40] Chunshu Y, Endoh K, Soutome M, Kawamura R, Kubota T. A patient with classic Rett syndrome with a novelmutation <strong>in</strong> MECP2 exon 1. Cl<strong>in</strong> Genet 2006;70:530e1.[41] Chen WG, Chang Q, L<strong>in</strong> Y, Meissner A, West AE, Griffith EC, Jaenisch R, Greenberg ME. Derepression of BDNFTranscription Involves Calcium-Dependent Phosphorylation of MeCP2. <strong>Science</strong> 2003;302:885e9.[42] Mart<strong>in</strong>owich K, Hattori D, Wu H, Fouse S, He F, Hu Y, Fan G, Sun YE. DNA methylation-related chromat<strong>in</strong>remodel<strong>in</strong>g <strong>in</strong> activity-dependent BDNF gene regulation. <strong>Science</strong> 2003;302:890e3.[43] Horike S, Cai S, Miyano M, Cheng JF, Kohwi-Shigematsu T. Loss of silent-chromat<strong>in</strong> loop<strong>in</strong>g and impairedimpr<strong>in</strong>t<strong>in</strong>g of DLX5 <strong>in</strong> Rett syndrome. Nat Genet 2005;37:31e40.201


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>202[44] Itoh M, Ide S, Takashima S, Kudo S, Nomura Y, Segawa M, et al. Methyl CpG-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> 2, whosemutation causes Rett syndrome, directly regulates Insul<strong>in</strong>-like Growth Factor B<strong>in</strong>d<strong>in</strong>g Prote<strong>in</strong> 3 <strong>in</strong> mouse andhuman Bra<strong>in</strong>s. J Neuropathol Exp Neurol 2007;66:117e23.[45] Miyake K, Hirasawa T, Soutome M, Itoh M, Goto Y, Endoh K, et al. The protocadher<strong>in</strong>s, PCDHB1 and PCDH7,are regulated by MeCP2 <strong>in</strong> neuronal cells and bra<strong>in</strong> tissues: implication for pathogenesis of Rett syndrome.BMC Neurosci 2011 (<strong>in</strong> press).[46] Ramocki MB, Peters SU, Tavyev YJ, Zhang F, Carvalho CM, Schaaf CP, et al. Autism and other neuropsychiatricsymptoms are prevalent <strong>in</strong> <strong>in</strong>dividuals with MeCP2 duplication syndrome. Ann Neurol 2009;66:771e82.[47] Bailey A, Le Couteur A, Gottesman I, Bolton P, Simonoff E, Yuzda E, Rutter M. Autism as a strongly geneticdisorder: evidence from a British tw<strong>in</strong> study. Psychol Med 1995;25:63e77.[48] Zafeiriou DI, Ververi A, Vargiami E. Childhood autism and associated comorbidities. Bra<strong>in</strong> Dev2007;29:257e72.[49] Burdge GC, Lillycrop KA, Phillips ES, Slater-Jefferies JL, Jackson AA, Hanson MA. Folic Acid Supplementationdur<strong>in</strong>g the Juvenile-Pubertal Period <strong>in</strong> Rats Modifies the Phenotype and Epigenotype Induced by PrenatalNutrition. J Nutr 2009;139:1054e60.[50] Lillycrop KA, Slater-Jefferies JL, Hanson MA, Godfrey KM, Jackson AA, Burdge GC. Induction of alteredepigenetic regulation of the hepatic glucocorticoid receptor <strong>in</strong> the offspr<strong>in</strong>g of rats fed a prote<strong>in</strong>-restricted dietdur<strong>in</strong>g pregnancy suggests that reduced DNA methyltransferase-1 expression is <strong>in</strong>volved <strong>in</strong> impaired DNAmethylation and changes <strong>in</strong> histone modifications. Br J Nutr 97:1064e73.[51] Jessberger S, Nakashima K, Clemenson Jr GD, Mejia E, Mathews E, Ure K, Ogawa S, S<strong>in</strong>ton CM, Gage FH,Hsieh J. Epigenetic Modulation of Seizure-Induced Neurogenesis and Cognitive Decl<strong>in</strong>e. J Neurosci2007;27:5967e75.[52] Weaver IC, Cervoni N, Champagne FA, D’Alessio AC, Sharma S, Seckl JR, Dymov S, Szyf M, Meaney MJ.Epigentic programm<strong>in</strong>g by maternal behavior. Nat Neurosci 2004;9:847e54.[53] Tsankova NM, Berton O, Renthal W, Kumar A, Neve RL, Nestler EJ. Susta<strong>in</strong>ed hippocampal chromat<strong>in</strong>regulation <strong>in</strong> a mouse model of depression and antidepressant action. Nat Neurosci 2006;9:519e25.[54] Dong E, Chen Y, Gav<strong>in</strong> DP, Grayson DR, Guidotti A. Valproate <strong>in</strong>duces DNA demethylation <strong>in</strong> nuclear extractsfrom adult mouse bra<strong>in</strong>. <strong>Epigenetics</strong> 2010;5:730e5.[55] Dong E, Nelson M, Grayson DR, Costa E, Guidotti A. Clozap<strong>in</strong>e and sulpiride but not haloperidol or olanzap<strong>in</strong>eactivate bra<strong>in</strong> DNA demethylation. Proc Natl Acad Sci USA 2008;105:13614e9.[56] Wang Q, Xu X, Li J, Liu J, Gu H, Zhang R, et al. Lithium, an anti-psychotic drug, greatly enhances thegeneration of <strong>in</strong>duced pluripotent stem cells. Cell Res 2011;21:1424e35.[57] Takayanagi Y, Yoshida M, Bielsky IF, Ross HE, Kawamata M, Onaka T, et al. Pervasive social deficits, but normalparturition, <strong>in</strong> oxytoc<strong>in</strong> receptor-deficient mice. Proc Natl Acad Sci USA 2005;102:16096e101.[58] Young LJ. Be<strong>in</strong>g human: love: neuroscience reveals all. Nature 2009;457:148.[59] Bartz JA, Zaki J, Bolger N, Ochsner KN. Social effects of oxytoc<strong>in</strong> <strong>in</strong> humans: context and person matter. TrendsCogn Sci 2011;15:301e9.[60] Yamasue H, Kuwabara H, Kawakubo Y, Kasai K. Oxytoc<strong>in</strong>, sexually dimorphic features of the social bra<strong>in</strong>, andautism. Psychiatry Cl<strong>in</strong> Neurosci 2010;63:129e40.[61] Andari E, Duhamel JR, Zalla T, Herbrecht E, Leboyer M, Sirigu A. Promot<strong>in</strong>g social behavior with oxytoc<strong>in</strong> <strong>in</strong>high-function<strong>in</strong>g autism spectrum disorders. Proc Natl Acad Sci USA 2010;107:4389e94.[62] Gregory SG, Connelly JJ, Towers AJ, Johnson J, Biscocho D, Markunas CA, et al. Genomic and epigeneticevidence for oxytoc<strong>in</strong> receptor deficiency <strong>in</strong> autism. BMC Med 2009;(7):62. 2009 Oct 22.[63] Ma DK, Jang MH, Guo JU, Kitabatake Y, Chang ML, Pow-Anpongkul N, Flavell RA, Lu B, M<strong>in</strong>g GL, Song H.Neuronal Activity-Induced Gadd45b Promotes Epigenetic DNA Demethylation and Adult Neurogenesis.<strong>Science</strong> 2009;323:1074e7.[64] Renthal W, Kumar A, Xiao G, Wilk<strong>in</strong>son M, Cov<strong>in</strong>gton 3rd HE, Maze I, et al. Genome-wide analysis ofchromat<strong>in</strong> regulation by coca<strong>in</strong>e reveals a role for sirtu<strong>in</strong>s. Neuron 2009;62:335e48.[65] Pascual M, Boix J, Felipo V, Guerri C. Repeated alcohol adm<strong>in</strong>istration dur<strong>in</strong>g adolescence causes changes <strong>in</strong>the mesolimbic dopam<strong>in</strong>ergic and glutamatergic systems and promotes alcohol <strong>in</strong>take <strong>in</strong> the adult rat. JNeurochem 2009;108:920e31.[66] Fraga MF, Ballestar E, Paz MF, Ropero S, Setien F, Ballestar ML, et al. Epigenetic differences arise dur<strong>in</strong>g thelifetime of monozygotic tw<strong>in</strong>s. Proc Natl Acad Sci USA 2005;102:10604e9.[67] Popp C, Dean W, Feng S, Cokus SJ, Andrews S, Pellegr<strong>in</strong>i M, Jacobsen SE, Reik W. Genome-wide erasure ofDNA methylation <strong>in</strong> mouse primordial germ cells is affected by AID deficiency. Nature 2011;463:1101e5.[68] Horsthemke B. Heritable germl<strong>in</strong>e epimutations <strong>in</strong> humans. Nat Genet 2007;39:573e4.[69] Dax<strong>in</strong>ger L, Whitelaw E. Transgenerational epigenetic <strong>in</strong>heritance: more questions than answers. Genome Res2010;20:1623e8.


CHAPTER 10Epigenetic Modulation of <strong>Human</strong> Neurobiological Disorders[70] Rakyan VK, Chong S, Champ ME, Cuthbert PC, Morgan HD, Luu KV, Whitelaw E. Transgenerational <strong>in</strong>heritanceof epigenetic states at the mur<strong>in</strong>e Ax<strong>in</strong>(Fu) allele occurs after maternal and paternal transmission.Proc Natl Acad Sci USA 2003;100:2538e43.[71] Seong KH, Li D, Shimizu H, Nakamura R, Ishii S. Inheritance of Stress-Induced, ATF-2-Dependent EpigeneticChange. Cell 2011;145:1049e61.[72] Weiss IC, Frankl<strong>in</strong> TB, Vizi S, Mansuy IM. Inheritable effect of unpredictable maternal separation on behavioralresponses <strong>in</strong> mice. Front Behav Neurosci. Front Behav Neurosci 2011;5:3.[73] Thayer ZM, Kuzawa CW. Biological memories of past environments: Epigenetic pathways to health disparities.<strong>Epigenetics</strong> 2011;6:798e803.[74] Arai JA, Feig LA. Long-last<strong>in</strong>g and transgenerational effects of an environmental enrichment on memoryformation. Bra<strong>in</strong> Res Bull 2011;85:30e5.[75] Watanabe H, Fukuoka H, Sugiyama T, Nagai Y, Ogasawara K, Yoshiike N. Dietary folate <strong>in</strong>take dur<strong>in</strong>g pregnancyand birth weight <strong>in</strong> Japan. Eur J Nutr 2008;47:341e7.[76] Park JH, Stoffers DA, Nicholls RD, Simmons RA. Development of type 2 diabetes follow<strong>in</strong>g <strong>in</strong>trauter<strong>in</strong>egrowth retardation <strong>in</strong> rats is associated with progressive epigenetic silenc<strong>in</strong>g of Pdx1. J Cl<strong>in</strong> Invest2008;118:2316e24.[77] Kucharski R, Maleszka J, Foret S, Maleszka R. Nutritional control of reproductive status <strong>in</strong> haneybees via DNAmethylation. <strong>Science</strong> 2008;319:1827e30.[78] Rimland B. Controversies <strong>in</strong> the Treatment of Autistic Children: Vitam<strong>in</strong> and Drug Therapy. J Child Neurol1998;3:68e72.[79] James SJ, Cutler P, Melnyk S, Jernigan S, Janak L, Gaylor DW, Neubrander JA. Metabolic biomarkers of<strong>in</strong>creased oxidative stress and impaired methylation capacity <strong>in</strong> children with autism. Am J Cl<strong>in</strong> Nutr2004;20:1611e7.[80] Moretti P, Sahoo T, Hyland K, Bottiglieri T, Peters S, del Gaudio D, et al. Cerebral folate deficiency withdevelopmental delay, autism, and response to fol<strong>in</strong>ic acid. Neurology 2005;64:1088e90.[81] Laird PW. Pr<strong>in</strong>ciples and challenges of genomewide DNA methylation analysis. Nat Rev Genet2005;11:191e203.[82] Varley KE, Mitra RD. Bisulfite Patch PCR enables multiplexed sequenc<strong>in</strong>g of promoter methylation acrosscancer samples. Genome Res 2010;20:1279e87.[83] Ohtsuki A, Kimura MT, M<strong>in</strong>oshima M, Suzuki T, Ikeda M, Bando T, Nagase H, Sh<strong>in</strong>ohara K, Sugiyama H.Synthesis and properties of PI polyamideeSAHA conjugate. Tetrahedron Lett 2009;50:7288e92.[84] Matsuda H, Fukuda N, Ueno T, Katakawa M, Wang X, Watanabe T, et al. Transcriptional <strong>in</strong>hibition ofprogressive renal disease by gene silenc<strong>in</strong>g pyrrole-imidazole polyamide target<strong>in</strong>g of the transform<strong>in</strong>g growthfactor-b1 promoter. Kidney Int 2011;79:46e56.[85] Coufal NG, Garcia-Perez JL, Peng GE, Yeo GW, Mu Y, Lovci MT, et al. L1 Retrotransposition <strong>in</strong> <strong>Human</strong> NeuralProgenitor Cells. Nature 2009;460:1127e31.[86] Muotri AR, Chu VT, Marchetto MC, Deng W, Moran JV, Gage FH. Somatic mosaicism <strong>in</strong> neuronal precursorcells mediated by L1 retrotransposition. Nature 2005;435:903e10.[87] Muotri AR, Marchetto MC, Coufal NG, Oefner R, Yeo G, Nakashima K, Gage FH. L1 retrotransposition <strong>in</strong>neurons is modulated by MeCP2. Nature 2010;468:903e10.[88] Muotri AR, Zhao C, Marchetto MC, Gage FH. Environmental Influence on L1 Retrotransposons <strong>in</strong> the AdultHippocampus. Hypoocumpus 2009;19:1002e7.[89] Lonetti G, Angelucci A, Morando L, Boggio EM, Giustetto M, Pizzorusso T. Early environmental enrichmentmoderates the behavioral and synaptic phenotype of MeCP2 null mice. Biol Psychiatry 2010;67:657e65.[90] Kerr B, Silva PA, Walz K, Young JI. Unconventional transcriptional response to environmental enrichment <strong>in</strong>a mouse model of Rett syndrome. PLoS One 2010;5:e11534.[91] Guy,.J, Gan J, Selfridge J, Cobb S, Bird A. Reversal of neurological defects <strong>in</strong> a mouse model of Rett syndrome.<strong>Science</strong> 2007;315:1143e7.203


This page <strong>in</strong>tentionally left blank


CHAPTER 11Epigenetic Basis ofAutoimmune Disorders <strong>in</strong><strong>Human</strong>sBiola M. Javierre, Lorenzo De La Rica, Esteban BallestarBellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, Barcelona,Spa<strong>in</strong>CHAPTER OUTLINE11.1 Immunity and Autoimmunity 20511.2 Epigenetic Deregulation <strong>in</strong>Autoimmunity 20611.2.1 Epigenetic Deregulation <strong>in</strong>SLE 207DNA Methylation ProfileAlteration <strong>in</strong> Systemic LupusErythematosus 208Histone Modification ProfileAlteration <strong>in</strong> Systemic LupusErythematosus 21011.2.2 Epigenetic Deregulation <strong>in</strong>RA 210DNA Methylation Alterations <strong>in</strong>Rheumatoid Arthritis 210Histone Modification ProfileAlteration <strong>in</strong> RheumatoidArthritis 21211.2.3 Epigenetic Deregulation <strong>in</strong>Multiple Sclerosis 21311.2.4 Epigenetic Deregulation <strong>in</strong>Sjögren’s Syndrome 21311.2.5 Epigenetic Deregulation <strong>in</strong>Vitiligo 21411.2.6 Epigenetic Deregulation <strong>in</strong>Progressive SystemicSclerosis 21411.2.7 Epigenetic Deregulation <strong>in</strong>Psoriasis 21411.2.8 Epigenetic Deregulation <strong>in</strong>Primary Biliary Cirrhosis 21511.2.9 Epigenetic Deregulation <strong>in</strong>Thyroid <strong>Disease</strong>s 21511.2.10 Epigenetic Deregulation <strong>in</strong>Inflammatory Bowel<strong>Disease</strong>s: Crohn’s <strong>Disease</strong>and Ulcerative Colitis 21611.2.11 Epigenetic Deregulation <strong>in</strong>Other Organ-SpecificAutoimmune Disorders 21711.3 Conclusions 217References 21820511.1 IMMUNITY AND AUTOIMMUNITYThe term immunity refers to the group of mechanisms developed by multicellular organismsto defend themselves aga<strong>in</strong>st potentially harmful agents. The development of an immunesystem occurred dur<strong>in</strong>g the evolution from unicellular to multicellular organisms. Autoimmunereactions emerged <strong>in</strong> parallel with the <strong>in</strong>creas<strong>in</strong>g complexity of the immune system torecognize and elim<strong>in</strong>ate pathogenic elements.T. Tollefsbol (Ed): <strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>. DOI: 10.1016/B978-0-12-388415-2.00011-1Copyright Ó 2012 Elsevier Inc. All rights reserved.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>To achieve specific recognition of harmful agents, it is essential first to identify their ownorganic components as someth<strong>in</strong>g <strong>in</strong>nocuous; <strong>in</strong> other words, the immune system must beautotolerant. When the f<strong>in</strong>e balance between recognition of self-components and defenseaga<strong>in</strong>st foreign agents is broken the immune system can react aga<strong>in</strong>st the body’s owncomponents, <strong>in</strong>duc<strong>in</strong>g cell destruction. If the immune system recognizes a particularcomponent located <strong>in</strong> a specific organ as harmful, and reacts solely aga<strong>in</strong>st this, it gives rise toa process of organ-specific autoimmunity. Examples <strong>in</strong>clude diabetes mellitus type 1, primarybiliary cirrhosis, Hashimoto’s thyroiditis, Grave’s disease, and celiac disease (Table 11.1).Conversely, if the reaction affects various organs and systems it is classified as systemic autoimmunity.This group of disorders <strong>in</strong>cludes systemic lupus erythematosus, rheumatoidarthritis, Sjögren’s syndrome, scleroderma, and psoriasis among others (Table 11.1).20611.2 EPIGENETIC DEREGULATION IN AUTOIMMUNITY<strong>Epigenetics</strong> is one of the most rapidly develop<strong>in</strong>g areas <strong>in</strong> the fields of molecular biology andbiomedic<strong>in</strong>e. This discipl<strong>in</strong>e has contributed to our understand<strong>in</strong>g of key aspects of cellbiology and the pathogenesis of a variety of diseases, among which cancer is the most widely<strong>in</strong>vestigated [1].<strong>Epigenetics</strong> is generally def<strong>in</strong>ed as the scientific discipl<strong>in</strong>e that studies the reversible andpotentially heritable changes <strong>in</strong> gene expression that do not affect the DNA sequence [2].Epigenetic mechanisms can also be described as those that register, signal, or perpetuate geneactivity states and <strong>in</strong>volve the chemical modification of chromat<strong>in</strong>. <strong>Epigenetics</strong> ma<strong>in</strong>ly focuseson two groups of chemical modifications: DNA methylation and histone post-translationalmodifications. Both groups of modifications have direct and/or <strong>in</strong>direct effects on geneexpression and nuclear structure, and ultimately, determ<strong>in</strong>e cell identity.There is a wide spectrum of critical biological phenomena regulated by epigenetic mechanisms.Epigenetic modifications are implicated <strong>in</strong> cell differentiation, cell cycle, apoptosis andsignal<strong>in</strong>g processes and are able to couple external signals to f<strong>in</strong>e regulation of gene expression.TABLE 11.1 Autoimmune Disorders: Types and Ma<strong>in</strong> Affected OrgansType <strong>Disease</strong> Ma<strong>in</strong> Affected Organ(s) ReferencesSystemic Systemic lupusSk<strong>in</strong>, jo<strong>in</strong>ts, heart, bra<strong>in</strong>, [4,5]erythematosuskidneys, otherRheumatoid arthritis Jo<strong>in</strong>ts, sk<strong>in</strong>, lungs, heart and [37]blood vessels, otherSjögren’s syndrome Salivary glands, tear glands, [85]jo<strong>in</strong>tsScleroderma Sk<strong>in</strong>, <strong>in</strong>test<strong>in</strong>e, lung [93]Psoriasis Sk<strong>in</strong>, jo<strong>in</strong>ts [97]Crohn’s disease Gastro<strong>in</strong>test<strong>in</strong>al tract, sk<strong>in</strong>, [113]jo<strong>in</strong>ts, eyeUlcerative colitis Gastro<strong>in</strong>test<strong>in</strong>al tract, eye, [113]muscle, sk<strong>in</strong>Ankylos<strong>in</strong>g spondylitis Sp<strong>in</strong>al and sacroiliac jo<strong>in</strong>ts, [137]eye, lung, heartOrgan-specific Multiple sclerosis Bra<strong>in</strong>, sp<strong>in</strong>al cord (CNS) [75]Vitiligo Sk<strong>in</strong> [88]Primary biliary cirrhosis Liver [103]Hashimoto’s thyroiditis Thyroid [110]Grave’s disease Thyroid [111]Diabetes mellitus type 1 Pancreatic islet cells [135]Celiac disease Gastro<strong>in</strong>test<strong>in</strong>al tract [136]


CHAPTER 11Epigenetic Basis of Autoimmune Disorders <strong>in</strong> <strong>Human</strong>sGiven the wide spectrum of processes where epigenetic regulation participates, deregulation isassociated with a wide variety of diseases, <strong>in</strong>clud<strong>in</strong>g cancer and genetically complex diseaseslike autoimmune disorders.In recent years, some laboratories have begun to elucidate the relevance of epigenetic alterations<strong>in</strong> autoimmune disorders. Research <strong>in</strong> epigenetic changes has ma<strong>in</strong>ly focused on autoimmunerheumatic disorders, primarily <strong>in</strong> systemic lupus erythematosus (SLE) and rheumatoid arthritis(RA). Results from these studies highlight the importance of <strong>in</strong>vest<strong>in</strong>g further efforts to identifythe full range of epigenetic alterations, different cell types <strong>in</strong>volved and to explore the potentialof these changes as targets for therapy. This chapter will summarize the most important aspectsof our knowledge about the <strong>in</strong>fluence of epigenetic deregulation <strong>in</strong> human autoimmunedisorders (Table 11.2) with special emphasis <strong>in</strong> SLE and RA.11.2.1 Epigenetic Deregulation <strong>in</strong> SLESLE is a systemic and chronic autoimmune disorder characterized by the production of nonorgan-specificautoantibodies, multisystem <strong>in</strong>flammation and damage to multiple organs [3].SLE patients experience multiple and systemic cl<strong>in</strong>ical manifestations and unpredictableTABLE 11.2 Concordance Rates <strong>in</strong> Monozygotic Tw<strong>in</strong>s for Different AutoimmuneDisorders as well as Some Examples of Epigenetic Deregulation Events <strong>in</strong>these Disorders<strong>Disease</strong>Systemic lupuserythematosusMZ CR*Examples of Epigenetic DeregulationEvents25% [6] e DNMT1 down-regulation [21]e Global DNA hypomethyaltion [20]e Focal hypomethylation (PRF1, CSF3R,TNFSF5, IFNGR2) [25,29,31]e Global histone H3 & H4hypoacetylation [35]e Focal hypoacetylation (IL-10) [33]Rheumatoid arthritis 15% [6] e Global DNA hypomethylation [44]e Focal hypomethylation (LINE-1,MAPK13, MET, hsa-mir-203) [45,46]e Focal hypermethylation (DR3) [49]Multiple sclerosis 25% [76] e Focal hypomethylation (PAD2) [81]Sjögren’s syndrome e Focal hypermethylation (BP230) [86]Scleroderma e DNMT1 overexpression [94]e Global DNA hypomethyaltion [19]e Focal hypermethylation (FL1) [95]Psoriasis 67% [97] e Focal hypomethylation (SHP1, p15,p16, p21) [98,99,101]e Focal hypermethylation (p14, p16)[100,102]Primary biliary cirrhosis 60% [6]Autoimmune thyroiddiseases30e50% [110,111] e Skewed X chromosome <strong>in</strong>activation[112]Crohn’s disease 60% [113]Ulcerative colitis 6% [113] e Global hypomethylation [114]e Focal hypermethylation (p14, p16,PAR2, MDR1, CDH1) [115,116,119e121]Diabetes mellitus type 1 21e70% [6]Celiac disease 75% [6]Ankylos<strong>in</strong>g spondylitis 50% [6]207*MZ, monozygotic; CR, concordance rate.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>exacerbations and relapses. These ma<strong>in</strong>ly affect the sk<strong>in</strong> and jo<strong>in</strong>ts, although there are alsocardiac, pulmonary, renal, neuropsychiatric, hematological, and reproductive alterations [4,5].The mechanisms responsible for the breakdown of immune tolerance to self-components andautoantibody production rema<strong>in</strong> unknown, however alterations <strong>in</strong> apoptosis, cytok<strong>in</strong>e levels,signal<strong>in</strong>g pathways, and immune cell behavior have been characterized <strong>in</strong> SLE patients [4].208SLE is characterized by a complex etiopathology, <strong>in</strong>clud<strong>in</strong>g not only genetic but also environmentalfactors, such as changes <strong>in</strong> hormone levels, viral <strong>in</strong>fection or exposure to chemicals.Recently, it has been proposed that epigenetic alterations <strong>in</strong>duced by the environment, andtherefore mysregulation of associated genes, could trigger autoimmunity when occurr<strong>in</strong>g <strong>in</strong>specific genetic backgrounds [3,4]. The observed concordance rates for monozygotic anddizygotic tw<strong>in</strong>s (around 25% and 2%, respectively), and the high heritability of SLE (morethan 66%), underscores the key <strong>in</strong>fluence of genetic factors <strong>in</strong> this autoimmune disorder [6,7].Currently, a variety of susceptibility genes for SLE, some of which are common to otherautoimmune disorders, have been identified [3,8,9]. In addition, different environmentalfactors are also key to the onset and the progression of SLE disease. Much evidence supportsthis contribution, <strong>in</strong>clud<strong>in</strong>g partial concordance between monozygotic tw<strong>in</strong>s and the <strong>in</strong>ductionof lupus-like syndromes follow<strong>in</strong>g adm<strong>in</strong>istration of certa<strong>in</strong> drugs. More than onehundred chemical substances, many of which <strong>in</strong>fluence the activity of epigenetic modifiers,have the ability to <strong>in</strong>duce SLE symptoms after long-term exposure. One clear example is that of5-azacytid<strong>in</strong>e and related compounds. 5-Azacytid<strong>in</strong>e is a chemical analog of cytid<strong>in</strong>e and is<strong>in</strong>corporated <strong>in</strong>to the DNA as cytos<strong>in</strong>e dur<strong>in</strong>g replication. Its demethylat<strong>in</strong>g properties arebased on the formation of stable complexes between DNA methyltransferases and DNA, wherethe analog is <strong>in</strong>corporated [10]. 5-Azacytid<strong>in</strong>e, also known under the commercial name ofVidaza Ò , has been used therapeutically s<strong>in</strong>ce 2004 to treat myelodysplastic syndrome andpatients receiv<strong>in</strong>g such treatment have frequently reported to develop lupus-like syndrome[11]. This autoimmune response has also been studied <strong>in</strong> mice. In both cases, lupus-likesymptoms disappear when the treatment is discont<strong>in</strong>ued. On the other hand, the <strong>in</strong> vitrotreatment of human or mur<strong>in</strong>e CD4þ T lymphocytes with 5-azacytid<strong>in</strong>e, or other demethylat<strong>in</strong>gagents, generates autoreactivity and the direct <strong>in</strong>jection of these treated cells <strong>in</strong>to healthymice <strong>in</strong>duces an autoimmune response with characteristics similar to those of SLE [12e18].DNA METHYLATION PROFILE ALTERATION IN SYSTEMIC LUPUSERYTHEMATOSUSEpigenetic dysregulation <strong>in</strong> SLE has been observed at the DNA methylation and histonemodification levels. In the particular case of DNA methylation, CD4 þ T lymphocytes of SLEpatients are characterized by a significant reduction <strong>in</strong> the total content of 5-methylcytos<strong>in</strong>eand the symptomatology is directly correlated with the loss of this epigenetic mark [19,20].DNA methyltransferases were early reported to be down-regulated T cells from lupus patients[21]. Alterations <strong>in</strong> DNA methylation levels have also been described <strong>in</strong> SLE mouse models.For example, T cells obta<strong>in</strong>ed from the mur<strong>in</strong>e model MRL/lpr are also characterized by lowerlevels of DNA methylation and reduced DNMT1 expression than control mice and this isdirectly correlated with ag<strong>in</strong>g and SLE progression [22,23].Global hypomethylation can potentially have different consequences, <strong>in</strong>clud<strong>in</strong>g altered geneexpression, erasure of impr<strong>in</strong>t<strong>in</strong>g signature, and reactivation of endoparasitic sequences,ultimately contribut<strong>in</strong>g to the loss of autotolerance as well as to SLE development. Alterations<strong>in</strong> DNA methylation occur at repetitive sequences and at gene promoters (Figure 11.1). Withrespect to the repetitive elements, a global decrease <strong>in</strong> the content of 5-methylcytos<strong>in</strong>e suggestshypomethylation <strong>in</strong> repetitive elements that are the major contributors of CpG d<strong>in</strong>ucleotidesto the genome 24. However, to date there is little <strong>in</strong>formation on the specific repetitiveelements that are affected. Recently, it has been demonstrated that the 18S and 28S regions ofthe ribosomal RNA genes that are present <strong>in</strong> the genome <strong>in</strong> several hundred copies, undergo


CHAPTER 11Epigenetic Basis of Autoimmune Disorders <strong>in</strong> <strong>Human</strong>sDNADEMETHYLATINGDRUGAUTOREACTIVITY[DNMT1][DNMTGENOMICINSTABILITY ?GLOBALHYPOMETHYLATIONGENE PROMOTERHYPOMETHYLATIONGENEOVEREXPRESSION18S & 28SrRNAREPEATSCD40LGCD70CSF3RIFNGR2IL4 & IL6LCN2LFA-1PRF1MMP14,…HDACINHIBITORSNORMAL GENEEXPRESSIONPROFILEGLOBALH3 & H4 HYPOACETYLATIONGENE EXPRESSION ALTEREDFIGURE 11.1A scheme depict<strong>in</strong>g some of the best-known epigenetic deregulation events <strong>in</strong> blood cells from SLE patients: changes <strong>in</strong> DNA methylation and histonemodifications, as well as the effects of epigenetic drugs.hypomethylation <strong>in</strong> white blood cells of SLE patients [25]. Hypomethylation at the ribosomalRNA gene repeats correlate with the overexpression of the pre-RNA and the 18S RNA. Thisf<strong>in</strong>d<strong>in</strong>g could potentially be associated with <strong>in</strong>creased production of ribosomal particles <strong>in</strong>SLE, perhaps associated with the production of the autoantibodies aga<strong>in</strong>st these particles thatare frequently detected <strong>in</strong> serum of SLE patients [26, 27].209With respect to promoters, various studies have reported the occurrence of hypomethylation atthe regulatory region of several genes <strong>in</strong> SLE. The majority of these studies have addressed thisissue by us<strong>in</strong>g candidate gene analysis. More recently, high-throughput analysis of monozygotictw<strong>in</strong>s discordant for SLE has led to the identification of a larger set of genes, as well asdissect the contribution of DNA methylation changes to this disease [25].Among the identified gene promoters, several of them are implicated <strong>in</strong> immune processes suchas cellular death (PRF1), B-cell co-stimulation (TNFSF7 and TNFSF5), immune synapse (ITGALand CSF3R) and <strong>in</strong>terleuk<strong>in</strong> signal<strong>in</strong>g (IL4 and IL6). One example is represented by PRF1, thegene that encodes perfor<strong>in</strong>, <strong>in</strong>volved <strong>in</strong> cellular death through generation of a pore by <strong>in</strong>sertion<strong>in</strong>to the cytoplasmic membrane [28]. InCD4 þ T lymphocytes of SLE patients, the promoterregion of the PRF1 gene is hypomethylated, the gene overexpressed and there is a consequent<strong>in</strong>crease <strong>in</strong> monocyte and macrophage death [29,30]. The treatment of healthy human CD4 þcells with DNA demethylat<strong>in</strong>g drugs <strong>in</strong>duces autoreactivity <strong>in</strong> vitro and monocyte kill<strong>in</strong>g ability.On the other hand, the use of concanamyc<strong>in</strong> A, a perfor<strong>in</strong> <strong>in</strong>hibitor, reduces monocyte kill<strong>in</strong>g <strong>in</strong>SLE [29]. Another example is the B-cell costimulat<strong>in</strong>g prote<strong>in</strong> CD40LG, also called CD154 orTNFSF5. TNFSF5 is encoded on the X-chromosome and the two alleles are methylated <strong>in</strong> healthywomen, whereas <strong>in</strong> men the s<strong>in</strong>gle copy is unmethylated. In CD4þ lymphocytes of women withSLE, the TNFSF5 promoter tends to become hypomethylated and, for this reason, the costimulat<strong>in</strong>gmolecule is overexpressed [31,32]. This specific epigenetic dysregulation could partiallyexpla<strong>in</strong> the higher <strong>in</strong>cidence of SLE <strong>in</strong> women than <strong>in</strong> men. These are just some examples ofgenes characterized by DNA methylation alteration <strong>in</strong> SLE. Undoubtedly, many other genes


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>rema<strong>in</strong> to be discovered that develop epigenetic alterations <strong>in</strong> SLE. In-depth studies based onfractionated cell populations and more powerful high-throughput technologies will provide uswith a better knowledge of the role of DNA methylation changes <strong>in</strong> SLE.HISTONE MODIFICATION PROFILE ALTERATION IN SYSTEMIC LUPUSERYTHEMATOSUSOur knowledge about the alterations <strong>in</strong> the histone modification patterns <strong>in</strong> SLE is also farfrom complete. Most of the evidence about the role of changes <strong>in</strong> histone modifications <strong>in</strong> SLEcomes from the use of epigenetic drugs. Specifically, the use of histone deacetylase (HDAC)<strong>in</strong>hibitors suggests that deacetylation is <strong>in</strong>volved <strong>in</strong> the skewed expression of certa<strong>in</strong> genes thatare associated with the disease. For <strong>in</strong>stance, SLE T-helper cells exhibit <strong>in</strong>creased and prolongedexpression of cell-surface CD40 ligand (CD154), spontaneously overproduce <strong>in</strong>terleuk<strong>in</strong>-10(IL-10), but underproduce <strong>in</strong>terferon-gamma (IFN-g). The histone deacetylase <strong>in</strong>hibitortrichostat<strong>in</strong> A (TSA) significantly reverses this skewed expression of these gene products [33](Figure 11.1). It is likely that this reversion is the result of modification of the histone acetylationstatus, although alteration of the acetylation levels of regulatory prote<strong>in</strong>s cannot bediscarded. This result not only suggests that histone acetylation might account for this aberrantexpression but also that this pharmacological agent may be a candidate for the treatment ofthis autoimmune disease.210Little <strong>in</strong>formation on the histone acetylation changes associated with SLE is available. Most ofthe <strong>in</strong>formation comes from the MRL/lpr mouse model, where histone hypoacetylation hasbeen demonstrated [34]. InCD4 þ T lymphocytes of SLE patients with active disease are alsocharacterized by global histone H3 and H4 hypoacetylation [35] (Figure 11.1). In contrast,monocytes from SLE patients have been reported to undergo histone H4 hyperacetylation atgene promoters and this alteration associates with aberrant overexpression of associated genes.Interest<strong>in</strong>gly, the 179 identified genes are characterized by the presence of potential IRF1b<strong>in</strong>d<strong>in</strong>g sites with<strong>in</strong> the 5 Kb upstream region and the IFNa treatment <strong>in</strong>creases geneexpression and histone acetylation [36]. The use of high-throughput approaches and analysisof specific cell types and more histone modification marks will surely lead to detailed <strong>in</strong>formationabout genomic sites that undergo histone modification changes <strong>in</strong> SLE.11.2.2 Epigenetic Deregulation <strong>in</strong> RARA is a systemic autoimmune disease characterized by synovial hyperplasia and jo<strong>in</strong>t<strong>in</strong>flammation and progressive destruction [37]. In the affected jo<strong>in</strong>t, there is an <strong>in</strong>flammatorymicroenvironment <strong>in</strong>volv<strong>in</strong>g many immune cells [37]. These <strong>in</strong>clude synovial cells, which arehyperactivated and hyper-reactive due to high concentrations of pro<strong>in</strong>flammatory cytok<strong>in</strong>es.In addition, there is bone and cartilage destruction by the ma<strong>in</strong> effector cell types, osteoclastsand rheumatoid arthritis synovial fibroblasts (RASF), respectively [38].RASFs constitute one of the cell types <strong>in</strong>volved <strong>in</strong> RA pathogenesis as well as one of the moreextensively studied. RASFs are more aggressive than their normal counterparts [39]. Theyoverexpress metalloprote<strong>in</strong>ases (MMPs) and cytok<strong>in</strong>es [40] and show tumoral behavior(<strong>in</strong>vasiveness [41], resistance to apoptosis [42], and anchorage-<strong>in</strong>dependent growth [43]).DNA METHYLATION ALTERATIONS IN RHEUMATOID ARTHRITISSynovial fibroblasts are the best-characterized cells for epigenetic alterations <strong>in</strong> RA. In theearly 1990s, global DNA hypomethylation was reported to occur <strong>in</strong> blood, synovial mononuclearcells, and synovial tissue of RA <strong>in</strong>dividuals [44] (Figure 11.2). In RASFs, hypomethylationwas associated with aberrant overexpression of retrotransposable L<strong>in</strong>e 1,affect<strong>in</strong>g the expression of other genes [45]. More recently, met proto-oncogene (MET),p38delta MAP k<strong>in</strong>ase (MAPK13), and galect<strong>in</strong> 3 b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> (LGALS3BP) have beenshown to be overexpressed, contribut<strong>in</strong>g to the aggressive phenotype of these effector cell


CHAPTER 11Epigenetic Basis of Autoimmune Disorders <strong>in</strong> <strong>Human</strong>sRA DNA METHYLATIONHDACi EFFECT IN RACELL ADHESIONEPHRINBEPHRINBIL6T-CELLGlobal DNAHypomethylationRASFAPOPTOSISDR3METMAPK13miR203IL6MMP1INFLAMMATIONHDACiT-CELLRASFNr AUTO-REACTIVE Th1Nr TregNr Th2• APOPTOSIS• ANGIOGENESIS• PROLIFERATION• INFLAMMATION• MATRIXDEGRADATIONMATRIXDEGRADATIONMONOCYTEOSTEOCLASTFIGURE 11.2Epigenetic changes <strong>in</strong> RA. There is global DNA hypomethylation <strong>in</strong> the context of T-cells [20] and RASFs [44]. Examples of gene promoters hypomethylated andoverexpressed <strong>in</strong> T-cells are EPHRINB [51] (cell adhesion molecule) and IL6 [52] (pro<strong>in</strong>flammatory cytok<strong>in</strong>e). LINE1 repetitive sequences [46] and hsa-mir-203[50] are hypomethylated <strong>in</strong> RASFs. Hsa-miR-203 deregulation leads to MMP1 (matrix degradat<strong>in</strong>g enzyme) and IL6 overexpression, while LINE1 deregulationcauses MET and MAPK13 up-regulation. There is also specific promoter hypermethylation, as it happens <strong>in</strong> DR3 (underl<strong>in</strong>ed) [49], and is related to higher resistanceto apoptosis. A pro<strong>in</strong>flammatory microenviroment is developed with<strong>in</strong> the affected jo<strong>in</strong>t. HDACi have shown several <strong>in</strong>hibitory effects <strong>in</strong> RA diseaseeffector cell types. They are able to decrease the number of autoreactive Th1 while <strong>in</strong>creas<strong>in</strong>g Treg and Th2, balanc<strong>in</strong>g T cells to a more protective status [68e71].In RASFs, HDACi are able to <strong>in</strong>duce apoptosis [60,61], <strong>in</strong>hibit proliferation, and decrease secretion of <strong>in</strong>flammatory cytok<strong>in</strong>es, matrix degradat<strong>in</strong>g prote<strong>in</strong>s [56] aswell as angiogenic factors [67]. It also achieves lower bone degradation by <strong>in</strong>hibit<strong>in</strong>g osteoclastogenesis [74]. This figure is reproduced <strong>in</strong> the color plate section.211type [46]. Neidhart and Gay’s team has identified that RASF display lower levels of DNMT1than their healthy counterparts. They have also shown that these alterations <strong>in</strong> DNMT1 andglobal DNA methylation levels associate with the aggressive phenotype of RASFs. Moreover,synovial fibroblasts obta<strong>in</strong>ed from healthy donors treated with DNA hypomethylat<strong>in</strong>g drugs,like 5-aza-2-desoxicytid<strong>in</strong>e, become as aggressive as RASFs [47].RASFs also display specific promoter hypermethylation, like cancer cells [48,49] (Figure 11.2).For <strong>in</strong>stance, the promoter of the tumor necrosis factor receptor superfamily, member 25 gene(TNFRSF25), also known as death receptor 3 (DR3), is hypermethylated, and consequentlydown-regulated. This could expla<strong>in</strong> the <strong>in</strong>creased resistance to apoptosis that this cell typeexhibits [49]. Some microRNAs are also deregulated <strong>in</strong> RASF by aberrant methylation changesat their promoters. As an example, hsa-miR-203 is up-regulated <strong>in</strong> RASFs relative to OASFs, andleads to an <strong>in</strong>crease <strong>in</strong> the production of MMP-1 and IL-6 [50].Blood cells are also reported to exhibit DNA methylation changes <strong>in</strong> RA. One example isEphr<strong>in</strong> B1 (EFNB1), which codes for a membrane prote<strong>in</strong> <strong>in</strong>volved <strong>in</strong> cell adhesion and<strong>in</strong>flammation process signal<strong>in</strong>g. EFNB1 mRNA and prote<strong>in</strong> levels are up-regulated <strong>in</strong> bloodand synovial T lymphocytes ow<strong>in</strong>g to the hypomethylation of its promoter [51] (Figure 11.2).Another example is the multifunctional <strong>in</strong>terleuk<strong>in</strong>-6 (IL-6). Loss of methylation of a s<strong>in</strong>gleCpG at the promoter of IL-6 results <strong>in</strong> overexpression, that probably results <strong>in</strong> immune cellhyperactivation [52].In RA CD4 þ T cells, as well as <strong>in</strong> RASFs, there is global DNA hypomethylation and loweractivity levels of DNMTs [20].There is a subset of “senescent” CD4 þ CD28 e T cells that are more frequent <strong>in</strong> the elderly andRA patients, and that, among other abnormalities, are autoreactive. In this subset, ERK and


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>JNK pathways are decreased and DNMT1 and DNMT3 down-regulated. These decreased levelsof these DNMTs are associated with demethylation of some gene promoters and subsequentprote<strong>in</strong> overexpression. Some examples are CD70, perfor<strong>in</strong> (PRF1), and KIR2DL4, whichcould contribute to the <strong>in</strong>flammatory phenotype [53].It has been speculated that there is an imbalance of helper lymphocyte differentiation <strong>in</strong> RA. Inthis disease, peripheral CD4 þ T cells are characterized by FOXP3 promoter hypermethylation.On the other hand, synovial CD4 þ T cells experience FOXP3 and IFNG promoter demethylation,possibly <strong>in</strong>dicat<strong>in</strong>g a higher representation of regulatory T and T helper 1 cells <strong>in</strong> theaffected jo<strong>in</strong>ts [54].HISTONE MODIFICATION PROFILE ALTERATION IN RHEUMATOID ARTHRITISIn RA, there is also evidence of a potential role of histone modifications. Although no specifichistone modification alterations have been described <strong>in</strong> this autoimmune disorder, theimportant role of this epigenetic mechanism is demonstrated by research based on HDAC<strong>in</strong>hibitors (Figure 11.2).In RA, synovial fibroblasts show higher levels of HDAC1, and these are positively regulated bythe concentration of TNF-alpha [55]. The depletion of HDAC1 and HDAC2 results <strong>in</strong>decreased cell proliferation and <strong>in</strong>hibition of certa<strong>in</strong> TNF-alpha cytok<strong>in</strong>es such as MMP1 [56].Nucleosomes located along the MMP1 promoter are hyperacetylated <strong>in</strong> RASFs, where this geneis overexpressed [57].212The first observation that HDACi could modulate the expression of RA-related genes wasmade <strong>in</strong> 2003 <strong>in</strong> an animal model of this autoimmune disorder. Adjuvant arthritis ratstreated with phenylbutyrate and trichostat<strong>in</strong> A (TSA) showed RA pathology suppressionand the isolated RASFs from these treated rats did not proliferate. This effect was due to theup-regulation of two CDK <strong>in</strong>hibitors (p16 INK4a and p21 WAF1/Cip1 ) and <strong>in</strong>hibition of TNFalpha,IL-1, andIL-6 <strong>in</strong> the affected jo<strong>in</strong>ts [58]. In 2004, systemic adm<strong>in</strong>istration of theHDACi FK228 succeeded <strong>in</strong> ameliorat<strong>in</strong>g synovial proliferation and jo<strong>in</strong>t destruction whenadm<strong>in</strong>istered to autoantibody-mediated arthritis mice by up-regulat<strong>in</strong>g the same CDK<strong>in</strong>hibitors [59]. It has also been shown that treatment of RASFs with HDACi <strong>in</strong>ducesapoptosis via Fas receptor and TRAIL [60,61]. Other HDACi, such as MS-275 or suberoylanilidehydroxamic acid (SAHA), have also demonstrated growth arrest activity, <strong>in</strong>hibitionof pro<strong>in</strong>flammatory cytok<strong>in</strong>es (TNF-alpha, IL-1beta) as well as down-regulation ofangiogenesis and MMP expression <strong>in</strong> synovial fibroblasts <strong>in</strong> several RA animal models andpatients [62e65]. The <strong>in</strong>hibition of HDACs, and thus, of MMPs, produces decreasedcartilage resorption [66].HDACi show strong anti-<strong>in</strong>flammatory effects <strong>in</strong> vivo that cannot only be expla<strong>in</strong>ed by theireffects on cell cycle and cell proliferation. These drugs are also <strong>in</strong>volved <strong>in</strong> the <strong>in</strong>hibition ofangiogenesis <strong>in</strong> the affected jo<strong>in</strong>t by down-regulat<strong>in</strong>g the hypoxia-<strong>in</strong>duced factors HIF-1a andVEGF, which play central roles <strong>in</strong> the angiogenic process [67].Apart from RASFs, the antiarthritic effects of HDACi have also been tested <strong>in</strong> T cells. In RApatients, regulatory T cells fail to suppress CD4 þ effector cells. TSA and valproic acid are able to<strong>in</strong>crease the function and number of FOXP3 express<strong>in</strong>g CD25 þ CD4 þ regulatory T lymphocytes,which are able to <strong>in</strong>hibit the proliferative response of effector T cells [68], lead<strong>in</strong>g tobetter cl<strong>in</strong>ical features [69].TSA also has the ability to suppress arthritis severity <strong>in</strong> collagen-<strong>in</strong>duced arthritis mice. This isachieved by <strong>in</strong>hibit<strong>in</strong>g autoantigen-specific T helper 1 cell proliferation and apoptosis<strong>in</strong>duction and also gives rise to less IFN-gamma release. On the other hand, it enhances Thelper 2 response by acetylat<strong>in</strong>g IL-4 promoter and thus its overexpression [70]. In conclusion,TSA is able to balance the relationship between T helper 1 and T helper 2 to produce


CHAPTER 11Epigenetic Basis of Autoimmune Disorders <strong>in</strong> <strong>Human</strong>sa protective status. Moreover, another HDACi, LAQ824, has previously been characterized bythe ability to regulate this balance <strong>in</strong> human healthy T lymphocytes [71].HDACi have also been shown to <strong>in</strong>duce osteoblast proliferation and maturation as well asaccelerat<strong>in</strong>g matrix m<strong>in</strong>eralization by regulat<strong>in</strong>g the expression of several genes, such as thosecod<strong>in</strong>g for growth factors and Wnt receptors [72,73].On the other hand, the HDACi FR901228 can <strong>in</strong>hibit osteoclastogenesis. It <strong>in</strong>hibits nucleartranslocation of NFATc1 (a crucial factor for osteoclastogenic differentiation) and boostsproduction of IFN-beta (an <strong>in</strong>hibitor of osteoclastogenesis). As a result, bone destruction issuppressed, and the cl<strong>in</strong>ical features of RA are improved [74].11.2.3 Epigenetic Deregulation <strong>in</strong> Multiple SclerosisMultiple sclerosis (MS) is a multisystemic <strong>in</strong>flammatory and chronic autoimmune disordercharacterized by demyel<strong>in</strong>ation and neurodegeneration of bra<strong>in</strong> and sp<strong>in</strong>al cord [75]. Itisa genetically complex disease, but its etiology depends on environmental factors <strong>in</strong>clud<strong>in</strong>gEpste<strong>in</strong>eBarr virus <strong>in</strong>fection. It is also thought that epigenetic dysregulation also participates<strong>in</strong> the onset of the disease driven by environmental factors. In fact, concordance rates ofaround 25% and 5% <strong>in</strong> monozygotic and dizygotic tw<strong>in</strong>s have been estimated [76]. Sunlightdeficiency, low vitam<strong>in</strong> D <strong>in</strong>gestion, radioactivity, geomagnetism, atmospheric contam<strong>in</strong>ants,tox<strong>in</strong>s, and nicot<strong>in</strong>e poison<strong>in</strong>g have been identified as environmental triggers of epigeneticderegulation <strong>in</strong> MS [77]. Various studies have demonstrated that some of these factors have theability to modify the epigenetic profile directly or <strong>in</strong>directly [78,79].Although the participation of epigenetic deregulation events <strong>in</strong> MS development has beenproposed, there is little experimental evidence. One key prote<strong>in</strong> <strong>in</strong> MS is myel<strong>in</strong> basic prote<strong>in</strong>(MBP), a major component of the myel<strong>in</strong> sheath of Schwann cells and oligodendrocytes <strong>in</strong> thenervous system. This prote<strong>in</strong> is citrullated by peptide arg<strong>in</strong><strong>in</strong>e deim<strong>in</strong>ase 2 (PAD2), generat<strong>in</strong>gloss of myel<strong>in</strong> <strong>in</strong>stability. In MS, the PAD2 promoter is hypomethylated and consequentlyPAD2 is overproduced and the myel<strong>in</strong> is more easily degraded [80,81]. Moreover, MBPcitrullation has the ability to <strong>in</strong>duce autocleavage, generat<strong>in</strong>g new potential autoantigens andallow<strong>in</strong>g molecular mimicry [82,83].213Recently, Baranz<strong>in</strong>i and collaborators studied the genome, the epigenome, and the transcriptomeof CD4 þ T cells from three pairs of monozygotic tw<strong>in</strong>s discordant for MS. Although,they did not detect any reproducible and significant difference at the DNA methylation profilebetween co-sibl<strong>in</strong>gs, this study represented the first high-throughput approach to <strong>in</strong>vestigateDNA methylation alterations <strong>in</strong> MS [84]. It is expected that future high-throughput studieswith larger sets of samples and perhaps us<strong>in</strong>g other sets of cell types will help to dissectepigenetic profiles specific to MS.11.2.4 Epigenetic Deregulation <strong>in</strong> Sjögren’s SyndromeSjögren’s syndrome (SjS) is a systemic autoimmune disease characterized by chronic <strong>in</strong>flammationof the exocr<strong>in</strong>e glands that produce saliva and tears [85].As <strong>in</strong> the previously described autoimmune disorders, SjS is a multifactorial disease <strong>in</strong> whichgenetic predisposition and environmental factors both feature. One report has describedBP230 promoter hypermethylation <strong>in</strong> labial salivary glands of SjS patients. BP230 is theepithelial splice isoform of the dyston<strong>in</strong> gene, a component of the plak<strong>in</strong> prote<strong>in</strong> family ofadhesion junction plaque molecules and it is <strong>in</strong>volved <strong>in</strong> basal lam<strong>in</strong>a anchorage. Thishypermethylation correlates with mRNA down-regulation, although paradoxically the prote<strong>in</strong>level is <strong>in</strong>creased [86]. The BP230 epigenetic deregulation could expla<strong>in</strong> the anchorage alterationsof salivary gland cells that characterize SjS patients. Also, <strong>in</strong> SjS patients’ salivary glands,up-regulation of two miRNAs has been detected and have been shown to be useful as markersof disease progression [87].


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>11.2.5 Epigenetic Deregulation <strong>in</strong> VitiligoVitiligo is a depigment<strong>in</strong>g autoimmune disorder characterized by melanocyte destruction andpatchy loss of sk<strong>in</strong> pigmentation. Unlike other autoimmune disorders, it equally affects bothsexes and all races, although it usually appears between the ages of 10 and 30 years [88].Interest<strong>in</strong>gly, vitiligo occurs <strong>in</strong> association with other autoimmune disorders <strong>in</strong>clud<strong>in</strong>g Addison’sdisease, autoimmune thyroiditis, SLE, RA, and psoriasis among others, highlight<strong>in</strong>g thatmechanisms of deregulation are shared between all these conditions [89,90]. The etiology ofvitiligo is a matter of a complex <strong>in</strong>teraction between genetic predisposition and epigeneticalteration [91]. Environmental factors are key regulators of the onset of vitiligo. In fact, sunburnand traumas are associated with the manifestation of this autoimmune disorder. There are fewdata about epigenetic deregulation events <strong>in</strong> this autoimmune disorder, although the use of theSmyth l<strong>in</strong>e (SL) chicken, an animal model for human vitiligo, has yielded some <strong>in</strong>formation.Specifically, <strong>in</strong> vivo 5-azacytid<strong>in</strong>e treatment of these chickens <strong>in</strong>duces the production of antibodiesaga<strong>in</strong>st the melanocyte-specific prote<strong>in</strong> TRP-1, sk<strong>in</strong> melanocyte depletion and consequentdepigmentation [92]. This study represents a clear example of how the adm<strong>in</strong>istration ofa DNA hypomethylat<strong>in</strong>g drug can <strong>in</strong>duce vitiligo disease <strong>in</strong> genetically predisposed <strong>in</strong>dividuals,re<strong>in</strong>forc<strong>in</strong>g the role of genetic and epigenetic <strong>in</strong>fluences on this autoimmune disorder.11.2.6 Epigenetic Deregulation <strong>in</strong> Progressive Systemic SclerosisProgressive systemic sclerosis (PSS), or scleroderma, is a rare disease characterized by excessivecollagen deposition, ma<strong>in</strong>ly <strong>in</strong> sk<strong>in</strong>, but also <strong>in</strong> other organs, and progressive vasculopathy. Itis considered an autoimmune disease because of the presence of autoantibodies, several ofwhich act aga<strong>in</strong>st nuclear epitopes. A greater <strong>in</strong>cidence <strong>in</strong> women and frequent autoimmunecomorbidities have been observed [93].214PSS is characterized by aberrant fibroblast activation <strong>in</strong> which there is greater deposition ofcollagen and lower secretion of MMPs, result<strong>in</strong>g <strong>in</strong> an elastic sk<strong>in</strong>. These fibroblasts ma<strong>in</strong>ta<strong>in</strong>their activated phenotype even when cultured <strong>in</strong> vitro, suggest<strong>in</strong>g that gene expression isepigenetically regulated. Specifically, PSS fibroblasts produce higher levels of some cytok<strong>in</strong>esand growth factors as well as lower levels of some MMPs. A correlation has been reportedbetween the scleroderma fibroblast phenotype and higher detected levels of DNMT1,emphasiz<strong>in</strong>g the key role of epigenetic deregulation <strong>in</strong> PSS [94]. For example, FL1, a transcriptionfactor that <strong>in</strong>hibits collagen production, undergoes hypermethylation <strong>in</strong> PSS fibroblastsand thus silenc<strong>in</strong>g, result<strong>in</strong>g <strong>in</strong> <strong>in</strong>creased production of collagen. This property can bereverted by treatment with epigenetic modifiers, <strong>in</strong>clud<strong>in</strong>g HDACi and DNA methylation<strong>in</strong>hibitors. These observations provide a bona fide example of how epigenetic mechanismsorchestrate the pathogenic phenotype of a cell <strong>in</strong>volved <strong>in</strong> an autoimmune disorder [95].T lymphocytes are another cell type <strong>in</strong>volved <strong>in</strong> the pathogenesis of this disease. In particular,this cell population displays lower levels of 5-methylcytos<strong>in</strong>e and down-regulation of themethyl-CpG b<strong>in</strong>d<strong>in</strong>g doma<strong>in</strong> prote<strong>in</strong> (MBD4) [19].On the other hand, the reported important role of skewed X-chromosome <strong>in</strong>activationmosaicism <strong>in</strong> the pathogenesis of scleroderma could expla<strong>in</strong> the great prevalence of PSS <strong>in</strong>females [96]. This geneedosage equilibration mechanism is also altered <strong>in</strong> autoimmunethyroid diseases (AITDs), as is expla<strong>in</strong>ed <strong>in</strong> more detail below.11.2.7 Epigenetic Deregulation <strong>in</strong> PsoriasisPsoriasis is a chronic systemic sk<strong>in</strong> disease characterized by precocious kerat<strong>in</strong>ocyte differentiationand hyperproliferation, triggered by autoimmune reactions.Psoriasis etiopathology consists of a complex comb<strong>in</strong>ation of genetic risk and epigeneticderegulation, which is reflected by the concordance rates for monozygotic and dizygotic tw<strong>in</strong>sfor this autoimmune disorder (around 67% and 15%, respectively) [97].


CHAPTER 11Epigenetic Basis of Autoimmune Disorders <strong>in</strong> <strong>Human</strong>sWith respect to the identification of epigenetic alterations <strong>in</strong> psoriasis, SHP-1 (also calledPTPN6) promoter 2 is hypomethylated <strong>in</strong> psoriatic sk<strong>in</strong> samples and the isoform II expressionlevels are <strong>in</strong>creased. Conversely, this promoter is hypermethylated <strong>in</strong> normal sk<strong>in</strong>, repress<strong>in</strong>ggene expression. This observation contrasts with the silenc<strong>in</strong>g of this gene observed <strong>in</strong> otherhyperproliferative syndromes, such as leukemias and lymphomas [98].Abnormal proliferative activity has been detected <strong>in</strong> the hematopoietic cells of patientssuffer<strong>in</strong>g psoriasis. The p16 INK4A tumor suppressor is a negative regulator of CDK4, a prote<strong>in</strong>that accelerates cell cycle progression. In hematopoietic cells obta<strong>in</strong>ed from psoriatic patients,p16 gene promoter is hypomethylated and p16 mRNA level is <strong>in</strong>creased. This overproductionresults <strong>in</strong> a reduced ability to form colonies <strong>in</strong> hematopoietic cells from psoriasis patients,show<strong>in</strong>g that bone marrow, and not only immunocytes, is dysfunctional <strong>in</strong> psoriatic patients[99]. Conversely, p16 is hypermethylated <strong>in</strong> epidermal samples from psoriatic patients [100].Consistent with this l<strong>in</strong>e of evidence, other research shows a lower proliferative potential ofhematopoietic cells <strong>in</strong> psoriatic patients, and is correlated with lower promoter methylation ofp15 and p21 genes, and thus higher expression levels [101].An <strong>in</strong>crease <strong>in</strong> DNA methylation has been reported <strong>in</strong> PBMCs from psoriatic patients as well as<strong>in</strong> psoriatic sk<strong>in</strong> lesions. This methylation is correlated positively with PASI (Psoriasis Area andSeverity Index) scores. Specifically, DNMT1 is up-regulated while the MBD2 and MeCP2 aredown-produced, <strong>in</strong>dicat<strong>in</strong>g that the methylation mach<strong>in</strong>ery of psoriatic patients is altered.Another example is that of the p14 gene promoter. This gene, a homologues of p16 INK4A ,ishypermethylated and its expression is consequently downregulated [102].11.2.8 Epigenetic Deregulation <strong>in</strong> Primary Biliary CirrhosisPrimary biliary cirrhosis (PBC) is a chronic and progressive organic-specific autoimmunedisease of the liver that it is characterized by the destruction of small-to-medium bile ducts,cholestasis fibrosis, and cirrhosis [103].The etiology rema<strong>in</strong>s unknown, although it is clearly related to a comb<strong>in</strong>ation of geneticpredisposition and environmental stimulation, as <strong>in</strong>dicated by the 60% concordance formonozygotic tw<strong>in</strong>s and the strik<strong>in</strong>g predom<strong>in</strong>ance <strong>in</strong> women (female:male ratio of 10:1)[104]. Various studies have demonstrated that familiar <strong>in</strong>cidence of PBC, recurrent ur<strong>in</strong>arytract <strong>in</strong>fections, use of nail polish, and hormone treatment are risk factors for PBC development[105]. Some of these external factors have the ability to <strong>in</strong>duce epigenetic changes.Despite the lack of studies, it is likely that dysregulation of epigenetic factors is implicated <strong>in</strong>PBC. For example, two X-chromosome genes are known to have lower expression <strong>in</strong> peripheralblood cells of PBC tw<strong>in</strong>s <strong>in</strong> comparison with their healthy counterparts [106]. These genes areCLIC2, which encodes a chloride channel <strong>in</strong>volved <strong>in</strong> stabilization of cell membrane potential,transepithelial transport, ma<strong>in</strong>tenance of <strong>in</strong>tracellular pH, and regulation of cell volume[107,108], and PIN4, a member of the parvul<strong>in</strong> subfamily of the peptidyl-prolyl cis/transisomerases [109]. Interest<strong>in</strong>gly, promoter methylation analysis revealed some dysregulationevents but not always correlated with the expression behavior [106]. This study emphasizes therelationship between sex, epigenetics, and PBC.21511.2.9 Epigenetic Deregulation <strong>in</strong> Thyroid <strong>Disease</strong>sAutoimmune thyroid diseases (AITDs) are the group of conditions <strong>in</strong> which the autoimmuneattack of the thyroid takes place by <strong>in</strong>filtration of lymphocytes of the glandule. It is an organspecificautoimmune disease that affects ma<strong>in</strong>ly women. This attack results <strong>in</strong> two oppositecl<strong>in</strong>ical outcomes: Hashimoto’s thyroiditis (HT) and Grave’s disease (GD). HT is a hypothyroidism<strong>in</strong> which there is apoptosis of thyroid cells, while GD <strong>in</strong>volves hyperactivation of thethyroid due to TSH receptor-stimulat<strong>in</strong>g antibodies caus<strong>in</strong>g hyperthyroidism.Many susceptibility loci have been identified <strong>in</strong>clud<strong>in</strong>g HLA-DR, CTLA-4, CD40, FOXP3,CD25, TSHR, and others. However, as with many other autoimmune diseases, genetics cannot


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>expla<strong>in</strong> all the factors <strong>in</strong>volved <strong>in</strong> AITDs. The concordance rates between monozygotic tw<strong>in</strong>sare around 30e50% [110,111], imply<strong>in</strong>g that other mechanisms such as epigenetic changescould trigger the onset of the disease.A mechanism of skewed X chromosome <strong>in</strong>activation (XCI) has been proposed <strong>in</strong> AITD. Underphysiological conditions, females have <strong>in</strong> their cells either the maternal or paternal X-chromosomeactive, at random and at a ratio of 50:50. When this ratio is modified (for example20:80) there is a process of skewed XCI. Much epigenetic mach<strong>in</strong>ery is <strong>in</strong>volved <strong>in</strong> the process ofXCI <strong>in</strong> order to methylate CpG on DNA, lys<strong>in</strong>es of histones, and deacetylate the silenced region.Mechanistically, the expression of antigens may not be sufficiently tolerated <strong>in</strong> some cells. Someself-antigens on one X-chromosome may not be present <strong>in</strong> the thymus or <strong>in</strong> other tolerancerelatedtissues, but may be highly expressed <strong>in</strong> other tissues, trigger<strong>in</strong>g immune responses [112].21611.2.10 Epigenetic Deregulation <strong>in</strong> Inflammatory Bowel <strong>Disease</strong>s:Crohn’s <strong>Disease</strong> and Ulcerative ColitisInflammatory bowel diseases (IBD) are a group of pathologies that affect the digestive tract,<strong>in</strong>clud<strong>in</strong>g the small <strong>in</strong>test<strong>in</strong>e and colon, and are characterized by <strong>in</strong>flammation and tissuedestruction. There are two ma<strong>in</strong> forms: Crohn’s disease (CD) and ulcerative colitis (UC). CD,also known as regional enteritis or ileitis, is an organ-specific autoimmune disorder thatcompromises the gastro<strong>in</strong>test<strong>in</strong>al epithelia, from mouth to anus. It has a concordance rateamong monozygotic tw<strong>in</strong>s of around 60% [113], show<strong>in</strong>g a stronger genetic component thanUC. UC is a type of <strong>in</strong>flammatory bowel disease <strong>in</strong> which only the large <strong>in</strong>test<strong>in</strong>e or colon isaffected by the autoimmune pathological mechanism. UC patients present ulcers and opensores <strong>in</strong> their gastric mucosa that cause abdom<strong>in</strong>al pa<strong>in</strong>, diarrhea, fever, and weight loss,among other symptoms. The concordance rate for monozygotic tw<strong>in</strong>s is as low as 6% [113],display<strong>in</strong>g the slight effect that genetic f<strong>in</strong>gerpr<strong>in</strong>ts have on this disease. The causes trigger<strong>in</strong>gthis condition are still unknown, but there are some l<strong>in</strong>es of evidence that po<strong>in</strong>t to thecontribution of epigenetic mechanisms to UC pathogenesis.With respect to DNA methylation, it has been reported that global hypomethylation correlateswith higher proliferative activity <strong>in</strong> the affected mucosa of the rectum from patients withlongstand<strong>in</strong>g UC <strong>in</strong> comparison with healthy <strong>in</strong>dividuals [114]. Moreover, CpG islandhypermethylation at specific gene promoters has been described, and this epigenetic signatureis <strong>in</strong>volved <strong>in</strong> UC-associated colorectal carc<strong>in</strong>ogenesis [115]. Specific promoter hypermethylationhas been assessed for many key sequences. Among these, CDKN2A at bothp16 INK4a [115] and p14 ARF [116] stand out, s<strong>in</strong>ce the promoters of both are commonly silencedby methylation <strong>in</strong> sporadic colorectal cancer [117,118].The methylation status of other genes, <strong>in</strong>clud<strong>in</strong>g PAR2, MDR1, CDH1,andGDNF, has beencorrelated with the severity of the disease. In particular, PAR2 promoter methylation correlateswith severe cl<strong>in</strong>ical phenotypes of UC [119] and the chronic cont<strong>in</strong>uous type and earlier onset ofUC are associated with MDR1 promoter hypermethylation [120]. In the parts of the gastricmucosa with active methylation, there is specific methylation of CDH1 and GDNF [121].On the other hand, patients suffer<strong>in</strong>g UC have an <strong>in</strong>creased <strong>in</strong>cidence of colorectal cancer(UC-associated colorectal carc<strong>in</strong>oma) and there are some genes that could be used as biomarkersfor early detection of cancer or dysplasia <strong>in</strong> UC. Some examples of these epigenetic biomarkers areCDH1, HPP1, EYA4, SFRP2, ESR1, RUNX3, MINT1, COX-2,andDAPK. Interest<strong>in</strong>gly, themethylation level of these regulatory sequences directly <strong>in</strong>creases with malignancy [122e128].It is remarkable that <strong>in</strong> a subset of IBD-associated colorectal carc<strong>in</strong>oma there is microsatellite<strong>in</strong>stability due to the hypermethylation and silenc<strong>in</strong>g of MLH1 gene and mutation <strong>in</strong> TGFbetaRII,whose prote<strong>in</strong>s are <strong>in</strong>volved <strong>in</strong> the DNA mismatch repair gene pathway [129,130].It has been speculated that UC is a disease of accelerated ag<strong>in</strong>g of the colon, an idea for whichthere is evidence from the short length of the telomeres and the level of DNA damage detected


CHAPTER 11Epigenetic Basis of Autoimmune Disorders <strong>in</strong> <strong>Human</strong>s<strong>in</strong> the colonocytes of these people [131]. In fact, <strong>in</strong> the mucosa of UC patients, the promoter ofsome genes, such as ER, MYOD, p16, and CSPG2 have accelerated age-related methylation[132]. These f<strong>in</strong>d<strong>in</strong>gs could help expla<strong>in</strong> the higher risk of UC-associated colorectal carc<strong>in</strong>ogenesisassociated with this autoimmune disorder.On the other hand, there are some polymorphisms, affect<strong>in</strong>g XRCC1 and GST genes amongothers, that have a protective effect aga<strong>in</strong>st methylation <strong>in</strong> UC. This could be useful to classifypatients with high or low risk of develop<strong>in</strong>g cancerous lesions <strong>in</strong> ulcerative colitis [133].UC and CD share epigenetic dysregulation features, as shown <strong>in</strong> a genome-wide methylationstudy <strong>in</strong> which many genes related to immune system were found to be differentiallymethylated <strong>in</strong> both diseases. Some examples of these commonly altered genes are STAT5A,TNFTSF1A, PECAM1, FABP3, FGF, and TNFSF8, among others [134].11.2.11 Epigenetic Deregulation <strong>in</strong> Other Organ-Specific AutoimmuneDisordersType 1 diabetes mellitus (T1DM), also known as juvenile diabetes, is a type of diabetes mellitusgenerated by the organ-specific autoimmune destruction of the <strong>in</strong>sul<strong>in</strong>-produc<strong>in</strong>g beta cells ofthe pancreas [135]. As <strong>in</strong> the diseases described above, T1DM is ma<strong>in</strong>ly determ<strong>in</strong>ed by epigeneticderegulation aga<strong>in</strong>st a background of genetic susceptibility, data corroborated by theconcordance rate of 21% to 70% <strong>in</strong> monozygotic tw<strong>in</strong>s and around 10% <strong>in</strong> dizygotic tw<strong>in</strong>s [6].Although there is no doubt about the contribution of the epigenetic component <strong>in</strong> this autoimmunedisorder, no specific examples of epigenetic dysregulation have yet been described.Celiac disease, also called celiac sprue or gluten-sensitive enteropathy, is another example oforgan-specific autoimmune disorder determ<strong>in</strong>ed by the environment and genetic susceptibility. It<strong>in</strong>volves chronic <strong>in</strong>flammation and destruction of the proximal small <strong>in</strong>test<strong>in</strong>e and consequentlythe alteration of nutrient absorption. Patients with this condition cannot tolerate gluten. In fact,a transglutam<strong>in</strong>ase enzyme modifies gliad<strong>in</strong>, a gluten component, generat<strong>in</strong>g an immune systemcrossreaction and destruction of the tissue of the small bowel [136]. In this case, the concordancerates for monozygotic and dizygotic tw<strong>in</strong>s are around 75% and 11%, respectively [6].217Ankylos<strong>in</strong>g spondylitis (AS), also known as Bekhterev syndrome or Marie-Strümpell disease, isan organ-specific autoimmune disease. This disorder is one of the spondyloarthropathieswhich ma<strong>in</strong>ly occurs <strong>in</strong> young men. This chronic arthritis affects the sp<strong>in</strong>al and sacroiliacjo<strong>in</strong>ts, generat<strong>in</strong>g <strong>in</strong>flammation, jo<strong>in</strong>t damage and, <strong>in</strong> severe cases, bone formation andconsequent sp<strong>in</strong>e fusion [137]. Despite its genetic contribution, environmentally drivenepigenetic changes play an important role <strong>in</strong> AS etiology, a fact reflected <strong>in</strong> the high percentageof concordance between monozygotic and dizygotic sibl<strong>in</strong>gs (50% and 20%, respectively) [6].11.3 CONCLUSIONSThe study of epigenetic alterations <strong>in</strong> autoimmune diseases still needs greater efforts todeterm<strong>in</strong>e its relevance and potential as targets for therapeutic approaches. Further studies aretherefore needed to understand the epigenetic contributions to the pathogenesis of autoimmunediseases. Many of these disorders share cl<strong>in</strong>ical and genetic features and are <strong>in</strong>fluencedby similar environmental factors. Epigenetic modifications are <strong>in</strong>fluenced by environmentalfactors and are known to directly determ<strong>in</strong>e gene function, therefore constitut<strong>in</strong>g a relevanttarget to <strong>in</strong>vestigate its participation <strong>in</strong> the etiology of these diseases. Most efforts to identifythe epigenetic alterations that occur <strong>in</strong> autoimmune disease have focused on SLE and RA andhave served to identify both global and sequence-specific hypomethylation and overexpressionof key genes <strong>in</strong> immune function. Several issues are now key to address: to makeextensive use of high-throughput approaches, to systematically analyze all potential specificcell types relevant to disease pathogenesis, and to f<strong>in</strong>d the best way of us<strong>in</strong>g this <strong>in</strong>formation <strong>in</strong>a cl<strong>in</strong>ical sett<strong>in</strong>g.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>218References[1] Portela A, Esteller M. Epigenetic modifications and their relevance to disease. Nat Biotech 2010.[2] Berger SL, Kouzarides T, Shiekhattar R, Shilatifard A. An operational def<strong>in</strong>ition of epigenetics. Genes Dev2009;23:781e3.[3] Rahman A, Isenberg DA. Systemic lupus erythematosus. N Engl J Med 2008;358:929e39.[4] D’Cruz DP, Khamashta MA, Hughes GR. Systemic lupus erythematosus. Lancet 2007;369:587e96.[5] Rothfield N, Sontheimer RD, Bernste<strong>in</strong> M. Lupus erythematosus: systemic and cutaneous manifestations. Cl<strong>in</strong>Dermatol 2006;24:348e62.[6] Ballestar E. <strong>Epigenetics</strong> lessons from tw<strong>in</strong>s: prospects for autoimmune disease. Cl<strong>in</strong> Rev Allergy Immunol2010;39:30e41.[7] Sullivan KE. Genetics of systemic lupus erythematosus. Cl<strong>in</strong>ical implications. Rheum Dis Cl<strong>in</strong> North Am2000;26:229e56. v-vi.[8] Moser KL, Kelly JA, Lessard CJ, Harley JB. Recent <strong>in</strong>sights <strong>in</strong>to the genetic basis of systemic lupus erythematosus.Genes Immun 2009;10:373e9.[9] Rhodes B, Vyse TJ. The genetics of SLE: an update <strong>in</strong> the light of genome-wide association studies. Rheumatology(Oxford) 2008;47:1603e11.[10] Cihak A. Biological effects of 5-azacytid<strong>in</strong>e <strong>in</strong> eukaryotes. Oncology 1974;30:405e22.[11] Kam<strong>in</strong>skas E, Farrell A, Abraham S, Baird A, Hsieh LS, Lee SL, et al. Approval summary: azacitid<strong>in</strong>e fortreatment of myelodysplastic syndrome subtypes. Cl<strong>in</strong> Cancer Res 2005;11:3604e8.[12] Richardson B. Effect of an <strong>in</strong>hibitor of DNA methylation on T cells. II. 5-Azacytid<strong>in</strong>e <strong>in</strong>duces self-reactivity <strong>in</strong>antigen-specific T4þ cells. Hum Immunol 1986;17:456e70.[13] Yung R, Powers D, Johnson K, Amento E, Carr D, La<strong>in</strong>g T, et al. Mechanisms of drug-<strong>in</strong>duced lupus. II. T cellsoverexpress<strong>in</strong>g lymphocyte function-associated antigen 1 become autoreactive and cause a lupuslike disease<strong>in</strong> syngeneic mice. J Cl<strong>in</strong> Invest 1996;97:2866e71.[14] Yung RL, Quddus J, Chrisp CE, Johnson KJ, Richardson BC. Mechanism of drug-<strong>in</strong>duced lupus. I. Cloned Th2cells modified with DNA methylation <strong>in</strong>hibitors <strong>in</strong> vitro cause autoimmunity <strong>in</strong> vivo. J Immunol1995;154:3025e35.[15] Yung R, Chang S, Hemati N, Johnson K, Richardson B. Mechanisms of drug-<strong>in</strong>duced lupus. IV. Comparisonof proca<strong>in</strong>amide and hydralaz<strong>in</strong>e with analogs <strong>in</strong> vitro and <strong>in</strong> vivo. Arthritis Rheum 1997;40:1436e43.[16] Richardson BC, Strahler JR, Pivirotto TS, Quddus J, Bayliss GE, Gross LA, et al. Phenotypic and functionalsimilarities between 5-azacytid<strong>in</strong>e-treated T cells and a T cell subset <strong>in</strong> patients with active systemic lupuserythematosus. Arthritis Rheum 1992;35:647e62.[17] Cornacchia E, Golbus J, Maybaum J, Strahler J, Hanash S, Richardson B. Hydralaz<strong>in</strong>e and proca<strong>in</strong>amide<strong>in</strong>hibit T cell DNA methylation and <strong>in</strong>duce autoreactivity. J Immunol 1988;140:2197e200.[18] Quddus J, Johnson KJ, Gavalch<strong>in</strong> J, Amento EP, Chrisp CE, Yung RL, et al. Treat<strong>in</strong>g activated CD4þ T cellswith either of two dist<strong>in</strong>ct DNA methyltransferase <strong>in</strong>hibitors, 5-azacytid<strong>in</strong>e or proca<strong>in</strong>amide, is sufficient tocause a lupus-like disease <strong>in</strong> syngeneic mice. J Cl<strong>in</strong> Invest 1993;92:38e53.[19] Lei W, Luo Y, Yan K, Zhao S, Li Y, Qiu X, et al. Abnormal DNA methylation <strong>in</strong> CD4þ T cells from patientswith systemic lupus erythematosus, systemic sclerosis, and dermatomyositis. Scand J Rheumatol 2009:1e6.[20] Richardson B, Sche<strong>in</strong>bart L, Strahler J, Gross L, Hanash S, Johnson M. Evidence for impaired T cell DNAmethylation <strong>in</strong> systemic lupus erythematosus and rheumatoid arthritis. Arthritis Rheum 1990;33:1665e73.[21] Deng C, Kaplan MJ, Yang J, Ray D, Zhang Z, McCune WJ, et al. Decreased Ras-mitogen-activated prote<strong>in</strong>k<strong>in</strong>ase signal<strong>in</strong>g may cause DNA hypomethylation <strong>in</strong> T lymphocytes from lupus patients. Arthritis Rheum2001;44:397e407.[22] Wu J, Zhou T, He J, Mountz JD. Autoimmune disease <strong>in</strong> mice due to <strong>in</strong>tegration of an endogenous retrovirus<strong>in</strong> an apoptosis gene. J Exp Med 1993;178:461e8.[23] Mizugaki M, Yamaguchi T, Ishiwata S, Sh<strong>in</strong>do H, Hish<strong>in</strong>uma T, Nozaki S, et al. Alteration of DNA methylationlevels <strong>in</strong> MRL lupus mice. Cl<strong>in</strong> Exp Immunol 1997;110:265e9.[24] Wilson AS, Power BE, Molloy PL. DNA hypomethylation and human diseases. Biochim Biophys Acta2007;1775:138e62.[25] Javierre BM, Fernandez AF, Richter J, Al-Shahrour F, Mart<strong>in</strong>-Subero JI, Rodriguez-Ubreva J, et al. Changes <strong>in</strong>the pattern of DNA methylation associate with tw<strong>in</strong> discordance <strong>in</strong> systemic lupus erythematosus. GenomeRes 2010;20:170e9.[26] Isenberg DA, Manson JJ, Ehrenste<strong>in</strong> MR, Rahman A. Fifty years of anti-ds DNA antibodies: are weapproach<strong>in</strong>g journey’s end? Rheumatology (Oxford) 2007;46:1052e6.[27] Sawalha AH, Harley JB. Ant<strong>in</strong>uclear autoantibodies <strong>in</strong> systemic lupus erythematosus. Curr Op<strong>in</strong> Rheumatol2004;16:534e40.


CHAPTER 11Epigenetic Basis of Autoimmune Disorders <strong>in</strong> <strong>Human</strong>s[28] van den Broek MF, Hengartner H. The role of perfor<strong>in</strong> <strong>in</strong> <strong>in</strong>fections and tumour surveillance. Exp Physiol2000;85:681e5.[29] Kaplan MJ, Lu Q, Wu A, Attwood J, Richardson B. Demethylation of promoter regulatory elementscontributes to perfor<strong>in</strong> overexpression <strong>in</strong> CD4 þ lupus T cells. J Immunol 2004;172:3652e61.[30] Luo Y, Zhang X, Zhao M, Lu Q. DNA demethylation of the perfor<strong>in</strong> promoter <strong>in</strong> CD4(þ) T cells from patientswith subacute cutaneous lupus erythematosus. J Dermatol Sci 2009;56:33e6.[31] Lu Q, Wu A, Tesmer L, Ray D, Yousif N, Richardson B. Demethylation of CD40LG on the <strong>in</strong>active X <strong>in</strong> T cellsfrom women with lupus. J Immunol 2007;179:6352e8.[32] Zhou Y, Yuan J, Pan Y, Fei Y, Qiu X, Hu N, et al. T cell CD40LG gene expression and the production of IgG byautologous B cells <strong>in</strong> systemic lupus erythematosus. Cl<strong>in</strong> Immunol 2009;132:362e70.[33] Hu N, Qiu X, Luo Y, Yuan J, Li Y, Lei W, et al. Abnormal histone modification patterns <strong>in</strong> lupus CD4 þ T cells.J Rheumatol 2008;35:804e10.[34] Garcia BA, Busby SA, Shabanowitz J, Hunt DF, Mishra N. Resett<strong>in</strong>g the epigenetic histone code <strong>in</strong> the MRLlpr/lprmouse model of lupus by histone deacetylase <strong>in</strong>hibition. J Proteome Res 2005;4:2032e42.[35] Mishra N, Brown DR, Olorenshaw IM, Kammer GM. Trichostat<strong>in</strong> A reverses skewed expression of CD154,<strong>in</strong>terleuk<strong>in</strong>-10, and <strong>in</strong>terferon-gamma gene and prote<strong>in</strong> expression <strong>in</strong> lupus T cells. Proc Natl Acad Sci USA2001;98:2628e33.[36] Zhang Z, Song L, Maurer K, Petri MA, Sullivan KE. Global H4 acetylation analysis by ChIP-chip <strong>in</strong> systemiclupus erythematosus monocytes. Genes Immun 2009.[37] Duke O, Panayi GS, Janossy G, Poulter LW. An immunohistological analysis of lymphocyte subpopulationsand their microenvironment <strong>in</strong> the synovial membranes of patients with rheumatoid arthritis us<strong>in</strong>gmonoclonal antibodies. Cl<strong>in</strong> Exp Immunol 1982;49:22e30.[38] Neumann E, Lefevre S, Zimmermann B, Gay S, Muller-Ladner U. Rheumatoid arthritis progression mediatedby activated synovial fibroblasts. Trends Mol Med 2010;16:458e68.[39] Tolboom TC, van der Helm-Van Mil AH, Nelissen RG, Breedveld FC, Toes RE, Huiz<strong>in</strong>ga TW. Invasiveness offibroblast-like synoviocytes is an <strong>in</strong>dividual patient characteristic associated with the rate of jo<strong>in</strong>t destruction<strong>in</strong> patients with rheumatoid arthritis. Arthritis Rheum 2005;52:1999e2002.[40] Distler JH, Jungel A, Huber LC, Seemayer CA, Reich 3rd CF, Gay RE, et al. The <strong>in</strong>duction of matrix metalloprote<strong>in</strong>aseand cytok<strong>in</strong>e expression <strong>in</strong> synovial fibroblasts stimulated with immune cell microparticles. ProcNatl Acad Sci USA 2005;102:2892e7.[41] Muller-Ladner U, Kriegsmann J, Frankl<strong>in</strong> BN, Matsumoto S, Geiler T, Gay RE, et al. Synovial fibroblasts ofpatients with rheumatoid arthritis attach to and <strong>in</strong>vade normal human cartilage when engrafted <strong>in</strong>to SCIDmice. Am J Pathol 1996;149:1607e15.[42] Baier A, Me<strong>in</strong>eckel I, Gay S, Pap T. Apoptosis <strong>in</strong> rheumatoid arthritis. Curr Op<strong>in</strong> Rheumatol 2003;15:274e9.[43] Lafyatis R, Remmers EF, Roberts AB, Yocum DE, Sporn MB, Wilder RL. Anchorage-<strong>in</strong>dependent growth ofsynoviocytes from arthritic and normal jo<strong>in</strong>ts. Stimulation by exogenous platelet-derived growth factor and<strong>in</strong>hibition by transform<strong>in</strong>g growth factor-beta and ret<strong>in</strong>oids. J Cl<strong>in</strong> Invest 1989;83:1267e76.[44] Corvetta A, Della Bitta R, Luchetti MM, Pomponio G. 5-Methylcytos<strong>in</strong>e content of DNA <strong>in</strong> blood, synovialmononuclear cells and synovial tissue from patients affected by autoimmune rheumatic diseases. J Chromatogr1991;566:481e91.[45] Neidhart M, Rethage J, Kuchen S, Kunzler P, Crowl RM, Bill<strong>in</strong>gham ME, et al. Retrotransposable L1 elementsexpressed <strong>in</strong> rheumatoid arthritis synovial tissue: association with genomic DNA hypomethylation and<strong>in</strong>fluence on gene expression. Arthritis Rheum 2000;43:2634e47.[46] Kuchen S, Seemayer CA, Rethage J, von Knoch R, Kuenzler P, Beat AM, et al. The L1 retroelement-related p40prote<strong>in</strong> <strong>in</strong>duces p38delta MAP k<strong>in</strong>ase. Autoimmunity 2004;37:57e65.[47] Karouzakis E, Gay RE, Michel BA, Gay S, Neidhart M. DNA hypomethylation <strong>in</strong> rheumatoid arthritis synovialfibroblasts. Arthritis Rheum 2009;60:3613e22.[48] Esteller M. <strong>Epigenetics</strong> <strong>in</strong> cancer. N Engl J Med 2008;358:1148e59.[49] Takami N, Osawa K, Miura Y, Komai K, Taniguchi M, Shiraishi M, et al. Hypermethylated promoter region ofDR3, the death receptor 3 gene, <strong>in</strong> rheumatoid arthritis synovial cells. Arthritis Rheum 2006;54:779e87.[50] Stanczyk J, Ospelt C, Karouzakis E, Filer A, Raza K, Koll<strong>in</strong>g C, et al. Altered expression of microRNA-203 <strong>in</strong>rheumatoid arthritis synovial fibroblasts and its role <strong>in</strong> fibroblast activation. Arthritis Rheum2011;63:373e81.[51] Kitamura T, Kabuyama Y, Kamataki A, Homma MK, Kobayashi H, Aota S, et al. Enhancement of lymphocytemigration and cytok<strong>in</strong>e production by ephr<strong>in</strong>B1 system <strong>in</strong> rheumatoid arthritis. Am J Physiol Cell Physiol2008;294:C189e96.[52] Nile CJ, Read RC, Akil M, Duff GW, Wilson AG. Methylation status of a s<strong>in</strong>gle CpG site <strong>in</strong> the IL6 promoter isrelated to IL6 messenger RNA levels and rheumatoid arthritis. Arthritis Rheum 2008;58:2686e93.219


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>220[53] Chen Y, Gorelik GJ, Strickland FM, Richardson BC. Decreased ERK and JNK signal<strong>in</strong>g contribute to geneoverexpression <strong>in</strong> "senescent" CD4þCD28- T cells through epigenetic mechanisms. J Leukoc Biol2010;87:137e45.[54] Janson PC, L<strong>in</strong>ton LB, Bergman EA, Marits P, Eberhardson M, Piehl F, et al. Profil<strong>in</strong>g of CD4 þ T cells withepigenetic immune l<strong>in</strong>eage analysis. J Immunol 2011;186:92e102.[55] Kawabata T, Nishida K, Takasugi K, Ogawa H, Sada K, Kadota Y, et al. Increased activity and expression ofhistone deacetylase 1 <strong>in</strong> relation to tumor necrosis factor-alpha <strong>in</strong> synovial tissue of rheumatoid arthritis.Arthritis Res Ther 2010;12:R133.[56] Horiuchi M, Mor<strong>in</strong>obu A, Ch<strong>in</strong> T, Sakai Y, Kurosaka M, Kumagai S. Expression and function of histonedeacetylases <strong>in</strong> rheumatoid arthritis synovial fibroblasts. J Rheumatol 2009;36:1580e9.[57] Maciejewska-Rodrigues H, Karouzakis E, Strietholt S, Hemmatazad H, Neidhart M, Ospelt C, et al. <strong>Epigenetics</strong>and rheumatoid arthritis: the role of SENP1 <strong>in</strong> the regulation of MMP-1 expression. J Autoimmun2010;35:15e22.[58] Chung YL, Lee MY, Wang AJ, Yao LF. A therapeutic strategy uses histone deacetylase <strong>in</strong>hibitors to modulate theexpression of genes <strong>in</strong>volved <strong>in</strong> the pathogenesis of rheumatoid arthritis. Mol Ther 2003;8:707e17.[59] Nishida K, Komiyama T, Miyazawa S, Shen ZN, Furumatsu T, Doi H, et al. Histone deacetylase <strong>in</strong>hibitorsuppression of autoantibody-mediated arthritis <strong>in</strong> mice via regulation of p16INK4a and p21(WAF1/Cip1)expression. Arthritis Rheum 2004;50:3365e76.[60] Jungel A, Baresova V, Ospelt C, Simmen BR, Michel BA, Gay RE, et al. Trichostat<strong>in</strong> A sensitises rheumatoidarthritis synovial fibroblasts for TRAIL-<strong>in</strong>duced apoptosis. Ann Rheum Dis 2006;65:910e2.[61] Mor<strong>in</strong>obu A, Wang B, Liu J, Yoshiya S, Kurosaka M, Kumagai S. Trichostat<strong>in</strong> A cooperates with Fas-mediatedsignal to <strong>in</strong>duce apoptosis <strong>in</strong> rheumatoid arthritis synovial fibroblasts. J Rheumatol 2006;33:1052e60.[62] Choo QY, Ho PC, Tanaka Y, L<strong>in</strong> HS. Histone deacetylase <strong>in</strong>hibitors MS-275 and SAHA <strong>in</strong>duced growth arrestand suppressed lipopolysaccharide-stimulated NF-kappaB p65 nuclear accumulation <strong>in</strong> human rheumatoidarthritis synovial fibroblastic E11 cells. Rheumatology (Oxford) 2010;49:1447e60.[63] Nasu Y, Nishida K, Miyazawa S, Komiyama T, Kadota Y, Abe N, et al. Trichostat<strong>in</strong> A, a histone deacetylase<strong>in</strong>hibitor, suppresses synovial <strong>in</strong>flammation and subsequent cartilage destruction <strong>in</strong> a collagen antibody<strong>in</strong>ducedarthritis mouse model. Osteoarthritis Cartilage 2008;16:723e32.[64] L<strong>in</strong> HS, Hu CY, Chan HY, Liew YY, Huang HP, Lepescheux L, et al. Anti-rheumatic activities of histonedeacetylase (HDAC) <strong>in</strong>hibitors <strong>in</strong> vivo <strong>in</strong> collagen-<strong>in</strong>duced arthritis <strong>in</strong> rodents. Br J Pharmacol2007;150:862e72.[65] Carta S, Tassi S, Sem<strong>in</strong>o C, Fossati G, Mascagni P, D<strong>in</strong>arello CA, et al. Histone deacetylase <strong>in</strong>hibitors preventexocytosis of <strong>in</strong>terleuk<strong>in</strong>-1beta-conta<strong>in</strong><strong>in</strong>g secretory lysosomes: role of microtubules. Blood2006;108:1618e26.[66] Young DA, Lakey RL, Penn<strong>in</strong>gton CJ, Jones D, Kevorkian L, Edwards DR, et al. Histone deacetylase <strong>in</strong>hibitorsmodulate metalloprote<strong>in</strong>ase gene expression <strong>in</strong> chondrocytes and block cartilage resorption. Arthritis ResTher 2005;7:R503e12.[67] Manabe H, Nasu Y, Komiyama T, Furumatsu T, Kitamura A, Miyazawa S, et al. Inhibition of histonedeacetylase down-regulates the expression of hypoxia-<strong>in</strong>duced vascular endothelial growth factor by rheumatoidsynovial fibroblasts. Inflamm Res 2008;57:4e10.[68] Tao R, de Zoeten EF, Ozkaynak E, Chen C, Wang L, Porrett PM, et al. Deacetylase <strong>in</strong>hibition promotes thegeneration and function of regulatory T cells. Nat Med 2007;13:1299e307.[69] Saouaf SJ, Li B, Zhang G, Shen Y, Furuuchi N, Hancock WW, et al. Deacetylase <strong>in</strong>hibition <strong>in</strong>creases regulatoryT cell function and decreases <strong>in</strong>cidence and severity of collagen-<strong>in</strong>duced arthritis. Exp Mol Pathol2009;87:99e104.[70] Zhou X, Hua X, D<strong>in</strong>g X, Bian Y, Wang X. Trichostat<strong>in</strong> Differentially Regulates Th1 and Th2 Responses andAlleviates Rheumatoid Arthritis <strong>in</strong> Mice. J Cl<strong>in</strong> Immunol 2011.[71] Brogdon JL, Xu Y, Szabo SJ, An S, Buxton F, Cohen D, et al. Histone deacetylase activities are required for<strong>in</strong>nate immune cell control of Th1 but not Th2 effector cell function. Blood 2007;109:1123e30.[72] Schroeder TM, Westendorf JJ. Histone deacetylase <strong>in</strong>hibitors promote osteoblast maturation. J Bone M<strong>in</strong>erRes 2005;20:2254e63.[73] Schroeder TM, Nair AK, Staggs R, Lambl<strong>in</strong> AF, Westendorf JJ. Gene profile analysis of osteoblast genesdifferentially regulated by histone deacetylase <strong>in</strong>hibitors. BMC Genomics 2007;8:362.[74] Nakamura T, Kukita T, Shobuike T, Nagata K, Wu Z, Ogawa K, et al. Inhibition of histone deacetylasesuppresses osteoclastogenesis and bone destruction by <strong>in</strong>duc<strong>in</strong>g IFN-beta production. J Immunol2005;175:5809e16.[75] Compston A, Coles A. Multiple sclerosis. Lancet 2008;372:1502e17.[76] Ebers GC, Sadovnick AD. The role of genetic factors <strong>in</strong> multiple sclerosis susceptibility. J Neuroimmunol1994;54:1e17.


CHAPTER 11Epigenetic Basis of Autoimmune Disorders <strong>in</strong> <strong>Human</strong>s[77] Kragt J, van Amerongen B, Killeste<strong>in</strong> J, Dijkstra C, Uitdehaag B, Polman C, et al. Higher levels of 25-hydroxyvitam<strong>in</strong> D are associated with a lower <strong>in</strong>cidence of multiple sclerosis only <strong>in</strong> women. Mult Scler2009;15:9e15.[78] Mikaeloff Y, Caridade G, Tardieu M, Suissa S. Parental smok<strong>in</strong>g at home and the risk of childhood-onsetmultiple sclerosis <strong>in</strong> children. Bra<strong>in</strong> 2007;130:2589e95.[79] Marrie RA. Environmental risk factors <strong>in</strong> multiple sclerosis aetiology. Lancet Neurol 2004;3:709e18.[80] Moscarello MA, Mastronardi FG, Wood DD. The role of citrull<strong>in</strong>ated prote<strong>in</strong>s suggests a novel mechanism <strong>in</strong>the pathogenesis of multiple sclerosis. Neurochem Res 2007;32:251e6.[81] Mastronardi FG, Noor A, Wood DD, Paton T, Moscarello MA. Peptidyl arg<strong>in</strong><strong>in</strong>edeim<strong>in</strong>ase 2 CpG island <strong>in</strong>multiple sclerosis white matter is hypomethylated. J Neurosci Res 2007;85:2006e16.[82] D’Souza CA, Wood DD, She YM, Moscarello MA. Autocatalytic cleavage of myel<strong>in</strong> basic prote<strong>in</strong>: an alternativeto molecular mimicry. Biochemistry 2005;44:12905e13.[83] Musse AA, Boggs JM, Harauz G. Deim<strong>in</strong>ation of membrane-bound myel<strong>in</strong> basic prote<strong>in</strong> <strong>in</strong> multiple sclerosisexposes an immunodom<strong>in</strong>ant epitope. Proc Natl Acad Sci USA 2006;103:4422e7.[84] Baranz<strong>in</strong>i SE, Mudge J, van Velk<strong>in</strong>burgh JC, Khankhanian P, Khrebtukova I, Miller NA, et al. Genome,epigenome and RNA sequences of monozygotic tw<strong>in</strong>s discordant for multiple sclerosis. Nature2010;464:1351e6.[85] Mariette X, Gottenberg JE. Pathogenesis of Sjogren’s syndrome and therapeutic consequences. Curr Op<strong>in</strong>Rheumatol 2010;22:471e7.[86] Gonzalez S, Aguilera S, Alliende C, Urzua U, Quest AF, Herrera L, Mol<strong>in</strong>a C, Hermoso M, Ewert P,Brito M, Romo R, Leyton C, Perez P, Gonzalez MJ. Alterations <strong>in</strong> type I hemidesmosome componentssuggestive of epigenetic control <strong>in</strong> the salivary glands of patients with Sjogren’s syndrome. ArthritisRheum 63:1106e15.[87] Lu Q, Renaud<strong>in</strong>eau Y, Cha S, Ilei G, Brooks WH, Selmi C, Tzioufas A, Pers JO, Bombardieri S, Gershw<strong>in</strong> ME,Gay S, You<strong>in</strong>ou P. <strong>Epigenetics</strong> <strong>in</strong> autoimmune disorders: highlights of the 10th Sjogren’s syndrome symposium.Autoimmun Rev 9:627e30.[88] Das SK, Majumder PP, Chakraborty R, Majumdar TK, Haldar B. Studies on vitiligo. I. Epidemiological profile<strong>in</strong> Calcutta, India. Genet Epidemiol 1985;2:71e8.[89] Laberge G, Mailloux CM, Gowan K, Holland P, Bennett DC, Fa<strong>in</strong> PR, et al. Early disease onset and <strong>in</strong>creasedrisk of other autoimmune diseases <strong>in</strong> familial generalized vitiligo. Pigment Cell Res 2005;18:300e5.[90] Alkhateeb A, Fa<strong>in</strong> PR, Thody A, Bennett DC, Spritz RA. Epidemiology of vitiligo and associated autoimmunediseases <strong>in</strong> Caucasian probands and their families. Pigment Cell Res 2003;16:208e14.[91] Spritz RA, The genetics of generalized vitiligo: autoimmune pathways and an <strong>in</strong>verse relationship withmalignant melanoma. Genome Med 2:78.[92] Sreekumar GP, Erf GF, Smyth Jr JR. 5-azacytid<strong>in</strong>e treatment <strong>in</strong>duces autoimmune vitiligo <strong>in</strong> parental controlstra<strong>in</strong>s of the Smyth l<strong>in</strong>e chicken model for autoimmune vitiligo. Cl<strong>in</strong> Immunol Immunopathol1996;81:136e44.[93] Arora-S<strong>in</strong>gh RK, Assassi S, del Junco DJ, Arnett FC, Perry M, Irfan U, et al. Autoimmune diseases andautoantibodies <strong>in</strong> the first degree relatives of patients with systemic sclerosis. J Autoimmun 2010;35:52e7.[94] Qi Q, Guo Q, Tan G, Mao Y, Tang H, Zhou C, et al. Predictors of the scleroderma phenotype <strong>in</strong> fibroblastsfrom systemic sclerosis patients. J Eur Acad Dermatol Venereol 2009;23:160e8.[95] Wang Y, Fan PS, Kahaleh B. Association between enhanced type I collagen expression and epigeneticrepression of the FLI1 gene <strong>in</strong> scleroderma fibroblasts. Arthritis Rheum 2006;54:2271e9.[96] Ozbalkan Z, Bagislar S, Kiraz S, Akyerli CB, Ozer HT, Yavuz S, et al. Skewed X chromosome <strong>in</strong>activation <strong>in</strong>blood cells of women with scleroderma. Arthritis Rheum 2005;52:1564e70.[97] Raval K, Lofland JH, Waters H, Piech CT. <strong>Disease</strong> and treatment burden of psoriasis: exam<strong>in</strong><strong>in</strong>g the impact ofbiologics. J Drugs Dermatol 10:189e96.[98] Ruchusatsawat K, Wongpiyabovorn J, Shuangshoti S, Hirankarn N, Mutirangura A. SHP-1 promoter 2methylation <strong>in</strong> normal epithelial tissues and demethylation <strong>in</strong> psoriasis. J Mol Med 2006;84:175e82.[99] Zhang K, Zhang R, Li X, Y<strong>in</strong> G, Niu X, Hou R. The mRNA expression and promoter methylation status of thep16 gene <strong>in</strong> colony-form<strong>in</strong>g cells with high proliferative potential <strong>in</strong> patients with psoriasis. Cl<strong>in</strong> ExpDermatol 2007;32:702e8.[100] Chen M, Chen ZQ, Cui PG, Yao X, Li YM, Li AS, et al. The methylation pattern of p16INK4a gene promoter <strong>in</strong>psoriatic epidermis and its cl<strong>in</strong>ical significance. Br J Dermatol 2008;158:987e93.[101] Zhang K, Zhang R, Li X, Y<strong>in</strong> G, Niu X. Promoter methylation status of p15 and p21 genes <strong>in</strong> HPP-CFCs ofbone marrow of patients with psoriasis. Eur J Dermatol 2009;19:141e6.[102] Zhang P, Su Y, Chen H, Zhao M, Lu Q. Abnormal DNA methylation <strong>in</strong> sk<strong>in</strong> lesions and PBMCs of patientswith psoriasis vulgaris. J Dermatol Sci 2010;60:40e2.221


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>222[103] Poupon R. Primary biliary cirrhosis: a. J Hepatol 2010;52:745e58.[104] Selmi C, Invernizzi P, Miozzo M, Podda M, Gershw<strong>in</strong> ME. Primary biliary cirrhosis: does X mark the spot?Autoimmun Rev 2004;3:493e9.[105] Gershw<strong>in</strong> ME, Selmi C, Worman HJ, Gold EB, Watnik M, Utts J, et al. Risk factors and comorbidities <strong>in</strong>primary biliary cirrhosis: a controlled <strong>in</strong>terview-based study of 1032 patients. Hepatology 2005;42:1194e202.[106] Mitchell MM, Lleo A, Zammataro L, Mayo MJ, Invernizzi P, Bach N, Shimoda S, Gordon S, Podda M, Gershw<strong>in</strong>ME, Selmi C, LaSalle JM. Epigenetic <strong>in</strong>vestigation of variably X chromosome <strong>in</strong>activated genes <strong>in</strong> monozygoticfemale tw<strong>in</strong>s discordant for primary biliary cirrhosis. <strong>Epigenetics</strong> 6:95e102.[107] Thiemann A, Grunder S, Pusch M, Jentsch TJ. A chloride channel widely expressed <strong>in</strong> epithelial and nonepithelialcells. Nature 1992;356:57e60.[108] Rogner UC, Heiss NS, Kioschis P, Wiemann S, Korn B, Poustka A. Transcriptional analysis of the candidateregion for <strong>in</strong>cont<strong>in</strong>entia pigmenti (IP2) <strong>in</strong> Xq28. Genome Res 1996;6:922e34.[109] Uchida T, Fujimori F, Tradler T, Fischer G, Rahfeld JU. Identification and characterization of a 14 kDa humanprote<strong>in</strong> as a novel parvul<strong>in</strong>-like peptidyl prolyl cis/trans isomerase. FEBS Lett 1999;446:278e82.[110] Brix TH, Kyvik KO, Hegedus L. A population-based study of chronic autoimmune hypothyroidism <strong>in</strong> Danishtw<strong>in</strong>s. J Cl<strong>in</strong> Endocr<strong>in</strong>ol Metab 2000;85:536e9.[111] Brix TH, Kyvik KO, Christensen K, Hegedus L. Evidence for a major role of heredity <strong>in</strong> Graves’ disease:a population-based study of two Danish tw<strong>in</strong> cohorts. J Cl<strong>in</strong> Endocr<strong>in</strong>ol Metab 2001;86:930e4.[112] Brix TH, Knudsen GP, Kristiansen M, Kyvik KO, Orstavik KH, Hegedus L. High frequency of skewedX-chromosome <strong>in</strong>activation <strong>in</strong> females with autoimmune thyroid disease: a possible explanation for thefemale predisposition to thyroid autoimmunity. J Cl<strong>in</strong> Endocr<strong>in</strong>ol Metab 2005;90:5949e53.[113] Tysk C, L<strong>in</strong>dberg E, Jarnerot G, Floderus-Myrhed B. Ulcerative colitis and Crohn’s disease <strong>in</strong> an unselectedpopulation of monozygotic and dizygotic tw<strong>in</strong>s. A study of heritability and the <strong>in</strong>fluence of smok<strong>in</strong>g. Gut1988;29:990e6.[114] Gloria L, Cravo M, P<strong>in</strong>to A, de Sousa LS, Chaves P, et al. DNA hypomethylation and proliferative activityare <strong>in</strong>creased <strong>in</strong> the rectal mucosa of patients with long-stand<strong>in</strong>g ulcerative colitis. Cancer 1996;78:2300e6.[115] Hsieh CJ, Klump B, Holzmann K, Borchard F, Gregor M, Porschen R. Hypermethylation of the p16INK4apromoter <strong>in</strong> colectomy specimens of patients with long-stand<strong>in</strong>g and extensive ulcerative colitis. Cancer Res1998;58:3942e5.[116] Sato F, Harpaz N, Shibata D, Xu Y, Y<strong>in</strong> J, Mori Y, et al. Hypermethylation of the p14(ARF) gene <strong>in</strong> ulcerativecolitis-associated colorectal carc<strong>in</strong>ogenesis. Cancer Res 2002;62:1148e51.[117] Gonzalez-Zulueta M, Bender CM, Yang AS, Nguyen T, Beart RW, Van Tornout JM, et al. Methylation of the 5’CpG island of the p16/CDKN2 tumor suppressor gene <strong>in</strong> normal and transformed human tissues correlateswith gene silenc<strong>in</strong>g. Cancer Res 1995;55:4531e5.[118] Esteller M, Tortola S, Toyota M, Capella G, Pe<strong>in</strong>ado MA, Bayl<strong>in</strong> SB, et al. Hypermethylation-associated<strong>in</strong>activation of p14(ARF) is <strong>in</strong>dependent of p16(INK4a) methylation and p53 mutational status. Cancer Res2000;60:129e33.[119] Tahara T, Shibata T, Nakamura M, Yamashita H, Yoshioka D, Okubo M, et al. Promoter methylation ofprotease-activated receptor (PAR2) is associated with severe cl<strong>in</strong>ical phenotypes of ulcerative colitis (UC).Cl<strong>in</strong> Exp Med 2009;9:125e30.[120] Tahara T, Shibata T, Nakamura M, Yamashita H, Yoshioka D, Okubo M, et al. Effect of MDR1 gene promotermethylation <strong>in</strong> patients with ulcerative colitis. Int J Mol Med 2009;23:521e7.[121] Saito S, Kato J, Hiraoka S, Horii J, Suzuki H, Higashi R, Kaji E, Kondo Y, Yamamoto K. DNA methylation ofcolon mucosa <strong>in</strong> ulcerative colitis patients: Correlation with <strong>in</strong>flammatory status. Inflamm Bowel Dis2011;17(9):1955e65.[122] Wheeler JM, Kim HC, Efstathiou JA, Ilyas M, Mortensen NJ, Bodmer WF. Hypermethylation of the promoterregion of the E-cadher<strong>in</strong> gene (CDH1) <strong>in</strong> sporadic and ulcerative colitis associated colorectal cancer. Gut2001;48:367e71.[123] Sato F, Shibata D, Harpaz N, Xu Y, Y<strong>in</strong> J, Mori Y, et al. Aberrant methylation of the HPP1 gene <strong>in</strong> ulcerativecolitis-associated colorectal carc<strong>in</strong>oma. Cancer Res 2002;62:6820e2.[124] Osborn NK, Zou H, Mol<strong>in</strong>a JR, Lesche R, Lew<strong>in</strong> J, Lofton-Day C, et al. Aberrant methylation of the eyesabsent 4 gene <strong>in</strong> ulcerative colitis-associated dysplasia. Cl<strong>in</strong> Gastroenterol Hepatol 2006;4:212e8.[125] Huang Z, Li L, Wang J. Hypermethylation of SFRP2 as a potential marker for stool-based detection ofcolorectal cancer and precancerous lesions. Dig Dis Sci 2007;52:2287e91.[126] Fujii S, Tom<strong>in</strong>aga K, Kitajima K, Takeda J, Kusaka T, Fujita M, et al. Methylation of the oestrogen receptorgene <strong>in</strong> non-neoplastic epithelium as a marker of colorectal neoplasia risk <strong>in</strong> longstand<strong>in</strong>g and extensiveulcerative colitis. Gut 2005;54:1287e92.


CHAPTER 11Epigenetic Basis of Autoimmune Disorders <strong>in</strong> <strong>Human</strong>s[127] Garrity-Park MM, Loftus Jr EV, Sandborn WJ, Bryant SC, Smyrk TC. Methylation status of genes <strong>in</strong> nonneoplasticmucosa from patients with ulcerative colitis-associated colorectal cancer. Am J Gastroenterol2010;105:1610e9.[128] Kuester D, Guenther T, Biesold S, Hartmann A, Bataille F, Ruemmele P, Peters B, Meyer F, Schubert D, BohrUR, Malferthe<strong>in</strong>er P, Lippert H, Silver AR, Roessner A,Schneider-Stock, R. Aberrant methylation of DAPK <strong>in</strong>long-stand<strong>in</strong>g ulcerative colitis and ulcerative colitis-associated carc<strong>in</strong>oma. Pathol Res Pract 206:616e24.[129] Fleisher AS, Esteller M, Harpaz N, Leyt<strong>in</strong> A, Rashid A, Xu Y, et al. Microsatellite <strong>in</strong>stability <strong>in</strong> <strong>in</strong>flammatorybowel disease-associated neoplastic lesions is associated with hypermethylation and dim<strong>in</strong>ished expressionof the DNA mismatch repair gene, hMLH1. Cancer Res 2000;60:4864e8.[130] Fujiwara I, Yashiro M, Kubo N, Maeda K, Hirakawa K. Ulcerative colitis-associated colorectal cancer isfrequently associated with the microsatellite <strong>in</strong>stability pathway. Dis Colon Rectum 2008;51:1387e94.[131] Risques RA, Lai LA, Brentnall TA, Li L, Feng Z, Gallaher J, et al. Ulcerative colitis is a disease of acceleratedcolon ag<strong>in</strong>g: evidence from telomere attrition and DNA damage. Gastroenterology 2008;135:410e8.[132] Issa JP, Ahuja N, Toyota M, Bronner MP, Brentnall TA. Accelerated age-related CpG island methylation <strong>in</strong>ulcerative colitis. Cancer Res 2001;61:3573e7.[133] Tahara T, Shibata T, Nakamura M, Okubo M, Yamashita H, Yoshioka D, Yonemura J, Hirata I, Arisawa T.Association between polymorphisms <strong>in</strong> the XRCC1 and GST genes, and CpG island methylation status <strong>in</strong>colonic mucosa <strong>in</strong> ulcerative colitis. Virchows Arch 458:205e11.[134] L<strong>in</strong> Z, Hegarty J, Cappel J, Yu W, Chen X, Faber P, et al. Identification of disease-associated DNA methylation<strong>in</strong> <strong>in</strong>test<strong>in</strong>al tissues from patients with <strong>in</strong>flammatory bowel disease. Cl<strong>in</strong> Genet 2011;80:59e67.[135] van Belle TL, Coppieters KT, von Herrath MG. Type 1 diabetes: etiology, immunology, and therapeuticstrategies. Physiol Rev 2011;91:79e118.[136] Moore JK, West SR, Rob<strong>in</strong>s G. Advances <strong>in</strong> celiac disease. Curr Op<strong>in</strong> Gastroenterol 2011;27:112e8.[137] Dougados M, Baeten D. Spondyloarthritis. Lancet 2011;377:2127e37.223


This page <strong>in</strong>tentionally left blank


CHAPTER 12Approaches toAutoimmune <strong>Disease</strong>sUs<strong>in</strong>g Epigenetic TherapyChristopher ChangThomas Jefferson University, Wilm<strong>in</strong>gton, DE, USACHAPTER OUTLINE12.1 Introduction 22512.2 Pathophysiologic Basis for theDevelopment of EpigeneticTreatments <strong>in</strong> Autoimmunity 22612.3 Pathology of Autoimmune <strong>Disease</strong>and Potential Targets forEpigenetic Drugs 22912.4 HDAC Inhibitors 23012.4.1 HDAC Inhibitors <strong>in</strong> BoneHomeostasis 23312.4.2 HDAC Inhibitors <strong>in</strong> theTreatment of InflammatoryBowel <strong>Disease</strong> 23412.4.3 HDAC Inhibitors <strong>in</strong> theTreatment of MultipleSclerosis 23512.4.4 Epigenetic Regulation of theGlucocorticoid Receptor andRelated ImmuneRegulation 23512.5 DNA Methylation and DNAMethyltransferases 23612.6 MicroRNA 23712.6.1 miRNAs <strong>in</strong> Systemic LupusErythematosus (SLE) 23912.6.2 miRNAs <strong>in</strong> Rheumatoid Arthritis(RA) 23912.6.3 miRNAs <strong>in</strong> Multiple Sclerosis,Scleroderma, Sjögren’sSyndrome, Polymyositis,Dermatomyositis, and PrimaryBiliary Cirrhosis 24012.7 Antagomirs 24012.8 Techniques to Measure EpigeneticAlterations e Application of<strong>Epigenetics</strong> as Biomarkers 24312.9 Potential Side Effects of Treatmentwith Epigenetic Drugs <strong>in</strong>Autoimmune <strong>Disease</strong>s 24312.10 Balanc<strong>in</strong>g Conventional Therapyand Epigenetic Therapy 24412.11 Where do we go from here? 24512.12 Discussion 245References 24622512.1 INTRODUCTIONAutoimmune diseases affect more than 23.5 million people <strong>in</strong> the United States and about anestimated 5% of the world’s population. The <strong>in</strong>cidence of pediatric systemic lupus erythematosusranges from 0.36 to 2.5 per 100 000 per year [1]. Rates vary <strong>in</strong> different parts of theworld, reflect<strong>in</strong>g a genetic basis for many of these conditions. There are “hotspots” where<strong>in</strong>cidence rates may <strong>in</strong>crease, and this is believed to be <strong>in</strong>fluenced by environmental or otherT. Tollefsbol (Ed): <strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>. DOI: 10.1016/B978-0-12-388415-2.00012-3Copyright Ó 2012 Elsevier Inc. All rights reserved.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>exposures. There is also a role for gender <strong>in</strong> autoimmunity, with the prevalence <strong>in</strong> womenbe<strong>in</strong>g significantly higher than <strong>in</strong> men [2]. In some conditions, such as scleroderma andautoimmune thyroiditis, this gender bias has been traced to an imbalance <strong>in</strong> X-chromosome<strong>in</strong>activation, known as the X chromosome <strong>in</strong>activation skew theory [3]. This is evidence thatnot only genetics, but epigenetics may play a role <strong>in</strong> the pathogenesis of autoimmune diseases.Phenotypic variation with<strong>in</strong> each of the autoimmune diseases may <strong>in</strong>deed be a function ofepigenetic <strong>in</strong>fluences on a basel<strong>in</strong>e level of gene expression [4e6]. Because epigenetic modificationsare reversible [7], this also opens the door for potential treatments to be developedthat will reverse the epigenetic changes that contribute to the pathogenesis of the disease.226The treatment of autoimmune diseases has undergone several very significant paradigmchanges over the past century. With a better understand<strong>in</strong>g of the mechanisms of this group ofdiseases have come newer and more <strong>in</strong>novative modes of therapy. The discovery of cortisone,<strong>in</strong>itially called “Compound E” <strong>in</strong> the 1940s was hailed as a wonder drug after the successfultreatment of a woman with rheumatoid arthritis at the Mayo Cl<strong>in</strong>ic. This would ultimately leadto a Noble Prize for Hench, Kendall, and Reichste<strong>in</strong>. Kendall, <strong>in</strong> 1964, proclaimed that it wouldbe “.highly improbable that any product will ever be found which can be used <strong>in</strong> place ofcortisone.” [8]. The steroids and non-steroidal <strong>in</strong>flammatory agents that were <strong>in</strong>itially used <strong>in</strong>the treatment of these diseases was followed by the discovery and subsequent development ofdisease-modify<strong>in</strong>g antirheumatic drugs (DMARDs). The difference is that the former groupwould be effective <strong>in</strong> treat<strong>in</strong>g symptoms but unlike DMARDs, would not slow progressionof the disease. DMARD drugs were <strong>in</strong>itially used to treat rheumatoid arthritis, hence the name,but their use was then extended to <strong>in</strong>clude other autoimmune diseases <strong>in</strong>clud<strong>in</strong>g systemic lupuserythematosus (SLE), myasthenia gravis, immune thrombocytopenic purpure (ITP), Crohn’sdisease, and many others. The earlier DMARDs consisted of traditional drugs that were of lowmolecular weight, but more recently another new class of DMARDs has emerged. These are thebiological agents, which are synthesized by genetic eng<strong>in</strong>eer<strong>in</strong>g and have proven to beextremely effective <strong>in</strong> the control of these diseases. The earliest biological agent to treat rheumatoidarthritis was rituximab, <strong>in</strong>troduced <strong>in</strong> 1986. Other biologics used to treat autoimmunediseases such as Crohn’s disease <strong>in</strong>clude the tumor necrosis factor alpha <strong>in</strong>hibitors. The firstanti-TNF drug was <strong>in</strong>fliximab, <strong>in</strong>troduced <strong>in</strong> 1998. Although generally considered safer thanchronic corticosteroid use, the potential for serious side effects can occur.More recently, a new strategy towards the treatment of autoimmune disease has been <strong>in</strong>troduced.This strategy is based on observations that epigenetics may play a role <strong>in</strong> the developmentof autoimmunity. The bulk of experience <strong>in</strong> the use of the epigenetic drugs has so farbeen <strong>in</strong> the treatment of cancer (Box 12.1). This experience has led to a great deal of promisefor a similar application <strong>in</strong> the treatment of autoimmunity. Interest<strong>in</strong>gly, the use of corticosteroids<strong>in</strong> the treatment of these illnesses may be <strong>in</strong>tertw<strong>in</strong>ed with the development ofepigenetic drugs because of the impact of epigenetic drugs on the glucocorticoid receptor[9,10]. Epigenetic drugs may also play a role <strong>in</strong> treatment of other <strong>in</strong>flammatory diseases statessuch as asthma [11,12] as well as other classes of disease, <strong>in</strong>clud<strong>in</strong>g neurologic [13] orpsychiatric [13,14] disorders. The challenges may be different, s<strong>in</strong>ce the target genes and cellsthat have gone awry may be different depend<strong>in</strong>g on disease states, but the pr<strong>in</strong>ciples that leadto the development of epigenetic drugs are similar. A historical and current timel<strong>in</strong>e for thedevelopment of drugs for autoimmune diseases is shown <strong>in</strong> Figure 12.1.12.2 PATHOPHYSIOLOGIC BASIS FOR THE DEVELOPMENT OFEPIGENETIC TREATMENTS IN AUTOIMMUNITYLike many diseases, the pathophysiology of autoimmune diseases may <strong>in</strong>clude both geneticand environmental factors. <strong>Epigenetics</strong> describes changes <strong>in</strong> gene expression which are stableand heritable, but reversible. One of the key features of epigenetics that makes it an attractivetarget for development of new drugs is that the changes <strong>in</strong> gene expression are occasionally


CHAPTER 12Approaches to Autoimmune <strong>Disease</strong>s Us<strong>in</strong>g Epigenetic TherapyBOX 12.1 CATEGORIES OF EPIGENETIC DRUGSHDAC InhibitorsPhenylbutyrateTrichostat<strong>in</strong> ASuberoylanilide hydroxamic acidMS275FK228Valproic acidDNA Methylat<strong>in</strong>g Agents5-azacytid<strong>in</strong>eDecitab<strong>in</strong>eZebular<strong>in</strong>eProca<strong>in</strong>amideProca<strong>in</strong>eHydralaz<strong>in</strong>eEpigallocatech<strong>in</strong>-3-gallate (EGCG) [126]DNA methyltransferase 1 antisense oligonucleotides (DNMT1 ASO) [127]MicroRNAlimited to certa<strong>in</strong> cell types [15], although genome-wide epigenetic changes <strong>in</strong> disease do exist[16]. On the other hand, the knowledge that we need to devise ways to specifically target thegene or cell responsible for the disease is still not available. While global, non-specificepigenetic changes may be easier to <strong>in</strong>duce by epigenetic drugs, and may yet prove to be227FIGURE 12.1Timel<strong>in</strong>e outl<strong>in</strong><strong>in</strong>g significant discoveries <strong>in</strong> autoimmunity drugs.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>cl<strong>in</strong>ically valuable <strong>in</strong> treat<strong>in</strong>g autoimmune diseases, a greater success would arise from theability to target the effect of epigenetic drugs directly to the cells <strong>in</strong> which dysregulation oftranscription occurs. The successful target<strong>in</strong>g of the control of a s<strong>in</strong>gle gene or cell type may beassociated with a lower risk of side effects, s<strong>in</strong>ce genes irrelevant to the disease will be spared.The fact that epigenetic changes are believed to be reversible <strong>in</strong>dicates that drugs known toaffect gene transcription may be used to restore normal transcription and lead to resolution ofcl<strong>in</strong>ical symptoms.The two target areas by which epigenetic modification <strong>in</strong>fluences gene expression are histonemodification and DNA methylation [17]. In autoimmune diseases such as SLE, it is suspectedthat DNA methylation plays a role <strong>in</strong> the disease process. Studies of identical tw<strong>in</strong>s with SLEhave suggested that there is a significant role for non-genetic factors as the highest concordancerate among identical tw<strong>in</strong>s fails to reach 60% [18]. The observation of a global hypomethylationof DNA <strong>in</strong> the T cells of patients with SLE [19] has <strong>in</strong>dicated epigenetic modificationsmay play a role <strong>in</strong> disease pathogenesis and progression. In general, DNA methylationoccurs at the 5 0 position of cytos<strong>in</strong>e <strong>in</strong> CpG d<strong>in</strong>ucleotides located <strong>in</strong> the promoter regions andleads to repression of transcription.228The existence of a role of chromat<strong>in</strong> and histone modification <strong>in</strong> the regulation of geneexpression is a common phenomenon of many cell types and genes. Epigenetic modification is<strong>in</strong>volved <strong>in</strong> the regulation of various pro<strong>in</strong>flammatory cascades responsible for many diseasestates, <strong>in</strong>clud<strong>in</strong>g <strong>in</strong>fection, cancer, and autoimmune diseases. The central event <strong>in</strong> this regulatorynetwork is the activation of nuclear factor kappa light cha<strong>in</strong> enhancer of activated B cells(NFkB). NFkB is a heterodimeric prote<strong>in</strong>, present <strong>in</strong> almost all mammalian cell types, thatregulates DNA transcription. It is at the core of most <strong>in</strong>flammatory processes and its activationis closely l<strong>in</strong>ked to a number of histone acetyltransferases. Histone modification enzymes<strong>in</strong>clude histone acetyltransferases (HAT) or histone deacetylases (HDAC). Histone deacetylasesremove acetyl groups from lys<strong>in</strong>e residues form<strong>in</strong>g compact and condensed chromat<strong>in</strong>which is transcriptionally silenced. Conversely, HAT enzymes acetylate lys<strong>in</strong>e residues andrender chromat<strong>in</strong> less compact and more transcriptionally active. Histone deacetylase <strong>in</strong>hibitors(HDACi) block the action of HDAC, and lead to <strong>in</strong>creased acetylation of nucleosome corehistones (see Figure 12.2). In some cases this leads to transcriptional activation, but manygenes are repressed by HDACi as well. It should also be noted that HDAC activity is not limitedto histone prote<strong>in</strong>s, and other prote<strong>in</strong>s can be deacetylated at the lys<strong>in</strong>e residue as well.DNA methylation and histone modification are processes that work <strong>in</strong> concert with each otherto determ<strong>in</strong>e transcriptional activity [20]. The hallmark of these processes is reversibility,although early on it was not believed to be so. Besides acetylation, histone prote<strong>in</strong>s canundergo other reactions to render the chromat<strong>in</strong> open or closed, and these <strong>in</strong>clude phosphorylation,methylation, ribosylation, sumoylation, and ubiquit<strong>in</strong>ation. The reactions arenot mutually exclusive. The primary site of action is at the histone tail, which is near the am<strong>in</strong>oterm<strong>in</strong>us of the prote<strong>in</strong>. In general, open<strong>in</strong>g the chromat<strong>in</strong>, as occurs through acetylation isassociated with <strong>in</strong>creased gene expression.There are at least 18 known HDACs distributed among four classes of histone deacetylases,I through IV. They act on a variety of cells and signal<strong>in</strong>g pathways to regulate chromat<strong>in</strong>architecture and immunologic function [21]. The class I, II, and IV HDAC enzyme activity isdependent on Zn 2þ ion. Class III HDACs are the sirtu<strong>in</strong>s, which also possess ability to <strong>in</strong>fluenceimmune function [22]. The class I HDAC enzymes are the most widely studied. These aregenerally found <strong>in</strong> the nucleus and regulate the production of <strong>in</strong>flammatory cytok<strong>in</strong>es. Class IIHDAC enzymes translocate from the nucleus to the cytoplasm <strong>in</strong> response to external stimuli.Their primary effect is <strong>in</strong> the regulation of lymphocyte differentiation and activation [23].HDAC enzymes have been found to be able to regulate <strong>in</strong>nate immunity by virtue of theireffects on Toll-like receptor (TLR) signal<strong>in</strong>g. In addition, HDAC can <strong>in</strong>crease the expression of


CHAPTER 12Approaches to Autoimmune <strong>Disease</strong>s Us<strong>in</strong>g Epigenetic TherapyFIGURE 12.2Molecular targets for epigenetic treatment of autoimmune diseases. This figure is reproduced <strong>in</strong> the color plate section.pro<strong>in</strong>flammatory mediators through suppression of <strong>in</strong>hibitors of NFkB such as IKK-b (<strong>in</strong>hibitorof nuclear factor kappa-B k<strong>in</strong>ase subunit beta). It has been shown that HDACs can <strong>in</strong>ducetype I <strong>in</strong>terferon production. HDACs play a role <strong>in</strong> regulatory T cell (Treg) homeostasisthrough cooperative modulation of forkhead box P3 (Foxp3) transcription factor. HDAC7,HDAC9, Tip60, and Foxp3 coexist <strong>in</strong> a complex and together regulate function and stability ofTreg cells [24]. HDACs appear to suppress Treg cell function while acetylation of Foxp3 seemsto augment it. On the other hand, HDACs appear to also suppress cytok<strong>in</strong>e production fromactivated T cells. In a mouse allergy model, deletion of Hdac1 led to exacerbation of airway<strong>in</strong>flammation, mucus hypersecretion, <strong>in</strong>creased airway resistance, and parenchymal lung<strong>in</strong>flammation [25]. Th2 cells stimulated <strong>in</strong> vitro had higher IL-4 production and eos<strong>in</strong>ophilrecruitment was observed as well. Clearly the <strong>in</strong>teraction between histone acetylation andimmune function is highly complex, with oppos<strong>in</strong>g forces act<strong>in</strong>g to ma<strong>in</strong>ta<strong>in</strong> balance <strong>in</strong>immune homeostasis. Abnormalities <strong>in</strong> HDAC activity may lead to either pro<strong>in</strong>flammatorycell activation or may lead to immune suppression. HDAC <strong>in</strong>hibitors may therefore play a role<strong>in</strong> revers<strong>in</strong>g these epigenetic changes <strong>in</strong>duced by HDACs.229In cancer, where most of the experience <strong>in</strong> the efficacy of HDAC <strong>in</strong>hibitors exists, HDAC<strong>in</strong>hibitors have been shown to <strong>in</strong>duce cell cycle arrest, cell differentiation, and apoptotic celldeath of “transformed cells”. If one views synovial hyperplasia <strong>in</strong> rheumatoid arthritis <strong>in</strong> thecontext of “tumor-like” synovial cells that have gone out of control, then many of the applicationsof HDAC <strong>in</strong>hibitors <strong>in</strong> cancer therapy may be extended to autoimmune diseases.12.3 PATHOLOGY OF AUTOIMMUNE DISEASE AND POTENTIALTARGETS FOR EPIGENETIC DRUGSThe pathophysiology of autoimmune diseases is complex and the immunological pathwaysthat are impacted vary depend<strong>in</strong>g on the specific autoimmune disease. However, there are


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>common features that may lend it to strategic target<strong>in</strong>g of epigenetic pharmacotherapeutics.Autoimmune diseases arise as a result of an imbalance <strong>in</strong> the immune system that leads to lossof tolerance to self antigens. The presence of autoreactive T cells and autoantibodies playsa role <strong>in</strong> the disease pathogenesis. Pathologic events that lead to the disease phenotype may<strong>in</strong>clude defects <strong>in</strong> important signal<strong>in</strong>g pathways for apoptosis, <strong>in</strong>flammatory cell activation,mediator release, immune tolerance, and regulatory cell function. T helper lymphocytes, Blymphocytes, dendritic cells, T regulatory cells, and other cell types play a role <strong>in</strong> the pathogenesisof various autoimmune diseases. For each of the diseases, specific cell types may playa role, for example synoviocytes <strong>in</strong> rheumatoid arthritis, lymphocytes <strong>in</strong> systemic lupuserythematosus, or neurons <strong>in</strong> multiple sclerosis.The cytok<strong>in</strong>e profile, which is <strong>in</strong>tricately l<strong>in</strong>ked to the selective activation of various cell types,is also important. Cytok<strong>in</strong>es that have been identified to play a role <strong>in</strong> the development ofautoimmune diseases <strong>in</strong>clude IL-6, TGF-b, IL-17, IL-12, IL-23, IL-2, TNFa, <strong>in</strong>terferon g and thechemok<strong>in</strong>es CXCL9, CXCL10, and CXCL11, which are all Th1-attract<strong>in</strong>g chemok<strong>in</strong>es [26].Recently Th1 and Th17 cells also have been found to play a potential role <strong>in</strong> autoimmunedisease pathogenesis [27,28]. While there may also be many known and as yet unknownpathways, cell l<strong>in</strong>es and humoral factors <strong>in</strong>volved <strong>in</strong> the pathogenesis of autoimmune diseases,the above illustrates the numerous potential po<strong>in</strong>ts of attack for epigenetic drugs. All of thefactors listed above are regulated <strong>in</strong> some way at the DNA level, and control of gene expressionwith epigenetic drugs may allow restoration of normal immune homeostasis and reverse theaberrant function seen <strong>in</strong> patients with autoimmune diseases. The sheer extent of the<strong>in</strong>volvement of multiple pathways, cells, regulatory factors, mediators, and signal<strong>in</strong>g molecules<strong>in</strong>dicates that there are probably numerous redundant pathways <strong>in</strong> the disease process.Box 12.2 shows potential targets for epigenetic drugs <strong>in</strong> autoimmunity.230At a different level, the field of biological modulators also targets the same factors, thus thedevelopment of drugs which antagonize the function of such pro<strong>in</strong>flammatory mediators suchas TNFa. <strong>Epigenetics</strong>, by target<strong>in</strong>g identical pathways, provides a means to regulate thegeneration of these mediators at the DNA transcription level, rather than block<strong>in</strong>g alreadyexcessive production of the mediator. <strong>Epigenetics</strong> may also have the potential to regulate theexpression of more than one <strong>in</strong>flammatory mediator at a time, thus help<strong>in</strong>g to account forredundancy of the immune system.An example of the potential utility of epigenetic drugs <strong>in</strong> treat<strong>in</strong>g autoimmune diseases can beillustrated by first appreciat<strong>in</strong>g the role of regulator T cells <strong>in</strong> the pathogenesis of autoimmunediseases. The development and function of regulatory T cells <strong>in</strong> the human is under the controlof a critical transcription factor known as foxp3. While the precise role of Treg cells <strong>in</strong> thepathogenesis of autoimmune diseases, such as SLE and rheumatoid arthritis, is still <strong>in</strong>completelyunderstood, there is some evidence that the levels of certa<strong>in</strong> phenotypes of Treg cellssuch as CD4þCD25þFoxp3þ T reg cells correlate <strong>in</strong>versely with disease activity [29,30],suggest<strong>in</strong>g a suppressive effect of these cells. Histone deacetylases have been shown to controlthe functions of Treg cells by alter<strong>in</strong>g transcription factors of the foxp3 gene. Acetylation leadsto improved DNA b<strong>in</strong>d<strong>in</strong>g and histone acetyltransferases (HATs) such as Tip60 have theopposite effect; acetylation would lead to <strong>in</strong>creased expression of Foxp3 [31] and resistance toprotease degradation, as would the use of HDAC <strong>in</strong>hibitors. Thus, HDAC <strong>in</strong>hibitors maypotentially augment Treg cell function, and potentially reverse disease progression <strong>in</strong> autoimmunedisorders. Figure 12.3 illustrates how an epigenetic treatment us<strong>in</strong>g HDAC <strong>in</strong>hibitorsmay regulate pro<strong>in</strong>flammatory cytok<strong>in</strong>es and autoimmune disease activity.12.4 HDAC INHIBITORSAs mentioned above, the cl<strong>in</strong>ical data regard<strong>in</strong>g HDAC <strong>in</strong>hibitors is derived primarily fromcancer research. Some previously known drugs have HDAC <strong>in</strong>hibition activity, <strong>in</strong>clud<strong>in</strong>g


CHAPTER 12Approaches to Autoimmune <strong>Disease</strong>s Us<strong>in</strong>g Epigenetic TherapyBOX 12.2 POSSIBLE TARGETS FOR EPIGENETIC REGULATION INLUPUS AND OTHER AUTOIMMUNE DISEASESPathwaysInflammatory signal<strong>in</strong>g pathwaysApoptosis [128]B-cell stimulatory pathways [129]T-cell stimulatory pathways [130]NFkB pathway [119]Glucocorticoid receptor activation [131]Complement systemToll-like receptorGene Clusters/SystemsMHC [132]FCgRsTCR x-cha<strong>in</strong> geneCytok<strong>in</strong>es and Signal<strong>in</strong>g MoleculesCTLA-4 [133]PCD-1 (programmed cell death-1)IL-2 [55]CD40 ligand [134]MMPs [81]IFN-gIL-10CD70 [72]LFA-1 [72]IL-12, IL-23 [35]IL-6 [135]IL-17 [136]TGF-bTNFa [137]IL-18 [138]Cycl<strong>in</strong>-dependent k<strong>in</strong>ase <strong>in</strong>hibitor p21 [139]Chemok<strong>in</strong>es and Chemok<strong>in</strong>e ReceptorsCXCL9 [35]CXCL10 [35]CXCL11 [35]CXCR12 [80]231valproic acid used <strong>in</strong> the treatment of seizures and trichostat<strong>in</strong> A used <strong>in</strong> the treatment offungal <strong>in</strong>fections. Other compounds with HDAC <strong>in</strong>hibition activity <strong>in</strong>clude butyrates such asphenylbutyrate, benzamides, and cyclic peptides such as depsipeptides and apicid<strong>in</strong> [32].Newer HDAC <strong>in</strong>hibitors such as suberoylanilide hydroxamic acid (SAHA) are available for thetreatment of cancers, but no <strong>in</strong>dication has been established for the use of these drugs <strong>in</strong>treat<strong>in</strong>g autoimmune diseases. However, as illustrated above, the role of HDACs <strong>in</strong> immunefunction is significant. In fact, multiple studies have documented effects of HDAC <strong>in</strong>hibitorson immune response. HDAC <strong>in</strong>hibitors have been shown to play a role <strong>in</strong> the reduction ofexpression of costimulatory molecules <strong>in</strong> antigen presentation by dendritic cells [33]. Othereffects of HDAC <strong>in</strong>hibitors <strong>in</strong>clude a reduction <strong>in</strong> IL12 and IL6 by dendritic cells andmacrophages [34]. Expression of Th cell chemok<strong>in</strong>es CXCL9 and CXCL10 is also reduced.HDAC <strong>in</strong>hibitors have also been found to impair differentiation of Th1 and Th17 cells [35].Suberoylanilide hydroxamic acid (SAHA) is a HDAC <strong>in</strong>hibitor that has been used successfully<strong>in</strong> the treatment of gliomas, cutaneous T-cell lymphoma, Sezary syndrome, and other solid


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>FIGURE 12.3Potential effects of epigenetic changes <strong>in</strong> autoimmune diseases. This figure is reproduced <strong>in</strong> the color plate section.232cancers [36]. A recent study also suggests a role <strong>in</strong> the treatment of lung fibrosis, <strong>in</strong>dicat<strong>in</strong>g thatSAHA may play a role <strong>in</strong> regulation of transcription of genes encod<strong>in</strong>g pro<strong>in</strong>flammatorycytok<strong>in</strong>es [37]. Both SAHA and MS275 have been shown to have antirheumatic properties <strong>in</strong>a rat model of collagen-<strong>in</strong>duced arthritis [38]. MS275, an HDAC1 selective <strong>in</strong>hibitor, wasfound to have the capability to delay onset of disease <strong>in</strong> rats and mice, and to prevent boneerosion [39].Trichostat<strong>in</strong> A is an HDAC <strong>in</strong>hibitor that has been studied as a potential anticancer drug [40].Trichostat<strong>in</strong> A <strong>in</strong>hibits HDACs 1, 3, 4, 6, and 10. From a global hypomethylation standpo<strong>in</strong>t,patients with SLE display an altered level of gene expression than healthy controls. Differences<strong>in</strong> DNA methylation are tied to activity of histone acetylation and methylation, DNA methylation,gene position<strong>in</strong>g, and their <strong>in</strong>teractions. The histone code, represent<strong>in</strong>g posttranslationalmodification and position<strong>in</strong>g of histones, is one of the master regulators of DNAmethylation and gene expression [41,42]. It has been demonstrated <strong>in</strong> a MRL-lpr/lpr mousemodel of SLE that trichostat<strong>in</strong> A can alter site-specific hypoacetylation of histones H3 and H4,and this is associated with an improvement <strong>in</strong> disease phenotype [43]. The authors <strong>in</strong>terpretedthis as a resett<strong>in</strong>g of an aberrant “histone code” that was present <strong>in</strong> diseased mice. It isimportant to emphasize that while these observations provide optimism for the potential useof this agent <strong>in</strong> treat<strong>in</strong>g SLE, much is still unknown, <strong>in</strong>clud<strong>in</strong>g how a global hypomethylationaffects suppression of anti-lupus genes and stimulation of pro-lupus genes.Trichostat<strong>in</strong> A has also been found to be able to stimulate overexpression of the genesencod<strong>in</strong>g for cycloxygenase-2 (Cox-2) and CXCL12 <strong>in</strong> mouse macrophages. On the otherhand, it suppresses expression of genes cod<strong>in</strong>g for other immunostimulatory molecules suchas TNFa, IL-6, IL-12p40, CCL2/MCP1, CCL17, and Endothel<strong>in</strong>-1 [44,45]. Along with SAHA, itwas also found to reduce costimulatory molecule expression <strong>in</strong> dendritic cells. Bosisio showedthat TSA could block the production of Th17-polariz<strong>in</strong>g cytok<strong>in</strong>es [35] and Tao showed thatTSA promotes the production and activity of regulatory T cells [23,46]. These two observations


CHAPTER 12Approaches to Autoimmune <strong>Disease</strong>s Us<strong>in</strong>g Epigenetic Therapysuggest that TSA acts on different cell l<strong>in</strong>es to suppress <strong>in</strong>flammation and may play a role <strong>in</strong>hyperactive immune states such as autoimmune diseases.The pathophysiology of rheumatoid arthritis ultimately leads to tissue <strong>in</strong>flammation andpannus, or synovial hyperplasia. This leads to destruction of cartilage and bone. The pathogenesisis mediated through antigen-dependent T-cell activation, followed by <strong>in</strong>filtration of Tand B lymphocytes <strong>in</strong>to synovium and also release of pro<strong>in</strong>flammatory mediators <strong>in</strong>tosynovium by macrophages and fibroblasts. These pro<strong>in</strong>flammatory cytok<strong>in</strong>es lead to proliferationof synovial cells. Historically, the treatment of rheumatoid arthritis has targeted the<strong>in</strong>flammatory response at various levels. Non-steroidal anti-<strong>in</strong>flammatory agents, steroids,DMARDs, and immunosuppressives are all non-selective medications which through their<strong>in</strong>dividual mechanisms of action, act to suppress this <strong>in</strong>flammatory response. Because of this,they each have their own set of adverse side effects, and some are serious, <strong>in</strong>clud<strong>in</strong>g gastro<strong>in</strong>test<strong>in</strong>albleed<strong>in</strong>g, hepatorenal toxicity, bone marrow suppression, etc. Will the use ofepigenetic agents be any different?The unrelated HDAC <strong>in</strong>hibitors trichostat<strong>in</strong> A (TSA) and phenylbutazone were assessed fortheir ability to modulate gene expression <strong>in</strong> rheumatoid arthritis and for their cl<strong>in</strong>ical efficacy<strong>in</strong> an adjuvant arthritis rat model for rheumatoid arthritis. The goal was to <strong>in</strong>vestigate themechanism of action of these agents and to determ<strong>in</strong>e if disease progression can besuppressed. Of <strong>in</strong>terest was the role of regulation of cell cycle <strong>in</strong>hibitors genes p16 INK4 andp21 Cip1 by epigenetic modification. Experiments on gene therapy had already shown that<strong>in</strong>jection of an adenovirus that carries p16 INK4 and p21 Cip1 can <strong>in</strong>hibit synovial fibroblastproliferation. The technique was deemed impractical because it must be adm<strong>in</strong>istered <strong>in</strong>to<strong>in</strong>dividual jo<strong>in</strong>ts and rheumatoid arthritis typically affects multiple jo<strong>in</strong>ts. Because of the roleof HDAC <strong>in</strong>hibitors as chromat<strong>in</strong> modifiers <strong>in</strong> cancer, and because of the role of histoneacetylation <strong>in</strong> up-regulat<strong>in</strong>g cell cycle <strong>in</strong>hibitors, down-regulat<strong>in</strong>g pro<strong>in</strong>flammatory cytok<strong>in</strong>essuch as IL-6, IL-1, TNF-a, and IL8, and because of the parallels between tumorigenesis andsynovial hyperplasia as result<strong>in</strong>g from a failure of gene regulation, it was believed that HDAC<strong>in</strong>hibition may play a role <strong>in</strong> the treatment of rheumatoid arthritis.233In the study by Chung et al., HDAC <strong>in</strong>hibitors were found to be able to up-regulate p16 INK4and p21 Cip1 . Levels of acetylated H3 and H4 were also <strong>in</strong>creased when cultured synovialfibroblasts were <strong>in</strong>cubated with TSA or phenylbutyrate. The effect for p21 Cip1 was reversible,but the <strong>in</strong>creased expression of p16 INK4 <strong>in</strong> synovial fibroblasts from rats with adjuvant arthritiswas susta<strong>in</strong>ed even when HDAC <strong>in</strong>hibitors were removed. Topically applied phenylbutyrateand TSA also <strong>in</strong>duced expression of p21 Cip1 and p16 INK4 <strong>in</strong> the synovium of rats with adjuvantarthritis, but not <strong>in</strong> normal synovial cells. In vivo studies showed that jo<strong>in</strong>t swell<strong>in</strong>g wasimproved <strong>in</strong> rats who were adm<strong>in</strong>istered TSA or PB topically. Other f<strong>in</strong>d<strong>in</strong>gs of this study<strong>in</strong>cluded an ability of 10% PB cream or 1% Trichostat<strong>in</strong> A o<strong>in</strong>tment to suppress paw swell<strong>in</strong>gand pannus formation, and to promote wound heal<strong>in</strong>g. The pro<strong>in</strong>flammatory cytok<strong>in</strong>e TNFawas also suppressed <strong>in</strong> affected tissues <strong>in</strong> the rats with adjuvant arthritis who were treatedwith both TSA and phenylbutyrate topically [47]. A summary of anti-<strong>in</strong>flammatory effects ofHDAC <strong>in</strong>hibitors and other epigenetic drugs <strong>in</strong> rheumatoid arthritis is shown <strong>in</strong> Box 12.3.12.4.1 HDAC Inhibitors <strong>in</strong> Bone HomeostasisThe role of HDAC <strong>in</strong>hibitors <strong>in</strong> the pathogenesis of bone loss that is frequently seen <strong>in</strong> chronicdiseases, <strong>in</strong>clud<strong>in</strong>g autoimmune diseases, is currently be<strong>in</strong>g <strong>in</strong>vestigated. Osteoimmunology isa relatively new field, but recent evidence has demonstrated that cell surface receptors may playa complex and common role <strong>in</strong> bone homeostasis and immunity. The ma<strong>in</strong>tenance of healthybone is a balance between bone formation and bone resorption, which is a dynamic processthat leads to replacement of about 10% of our bone matrix every year. The regulatory factorsthat govern this balance <strong>in</strong>clude growth factors such as Receptor Activator for Nuclear FactorkB Ligand (RANKL) and Colony Stimulat<strong>in</strong>g Factor 1 (CSF-1) 48 . RANKL is necessary for the


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>BOX 12.3 EPIGENETIC AGENT-INDUCED ANTI-INFLAMMATORYEFFECTS IN RHEUMATOID ARTHRITIS*HDAC Inhibitors [140]1. Suppression of IL-6 and TNFa [47]2. Suppression of cytok<strong>in</strong>e gene expression [44]3. Block<strong>in</strong>g of cytok<strong>in</strong>e production from synovial macrophages [140]4. Induction of apoptosis <strong>in</strong> human macrophages [140]5. Induction of apoptosis <strong>in</strong> rheumatoid arthritis synovial macrophages [140]6. Suppression of macrophage expression of Bfl-1 prote<strong>in</strong> [140]DNA Methylation Agents1. Increased expression of CXCL12 [141]2. Potential <strong>in</strong>hibition of CXCL12-<strong>in</strong>duced expression of matrix metalloprote<strong>in</strong>ases [81]3. Potential reduction <strong>in</strong> CXCL12-<strong>in</strong>duced collagenase activity and bone destruction [81]* Other effects seen <strong>in</strong> lupus cells are not shown here.formation of osteoclasts and is thus a promoter of bone resorption. The first monoclonalantibody aga<strong>in</strong>st RANKL, denosumab, is FDA-approved to treat osteoporosis. RANKL is alsopresent on T cells and is believed to be a key factor <strong>in</strong> dendritic cell maturation.234RANKL has been shown to cause substantial bone resorption when adm<strong>in</strong>istered locally tomouse calvariae, but TSA, when coadm<strong>in</strong>istered, was found to <strong>in</strong>hibit this effect by promot<strong>in</strong>gp21-WAF-dependent apoptosis of osteoclasts [49]. TSA and sodium butyrate were also foundto <strong>in</strong>hibit <strong>in</strong> vitro differentiation of mur<strong>in</strong>e bone marrow cultures <strong>in</strong>to osteoclast-like cells[50]. SAHA also <strong>in</strong>hibited RANKL-mediated osteoclastogenesis and potentiated apoptosis bysuppress<strong>in</strong>g NF-kB activation <strong>in</strong> a RAW264 mouse macrophage system. In vitro, SAHA was alsoable to suppress class I and II histone deacetylase activity <strong>in</strong> human osteoblasts [51]. Not allHDAC <strong>in</strong>hibitors were found to suppress osteoclast activity. Interest<strong>in</strong>gly, MS275, which isa class I HDAC <strong>in</strong>hibitor, and 2664.12, a class II HDAC <strong>in</strong>hibitor, failed to show osteoclastactivity suppression by themselves, but when comb<strong>in</strong>ed, were able to show marked levels of<strong>in</strong>hibition. This effect was similar to that seen <strong>in</strong> the broad-spectrum HDAC <strong>in</strong>hibitor 1179.4b,suggest<strong>in</strong>g that both HDAC class I and HDAC class II enzymes need to be suppressed <strong>in</strong> orderfor bone resorption to be <strong>in</strong>hibited.In addition to the effects of HDAC <strong>in</strong>hibitors on osteoclast activity, there is also evidence thatthey may impact osteoblasts as well. The HDAC <strong>in</strong>hibitors valproic acid, sodium butyrate, andMS275 all promoted osteoblast maturation. TSA was shown to enhance the function of matureosteoblasts and enhance the expression of the osteoblast genes, type I collagen, osteopont<strong>in</strong>,osteocalc<strong>in</strong>, and bone sialoprote<strong>in</strong> <strong>in</strong> the calvarial-derived primary osteoblast cell l<strong>in</strong>e MC3T3-E1 52 . Runx2 transcription was also <strong>in</strong>creased, and TSA accelerated matrix m<strong>in</strong>eralization andalkal<strong>in</strong>e phosphatase production <strong>in</strong> MC3T3-E1 cells.These data suggest that HDAC <strong>in</strong>hibitors may play a role <strong>in</strong> treat<strong>in</strong>g osteoporosis related toautoimmune and other diseases. In particular, bone loss <strong>in</strong> rheumatoid arthritis may be anarea where HDAC <strong>in</strong>hibitors may provide the most benefit because of the duplicity of action <strong>in</strong>also re<strong>in</strong><strong>in</strong>g <strong>in</strong> the hyperactive immune response <strong>in</strong> this disease. Much research still needs to bedone to elucidate the efficacy and safety of these agents <strong>in</strong> the treatment of autoimmunedisease and their related morbidities.12.4.2 HDAC Inhibitors <strong>in</strong> the Treatment of Inflammatory Bowel <strong>Disease</strong>The HDAC <strong>in</strong>hibitor butyrate was found to <strong>in</strong>hibit NF-kB-mediated <strong>in</strong>flammation <strong>in</strong>peripheral blood mononuclear cell (PBMC) cultures from <strong>in</strong>test<strong>in</strong>al biopsy specimens ofpatients with Crohn’s disease. Butyrate <strong>in</strong>hibited the production of TNF and LPS <strong>in</strong>duced


CHAPTER 12Approaches to Autoimmune <strong>Disease</strong>s Us<strong>in</strong>g Epigenetic TherapymRNA expression of pro<strong>in</strong>flammatory cytok<strong>in</strong>es, and also blocked translocation of NFkB fromthe cytoplasm to the nucleus. This was believed to be mediated by an <strong>in</strong>hibition of degradationof the NFkB <strong>in</strong>hibitory prote<strong>in</strong> IkB, as the decrease <strong>in</strong> IkB normally seen <strong>in</strong> LPS-<strong>in</strong>duced NFkBactivation was not seen <strong>in</strong> the presence of HDAC <strong>in</strong>hibitor. An additional f<strong>in</strong>d<strong>in</strong>g was that<strong>in</strong> vivo adm<strong>in</strong>istration of butyrate to a tr<strong>in</strong>itrobenzene sulfonic acid (TNBS) experimentalcolitis model <strong>in</strong> rats reduced <strong>in</strong>flammation [53]. The authors were not certa<strong>in</strong> this effect wasdue to HDAC <strong>in</strong>hibitor activity because the same effect was not seen with TSA.12.4.3 HDAC Inhibitors <strong>in</strong> the Treatment of Multiple SclerosisThe efficacy of HDAC <strong>in</strong>hibitors <strong>in</strong> treat<strong>in</strong>g multiple sclerosis may be related to severaldifferent mechanisms. The existence of the experimental autoimmune encephalomyelitis(EAE) animal model for MS is particularly useful <strong>in</strong> study<strong>in</strong>g various novel treatments. It hasbeen demonstrated that <strong>in</strong>terferon-g plays a role <strong>in</strong> pathogenesis by virtue of its ability to<strong>in</strong>duce antigen-present<strong>in</strong>g cells to secrete IL-12, which facilitates the differentiation of naïveTh0 cells to Th1 cells [54]. As mentioned above, Th1 cells may play a role <strong>in</strong> autoimmunediseases, and they are known to secrete IL-2 , a potent pro<strong>in</strong>flammatory cytok<strong>in</strong>e. HDAC<strong>in</strong>hibitors, particularly TSA, <strong>in</strong>hibit gene expression of IL-2 [55,56]. HDAC <strong>in</strong>hibitors have alsobeen found to <strong>in</strong>crease expression of IL-10 <strong>in</strong> Th2 cells <strong>in</strong> EAE, but the effect was opposite <strong>in</strong>SLE cells. When NFkB is associated with HAT, gene expression <strong>in</strong>creases, but it decreases whenassociated with HDACs [57]. NFkB is normally bound to IkB <strong>in</strong> the cytoplasm, and upondegradation of the <strong>in</strong>hibitory prote<strong>in</strong>, NFkB is released to translocate to the nucleus. HDAC<strong>in</strong>hibitors such as butyrate <strong>in</strong>terfere with degradation of IkB, lead<strong>in</strong>g to sequestration of NFkB<strong>in</strong> the cytoplasm and <strong>in</strong>hibition of NF-kB-mediated <strong>in</strong>flammatory responses [53,58,59].12.4.4 Epigenetic Regulation of the Glucocorticoid Receptor andRelated Immune RegulationGlucocorticoids have been used s<strong>in</strong>ce the 1950s <strong>in</strong> the treatment of many diseases. Glucocorticoidshave been shown to have anti-<strong>in</strong>flammatory effects <strong>in</strong> the treatment of asthma,autoimmune diseases, neoplastic diseases, adverse effects of transplantation and many others.The mechanism of action is through reversal of histone acetylation of activated <strong>in</strong>flammatorygenes. The effects of glucocorticoids on the glucocorticoid receptor (GR) are complex anddose-dependent. The anti-<strong>in</strong>flammatory effect of glucocorticoids is mediated through b<strong>in</strong>d<strong>in</strong>gof liganded GRs to coactivators and by recruitment of the class I HDAC1 and HDAC2, to theactivated transcription complex. The result is activation of GR which translocates to thenucleus, and <strong>in</strong> the nucleus, the GR can either stimulate or <strong>in</strong>hibit gene transcription. Studieshave shown that at low concentrations, GR reduces gene transcription of NFkB and AP-1associated <strong>in</strong>flammatory genes. GR can also b<strong>in</strong>d to specific DNA elements known as glucocorticoidresponse elements (GREs), which then results <strong>in</strong> <strong>in</strong>hibition of pro<strong>in</strong>flammatorygenes such as IL-6 [60].235This mechanism has been shown to play a role <strong>in</strong> oxidative stress-mediated resistance to theanti-<strong>in</strong>flammatory effects of glucocorticoids, such as <strong>in</strong> smok<strong>in</strong>g. Because glucocorticoids acton the NFkB pathway to mediate their anti-<strong>in</strong>flammatory effects through HDAC2 recruitment,the possibility of us<strong>in</strong>g HDAC2 to restore glucocorticoid sensitivity <strong>in</strong> conditions of glucocorticoidresistance may prove to be a way of limit<strong>in</strong>g glucocorticoid side effects.The question rema<strong>in</strong>s as to whether or not these epigenetic drugs can be used safely as anti<strong>in</strong>flammatoryagents <strong>in</strong> the treatment of autoimmune and other immunological diseases.Trichostat<strong>in</strong> A has been found to attenuate airway <strong>in</strong>flammation <strong>in</strong> a mouse model for asthma[61], and SAHA antirheumatic effects further suggest that this is possible, but aga<strong>in</strong>, the actuallikelihood of side effects of these agents is completely unknown at this time.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>The world of HDAC <strong>in</strong>hibitors is a rapidly chang<strong>in</strong>g one, with new agents cont<strong>in</strong>ually be<strong>in</strong>gdeveloped. Most cl<strong>in</strong>ical trials currently be<strong>in</strong>g conducted are focused on the efficacy and safetyof these drugs <strong>in</strong> treat<strong>in</strong>g cancer. Some of the HDAC <strong>in</strong>hibitors undergo<strong>in</strong>g laboratory<strong>in</strong>vestigation or cl<strong>in</strong>ical trials <strong>in</strong>clude romideps<strong>in</strong> and panob<strong>in</strong>ostat <strong>in</strong> the treatment ofcytotoxic T-cell lymphomas [62,63], CI-994 <strong>in</strong> the treatment of cancers, MS-275 <strong>in</strong> thetreatment of myelodysplastic syndromes [64], BML-210 and NVP-LAQ824, both antileukemicagents, M344, a benzamide with potential application for the treatment of sp<strong>in</strong>al muscularatrophy, Mocet<strong>in</strong>ostat <strong>in</strong> solid tumors and hematologic cancers [65], and PXD101, a hydroxamatetype HDAC <strong>in</strong>hibitor undergo<strong>in</strong>g cl<strong>in</strong>ical trials <strong>in</strong> the treatment of ovarian and othersolid tumors [66]. This list is by no means all-<strong>in</strong>clusive. The development of HDAC <strong>in</strong>hibitors<strong>in</strong> the treatment of autoimmune diseases is <strong>in</strong> its early phases, and lags beh<strong>in</strong>d progress made<strong>in</strong> the oncologic and hematologic diseases.12.5 DNA METHYLATION AND DNA METHYLTRANSFERASESThe other potential target for epigenetic drugs is at the level of DNA methylation. In thegenome of most vertebrates, <strong>in</strong>clud<strong>in</strong>g humans, most CG di-nucleotide sequences, the primarytarget of DNA methylation, are methylated. DNA methylation is associated <strong>in</strong> vertebrates withchromat<strong>in</strong> structure. In general, hypomethylation is associated with active chromat<strong>in</strong> [67]. Themethylation of genes has been shown to be an important mechanism by which genes areregulated or silenced.236DNA methylat<strong>in</strong>g agents have been used successfully <strong>in</strong> the treatment of cancer. These <strong>in</strong>clude5-aza-cytid<strong>in</strong>e, decitab<strong>in</strong>e, zebular<strong>in</strong>e, proca<strong>in</strong>amide, proca<strong>in</strong>e, EGCG, and DNMT1 ASO [68].DNA methylation is generally thought to be associated with transcription repression, whilehypomethylation usually has a stimulatory role and leads to activation of genes. The consequenceof this gene activation <strong>in</strong> the context of autoimmune diseases is that this may lead toautoreactivity. But not all genes that are overexpressed as a result of DNA hypomethylation areimmunostimulatory <strong>in</strong> nature.The methylat<strong>in</strong>g enzymes that ma<strong>in</strong>ta<strong>in</strong> global methylation of DNA <strong>in</strong>clude theDNA(Cytos<strong>in</strong>e-5) methyltransferases DNMT1, DNMT2, and DNMT3A and 3B. DNMT1 is theone that is most well studied. It is constitutively expressed and it has been shown that itsactivity is decreased <strong>in</strong> the CD4þ cells of patients with SLE. Correspond<strong>in</strong>g low levels ofDNMT mRNA are also seen <strong>in</strong> SLE patients [69]. Inhibition of DNA methylation can lead tooverexpression of genes that may be <strong>in</strong>volved <strong>in</strong> the pathogenesis of lupus, <strong>in</strong>clud<strong>in</strong>g ITGAL[70], perfor<strong>in</strong> [71] and CD70 [72], perhaps via a mechanism that <strong>in</strong>volves decreased T-cell ERKpathway signal<strong>in</strong>g [73]. It is presumed that while the demethylation seen <strong>in</strong> SLE patients isa global phenomenon, it is genes that play a role <strong>in</strong> the pathogenesis of SLE that are of <strong>in</strong>terestwhen develop<strong>in</strong>g methylat<strong>in</strong>g drugs as pharmacotherapeutic agents.Besides DNA methyltransferases, other prote<strong>in</strong>s may play a role <strong>in</strong> the active methylation ofDNA. These <strong>in</strong>clude the methyl-CpG-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s MBD2 and MBD4. It has beensuggested that these “demethylat<strong>in</strong>g” prote<strong>in</strong>s may <strong>in</strong> fact be the ma<strong>in</strong> reason for the DNAhypomethylation <strong>in</strong> SLE patients, as it has been observed that MBD2 mRNA transcript levelswere elevated <strong>in</strong> SLE patients [74]. MBD4 transcripts were observed to be elevated <strong>in</strong> SLEpatients. Thus, MBD2 and MBD4 may be the primary regulators of DNA hypomethylation <strong>in</strong>patients with SLE [75,76].Certa<strong>in</strong> drugs can cause a condition known as drug-<strong>in</strong>duced lupus. The mechanism by whichDNA methylation occurs <strong>in</strong> this group of conditions may vary from drug to drug [77,78].Proca<strong>in</strong>amide, one of the earlier drugs associated with lupus, is a competitive DNA methyltransferase(DNMT) <strong>in</strong>hibitor. Another demethylat<strong>in</strong>g drug is hydralaz<strong>in</strong>e, though viaa different mechanism. Hydralaz<strong>in</strong>e <strong>in</strong>hibits ERK pathway signal<strong>in</strong>g and thereby <strong>in</strong>hibitsDNMT activity, lead<strong>in</strong>g to the DNA hypomethylation that is also seen <strong>in</strong> idiopathic lupus [78].


CHAPTER 12Approaches to Autoimmune <strong>Disease</strong>s Us<strong>in</strong>g Epigenetic TherapyThe DNA hypomethylation result<strong>in</strong>g from both pathways leads to <strong>in</strong>creased transcription ofgenes cod<strong>in</strong>g for <strong>in</strong>flammatory prote<strong>in</strong>s, <strong>in</strong>clud<strong>in</strong>g LFA-1 and CD70, which <strong>in</strong> turn leads to<strong>in</strong>creased T-cell autoreactivity and a breakdown <strong>in</strong> peripheral tolerance.In mice, 5-azacytid<strong>in</strong>e can <strong>in</strong>duce lupus. Azacytid<strong>in</strong>e is a cytos<strong>in</strong>e analog that is <strong>in</strong>corporated<strong>in</strong>to DNA dur<strong>in</strong>g synthesis, and treatment of mice with this drug leads to a genome-widehypomethylation. Karouzakis has shown that 5-azacytid<strong>in</strong>e <strong>in</strong>creases expression of CXCL12and decreased methylation of CpG nucleotides <strong>in</strong> the promoter region of CXCL12 <strong>in</strong> rheumatoidarthritis synovial fibroblasts. CXCL12, or stromal-derived factor-1 (SDF-1) plays a role<strong>in</strong> the traffick<strong>in</strong>g of progenitor cells and is activated <strong>in</strong> response to tissue damage. It isa mediator of chronic <strong>in</strong>flammation by attraction of lymphocytes and monocytes <strong>in</strong>to jo<strong>in</strong>ts <strong>in</strong>patients with rheumatoid arthritis [79,80]. It also appears to <strong>in</strong>crease expression of matrixmetalloprote<strong>in</strong>ases, which <strong>in</strong>duce collagenase activity and may be significant mediators ofjo<strong>in</strong>t destruction <strong>in</strong> rheumatoid arthritis [81].miRNAs, which will be discussed later <strong>in</strong> the article, may be <strong>in</strong>tricately l<strong>in</strong>ked with DNAmethylation patterns as well. miRNAs have been l<strong>in</strong>ked to regulation of DNA <strong>in</strong> the cells ofpatients with lupus. Two microRNAs which show an ability to down-regulate prote<strong>in</strong> levels ofDNMT1. This leads to relatively hypomethylated CD4 þ T cells. S<strong>in</strong>ce these two miRNAs,miRNA-21 and miRNA-148 are both found <strong>in</strong> abundance <strong>in</strong> the CD4 þ T cells of animals andhumans with lupus, the hypomethylated state may expla<strong>in</strong> the <strong>in</strong>crease <strong>in</strong> autoimmunerelatedcellular markers such as CD70 and LFA-1. In this study, <strong>in</strong>hibition of miRNA-148 andmiRNA-21 resulted <strong>in</strong> reversal of the hypomethylation <strong>in</strong> CD4 þ T cells [82,83].This observation that drugs can lead to the global hypomethylation seen <strong>in</strong> drug-<strong>in</strong>ducedlupus has driven research <strong>in</strong>to develop<strong>in</strong>g drugs that can do the opposite, i.e. deactivatetranscription of those genes that lead to autoreactivity. But the epigenetic dysregulation ofDNA methylation <strong>in</strong> the T cells of patients with SLE is <strong>in</strong>deed a complex process. Many genescan be affected, some with more cl<strong>in</strong>ical significance than others.23712.6 MicroRNAMicroRNA is another avenue of research with a potential impact on epigenetic therapeutics.While microRNAs are not recognized universally as an epigenetic phenomenon, it has beenshown that miRNAs can regulate DNA methylation. Autoimmune diseases <strong>in</strong>volve a dysregulationof the immune system lead<strong>in</strong>g to the production of antibodies to self-antigens. Theseantibodies, along with other changes <strong>in</strong> cellular immunity generate an aberrant <strong>in</strong>flammatoryresponse. The result of this response is the destruction or abnormal function of tissues andorgans to which these antibodies are directed. MicroRNAs are non-cod<strong>in</strong>g RNAs about 21e23nucleotides long that function as master regulators or post-transcriptional regulators of geneexpression. These extremely important molecules were discovered <strong>in</strong> the early 1990s and playa role <strong>in</strong> the regulation of approximately half of all prote<strong>in</strong>-cod<strong>in</strong>g genes <strong>in</strong> mammals [84].miRNAs exert their action by b<strong>in</strong>d<strong>in</strong>g to partially complementary nucleotides <strong>in</strong> the 3 0 -untranslated region of messenger RNA, and exert their action by <strong>in</strong>hibition of translation ordegradation of RNA [82]. In order to facilitate this b<strong>in</strong>d<strong>in</strong>g, “mature” miRNAs are loaded ontomiRNA-<strong>in</strong>duced silenc<strong>in</strong>g complexes (miRISCs), which will then f<strong>in</strong>d a target mRNAcomplementary to the “seed sequence” on the mature RNA. This sequence generally recognizescomplementary RNA at the 3 0 UTR region of the messenger RNA.miRNAs may be degraded after exert<strong>in</strong>g their effects on gene expression, but it has beenrecently reported that miRNAs may be stably expressed [85]. Because they are <strong>in</strong>volved <strong>in</strong> thereversible modification of gene expression, they may be considered an epigenetic phenomenon.S<strong>in</strong>ce their discovery and the recognition of their importance <strong>in</strong> gene expression, thetargets of miRNA have been hotly sought after. The number of miRNAs identified has grown toover 800, and one mRNA may have multiple gene targets. They are now known to play a role <strong>in</strong>


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>the regulation of every facet of cellular activity, affect<strong>in</strong>g all cell types and tissues. Their role <strong>in</strong>autoimmune diseases has been supported by the f<strong>in</strong>d<strong>in</strong>g that there is aberrant expression ofmiRNAs <strong>in</strong> many autoimmune diseases. The normal up-regulation or down-regulation of theexpression of several specific miRNAs appears to be critical for the ma<strong>in</strong>tenance of immunehomeostasis. Defects <strong>in</strong> balanc<strong>in</strong>g elements of our immune system can either lead toa hyperactive or hypoactive immune system, the former lead<strong>in</strong>g to allergies or autoimmunity,and the latter lead<strong>in</strong>g to defects <strong>in</strong> immune surveillance and <strong>in</strong> protection aga<strong>in</strong>st <strong>in</strong>fection.238A defect <strong>in</strong> a particular miRNA may be responsible for a target gene cod<strong>in</strong>g for prote<strong>in</strong>s that are<strong>in</strong>volved <strong>in</strong> the pathogenesis of multiple autoimmune diseases, thus this may be a potentialreason for the “overlap” syndrome, whereby <strong>in</strong>dividuals with one autoimmune disease aresusceptible to others as well. Indeed, certa<strong>in</strong> miRNAs have been identified to be directedaga<strong>in</strong>st targets that code for mediators and signal<strong>in</strong>g molecules responsible for the pathogenesisof autoimmune diseases. The computer age has ushered <strong>in</strong> new methodologies andtechnologies to identify miRNA targets. The development of algorithms that are based onevolutionary conservation and seed sequences, leads to the “narrow<strong>in</strong>g down” of predictedtargets for a particular miRNA [86]. But this still leaves hundreds or thousands of possibilities.The identification of biologically relevant prote<strong>in</strong>s can further be facilitated by additionalcomputational approaches that utilize known <strong>in</strong>formation on gene ontology and study ofpotential target genes [87]. This can be followed by laboratory studies that utilize microarrayanalysis techniques <strong>in</strong> conjunction with miRNA knockdown or overexpression to identifyfunction. An example would be a study by Zhao et al., who <strong>in</strong>vestigated the relationshipbetween miRNA-126, DNA methylation, and immune activity [88]. They were able to showthat <strong>in</strong> CD4 þ T cells from patients with systemic lupus erythematosus, miRNA-126 is highlyoverexpressed, and this correlated <strong>in</strong>versely with DNMT1 prote<strong>in</strong> levels. Lower DNMT prote<strong>in</strong>levels leads to DNA hypomethylation and <strong>in</strong>creased gene transcription of immune-relatedgenes LFA-1 and CD20. The authors showed that <strong>in</strong> vitro knockdown of miRNA-126 <strong>in</strong> CD4 þcells from lupus patients resulted <strong>in</strong> decreased autoimmune activity, and decreased IgGproduction from B cells.Other methods of identify<strong>in</strong>g miRNA targets <strong>in</strong>clude proteomic analysis. The technique<strong>in</strong>volves stable isotope label<strong>in</strong>g with am<strong>in</strong>o acids <strong>in</strong> cell culture (SILAC) and analysis of thenuclear proteome after overexpression of a particular miRNA. Match<strong>in</strong>g of changes <strong>in</strong> miRNAexpression with prote<strong>in</strong> levels can provide clues to the target genes of miRNAs. Othercomputational and laboratory techniques exist to correlate miRNA with gene targets. These<strong>in</strong>clude gene network analysis, identification of miRISC-associated miRNA targets, identificationof miRISC-bound MREs and software algorithms utiliz<strong>in</strong>g gene ontology and <strong>in</strong>teractomeanalysis. Further discussion of these techniques is beyond the scope of this chapter.An understand<strong>in</strong>g of the factors that play a role <strong>in</strong> disease pathogenesis is important to thedevelopment of treatment strategies. S<strong>in</strong>ce it is now likely that miRNAs may play a role <strong>in</strong>disease pathogenesis of autoimmune diseases, one might consider treatments that can eitherreverse the disease process or replenish miss<strong>in</strong>g regulatory factors. The adm<strong>in</strong>istration ofdisease-relevant miRNAs to treat patients with autoimmune disease has been <strong>in</strong>vestigated <strong>in</strong>animal models. In mice with autoantibody-mediated arthritis, up-regulation of Bcl-2 <strong>in</strong>terfereswith normal apoptosis. It was shown that adm<strong>in</strong>istration of <strong>in</strong>trarticular miRNA-15a <strong>in</strong>to thesynovium leads to <strong>in</strong>duction of apoptosis. In this study, miRNA was found to be able to reverseBcl-2 <strong>in</strong>duced suppression of normal apoptosis [89]. The cl<strong>in</strong>ical utility of this methodology isstill under <strong>in</strong>vestigation.A number of miRNAs play an important role <strong>in</strong> autoimmune diseases. MiRNA-181 plays a role<strong>in</strong> the development of immune tolerance by sett<strong>in</strong>g the threshold for TCR signal<strong>in</strong>g dur<strong>in</strong>g T cellmaturation <strong>in</strong> the thymus [90]. Positive and negative selection <strong>in</strong> the thymus dictates thesubsequent T-cell reactivity towards that antigen. If the threshold is set low, as seen with highmiRNA-181 expression, then the T-cell response will be enhanced. Conversely low miRNA-181


CHAPTER 12Approaches to Autoimmune <strong>Disease</strong>s Us<strong>in</strong>g Epigenetic Therapyexpression leads to tolerance. miRNA-181 also has effects on B cells, its expression lead<strong>in</strong>g to an<strong>in</strong>creased fraction of B l<strong>in</strong>eage cells <strong>in</strong> vitro <strong>in</strong> tissue culture differentiation systems and <strong>in</strong> vivo <strong>in</strong>a mouse model [91]. Other miRNAs <strong>in</strong>volved <strong>in</strong> immune function <strong>in</strong>clude miRNA-101, miRNA-146a, and miRNA-155. miRNA-146a is a master regulator of cytok<strong>in</strong>e production, <strong>in</strong>clud<strong>in</strong>gTNF-a, which is elevated <strong>in</strong> SLE. miRNA-155 is an important factor <strong>in</strong> autoimmunity, and playsa role <strong>in</strong> promot<strong>in</strong>g the development of T helper cells Th1 and Th17. MiRNA-155 <strong>in</strong>creasesthe presence of CD4 þ CD25 þ Foxp3 þ T reg cells <strong>in</strong> a mouse model (MRL/lpr) of systemic lupuserythematosus. These Treg cells have reduced suppressive ability, and one of the targets ofmiRNA-155 is CD62L. The effects of miRNA-155 <strong>in</strong> RA patients are discussed below.12.6.1 miRNAs <strong>in</strong> Systemic Lupus Erythematosus (SLE)A study identify<strong>in</strong>g abnormal expression <strong>in</strong> CD4 þ T cells <strong>in</strong> SLE revealed 11 miRNA candidates,some of which were up-regulated and some of which were down-regulated. MiRNA-126 <strong>in</strong>particular was overexpressed. The overexpression correlated with decreased DNMT1 levels.This was demonstrated to be a direct effect. miRNA-126 was found to <strong>in</strong>hibit DNMT1 translation.In CD4þ T cells from patients with SLE, up-regulation of miRNA-126 was associatedwith a reduced activity of DNMT1. Reduced activity of DNMT1 leads to reduced methylationof the immune-related genes TNFSF7 and ITGAL, which code for CD70 and CD11a respectively.This leads to <strong>in</strong>creased expression of CD70 and CD11a and subsequently to <strong>in</strong>creased B-and T-cell autoreactivity [70]. CD70 is the ligand for CD27, which <strong>in</strong>duces B-cell costimulatoryfunction and results <strong>in</strong> an <strong>in</strong>crease <strong>in</strong> IgG. CD11a is also known as lymphocyte functionassociatedantigen and plays a role <strong>in</strong> adhesion of T cells to other immune cells [92].More <strong>in</strong>terest<strong>in</strong>gly, <strong>in</strong>hibition of the overexpression of miRNA-126 <strong>in</strong> SLE patients led torestoration of DNMT1 levels and <strong>in</strong>creased methylation of the ITGAL and TNFSF7 promoter.The consequence of this is a reduction <strong>in</strong> the generation of CD11a and CD70 promoter activity,decreas<strong>in</strong>g T- and B-cell autoreactivity and a potential therapy for SLE [93]. While the earlyresults <strong>in</strong> these <strong>in</strong> vitro studies are promis<strong>in</strong>g, and demonstrate that DNMT1 is a direct target ofmiRNA-126, it is still a long way from us<strong>in</strong>g miRNA-126 or an <strong>in</strong>hibitor of miRNA-126 forcl<strong>in</strong>ical applications. The effects on DNA hypomethylation are not limited to microRNA-126.Other miRNA can also directly <strong>in</strong>duce hypomethylation, <strong>in</strong>clud<strong>in</strong>g miRNA-21 and miRNA-148a [94].239Another miRNA that is <strong>in</strong>volved <strong>in</strong> SLE is miRNA-146a, which is down-regulated <strong>in</strong> SLE andwhich targets TNF receptor-associated factor 6 (TRAF6) and IL-1 receptor associated k<strong>in</strong>ase 1(IRAK1), both of which are <strong>in</strong>volved <strong>in</strong> the activation of NF-kB. Type-1 <strong>in</strong>terferons (IFN) havebeen shown to play a role <strong>in</strong> the pathogenesis of SLE [95]. An IFN score is formulated by theactivity of IFN signature genes, MX1, OAS1, and LY6E [96]. Tang et al. showed a correlationbetween low miR-146a and high <strong>in</strong>terferon expression, which are both seen <strong>in</strong> patients withhigh SLE disease activity, especially with <strong>in</strong>volvement of renal disease [96]. miRNA-146a is also<strong>in</strong>volved <strong>in</strong> many other immune regulatory effects, play<strong>in</strong>g a role <strong>in</strong> Treg suppressor functions.miRNA146a is also a suppressor of apoptosis <strong>in</strong> T cells, and regulates levels of pro<strong>in</strong>flammatorycytok<strong>in</strong>es such as IL-2 [97].miRNA-125a has also been found to be relevant <strong>in</strong> the pathogenesis of SLE. Levels of miRNA-125 are reduced <strong>in</strong> the mononuclear cells <strong>in</strong> the blood of patients with SLE, result<strong>in</strong>g <strong>in</strong>elevated expression of KLF13 and subsequent overexpression of CCL5. This chemok<strong>in</strong>e isassociated with <strong>in</strong>flammatory activity <strong>in</strong> SLE, <strong>in</strong>clud<strong>in</strong>g nephritis and arthritis [98].12.6.2 miRNAs <strong>in</strong> Rheumatoid Arthritis (RA)One of the miRNAs with the greatest potential for evolv<strong>in</strong>g <strong>in</strong>to a therapeutic for the treatment ofrheumatoid arthritis is miRNA-155 [99]. The immune effects attributed to miRNA-155 <strong>in</strong>cludea lack of develop<strong>in</strong>g collagen-<strong>in</strong>duced arthritis when miRNA-155 is knocked out <strong>in</strong> mice.MiRNA-155 knockout mice <strong>in</strong> a K/BxN serum transfer arthritis model exhibited a reduction <strong>in</strong>


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>prote<strong>in</strong> expression of IL-6, IL-17, and IL-22, along with a reduction <strong>in</strong> pathogenic T and B cells[100]. Decreased numbers of osteoclasts result <strong>in</strong> less bone destruction. This observation suggeststhat <strong>in</strong>hibition of miRNA-155 may play a potential role <strong>in</strong> the treatment of RA.miRNAs that have been shown to be up-regulated <strong>in</strong> RA <strong>in</strong>clude miRNA-223 and miRNA-346.Up-regulation of miRNA-146a is also observed <strong>in</strong> RA, but the targets of this miRNA, TRAF6and IRAK1, are not overexpressed <strong>in</strong> patients compared with controls. On the other hand,TNF-a regulation appears to be impaired <strong>in</strong> these patients [101]. It has been noted also that upregulationof miRNA146a/b is associated with an overexpression of IL-17 [102]. OthermiRNAs that may play a role <strong>in</strong> RA <strong>in</strong>clude miRNA-124a and miRNA-203.12.6.3 miRNAs <strong>in</strong> Multiple Sclerosis, Scleroderma, Sjögren’s Syndrome,Polymyositis, Dermatomyositis, and Primary Biliary CirrhosisThe pathogenesis of scleroderma <strong>in</strong>volves the replacement of normal tissue with collagen-richextracellular matrix by aberrant function<strong>in</strong>g of fibroblasts. Important cytok<strong>in</strong>es that lead to<strong>in</strong>creased proliferation and activation of fibroblasts <strong>in</strong>clude IL-4, platelet-derived growth factor(PDGF)-b, and transform<strong>in</strong>g growth factor (TGF)- b[103]. The activity of these cytok<strong>in</strong>escorrelates <strong>in</strong>versely with levels of miR-29a. miRNAs found to be important <strong>in</strong> polymyositis ordermatomyositis <strong>in</strong>clude miRNA-146b, miRNA-155, miRNA-214, miRNA-221, and miRNA222 [104].240Relevant miRNAs <strong>in</strong> Sjögren’s syndrome <strong>in</strong>clude the miRNA-17-92 cluster. MiRNAs are alreadybe<strong>in</strong>g used as biomarkers <strong>in</strong> Sjögren’s syndrome [105]. miRNA-146a is also important <strong>in</strong>Sjögren’s syndrome <strong>in</strong> that levels are <strong>in</strong>creased <strong>in</strong> Sjögren’s syndrome compared with healthcontrols. The effects of miRNA-146a have already been discussed, but functional assaysrevealed that miRNA-146a significantly up-regulated phagocytic activity <strong>in</strong> human monocyticTHP-1 cells and negatively regulated production of TNF-a, IL-1b, MIP-1a, IP-10, and IL-6[106].While some might consider that miRNAs are not strictly epigenetic agents, they meritdiscussion here as epigenetic approaches to treatment of autoimmune diseases because of thesignificant role they play <strong>in</strong> autoimmunity, and because of recent evidence show<strong>in</strong>g thatvariations <strong>in</strong> miRNA expression are directly related to DNMT expression and alterations <strong>in</strong>DNA methylation. The mechanism for this association is still not entirely clear.In multiple sclerosis, miRNA-326 is up-regulated <strong>in</strong> patients with relaps<strong>in</strong>geremitt<strong>in</strong>gdisease, and studies us<strong>in</strong>g the experimental autoimmune encephalomyelitis (EAE) mousemodel showed that up-regulation of miRNA-326 is l<strong>in</strong>ked to <strong>in</strong>creased Th-17 cell number,and cl<strong>in</strong>ically to <strong>in</strong>creased severity of EAE symptoms [107]. Other miRNAs that are upregulated<strong>in</strong> active multiple sclerosis lesions <strong>in</strong>clude miRNA-34a and miRNA-155, whichis also up-regulated <strong>in</strong> a number of other autoimmune diseases. A recent report suggeststhat these miRNAs function by <strong>in</strong>duc<strong>in</strong>g CD47 to reduce <strong>in</strong>hibitory control signal<strong>in</strong>g ofmacrophages [108].In end-stage PBC, 35 different miRNAs were expressed <strong>in</strong> the liver. Down-regulation ofmiRNA-122a and miRNA-26a was detected, as was up-regulation of miRNA-328 and miRNA-299-5p. These miRNAs act upon targets that control cell proliferation, <strong>in</strong>flammation, andapoptosis [109]. Table 12.1 illustrates the effect of miRNAs <strong>in</strong> various autoimmune diseasestates.12.7 ANTAGOMIRSAntagomirs are synthetic antagonists of miRNAs which were first developed <strong>in</strong> 2005 assilenc<strong>in</strong>g agents of miRNAs. One of the earlier studies of the use of antagomirs to silencemiRNA <strong>in</strong>volved miRNA-122 <strong>in</strong> mice [110]. Antagomir-122 was an effective miRNA silenc<strong>in</strong>g


CHAPTER 12Approaches to Autoimmune <strong>Disease</strong>s Us<strong>in</strong>g Epigenetic TherapyTABLE 12.1 miRNAs Involved <strong>in</strong> Autoimmune <strong>Disease</strong>sRA SLE SS SC MS PM/DM PBC Psoriasis IBD ITP17-9216 [17 Y17-5p [18b [20 Y21 [ [26aY2934a [96 [122aY124a Y125aY126 [ [132145 [146 [ [146a [ Y148a [155 [ [184 Y192 Y198 [203 [ [206 Y207 Y214 Y221222223 [296 Y299-5p [326 [328 [346371-5p [375 [379 Y423-5p [513 Y599 [1224-3p Y241PM, polymyositis; DM, dermatomyositis; SLE, systemic lupus erythematosus; RA, rheumatoid arthritis; MS, multiple sclerosis;PBC, primary biliary cirrhosis; IBD, <strong>in</strong>flammatory bowel disease; ITP, idiopathic thrombocytopenic purpura.agent that was able to reduce miRNA levels which are normally abundant <strong>in</strong> the livers of mice.While many functions of miRNAs themselves are still not known, miRNA-122 is thought to be<strong>in</strong>volved <strong>in</strong> cholesterol biosynthesis, and <strong>in</strong>deed, antagomir-122-treated mice demonstratedlower plasma cholesterol levels.A study compar<strong>in</strong>g the efficacy of an antagomir to miRNA-145 and glucocorticoid therapy<strong>in</strong> the treatment of mice with allergy to dust mite showed that the antagomir-treated group


242TABLE 12.2 A comparison of Various forms of Current and Future Therapy <strong>in</strong> RATherapyConventional(Non-Steroidal)Conventional(Steroids)Mechanism Variable Regulation of<strong>in</strong>flammatorypathwaysGoals andexpectationsTreat<strong>in</strong>g symptoms Treat<strong>in</strong>g symptomsDMARDS (Non-Biologics) Biologics Gene Therapy Epigenetic TherapyVariableTreat<strong>in</strong>g symptomsand slow<strong>in</strong>g/ceas<strong>in</strong>gprogression ofdiseaseBlock<strong>in</strong>g of action ofmediatorsTreat<strong>in</strong>g symptomsand slow<strong>in</strong>g/ceas<strong>in</strong>gprogression ofdiseaseInduction ofimmunologicaltoleranceRegulation of geneexpression,regulation of adaptiveand <strong>in</strong>nate immunity<strong>Disease</strong> modification <strong>Disease</strong> modificationCurrently Available Yes Yes Yes Yes Yes YesApproved <strong>in</strong>dicationfor autoimmunediseasesYes Yes Yes Yes Yes NoSide effects (knownor potential)Gastro<strong>in</strong>test<strong>in</strong>albleed<strong>in</strong>g, asthmaImmunosuppression,Cush<strong>in</strong>g’s syndrome,hypertension,diabetes,osteonecrosis,osteoporosisBone marrowsuppression,hypersensitivityreactions,immunosuppressionAnaphylaxis,<strong>in</strong>fection,hypersensitivityreactions,immunosuppressionCancer, <strong>in</strong>fection,immunosuppressionBenefit/risk ratio þ/e þ/e þ þþþ Unknown UnknownCancer, <strong>in</strong>fection,immunosuppression<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>


CHAPTER 12Approaches to Autoimmune <strong>Disease</strong>s Us<strong>in</strong>g Epigenetic Therapyhad similar reductions <strong>in</strong> airway hyper-responsiveness, mucous secretion, eos<strong>in</strong>ophilic<strong>in</strong>flammation, and Th2 cytok<strong>in</strong>e levels [111,112]. miRNA-126 is another miRNA which hasimmunologically related regulatory function, and an antagomir to miRNA-126 has beenshown to reduce Th2 cell function when adm<strong>in</strong>istered to mice with house dust miteallergy [105].The impact of antagomirs on immunologic diseases such as allergic rh<strong>in</strong>itis portends the rapiddevelopment of new <strong>in</strong>novative treatment for allergic and autoimmune diseases. Given thewidespread and constantly <strong>in</strong>creas<strong>in</strong>g association between various miRNAs and autoimmunedisease, it is only a matter of time before antagomirs will be used <strong>in</strong> cl<strong>in</strong>ical trials to treatdiseases such as SLE, rheumatoid arthritis, scleroderma, etc.12.8 TECHNIQUES TO MEASURE EPIGENETIC ALTERATIONS eAPPLICATION OF EPIGENETICS AS BIOMARKERSS<strong>in</strong>ce the observation of <strong>in</strong>creased DNA hypomethylation seen <strong>in</strong> SLE patients, the possibilityof us<strong>in</strong>g epigenetic modifications or activity as biomarkers has been proposed. Measurementof epigenetic alteration techniques has been developed. Differential methylation hybridizationtakes advantage of microarray technology and oligonucleotide probes to measure changes <strong>in</strong>gene expression levels us<strong>in</strong>g picogram levels of DNA attached to a solid surface [113]. Thistechnique has been utilized as a potential screen<strong>in</strong>g technique to identify biomarkers <strong>in</strong> lungcancer [113]. Chromat<strong>in</strong> immunoprecipitation assays coupled to hybridization of genomicmicroarrays (ChIP-on-chip) is a method to <strong>in</strong>vestigate the <strong>in</strong>teraction between DNA andprote<strong>in</strong>. Because Chip-on-chip deals with chromat<strong>in</strong>, one might use this techniques to preferentiallyidentify genes with a regulatory component, <strong>in</strong>clud<strong>in</strong>g replication factors, promoterregions, repressors and silenc<strong>in</strong>g elements, enhancers, and other functional elements [114].Another method of quantify<strong>in</strong>g global DNA methylation is fluorescence correlation spectroscopy,which takes advantage of the high aff<strong>in</strong>ity of MBD for methylated DNA [115].243miRNAs can also be used for disease monitor<strong>in</strong>g and prognostication. This has beendemonstrated <strong>in</strong> cancer, for example, where miRNA29 targets DNMT3A and DNMT3B, and isdown-regulated <strong>in</strong> lung cancer tissue [116]. But expression levels of other miRNAs such asmiRNA-155 and miRNA 101, which are implicated <strong>in</strong> the pathogenesis of SLE, may potentiallybe developed as a diagnostic marker or used to monitor disease activity. Other miRNAs thatmay someday be utilized as diagnostic markers <strong>in</strong>clude miRNA-181a, miRNA-146, miRNA-223, and the miRNA17-92 gene cluster [117].12.9 POTENTIAL SIDE EFFECTS OF TREATMENT WITHEPIGENETIC DRUGS IN AUTOIMMUNE DISEASESOur knowledge of the effects of revers<strong>in</strong>g epigenetic changes that have been shown to bepresent <strong>in</strong> autoimmune disease is <strong>in</strong>complete. Chang<strong>in</strong>g the histone code, or chang<strong>in</strong>g levelsof global demethylation, may provide relief from the disease by alter<strong>in</strong>g prote<strong>in</strong> synthesis of<strong>in</strong>flammatory mediators that are responsible for the disease characteristics, but may affectother cell l<strong>in</strong>es and genes that can be crucial to normal function elsewhere <strong>in</strong> the organism.The fact that about 50% of promoter regions are located <strong>in</strong> CpG islands [118] may provide an<strong>in</strong>novative means for the design of drug or delivery systems to provide improved specificity,but whether this will yield the desired outcomes is unknown.The use of miRNA provides slightly more specificity as to what is targeted, but even then,a s<strong>in</strong>gle miRNA may target multiple genes and vice versa. This lack of target<strong>in</strong>g directly to thegene of <strong>in</strong>terest on the cell type of <strong>in</strong>terest suggests that there will be as yet unidentified sideeffects. These side effects could potentially be short term or long term, lead<strong>in</strong>g to significantdrug-<strong>in</strong>duced morbidity. The field of epigenetic therapy <strong>in</strong> treat<strong>in</strong>g autoimmune diseases is still


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>BOX 12.4 POTENTIAL SIDE EFFECTS OF EPIGENETIC DRUGS1. Unexpected activation or repression of bystander genes2. Bacterial <strong>in</strong>fection3. Reactivation of viral <strong>in</strong>fections4. Embryogenic side effects e teratogenicity5. Cytok<strong>in</strong>e storm6. Hypersensitivity to the drug7. Cancer e reactivation of cancer genes, <strong>in</strong>hibition of tumor suppressive genes8. Autoimmune diseases9. Interaction with other concurrent medications or treatments<strong>in</strong> its <strong>in</strong>fancy, and a search of “epigenetic therapy” and “side effects” on Pubmed revealed noarticles discuss<strong>in</strong>g side effects.244A further consideration is that epigenetic changes are, by nature, hereditary. When oneconsiders the use of epigenetic modification <strong>in</strong> the treatment of autoimmune diseases, oneshould also consider what effects such changes may have on offspr<strong>in</strong>g. It is not known if DNAmethylation levels, or methylation <strong>in</strong>dices change dur<strong>in</strong>g an <strong>in</strong>dividual’s lifetime, or even ifthey are different embryonically. Will the use of these drugs lead to teratogenicity? Genes areturned on and off constantly dur<strong>in</strong>g development <strong>in</strong> response to host and environmentalstimuli. Will epigenetic drugs disrupt the normal regulation dur<strong>in</strong>g development? These are allimportant considerations <strong>in</strong> the development of this potentially very effective treatment ofautoimmunity and other diseases. Specificity is another important issue. There is always thepossibility that use of HDAC <strong>in</strong>hibitors, DNA methylat<strong>in</strong>g enzymes and microRNA to treatautoimmune disease can also lead to effects of gene expression of constituent and necessarygenes. In adm<strong>in</strong>ister<strong>in</strong>g any treatment to a patient, the riskebenefit ratio must be consideredcarefully and conveyed to the patient. In the case of epigenetics, knowledge or risk is limited.Box 12.4 shows potential side effects of epigenetic drugs.12.10 BALANCING CONVENTIONAL THERAPYAND EPIGENETIC THERAPYAs we move well <strong>in</strong>to the 21st century, new approaches and new drugs are be<strong>in</strong>g developed forthe treatment of autoimmune diseases. Older drugs may become obsolete, or may still bevaluable tools for treatment. Avenues for comb<strong>in</strong>ed use of conventional and <strong>in</strong>novativetherapies will undoubtedly be a topic of debate as new drugs appear. A comparison of thevarious therapies for autoimmune diseases is outl<strong>in</strong>ed <strong>in</strong> Table 12.2.The effects of HDAC on the glucocorticoid receptor has been discussed earlier, and this must betaken <strong>in</strong>to consideration when comb<strong>in</strong><strong>in</strong>g HDAC <strong>in</strong>hibitors and glucocorticoids to treatautoimmune diseases. Other examples of possible conflicts of drugs exist. Methotrexate, forexample is a dihydrofolate reductase <strong>in</strong>hibitor and limits the action of DNA methyltransferases[119]. Another example of how two therapeutic modalities may affect each other <strong>in</strong>volves IL-6and HDAC <strong>in</strong>hibitors. IL-6 is a cytok<strong>in</strong>e that is <strong>in</strong>volved <strong>in</strong> DNA methylation [120]. Tocilizumabis a monoclonal antibody aga<strong>in</strong>st IL-6, and therefore it may have epigenetic propertiesitself, but whether or not it can be comb<strong>in</strong>ed with HDAC <strong>in</strong>hibitors as a treatment for autoimmunediseases has not been studied.Comb<strong>in</strong>ation of DNA methyltransferases with HDIC <strong>in</strong>hibitors has been attempted <strong>in</strong> cancerbut not autoimmune diseases. 5-aza-CdR has been used <strong>in</strong> conjunction with valproic acid andret<strong>in</strong>oic acid, two HDAC <strong>in</strong>hibitors, and reversed silenc<strong>in</strong>g of regulatory genes <strong>in</strong> breast cancercells [121]. This approach has also been successful <strong>in</strong> the treatment of myeloplastic syndromes[122]. Another strategy may be to use HDAC <strong>in</strong>hibitors or DNA methylation agents to restore


CHAPTER 12Approaches to Autoimmune <strong>Disease</strong>s Us<strong>in</strong>g Epigenetic Therapyexpression of certa<strong>in</strong> receptors <strong>in</strong> order to enhance the response to drugs that target thosereceptors. In breast cancer studies, 5-aza-CdR and TSA have been used <strong>in</strong> comb<strong>in</strong>ation to<strong>in</strong>crease expression of estrogen receptors to render the conventional drug, tamoxifen, moreeffective [123].12.11 WHERE DO WE GO FROM HERE?The future holds great promise for the utilization of epigenetic drugs <strong>in</strong> the treatment ofautoimmune diseases. There is also potential for great danger. In an article <strong>in</strong> 2008, Ballaswrote that the last two decades of the 20th century would be known for the development ofbiologics and was a venture <strong>in</strong>to a “brave new world” [124]. Perhaps epigenetic drug developmentwill similarly be the highlight of the early- to mid-21st century and should be labeleda “braver new world”, for the simple reason that with epigenetic drugs, side effects may extendto future generations. Some considerations <strong>in</strong> the development of epigenetic drugs must<strong>in</strong>clude the follow<strong>in</strong>g:1. How to target specific genes?2. Will the alteration of global DNA hypomethylation <strong>in</strong> autoimmune diseases shut offnecessary genes?3. How reversible may side effects of the next generation of epigenetic drugs be?4. What is the cost of development of epigenetic drugs?5. Are miRNAs the answer?6. What is the specificity of miRNAs used as therapeutic drugs?7. What are the potential adverse effects of epigenetic treatment of autoimmune diseases?8. Are there other ways than pharmaceutical development to utilize epigenetics <strong>in</strong> thetreatment of autoimmune diseases?9. What is the most effective and safest delivery method for adm<strong>in</strong>istration of these agents tothe patient?10. Will epigenetic modification have an effect <strong>in</strong> pregnancy or future offspr<strong>in</strong>g?245Our current goals should be focused on answer<strong>in</strong>g the above questions. Drummond hasstudied the delivery of HDAC <strong>in</strong>hibitors by packag<strong>in</strong>g <strong>in</strong> liposomes [125]. The use of siRNAs ormiRNAs <strong>in</strong> the treatment of diseases is be<strong>in</strong>g <strong>in</strong>vestigated by several commercial entities. Oneof the immediate goals is to identify all of the target genes for the known miRNAs. In caseswhere a specific miRNA overexpression is pathologic, the development of antagomirs [110],synthetic oligonucleotides that target specific miRNAs, may be helpful as a method ofenhanc<strong>in</strong>g the specificity of epigenetic drugs <strong>in</strong> the treatment of autoimmunity.12.12 DISCUSSIONAs of today, there is no Food and Drug Adm<strong>in</strong>istration (FDA)-approved epigenetic drug for thetreatment of autoimmune diseases. Several drugs are approved for treatment of cancer andmyeloplastic syndromes (MDS), and are therefore available commercially. The target genes<strong>in</strong>volved <strong>in</strong> the epigenetic treatment of cancer are typically tumor-suppressive genes. Inautoimmunity, the target genes, prote<strong>in</strong>s, and cell types are <strong>in</strong>volved <strong>in</strong> normal immunehomeostasis as well, render<strong>in</strong>g the treatment with epigenetics much more complex. Wellknownepigenetic targets <strong>in</strong>clude DNA methylation and histone/chromat<strong>in</strong> structure. Ourknowledge of epigenetics is <strong>in</strong> its <strong>in</strong>fancy. We are just beg<strong>in</strong>n<strong>in</strong>g to characterize the mechanismsof action of potential epigenetic drugs. Even less <strong>in</strong>formation is available about unexpectedor unwanted side effects associated with the use of these drugs. It is important toconsider these important issues when develop<strong>in</strong>g new targets for the treatment ofautoimmunity.MicroRNAs can impact epigenetics by virtue of their effects on DNA methylation <strong>in</strong> autoimmunediseases. MicroRNAs can be up-regulated or down-regulated <strong>in</strong> autoimmune diseases.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>The use of microRNAs or <strong>in</strong>hibitors of miRNA to impact DNA methylation is a method oftarget<strong>in</strong>g post-transcriptional gene transcription changes <strong>in</strong> CD4þ T cells <strong>in</strong> SLE patients, andthus may be a new method of <strong>in</strong>fluenc<strong>in</strong>g epigenetics to treat autoimmune diseases. Micro-RNAs are directed aga<strong>in</strong>st specific genes and thus may be associated with a higher specificity ofaction than global demethylat<strong>in</strong>g agents or HDAC <strong>in</strong>hibitors. Some of the relevant microRNAs<strong>in</strong> various autoimmune diseases such as SLE or RA have been identified and <strong>in</strong>clude miRNAsthat target pro<strong>in</strong>flammatory genes, genes that regulate T-cell function <strong>in</strong>clud<strong>in</strong>g Treg function,and genes that regulate apoptosis or the cell cycle.Besides pharmaceutical development, epigenetics may have other uses as potential biomarkers<strong>in</strong> monitor<strong>in</strong>g the effectiveness of therapy. In SLE, disease activity is associated with levels ofmethylation and of expression of genes such as TNFSF7 and ITGAL. While our treatment mayas yet not <strong>in</strong>volve the use of epigenetic manipulation, levels of gene expression can bepotentially used to monitor the success of other forms of therapy. For example, miRNAs can beused to assist <strong>in</strong> the diagnosis and determ<strong>in</strong>e prognosis and therapeutic outcome as well. Thiscan be accomplished with the help of computer analysis of the activity of multiple miRNAsalong with levels of hypomethylation and histone deacetylase activity.In summary, epigenetics is an area of research with tremendous potential. As our knowledge<strong>in</strong>creases, we will learn how to control expression of the critical factors that lead to autoimmunedisease, and how to do it <strong>in</strong> a selective manner than does not endanger the patient.Treatments of the past may be abandoned <strong>in</strong> favor of these more effective and potentially safertherapeutic methods. Morbidity and mortality will decrease, and patients with these disorderswill be able to enjoy a higher quality of life.246References[1] P<strong>in</strong>eles D, Valente A, Warren B, Peterson MG, Lehman TJ, Moorthy LN. Worldwide <strong>in</strong>cidence and prevalenceof pediatric onset systemic lupus erythematosus. Lupus 2011;20:1187e92.[2] Cooper GS, Miller FW, Germolec DR. Occupational exposures and autoimmune diseases. Int Immunopharmacol2002;2:303e13.[3] Morey C, Avner P. Genetics and epigenetics of the X chromosome. Ann N Y Acad Sci 2010;1214:E18e33.[4] Crow MK, Kirou KA, Wohlgemuth J. Microarray analysis of <strong>in</strong>terferon-regulated genes <strong>in</strong> SLE. Autoimmunity2003;36:481e90.[5] Banchereau J, Pascual V, Palucka AK. Autoimmunity through cytok<strong>in</strong>e-<strong>in</strong>duced dendritic cell activation.Immunity 2004;20:539e50.[6] Baechler EC, Batliwalla FM, Karypis G, et al. Interferon-<strong>in</strong>ducible gene expression signature <strong>in</strong> peripheralblood cells of patients with severe lupus. Proc Natl Acad Sci USA 2003;100:2610e5.[7] Sakao S, Tatsumi K. The importance of epigenetics <strong>in</strong> the development of chronic obstructive pulmonarydisease. Respirology 2011;16:1056e63.[8] Kendall E. The development of cortisone as a therapeutic agent. Amsterdam: Elsevier Publish<strong>in</strong>g Company;1964.[9] Barnes PJ, Adcock IM, Ito K. Histone acetylation and deacetylation: importance <strong>in</strong> <strong>in</strong>flammatory lungdiseases. Eur Respir J 2005;25:552e63.[10] Barnes PJ. Target<strong>in</strong>g histone deacetylase 2 <strong>in</strong> chronic obstructive pulmonary disease treatment. Expert Op<strong>in</strong>Ther Targets 2005;9:1111e21.[11] Koppelman GH, Nawijn MC. Recent advances <strong>in</strong> the epigenetics and genomics of asthma. Curr Op<strong>in</strong> AllergyCl<strong>in</strong> Immunol 2011;11:414e9.[12] Durham AL, Wiegman C, Adcock IM. <strong>Epigenetics</strong> of asthma. Biochim Biophys Acta 2011;1810:1103e9.[13] Abel T, Zuk<strong>in</strong> RS. Epigenetic targets of HDAC <strong>in</strong>hibition <strong>in</strong> neurodegenerative and psychiatric disorders. CurrOp<strong>in</strong> Pharmacol 2008;8:57e64.[14] Ptak C, Petronis A. Epigenetic approaches to psychiatric disorders. Dialogues Cl<strong>in</strong> Neurosci 2010;12:25e35.[15] Bell CG, Beck S. The epigenomic <strong>in</strong>terface between genome and environment <strong>in</strong> common complex diseases.Brief Funct Genomics 2010;9:477e85.[16] Park YJ, Claus R, Weichenhan D, Plass C. Genome-wide epigenetic modifications <strong>in</strong> cancer. Prog Drug Res2011;67:25e49.


CHAPTER 12Approaches to Autoimmune <strong>Disease</strong>s Us<strong>in</strong>g Epigenetic Therapy[17] Janson PC, W<strong>in</strong>qvist O. <strong>Epigenetics</strong> - the key to understand immune responses <strong>in</strong> health and disease. Am JReprod Immunol 2011;66(Suppl. 1):72e4.[18] Meda F, Folci M, Baccarelli A, Selmi C. The epigenetics of autoimmunity. Cell Mol Immunol 2011;8:226e36.[19] Renaud<strong>in</strong>eau Y, You<strong>in</strong>ou P. <strong>Epigenetics</strong> and autoimmunity, with special emphasis on methylation. Keio J Med2011;60:10e6.[20] Ooi SK, Qiu C, Bernste<strong>in</strong> E, et al. DNMT3L connects unmethylated lys<strong>in</strong>e 4 of histone H3 to de novomethylation of DNA. Nature 2007;448:714e7.[21] de Ruijter AJ, van Gennip AH, Caron HN, Kemp S, van Kuilenburg AB. Histone deacetylases (HDACs):characterization of the classical HDAC family. Biochem J 2003;370:737e49.[22] Stunkel W, Campbell RM, Sirtu<strong>in</strong> 1 (SIRT1). The Misunderstood HDAC. J Biomol Screen 2011;16:1153e69.[23] Wang L, Tao R, Hancock WW. Us<strong>in</strong>g histone deacetylase <strong>in</strong>hibitors to enhance Foxp3(þ) regulatory T-cellfunction and <strong>in</strong>duce allograft tolerance. Immunol Cell Biol 2009;87:195e202.[24] Beier UH, Akimova T, Liu Y, Wang L, Hancock WW. Histone/prote<strong>in</strong> deacetylases control Foxp3 expressionand the heat shock response of T-regulatory cells. Curr Op<strong>in</strong> Immunol 2011;23(5):670e8.[25] Grausenburger R, Bilic I, Boucheron N, et al. Conditional deletion of histone deacetylase 1 <strong>in</strong> T cells leads toenhanced airway <strong>in</strong>flammation and <strong>in</strong>creased Th2 cytok<strong>in</strong>e production. J Immunol 2010;185:3489e97.[26] Sozzani S. Dendritic cell traffick<strong>in</strong>g: more than just chemok<strong>in</strong>es. Cytok<strong>in</strong>e Growth Factor Rev 2005;16:581e92.[27] Annunziato F, Cosmi L, Santarlasci V, et al. Phenotypic and functional features of human Th17 cells. J ExpMed 2007;204:1849e61.[28] Yen D, Cheung J, Scheerens H, et al. IL-23 is essential for T cell-mediated colitis and promotes <strong>in</strong>flammationvia IL-17 and IL-6. J Cl<strong>in</strong> Invest 2006;116:1310e6.[29] Valencia X, Yarboro C, Illei G, Lipsky PE. Deficient CD4þCD25high T regulatory cell function <strong>in</strong> patients withactive systemic lupus erythematosus. J Immunol 2007;178:2579e88.[30] Lyssuk EY, Torgash<strong>in</strong>a AV, Soloviev SK, Nassonov EL, Bykovskaia SN. Reduced number and function ofCD4þCD25highFoxP3þ regulatory T cells <strong>in</strong> patients with systemic lupus erythematosus. Adv Exp Med Biol2007;601:113e9.[31] Li B, Samanta A, Song X, et al. FOXP3 <strong>in</strong>teractions with histone acetyltransferase and class II histonedeacetylases are required for repression. Proc Natl Acad Sci USA 2007;104:4571e6.[32] Halili MA, Andrews MR, Sweet MJ, Fairlie DP. Histone deacetylase <strong>in</strong>hibitors <strong>in</strong> <strong>in</strong>flammatory disease. CurrTop Med Chem 2009;9:309e19.[33] Nencioni A, Beck J, Werth D, et al. Histone deacetylase <strong>in</strong>hibitors affect dendritic cell differentiation andimmunogenicity. Cl<strong>in</strong> Cancer Res 2007;13:3933e41.[34] Brogdon JL, Xu Y, Szabo SJ, et al. Histone deacetylase activities are required for <strong>in</strong>nate immune cell control ofTh1 but not Th2 effector cell function. Blood 2007;109:1123e30.[35] Bosisio D, Vulcano M, Del Prete A, et al. Block<strong>in</strong>g TH17-polariz<strong>in</strong>g cytok<strong>in</strong>es by histone deacetylase <strong>in</strong>hibitors<strong>in</strong> vitro and <strong>in</strong> vivo. J Leukoc Biol 2008;84:1540e8.[36] Kelly WK, Marks PA, Drug <strong>in</strong>sight. Histone deacetylase <strong>in</strong>hibitorsedevelopment of the new targeted anticanceragent suberoylanilide hydroxamic acid. Nat Cl<strong>in</strong> Pract Oncol 2005;2:150e7.[37] Wang Z, Chen C, F<strong>in</strong>ger SN, et al. Suberoylanilide hydroxamic acid: a potential epigenetic therapeutic agentfor lung fibrosis? Eur Respir J 2009;34:145e55.[38] L<strong>in</strong> T, Chen H, Koustova E, et al. Histone deacetylase as therapeutic target <strong>in</strong> a rodent model of hemorrhagicshock: effect of different resuscitation strategies on lung and liver. Surgery 2007;141:784e94.[39] L<strong>in</strong> HS, Hu CY, Chan HY, et al. Anti-rheumatic activities of histone deacetylase (HDAC) <strong>in</strong>hibitors <strong>in</strong> vivo <strong>in</strong>collagen-<strong>in</strong>duced arthritis <strong>in</strong> rodents. Br J Pharmacol 2007;150:862e72.[40] Drummond DC, Noble CO, Kirpot<strong>in</strong> DB, Guo Z, Scott GK, Benz CC. Cl<strong>in</strong>ical development of histonedeacetylase <strong>in</strong>hibitors as anticancer agents. Annu Rev Pharmacol Toxicol 2005;45:495e528.[41] Morley M, Molony CM, Weber TM, et al. Genetic analysis of genome-wide variation <strong>in</strong> human geneexpression. Nature 2004;430:743e7.[42] Sims 3rd RJ, Re<strong>in</strong>berg D. From chromat<strong>in</strong> to cancer: a new histone lys<strong>in</strong>e methyltransferase enters the mix.Nat Cell Biol 2004;6:685e7.[43] Garcia BA, Busby SA, Shabanowitz J, Hunt DF, Mishra N. Resett<strong>in</strong>g the epigenetic histone code <strong>in</strong> the MRLlpr/lprmouse model of lupus by histone deacetylase <strong>in</strong>hibition. J Proteome Res 2005;4:2032e42.[44] Aung HT, Schroder K, Himes SR, et al. LPS regulates pro<strong>in</strong>flammatory gene expression <strong>in</strong> macrophages byalter<strong>in</strong>g histone deacetylase expression. FASEB J 2006;20:1315e27.[45] Bode KA, Schroder K, Hume DA, et al. Histone deacetylase <strong>in</strong>hibitors decrease Toll-like receptor-mediatedactivation of pro<strong>in</strong>flammatory gene expression by impair<strong>in</strong>g transcription factor recruitment. Immunology2007;122:596e606.247


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>248[46] Tao R, de Zoeten EF, Ozkaynak E, et al. Deacetylase <strong>in</strong>hibition promotes the generation and function ofregulatory T cells. Nat Med 2007;13:1299e307.[47] Chung YL, Lee MY, Wang AJ, Yao LF. A therapeutic strategy uses histone deacetylase <strong>in</strong>hibitors to modulate theexpression of genes <strong>in</strong>volved <strong>in</strong> the pathogenesis of rheumatoid arthritis. Mol Ther 2003;8:707e17.[48] Blair HC, Zaidi M. Osteoclastic differentiation and function regulated by old and new pathways. Rev EndocrMetab Disord 2006;7:23e32.[49] Yi T, Baek JH, Kim HJ, et al. Trichostat<strong>in</strong> A-mediated upregulation of p21(WAF1) contributes to osteoclastapoptosis. Exp Mol Med 2007;39:213e21.[50] Rahman MM, Kukita A, Kukita T, Shobuike T, Nakamura T, Kohashi O. Two histone deacetylase <strong>in</strong>hibitors,trichostat<strong>in</strong> A and sodium butyrate, suppress differentiation <strong>in</strong>to osteoclasts but not <strong>in</strong>to macrophages. Blood2003;101:3451e9.[51] Cantley MD, Fairlie DP, Bartold PM, et al. Inhibitors of histone deacetylases <strong>in</strong> class I and class II suppresshuman osteoclasts <strong>in</strong> vitro. J Cell Physiol 2011;226:3233e41.[52] Schroeder TM, Westendorf JJ. Histone deacetylase <strong>in</strong>hibitors promote osteoblast maturation. J Bone M<strong>in</strong>erRes 2005;20:2254e63.[53] Sega<strong>in</strong> JP, Ra<strong>in</strong>geard de la Bletiere D, Bourreille A, et al. Butyrate <strong>in</strong>hibits <strong>in</strong>flammatory responses throughNFkappaB <strong>in</strong>hibition: implications for Crohn’s disease. Gut 2000;47:397e403.[54] Bright JJ, Musuro BF, Du C, Sriram S. Expression of IL-12 <strong>in</strong> CNS and lymphoid organs of mice withexperimental allergic encephalitis. J Neuroimmunol 1998;82:22e30.[55] Takahashi I, Miyaji H, Yoshida T, Sato S, Mizukami T. Selective <strong>in</strong>hibition of IL-2 gene expression bytrichostat<strong>in</strong> A, a potent <strong>in</strong>hibitor of mammalian histone deacetylase. J Antibiot (Tokyo) 1996;49:453e7.[56] Camelo S, Iglesias AH, Hwang D, et al. Transcriptional therapy with the histone deacetylase <strong>in</strong>hibitortrichostat<strong>in</strong> A ameliorates experimental autoimmune encephalomyelitis. J Neuroimmunol 2005;164:10e21.[57] Gray SG, De Meyts P. Role of histone and transcription factor acetylation <strong>in</strong> diabetes pathogenesis. DiabetesMetab Res Rev 2005;21:416e33.[58] Luhrs H, Gerke T, Boxberger F, et al. Butyrate <strong>in</strong>hibits <strong>in</strong>terleuk<strong>in</strong>-1-mediated nuclear factor-kappa B activation<strong>in</strong> human epithelial cells. Dig Dis Sci 2001;46:1968e73.[59] Y<strong>in</strong> L, Laevsky G, Giard<strong>in</strong>a C. Butyrate suppression of colonocyte NF-kappa B activation and cellularproteasome activity. J Biol Chem 2001;276:44641e6.[60] Ray A, Prefonta<strong>in</strong>e KE, Ray P. Down-modulation of <strong>in</strong>terleuk<strong>in</strong>-6 gene expression by 17 beta-estradiol <strong>in</strong> theabsence of high aff<strong>in</strong>ity DNA b<strong>in</strong>d<strong>in</strong>g by the estrogen receptor. J Biol Chem 1994;269:12940e6.[61] Choi JH, Oh SW, Kang MS, Kwon HJ, Oh GT, Kim DY. Trichostat<strong>in</strong> A attenuates airway <strong>in</strong>flammation <strong>in</strong>mouse asthma model. Cl<strong>in</strong> Exp Allergy 2005;35:89e96.[62] Whittaker SJ, Demierre MF, Kim EJ, et al. F<strong>in</strong>al results from a multicenter, <strong>in</strong>ternational, pivotal study ofromideps<strong>in</strong> <strong>in</strong> refractory cutaneous T-cell lymphoma. J Cl<strong>in</strong> Oncol 2010;28:4485e91.[63] Shao W, Growney JD, Feng Y, et al. Activity of deacetylase <strong>in</strong>hibitor panob<strong>in</strong>ostat (LBH589) <strong>in</strong> cutaneous T-cell lymphoma models: Def<strong>in</strong><strong>in</strong>g molecular mechanisms of resistance. Int J Cancer 2010;127:2199e208.[64] Knipste<strong>in</strong> J, Gore L. Ent<strong>in</strong>ostat for treatment of solid tumors and hematologic malignancies. Expert Op<strong>in</strong>Investig Drugs 2011;20:1455e67.[65] Boumber Y, Younes A, Garcia-Manero G. Mocet<strong>in</strong>ostat (MGCD0103): a review of an isotype-specific histonedeacetylase <strong>in</strong>hibitor. Expert Op<strong>in</strong> Investig Drugs 2011;20:823e9.[66] Ramal<strong>in</strong>gam SS, Belani CP, Ruel C, et al. Phase II study of bel<strong>in</strong>ostat (PXD101), a histone deacetylase<strong>in</strong>hibitor, for second l<strong>in</strong>e therapy of advanced malignant pleural mesothelioma. J Thorac Oncol2009;4:97e101.[67] Raz<strong>in</strong> A, Cedar H. Distribution of 5-methylcytos<strong>in</strong>e <strong>in</strong> chromat<strong>in</strong>. Proc Natl Acad Sci USA 1977;74:2725e8.[68] Peedicayil J. Epigenetic therapyea new development <strong>in</strong> pharmacology. Indian J Med Res 2006;123:17e24.[69] Deng C, Kaplan MJ, Yang J, et al. Decreased Ras-mitogen-activated prote<strong>in</strong> k<strong>in</strong>ase signal<strong>in</strong>g may cause DNAhypomethylation <strong>in</strong> T lymphocytes from lupus patients. Arthritis Rheum 2001;44:397e407.[70] Lu Q, Kaplan M, Ray D, Zacharek S, Gutsch D, Richardson B. Demethylation of ITGAL (CD11a) regulatorysequences <strong>in</strong> systemic lupus erythematosus. Arthritis Rheum 2002;46:1282e91.[71] Kaplan MJ, Lu Q, Wu A, Attwood J, Richardson B. Demethylation of promoter regulatory elementscontributes to perfor<strong>in</strong> overexpression <strong>in</strong> CD4þ lupus T cells. J Immunol 2004;172:3652e61.[72] Oelke K, Lu Q, Richardson D, et al. Overexpression of CD70 and overstimulation of IgG synthesis by lupus Tcells and T cells treated with DNA methylation <strong>in</strong>hibitors. Arthritis Rheum 2004;50:1850e60.[73] Oelke K, Richardson B. Decreased T cell ERK pathway signal<strong>in</strong>g may contribute to the development of lupusthrough effects on DNA methylation and gene expression. Int Rev Immunol 2004;23:315e31.


CHAPTER 12Approaches to Autoimmune <strong>Disease</strong>s Us<strong>in</strong>g Epigenetic Therapy[74] Detich N, Theberge J, Szyf M. Promoter-specific activation and demethylation by MBD2/demethylase. J BiolChem 2002;277:35791e4.[75] Zhu B, Zheng Y, Angliker H, et al. 5-Methylcytos<strong>in</strong>e DNA glycosylase activity is also present <strong>in</strong> the humanMBD4 (G/T mismatch glycosylase) and <strong>in</strong> a related avian sequence. Nucleic Acids Res 2000;28:4157e65.[76] Vila<strong>in</strong> A, Vogt N, Dutrillaux B, Malfoy B. DNA methylation and chromosome <strong>in</strong>stability <strong>in</strong> breast cancer celll<strong>in</strong>es. FEBS Lett 1999;460:231e4.[77] Chang C, Gershw<strong>in</strong> ME. Drug-<strong>in</strong>duced lupus erythematosus: <strong>in</strong>cidence, management and prevention. DrugSaf 2011;34:357e74.[78] Chang C, Gershw<strong>in</strong> ME. Drugs and autoimmunityea contemporary review and mechanistic approach.J Autoimmun 2010;34:J266e75.[79] Nanki T, Hayashida K, El-Gabalawy HS, et al. Stromal cell-derived factor-1-CXC chemok<strong>in</strong>e receptor 4<strong>in</strong>teractions play a central role <strong>in</strong> CD4þ T cell accumulation <strong>in</strong> rheumatoid arthritis synovium. J Immunol2000;165:6590e8.[80] Blades MC, Ingegnoli F, Wheller SK, et al. Stromal cell-derived factor 1 (CXCL12) <strong>in</strong>duces monocytemigration <strong>in</strong>to human synovium transplanted onto SCID Mice. Arthritis Rheum 2002;46:824e36.[81] Kontt<strong>in</strong>en YT, A<strong>in</strong>ola M, Valleala H, et al. Analysis of 16 different matrix metalloprote<strong>in</strong>ases (MMP-1 to MMP-20) <strong>in</strong> the synovial membrane: different profiles <strong>in</strong> trauma and rheumatoid arthritis. Ann Rheum Dis1999;58:691e7.[82] Ceribelli A, Yao B, Dom<strong>in</strong>guez-Gutierrez PR, Chan EK. Lupus T cells switched on by DNA hypomethylationvia microRNA? Arthritis Rheum 2011;63:1177e81.[83] Pan W, Zhu S, Yuan M, et al. MicroRNA-21 and microRNA-148a contribute to DNA hypomethylation <strong>in</strong>lupus CD4þ T cells by directly and <strong>in</strong>directly target<strong>in</strong>g DNA methyltransferase 1. J Immunol2010;184:6773e81.[84] Krol J, Loedige I, Filipowicz W. The widespread regulation of microRNA biogenesis, function and decay. NatRev Genet 2010;11:597e610.[85] Fabian MR, Sonenberg N, Filipowicz W. Regulation of mRNA translation and stability by microRNAs. AnnuRev Biochem 2010;79:351e79.[86] Miranda KC, Huynh T, Tay Y, et al. A pattern-based method for the identification of MicroRNA b<strong>in</strong>d<strong>in</strong>g sitesand their correspond<strong>in</strong>g heteroduplexes. Cell 2006;126:1203e17.[87] John B, Enright AJ, Arav<strong>in</strong> A, Tuschl T, Sander C, Marks DS. <strong>Human</strong> MicroRNA targets. PLoS Biol 2004;2. e363.[88] Zhao S, Wang Y, Liang Y, et al. MicroRNA-126 regulates DNA methylation <strong>in</strong> CD4þ T cells and contributes tosystemic lupus erythematosus by target<strong>in</strong>g DNA methyltransferase 1. Arthritis Rheum 2011;63:1376e86.[89] Nagata Y, Nakasa T, Mochizuki Y, et al. Induction of apoptosis <strong>in</strong> the synovium of mice with autoantibodymediatedarthritis by the <strong>in</strong>traarticular <strong>in</strong>jection of double-stranded MicroRNA-15a. Arthritis Rheum2009;60:2677e83.[90] Li QJ, Chau J, Ebert PJ, et al. miR-181a is an <strong>in</strong>tr<strong>in</strong>sic modulator of T cell sensitivity and selection. Cell2007;129:147e61.[91] Chen CZ, Li L, Lodish HF, Bartel DP. MicroRNAs modulate hematopoietic l<strong>in</strong>eage differentiation. <strong>Science</strong>2004;303:83e6.[92] Hogg N, Leit<strong>in</strong>ger B. Shape and shift changes related to the function of leukocyte <strong>in</strong>tegr<strong>in</strong>s LFA-1 and Mac-1.J Leukoc Biol 2001;69:893e8.[93] Zhao S, Wang Y, Liang Y, et al. MicroRNA-126 regulates DNA methylation <strong>in</strong> CD4+ T cells and contributes tosystemic lupus erythematosus by target<strong>in</strong>g DNA methyltransferase 1. Arthritis Rheum 2010;63:1376e86.[94] Pan W, Zhu S, Yuan M, et al. MicroRNA-21 and microRNA-148a contribute to DNA hypomethylation <strong>in</strong>lupus CD4þ T cells by directly and <strong>in</strong>directly target<strong>in</strong>g DNA methyltransferase 1. J Immunol 184:6773e81.[95] Higgs BW, Liu Z, White B, et al. Patients with systemic lupus erythematosus, myositis, rheumatoid arthritisand scleroderma share activation of a common type I <strong>in</strong>terferon pathway. Ann Rheum Dis 2011;70:2029e36.[96] Tang Y, Luo X, Cui H, et al. MicroRNA-146A contributes to abnormal activation of the type I <strong>in</strong>terferonpathway <strong>in</strong> human lupus by target<strong>in</strong>g the key signal<strong>in</strong>g prote<strong>in</strong>s. Arthritis Rheum 2009;60:1065e75.[97] Curtale G, Citarella F, Carissimi C, et al. An emerg<strong>in</strong>g player <strong>in</strong> the adaptive immune response: microRNA-146a is a modulator of IL-2 expression and activation-<strong>in</strong>duced cell death <strong>in</strong> T lymphocytes. Blood2010;115:265e73.[98] Zhao X, Tang Y, Qu B, et al. MicroRNA-125a contributes to elevated <strong>in</strong>flammatory chemok<strong>in</strong>e RANTES levelsvia target<strong>in</strong>g KLF13 <strong>in</strong> systemic lupus erythematosus. Arthritis Rheum 62:3425e35.[99] Stanczyk J, Pedrioli DM, Brentano F, et al. Altered expression of MicroRNA <strong>in</strong> synovial fibroblasts andsynovial tissue <strong>in</strong> rheumatoid arthritis. Arthritis Rheum 2008;58:1001e9.[100] Leng RX, Pan HF, Q<strong>in</strong> WZ, Chen GM, Ye DQ. Role of microRNA-155 <strong>in</strong> autoimmunity. Cytok<strong>in</strong>e GrowthFactor Rev 2011;22:141e7.249


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>250[101] Pauley KM, Satoh M, Chan AL, Bubb MR, Reeves WH, Chan EK. Upregulated miR-146a expression <strong>in</strong>peripheral blood mononuclear cells from rheumatoid arthritis patients. Arthritis Res Ther 2008;10.R101.[102] Niimoto T, Nakasa T, Ishikawa M, et al. MicroRNA-146a expresses <strong>in</strong> <strong>in</strong>terleuk<strong>in</strong>-17 produc<strong>in</strong>g T cells <strong>in</strong>rheumatoid arthritis patients. BMC Musculoskelet Disord 2010;11:209.[103] Maurer B, Stanczyk J, Jungel A, et al. MicroRNA-29, a key regulator of collagen expression <strong>in</strong> systemic sclerosis.Arthritis Rheum 2010;62:1733e43.[104] Eisenberg I, Eran A, Nish<strong>in</strong>o I, et al. Dist<strong>in</strong>ctive patterns of microRNA expression <strong>in</strong> primary musculardisorders. Proc Natl Acad Sci USA 2007;104:17016e21.[105] Michael A, Bajracharya SD, Yuen PS, et al. Exosomes from human saliva as a source of microRNA biomarkers.Oral Dis 2010;16:34e8.[106] Pauley KM, Stewart CM, Gauna AE, et al. Altered miR-146a expression <strong>in</strong> Sjogren’s syndrome and its functionalrole <strong>in</strong> <strong>in</strong>nate immunity. Eur J Immunol 2011;41:2029e39.[107] Du C, Liu C, Kang J, et al. MicroRNA miR-326 regulates TH-17 differentiation and is associated with thepathogenesis of multiple sclerosis. Nat Immunol 2009;10:1252e9.[108] Junker A, Krumbholz M, Eisele S, et al. MicroRNA profil<strong>in</strong>g of multiple sclerosis lesions identifies modulatorsof the regulatory prote<strong>in</strong> CD47. Bra<strong>in</strong> 2009;132:3342e52.[109] Padgett KA, Lan RY, Leung PC, et al. Primary biliary cirrhosis is associated with altered hepatic microRNAexpression. J Autoimmun 2009;32:246e53.[110] Krutzfeldt J, Rajewsky N, Braich R, et al. Silenc<strong>in</strong>g of microRNAs <strong>in</strong> vivo with ’antagomirs’. Nature2005;438:685e9.[111] Collison A, Mattes J, Plank M, Foster PS. Inhibition of house dust mite-<strong>in</strong>duced allergic airways disease byantagonism of microRNA-145 is comparable to glucocorticoid treatment. J Allergy Cl<strong>in</strong> Immunol2011;128:160e7:e4.[112] Mattes J, Collison A, Plank M, Phipps S, Foster PS. Antagonism of microRNA-126 suppresses the effector functionof TH2 cells and the development of allergic airways disease. Proc Natl Acad Sci USA 2009;106:18704e9.[113] Son JW, Jeong KJ, Jean WS, et al. Genome-wide comb<strong>in</strong>ation profil<strong>in</strong>g of DNA copy number and methylationfor decipher<strong>in</strong>g biomarkers <strong>in</strong> non-small cell lung cancer patients. Cancer Lett 2011;311:29e37.[114] Ballestar E, Paz MF, Valle L, et al. Methyl-CpG b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s identify novel sites of epigenetic <strong>in</strong>activation<strong>in</strong> human cancer. EMBO J 2003;22:6335e45.[115] Umezu T, Ohyashiki K, Ohyashiki JH. Detection method for quantify<strong>in</strong>g global DNA methylation by fluorescencecorrelation spectroscopy. Anal Biochem 2011;415:145e50.[116] Dai Y, Sui W, Lan H, Yan Q, Huang H, Huang Y. Comprehensive analysis of microRNA expression patterns <strong>in</strong>renal biopsies of lupus nephritis patients. Rheumatol Int 2009;29:749e54.[117] Zhao S, Long H, Lu Q. Epigenetic perspectives <strong>in</strong> systemic lupus erythematosus: pathogenesis, biomarkers,and therapeutic potentials. Cl<strong>in</strong> Rev Allergy Immunol 2010;39:3e9.[118] Bird AP. CpG-rich islands and the function of DNA methylation. Nature 1986;321:209e13.[119] Wu J, Wood GS. Reduction of Fas/CD95 promoter methylation, upregulation of Fas prote<strong>in</strong>, and enhancementof sensitivity to apoptosis <strong>in</strong> cutaneous T-cell lymphoma. Arch Dermatol 2011;147:443e9.[120] Gasche JA, Hoffmann J, Boland CR, Goel A. Interleuk<strong>in</strong>-6 promotes tumorigenesis by alter<strong>in</strong>g DNA methylation<strong>in</strong> oral cancer cells. Int J Cancer 2011;129:1053e63.[121] Mongan NP, Gudas LJ. Valproic acid, <strong>in</strong> comb<strong>in</strong>ation with all-trans ret<strong>in</strong>oic acid and 5-aza-2’-deoxycytid<strong>in</strong>e,restores expression of silenced RARbeta2 <strong>in</strong> breast cancer cells. Mol Cancer Ther 2005;4:477e86.[122] Soriano AO, Yang H, Faderl S, et al. Safety and cl<strong>in</strong>ical activity of the comb<strong>in</strong>ation of 5-azacytid<strong>in</strong>e, valproicacid, and all-trans ret<strong>in</strong>oic acid <strong>in</strong> acute myeloid leukemia and myelodysplastic syndrome. Blood2007;110:2302e8.[123] Fan J, Y<strong>in</strong> WJ, Lu JS, et al. ER alpha negative breast cancer cells restore response to endocr<strong>in</strong>e therapy by comb<strong>in</strong>ationtreatment with both HDAC <strong>in</strong>hibitor and DNMT <strong>in</strong>hibitor. J Cancer Res Cl<strong>in</strong> Oncol 2008;134:883e90.[124] Ballas ZK. Immunomodulators: a brave new world. J Allergy Cl<strong>in</strong> Immunol 2008;121:331e3.[125] Drummond DC, Marx C, Guo Z, et al. Enhanced pharmacodynamic and antitumor properties of a histonedeacetylase <strong>in</strong>hibitor encapsulated <strong>in</strong> liposomes or ErbB2-targeted immunoliposomes. Cl<strong>in</strong> Cancer Res2005;11:3392e401.[126] Nandakumar V, Vaid M, Katiyar SK. (-)-Epigallocatech<strong>in</strong>-3-gallate reactivates silenced tumor suppressorgenes, Cip1/p21 and p16INK4a, by reduc<strong>in</strong>g DNA methylation and <strong>in</strong>creas<strong>in</strong>g histones acetylation <strong>in</strong> humansk<strong>in</strong> cancer cells. Carc<strong>in</strong>ogenesis 2011;32:537e44.[127] Yan L, Nass SJ, Smith D, Nelson WG, Herman JG, Davidson NE. Specific <strong>in</strong>hibition of DNMT1 by antisenseoligonucleotides <strong>in</strong>duces re-expression of estrogen receptor-alpha (ER) <strong>in</strong> ER-negative human breast cancercell l<strong>in</strong>es. Cancer Biol Ther 2003;2:552e6.


CHAPTER 12Approaches to Autoimmune <strong>Disease</strong>s Us<strong>in</strong>g Epigenetic Therapy[128] Kaplan MJ. Apoptosis <strong>in</strong> systemic lupus erythematosus. Cl<strong>in</strong> Immunol 2004;112:210e8.[129] Loh C, Pau E, Chang NH, Wither JE. An <strong>in</strong>tr<strong>in</strong>sic B-cell defect supports autoimmunity <strong>in</strong> New Zealand blackchromosome 13 congenic mice. Eur J Immunol 2011;41:527e36.[130] Church LD, Filer AD, Hidalgo E, et al. Rheumatoid synovial fluid <strong>in</strong>terleuk<strong>in</strong>-17-produc<strong>in</strong>g CD4 T cells haveabundant tumor necrosis factor-alpha co-expression, but little <strong>in</strong>terleuk<strong>in</strong>-22 and <strong>in</strong>terleuk<strong>in</strong>-23R expression.Arthritis Res Ther 2010;12:R184.[131] Usmani OS, Ito K, Maneechotesuwan K, et al. Glucocorticoid receptor nuclear translocation <strong>in</strong> airway cellsafter <strong>in</strong>haled comb<strong>in</strong>ation therapy. Am J Respir Crit Care Med 2005;172:704e12.[132] Kitamura H, Torigoe T, Asanuma H, Honma I, Sato N, Tsukamoto T. Down-regulation of HLA class Iantigens <strong>in</strong> prostate cancer tissues and up-regulation by histone deacetylase <strong>in</strong>hibition. J Urol2007;178:692e6.[133] Yang HX, Zhang W, Zhao LD, et al. Are CD4þCD25-Foxp3þ cells <strong>in</strong> untreated new-onset lupus patientsregulatory T cells? Arthritis Res Ther 2009;11:R153.[134] Pau E, Chang NH, Loh C, Lajoie G, Wither JE. Abrogation of pathogenic IgG autoantibody production <strong>in</strong>CD40L gene-deleted lupus-prone New Zealand Black mice. Cl<strong>in</strong> Immunol 2011;139:215e27.[135] Gillespie J, Savic S, Wong C, et al. Histone deacetylases are dysregulated <strong>in</strong> rheumatoid arthritis and a novelHDAC3-selective <strong>in</strong>hibitor reduces IL-6 production by PBMC of RA patients. Arthritis Rheum2012;64:418e22.[136] Jie G, Jiang Q, Rui Z, Yifei Y. Expression of <strong>in</strong>terleuk<strong>in</strong>-17 <strong>in</strong> autoimmune dacryoadenitis <strong>in</strong> MRL/lpr mice.Curr Eye Res 2010;35:865e71.[137] Cui LF, Guo XJ, Wei J, et al. Overexpression of TNF-alpha and TNFRII <strong>in</strong> <strong>in</strong>vasive micropapillary carc<strong>in</strong>oma ofthe breast: cl<strong>in</strong>icopathological correlations. Histopathology 2008;53:381e8.[138] Majumdar G, Rooney RJ, Johnson IM, Raghow R. Pan-histone deacetylase <strong>in</strong>hibitors <strong>in</strong>hibit pro<strong>in</strong>flammatorysignal<strong>in</strong>g pathways to ameliorate <strong>in</strong>terleuk<strong>in</strong>-18-<strong>in</strong>duced cardiac hypertrophy. Physiol Genomics2011;16:1319e33.[139] Zupkovitz G, Grausenburger R, Brunmeir R, et al. The cycl<strong>in</strong>-dependent k<strong>in</strong>ase <strong>in</strong>hibitor p21 is a crucial targetfor histone deacetylase 1 as a regulator of cellular proliferation. Mol Cell Biol 2010;30:1171e81.[140] Grabiec AM, Krausz S, de Jager W, et al. Histone deacetylase <strong>in</strong>hibitors suppress <strong>in</strong>flammatory activation ofrheumatoid arthritis patient synovial macrophages and tissue. J Immunol 2010;184:2718e28.[141] Karouzakis E, Rengel Y, Jungel A, et al. DNA methylation regulates the expression of CXCL12 <strong>in</strong> rheumatoidarthritis synovial fibroblasts. Genes Immun 2011;12:643e52.251


This page <strong>in</strong>tentionally left blank


CHAPTER 13Epigenetic Mechanisms of<strong>Human</strong> Impr<strong>in</strong>t<strong>in</strong>gDisordersRichard H. Scott 1,2 , Gudrun E. Moore 11 Institute of Child Health, University College London, London, UK2 Great Ormond Street Hospital, London, UKCHAPTER OUTLINE13.1 Introduction 25413.2 Chromat<strong>in</strong> Structure ReflectsEpigenetic Modifications 25413.2.1 DNA Methylation 25513.3 DNA Methylation andTranscriptional Silenc<strong>in</strong>g 25513.4 Ma<strong>in</strong>tenance and Establishment ofDNA Methylation Dur<strong>in</strong>gDevelopment 25513.4.1 Histone Modifications 25613.4.2 Non-Histone DNA-B<strong>in</strong>d<strong>in</strong>gProte<strong>in</strong>s 25713.4.3 Non-Cod<strong>in</strong>g RNAs 25713.4.4 Cross-Talk Between EpigeneticSystems 25713.5 Genomic Impr<strong>in</strong>t<strong>in</strong>g 25713.5.1 Impr<strong>in</strong>ted Loci 25813.5.2 Impr<strong>in</strong>t<strong>in</strong>g Centers ControlImpr<strong>in</strong>t<strong>in</strong>g <strong>in</strong> cis 25813.5.3 Establishment of DNAMethylation at Impr<strong>in</strong>ted LociDur<strong>in</strong>g Development 26013.5.4 The 11p15 Impr<strong>in</strong>tedRegion 26013.5.5 Impr<strong>in</strong>ted Doma<strong>in</strong> 1 261H19 DMR Controls Impr<strong>in</strong>tedDoma<strong>in</strong> 1 262Chromat<strong>in</strong> Loop<strong>in</strong>g <strong>in</strong> theControl of Impr<strong>in</strong>tedDoma<strong>in</strong> 1 262The IGF2 Differentially MethylatedRegions 26213.5.6 Impr<strong>in</strong>ted Doma<strong>in</strong> 2 262KvDMR1 Controls Impr<strong>in</strong>t<strong>in</strong>g atImpr<strong>in</strong>ted Doma<strong>in</strong> 2 26313.5.7 Establishment of Impr<strong>in</strong>t<strong>in</strong>g atthe 11p15 GRR 26313.5.8 Abnormalities at Impr<strong>in</strong>tedLoci 26313.6 Uniparental Disomy 26313.7 Epimutations 26413.8 Impr<strong>in</strong>t<strong>in</strong>g Center Mutations 26413.9 Mutations <strong>in</strong> Impr<strong>in</strong>tedGenes 26413.10 Copy Number AbnormalitiesEncompass<strong>in</strong>g Impr<strong>in</strong>tedGenes 26413.11 Mutations <strong>in</strong> Impr<strong>in</strong>t<strong>in</strong>gEstablishment or Ma<strong>in</strong>tenanceMach<strong>in</strong>ery 26413.11.1 Phenotypes Associated withConstitutional Abnormalitiesat Impr<strong>in</strong>ted Loci 26513.12 Chromosome 6q24 26513.13 Chromosome 7 26513.14 Chromosome 11p15 26513.15 Chromosome 14q32.2 26713.16 Chromosome 15q11-q13 26813.17 Chromosome 20q13.32 26813.18 Hypomethylation at MultipleImpr<strong>in</strong>ted Loci 26813.19 Conclusion 269References 269253T. Tollefsbol (Ed): <strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>. DOI: 10.1016/B978-0-12-388415-2.00013-5Copyright Ó 2012 Elsevier Inc. All rights reserved.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>13.1 INTRODUCTION<strong>Epigenetics</strong> literally means “above genetics” and refers to the biological mechanisms otherthan alterations <strong>in</strong> DNA sequence that <strong>in</strong>fluence gene expression and that are stable throughcell division [1,2]. The word epigenetics was orig<strong>in</strong>ally co<strong>in</strong>ed by Wadd<strong>in</strong>gton <strong>in</strong> 1942 asa portmanteau of “epigenesis” and “genetics” to describe the process by which the genotypesgive rise to phenotypes dur<strong>in</strong>g development [3]. Nowadays, Wadd<strong>in</strong>gton’s def<strong>in</strong>ition would beconsidered to apply to the field of developmental biology <strong>in</strong> general whereas the mean<strong>in</strong>g ofthe word epigenetics has narrowed to specifically refer to non-genetic factors that <strong>in</strong>fluencegene expression.There are three widely accepted and closely <strong>in</strong>teract<strong>in</strong>g epigenetic mechanisms: (1) DNAmethylation; (2) histone modifications; (3) DNA b<strong>in</strong>d<strong>in</strong>g of Polycomb/Trithorax prote<strong>in</strong>s(Table 13.1). Many other specific factors as well as general alterations <strong>in</strong> chromat<strong>in</strong> structurealso correlate with different states of gene activity but are not considered primary epigeneticmodifications as they are not stable through cell division <strong>in</strong>dependent of their <strong>in</strong>itial trigger.This caveat elim<strong>in</strong>ates, for example, the DNA b<strong>in</strong>d<strong>in</strong>g of transcription factors from considerationas truly epigenetic. However, it also leads some to question the use of the term forsystems widely referred to as epigenetic such as histone modification, whose <strong>in</strong>dependentheritability through cell division is uncerta<strong>in</strong>.25413.2 CHROMATIN STRUCTURE REFLECTS EPIGENETICMODIFICATIONSChromat<strong>in</strong> is the complex of DNA, histones, and other DNA-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s and RNA thattogether make up chromosomes. Differences <strong>in</strong> chromat<strong>in</strong> structure are seen between genes <strong>in</strong>active and <strong>in</strong>active states and reflect underly<strong>in</strong>g epigenetic modifications. At its most extreme,chromat<strong>in</strong> can be considered to be either <strong>in</strong> an open, active conformation (euchromat<strong>in</strong>) orTABLE 13.1 Features of Transcriptionally Active and Inactive Chromat<strong>in</strong>Feature Active InactiveChromat<strong>in</strong> structure Open, extended Closed, condensedDNA methylation atNoYespromoterHistone methylatione H3K4 mono-/YesNotrimethylatione H3K4NoYesdimethylatione H3K9YesNomonomethylatione H3K9NoYestrimethylatione H3K27YesNomonomethylatione H3K27NoYestrimethylationHistone acetylatione H3K9 acetylation Yes Noe H3K14 acetylation Yes NoPolycomb complexb<strong>in</strong>d<strong>in</strong>gNoYesH3K4, Histone 3 lys<strong>in</strong>e 4; H3K9, Histone 3 lys<strong>in</strong>e 9; H3K14, Histone 3 lys<strong>in</strong>e 14; H3K27, Histone 3 lys<strong>in</strong>e 27.


CHAPTER 13Epigenetic Mechanisms of <strong>Human</strong> Impr<strong>in</strong>t<strong>in</strong>g Disordersa closed, <strong>in</strong>active conformation (heterochromat<strong>in</strong>). Other alterations observed <strong>in</strong>cludechanges <strong>in</strong> large-scale chromat<strong>in</strong> conformation and physical <strong>in</strong>teractions between normallydistant regions of chromat<strong>in</strong>.13.2.1 DNA MethylationDNA methylation was the first epigenetic modification to be identified and is perhaps the beststudied. In mammals, it is well established to have a mitotically stable silenc<strong>in</strong>g effect on geneswhen present at CpG dense promoter sequences [4]. In mammals, DNA methylation occurs atcytos<strong>in</strong>e residues to form 5-methylcytos<strong>in</strong>e and almost exclusively affects CpG d<strong>in</strong>ucleotides[4]. DNA methylation affects the large majority of CpG d<strong>in</strong>ucleotides <strong>in</strong> the genome and isfound broadly across <strong>in</strong>ter- and <strong>in</strong>tragenic sequences <strong>in</strong>clud<strong>in</strong>g the gene bodies of active genes.Unmethylated doma<strong>in</strong>s account for only 1e2% of the genome, the majority of which are CpGislands, short CG-rich stretches of sequence found preferentially at gene promoter regions.Genetic knock-out experiments have demonstrated that DNA methylation is essential forembryonic development, genomic impr<strong>in</strong>t<strong>in</strong>g and X-<strong>in</strong>activation and may be <strong>in</strong>volved <strong>in</strong> thesilenc<strong>in</strong>g of transposons [5e10].13.3 DNA METHYLATION AND TRANSCRIPTIONAL SILENCINGDNA methylation can directly reduce b<strong>in</strong>d<strong>in</strong>g of transcription factors, but its pr<strong>in</strong>cipal meansof transcriptional repression is thought to be via the recruitment of methyl-CpG b<strong>in</strong>d<strong>in</strong>gdoma<strong>in</strong> (MBD) prote<strong>in</strong>s which effect alterations <strong>in</strong> chromat<strong>in</strong> conformation, for exampleMBD1 and MECP2, which both result <strong>in</strong> histone modification [11].The silenc<strong>in</strong>g effect of DNA methylation is well established when it is present <strong>in</strong> CpG densepromoter regions. However, the large majority of silent genes do not have a methylated CpGisland at their promoter, <strong>in</strong>dicat<strong>in</strong>g that other means of epigenetic control must exist. Theeffect of methylation at promoters with low CpG density is not established. Furthermore, <strong>in</strong>most tissue types, DNA methylation is normally stably present through cell division andrelatively uniform between most cell types. The contribution of dynamic/tissue-specificchanges <strong>in</strong> methylation <strong>in</strong> the control of gene expression rema<strong>in</strong>s unclear [12,13].25513.4 MAINTENANCE AND ESTABLISHMENT OF DNAMETHYLATION DURING DEVELOPMENTDNA methylation is ma<strong>in</strong>ta<strong>in</strong>ed through cell division by the DNA methyltransferase, DNMT1.The symmetry of the CpG sequence means that both strands of DNA have a CpG d<strong>in</strong>ucleotide.The two strands typically share the same methylation status and this is crucial to the ma<strong>in</strong>tenanceof stable DNA methylation through mitotic division. Follow<strong>in</strong>g DNA replication, thetwo daughter double-stranded DNA molecules are hemimethylated (i.e. methylated on onestrand only). Methylation of the new strand of each daughter molecule is then performed byDNMT1 [14].In contrast to its relative stability <strong>in</strong> differentiated cells, dramatic changes <strong>in</strong> DNA methylationoccur dur<strong>in</strong>g mammalian development. This epigenetic reprogramm<strong>in</strong>g occurs <strong>in</strong> twostages; (1) reprogramm<strong>in</strong>g of germ cells; and (2) reprogramm<strong>in</strong>g of early embryonic cells(Figure 13.1). Each stage <strong>in</strong>volves a round of demethylation and a round of de novomethylation. De novo methylation is carried out by a variety of DNA methyltransferases<strong>in</strong>clud<strong>in</strong>g DNMT1, DNMT3A, and DNMT3B, some of which have germ cell and sex-specificisoforms [15].Primordial germ cells undergo genome-wide demethylation early <strong>in</strong> development, like otherpost-zygotic cell types and are largely demethylated until gonadal differentiation. Aftergonadal differentiation, de novo methylation occurs and leads to substantial methylation <strong>in</strong>both sperm and eggs, pr<strong>in</strong>cipally target<strong>in</strong>g transposons and repeat sequences but also


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>(A)(B)256FIGURE 13.1Changes <strong>in</strong> the overall level of DNA methylationdur<strong>in</strong>g mammalian development. (A) Changes <strong>in</strong>DNA methylation <strong>in</strong> germ cells. (B) Changes <strong>in</strong>DNA methylation follow<strong>in</strong>g fertilization. In bothpanels the level of DNA methylation is shown onthe vertical axis and developmental time on thehorizontal axis. Adapted from [16]. This figure isreproduced <strong>in</strong> the color plate section.impr<strong>in</strong>ted loci [15,16]. The overall level of methylation is somewhat higher <strong>in</strong> sperm than eggsand sex-specific differences occur at impr<strong>in</strong>ted loci. The tim<strong>in</strong>g of de novo methylation alsodiffers between the sexes. It occurs before meiosis <strong>in</strong> male germ cells and dur<strong>in</strong>g meioticprophase arrest I <strong>in</strong> female germ cells.After fertilization a further round of genome-wide demethylation then occurs. Shortly beforegastrulation, de novo methylation occurs. Follow<strong>in</strong>g this, somatic embryonic cells show thehigh level of methylation at sequences other than CpG islands that are seen <strong>in</strong> maturity.Trophoblast cells undergo de novo methylation but rema<strong>in</strong> relatively less methylated.Primordial germ cells rema<strong>in</strong> largely unmethylated until after gastrulation [16].13.4.1 Histone ModificationsHistones are the chief prote<strong>in</strong> component of chromat<strong>in</strong> and are assembled as octamericparticles made up of two copies of each of the four classes of core histone molecule, H2A, H2B,H3, and H4. One hundred and forty-six base-pairs of DNA are wound around each histoneoctamer form<strong>in</strong>g the basic build<strong>in</strong>g block of chromat<strong>in</strong> e the nucleosome. Many posttranslationalmodifications <strong>in</strong>volve histones, often <strong>in</strong> comb<strong>in</strong>ation with one another, andexert epigenetic control on gene expression. Foremost amongst these are the methylation andacetylation of lys<strong>in</strong>e residues <strong>in</strong> the N-term<strong>in</strong>al tails of histones H3 and H4 (Table 13.1).


CHAPTER 13Epigenetic Mechanisms of <strong>Human</strong> Impr<strong>in</strong>t<strong>in</strong>g DisordersModifications most commonly associated with active gene expression <strong>in</strong>clude monomethylationat H3K9 and H3K27 (i.e. H3 lys<strong>in</strong>e 9 and H3 lys<strong>in</strong>e 27), trimethylation of H3K4,and acetylation at H3K9 and H3K14. Repressive modifications <strong>in</strong>clude dimethylation of H3K4and trimethylation of H3K9, H3K27. Control of these modifications is exerted by a widevariety of histone methyltransferase, histone demethylase, histone acetylase, and histonedeacetylase enzymes [17,18].Several models have been proposed to expla<strong>in</strong> the heritability of histone modificationsthrough cell division, but none is proven (reviewed <strong>in</strong> [19]). Indeed it is unclear whetherhistone modifications themselves are transmitted to daughter chromat<strong>in</strong> strands follow<strong>in</strong>gDNA replication or if this is transmitted via a separate system, for example DNA methylationor the b<strong>in</strong>d<strong>in</strong>g of non-histone prote<strong>in</strong>s. While this uncerta<strong>in</strong>ty rema<strong>in</strong>s, some authors arguethat histone modifications should not be regarded as true epigenetic modifications.13.4.2 Non-Histone DNA-B<strong>in</strong>d<strong>in</strong>g Prote<strong>in</strong>sA variety of non-histone prote<strong>in</strong>s also b<strong>in</strong>d to DNA to affect chromat<strong>in</strong> structure and exertepigenetic control on gene expression. The best established of these are the Polycomb andTrithorax group prote<strong>in</strong>s which promote transcriptional repression and activation respectively,and both of which act stably through cell division. The two systems <strong>in</strong>teract closely with oneanother and with other epigenetic systems and have been implicated <strong>in</strong> the regulation of genes<strong>in</strong> early development and stem cell renewal. They are also <strong>in</strong>volved <strong>in</strong> X-<strong>in</strong>activation andgenomic impr<strong>in</strong>t<strong>in</strong>g [20,21].The Polycomb system consists of two prote<strong>in</strong> complexes. The Polycomb repressor complex 2,the so-called <strong>in</strong>itiation complex, b<strong>in</strong>ds to target DNA sequences and, through the action of itscomponent prote<strong>in</strong> EZH2, results <strong>in</strong> the repressive histone modification H3K27 trimethylation(Table 13.1). The Polycomb repressor complex 1, the so-called ma<strong>in</strong>tenance complex,recognizes this repressive mark and is crucial <strong>in</strong> the resultant transcriptional repression. Themechanism by which the complex causes repression is unknown [21]. The Trithorax systemalso acts through histone modification with the Trithorax prote<strong>in</strong> MLL caus<strong>in</strong>g the activat<strong>in</strong>ghistone modification H3K4 methylation. Polycomb/Trithorax-<strong>in</strong>duced epigenetic states arestable through cell division, and the Polycomb repressor complex 1 has recently been shownto rema<strong>in</strong> bound to DNA dur<strong>in</strong>g DNA replication [22].25713.4.3 Non-Cod<strong>in</strong>g RNAsNon-cod<strong>in</strong>g RNAs have been shown to contribute to epigenetic control of gene expression, forexample the Xist non-cod<strong>in</strong>g RNA which is central to X-<strong>in</strong>activation [23,24].13.4.4 Cross-Talk Between Epigenetic SystemsThere is extensive correlation between activat<strong>in</strong>g and repressive modifications <strong>in</strong> differentepigenetic systems so that they often appear to work <strong>in</strong> a concerted manner rather than asseparate systems (Table 13.1). In addition to simple correlations, an <strong>in</strong>creas<strong>in</strong>g number ofspecific <strong>in</strong>teractions are be<strong>in</strong>g identified between the systems, for example between DNAmethylation and histone deacetylation through the recruitment of methyl cytos<strong>in</strong>e b<strong>in</strong>d<strong>in</strong>gprote<strong>in</strong>s such as MECP2 and MBD1 and through the <strong>in</strong>hibition of b<strong>in</strong>d<strong>in</strong>g of DNMT3A and itsco-factor DNMT3L to H3 by trimethylation at H3K4. A detailed exposition of these <strong>in</strong>teractionsis beyond the scope of this chapter. They are reviewed by Reik [25] and Cedar andBergman [11].13.5 GENOMIC IMPRINTINGGenomic impr<strong>in</strong>t<strong>in</strong>g is an epigenetic phenomenon observed <strong>in</strong> mammals, seeded plants, andsome <strong>in</strong>sects <strong>in</strong> which certa<strong>in</strong> genes show parent-of-orig<strong>in</strong>-specific patterns of expression.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>Around 60 genes have been shown to be consistently impr<strong>in</strong>ted <strong>in</strong> man (Geneimpr<strong>in</strong>t database2008; Catalogue of Parent of Orig<strong>in</strong> Effects 2009) [74,75]. Some are impr<strong>in</strong>ted <strong>in</strong> all celltypes exam<strong>in</strong>ed, while others show tissue-specific impr<strong>in</strong>t<strong>in</strong>g, or are only impr<strong>in</strong>ted at certa<strong>in</strong>stages of development. Impr<strong>in</strong>ted genes are often arranged <strong>in</strong> clusters, each cluster spann<strong>in</strong>gup to several megabases (Table 13.2).One theory that perhaps best expla<strong>in</strong>s the evolution of impr<strong>in</strong>t<strong>in</strong>g is that of parental genomeconflict. This theory suggests that there is a conflict of evolutionary advantage between thepaternal genome with the maternal genome which is as a result of the mother carry<strong>in</strong>g theoffspr<strong>in</strong>g <strong>in</strong> utero. The maternal genome must preserve herself and resources for futureoffspr<strong>in</strong>g so limits supplies to the baby, whereas the paternal genome only needs to considerthe baby and encourages growth. Proponents of the theory po<strong>in</strong>t to the existence of a numberof impr<strong>in</strong>ted genes that regulate growth and the tendency for paternally expressed genes topromote growth and for maternally expressed genes to suppress growth [26,27].25813.5.1 Impr<strong>in</strong>ted LociFormal demonstration of impr<strong>in</strong>t<strong>in</strong>g requires demonstration of parent-of-orig<strong>in</strong>-specific geneexpression. This can be technically challeng<strong>in</strong>g as human tissues are difficult to obta<strong>in</strong>, limit<strong>in</strong>gsystematic expression analysis of the human genes. Instead, known impr<strong>in</strong>ted loci have oftenbeen identified follow<strong>in</strong>g the observation of features suggestive of impr<strong>in</strong>t<strong>in</strong>g, <strong>in</strong>clud<strong>in</strong>g:1. Phenotypic abnormality result<strong>in</strong>g from uniparental disomy (UPD)2. Parent-of-orig<strong>in</strong>-specific effects of mutation, copy number abnormality or chromosomalrearrangement3. Parent-of-orig<strong>in</strong> effects of allelic loss <strong>in</strong> cancers4. Parent-of-orig<strong>in</strong>-specific epigenetic modifications <strong>in</strong> the region (for example differentialmethylation)5. DNA sequence characteristics similar to other impr<strong>in</strong>ted loci6. Evidence of impr<strong>in</strong>t<strong>in</strong>g <strong>in</strong> other species, e.g. mouse.Major clusters of impr<strong>in</strong>ted genes have been identified at 7q21.3, 7q32.2, 11p15, 15q11.2,19q13.4, and 20q13.32 (Table 13.2).13.5.2 Impr<strong>in</strong>t<strong>in</strong>g Centers Control Impr<strong>in</strong>t<strong>in</strong>g <strong>in</strong> cisImpr<strong>in</strong>t<strong>in</strong>g is controlled by epigenetic modifications at cis-act<strong>in</strong>g regulatory sequence elementstermed impr<strong>in</strong>t<strong>in</strong>g centers. The mechanism of control has been elucidated only <strong>in</strong> a subset ofimpr<strong>in</strong>ted loci. Even <strong>in</strong> this small number of loci, the variety of different mechanisms operat<strong>in</strong>gis strik<strong>in</strong>g. A shared feature is the presence of areas of parent-of-orig<strong>in</strong>-specific DNAmethylation termed differentially methylated regions (DMRs). These DMRs are CpG-richsequences located either at the promoter of an impr<strong>in</strong>ted gene or <strong>in</strong> a more distant regulatorysequence called an impr<strong>in</strong>t<strong>in</strong>g center. This differential methylation is associated with ma<strong>in</strong>tenanceof differential (i.e. impr<strong>in</strong>ted) expression of genes <strong>in</strong> the region. A s<strong>in</strong>gle differentiallymethylated impr<strong>in</strong>t<strong>in</strong>g center often appears to control impr<strong>in</strong>t<strong>in</strong>g of multiple genes <strong>in</strong>a cluster. This is sometimes termed a primary DMR. Differential methylation is sometimes seenat other nearby “secondary” DMRs which are established as a result of the action of theimpr<strong>in</strong>t<strong>in</strong>g center/primary DMR and which <strong>in</strong> some cases have been shown to be <strong>in</strong>volved <strong>in</strong>the ma<strong>in</strong>tenance of impr<strong>in</strong>t<strong>in</strong>g <strong>in</strong> the region [28].At some loci, differences <strong>in</strong> other epigenetic modifications have been demonstrated betweenthe two parental alleles <strong>in</strong>clud<strong>in</strong>g differential histone modification, Polycomb complexb<strong>in</strong>d<strong>in</strong>g as well as the differential b<strong>in</strong>d<strong>in</strong>g of the CCCTC-b<strong>in</strong>d<strong>in</strong>g factor CTCF and differences<strong>in</strong> high-order chromat<strong>in</strong> structure such as loop<strong>in</strong>g of chromat<strong>in</strong> to allow access of genes todistant enhancer elements. A further feature shared by a number of loci is the presence ofmultiple overlapp<strong>in</strong>g, often untranslated, transcripts that may play a regulatory function.


CHAPTER 13Epigenetic Mechanisms of <strong>Human</strong> Impr<strong>in</strong>t<strong>in</strong>g DisordersTABLE 13.2 Genes/Transcripts Reported to Show Impr<strong>in</strong>ted Expression <strong>in</strong> ManGene Location Expressed AlleleTP73 1p36.3 MaternalDIRAS3 1p31.3 PaternalLRRTM1 2p12 PaternalNAP1L5 4q22.1 PaternalPRIM2 6p11.2 MaternalPLAGL1 6q24.2 PaternalHYMAI 6q24.2 PaternalIGF2R 6q25.3 MaternalSLC22A2 6q25.3 MaternalSLC22A3 6q25.3 MaternalDDC 7p12.2 Isoform dependentGRB10 7p12.2 Isoform dependentCALCR 7q21.3 MaternalTFPI2 7q21.3 MaternalSGCE 7q21.3 PaternalPEG10 7q21.3 PaternalPPP1R9A 7q21.3 MaternalDLX5 7q21.3 MaternalCPA4 7q32.2 MaternalMEST 7q32.2 PaternalMESTIT1 7q32.2 PaternalCOPG2 7q32.2 PaternalCOPG2IT1 7q32.2 PaternalKLF14 7q32.2 MaternalDLGAP2 8p23.3 PaternalKCNK9 8q24.3 MaternalINPP5F V2 10q26.11 PaternalH19 11p15.5 MaternalIGF2 11p15.5 PaternalIGF2AS 11p15.5 PaternalINS 11p15.5 PaternalKCNQ1 11p15.5 MaternalKCNQ1OT1 11p15.5 PaternalKCNQ1DN 11p15.5 MaternalCDKN1C 11p15.5 MaternalSLC22A18AS 11p15.5 MaternalSLC22A18 11p15.5 MaternalPHLDA2 11p15.5 MaternalOSBPL5 11p15.4 MaternalZNF215 11p15.4 MaternalAWT1 11p13 PaternalWT1-AS 11p13 PaternalWIF1 12q14.3 PaternalDLK1 14q32.2 PaternalMEG3 14q32.2 MaternalRTL1 14q32.2 PaternalRTLas 14q32.2 MaternalMKRN3 15q11.2 PaternalMAGEL2 15q11.2 PaternalNDN 15q11.2 PaternalW89101 15q11.2 PaternalSNRPN 15q11.2 PaternalSNURF 15q11.2 PaternalSNORD107 15q11.2 PaternalSNORD64 15q11.2 PaternalSNORD108 15q11.2 Paternal259Cont<strong>in</strong>ued


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>TABLE 13.2Genes/Transcripts Reported to Show Impr<strong>in</strong>ted Expression <strong>in</strong>Mandcont<strong>in</strong>uedGene Location Expressed AlleleSNORD109A 15q11.2 PaternalPWCR1 15q11.2 PaternalSNORD115 cluster 15q11.2 PaternalSNORD116 15q11.2 PaternalSNORD109B 15q11.2 PaternalUBE3A 15q11.2 MaternalATP10A 15q11.2 MaternalH73492 15q13 PaternalZNF597 16p13.3 MaternalTCEB3C 18q21.1 MaternalZNF331 19q13.41 MaternalZIM2 19q13.43 PaternalPEG3 19q13.43 PaternalITUP1 19q13.43 PaternalZNF264 19q13.43 MaternalPSIMCT-1 20q11.21 PaternalNNAT 20q11.23 PaternalBLCAP 20q11.2 Isoform dependentL3MBTL 20q13.12 PaternalGNAS 20q13.32 MaternalGNAS Exon A/B 20q13.32 PaternalGNASXL 20q13.32 PaternalNESP 20q13.32 MaternalNESPAS 20q13.32 Paternal26013.5.3 Establishment of DNA Methylation at Impr<strong>in</strong>ted Loci Dur<strong>in</strong>gDevelopmentThe establishment of impr<strong>in</strong>t<strong>in</strong>g also <strong>in</strong>volves DNA methylation, histone modification, and/orPolycomb gene b<strong>in</strong>d<strong>in</strong>g, although it should be noted that many of the data <strong>in</strong> this area derivefrom analyses <strong>in</strong> the mouse and are assumed to apply also <strong>in</strong> man. As with other sequences,DNA methylation at impr<strong>in</strong>ted loci is reprogrammed dur<strong>in</strong>g germ cell development. However,the pattern of methylation differs between the male and female germ cells, result<strong>in</strong>g <strong>in</strong> theestablishment of primary DMRs and therefore of impr<strong>in</strong>t<strong>in</strong>g. A further difference fromreprogramm<strong>in</strong>g of non-impr<strong>in</strong>ted loci is that DMRs escape the second round of demethylationand de novo remethylation <strong>in</strong> early embryonic development.Impr<strong>in</strong>ted regions undergo DNA demethylation similar to that at non-impr<strong>in</strong>ted regions <strong>in</strong>primordial germ cells and are almost entirely demethylated when they enter the gonads shortlyafter gastrulation. Methylation of impr<strong>in</strong>ted genes <strong>in</strong> the male germl<strong>in</strong>e occurs at only a smallnumber of loci, the best-studied be<strong>in</strong>g the H19 DMR at 11p15. As with non-impr<strong>in</strong>ted loci,methylation at paternally methylated DMRs such as H19 occurs before meiosis [29]. Methylationat a larger number of impr<strong>in</strong>ted loci occurs <strong>in</strong> the female germl<strong>in</strong>e. As with methylationat other loci, this occurs after meiosis I [30]. Unlike the majority of other sequences, impr<strong>in</strong>tedloci appear to escape the genome-wide demethylation that occurs after fertilization, allow<strong>in</strong>gthem to reta<strong>in</strong> the differential methylation of the paternal and maternal alleles establisheddur<strong>in</strong>g germ cell development.13.5.4 The 11p15 Impr<strong>in</strong>ted RegionThe detailed description of the structure and control mechanisms of each of the knownimpr<strong>in</strong>ted regions is beyond the scope of this chapter. However, <strong>in</strong> order to illustrate a number


CHAPTER 13Epigenetic Mechanisms of <strong>Human</strong> Impr<strong>in</strong>t<strong>in</strong>g Disordersof the shared features of impr<strong>in</strong>ted regions, we set out the normal structure and function of the11p15 impr<strong>in</strong>ted region. The region has been studied extensively <strong>in</strong> man and is disrupted <strong>in</strong>the human disorders BeckwitheWiedemann syndrome and SilvereRussell syndrome.In the simplest terms, the paternal 11p15 allele promotes growth through the expression ofgrowth-promot<strong>in</strong>g genes and the silenc<strong>in</strong>g of growth-suppress<strong>in</strong>g genes and the maternal11p15 allele suppresses growth through the expression of growth-suppress<strong>in</strong>g genes and thesilenc<strong>in</strong>g of growth-promot<strong>in</strong>g genes. In fact, the 11p15 Growth Regulatory Region (GRR)consists of two, apparently <strong>in</strong>dependent, impr<strong>in</strong>ted doma<strong>in</strong>s each controlled <strong>in</strong> cis by its owndifferentially methylated impr<strong>in</strong>t<strong>in</strong>g center (Figure 13.2). Each doma<strong>in</strong> conta<strong>in</strong>s a cluster ofimpr<strong>in</strong>ted genes which <strong>in</strong>clude growth promoters and growth suppressors.13.5.5 Impr<strong>in</strong>ted Doma<strong>in</strong> 1Impr<strong>in</strong>ted doma<strong>in</strong> 1 is the more telomeric of the two doma<strong>in</strong>s and conta<strong>in</strong>s the paternallyexpressed growth promoter IGF2 (<strong>in</strong>sul<strong>in</strong>-like growth factor 2) and the maternally expressednon-cod<strong>in</strong>g H19 (Figure 13.2) [31]. Impr<strong>in</strong>t<strong>in</strong>g at H19 and IGF2 is observed <strong>in</strong> both placentaland embryonic tissues and is ma<strong>in</strong>ta<strong>in</strong>ed <strong>in</strong> maturity <strong>in</strong> many differentiated tissues.Impr<strong>in</strong>t<strong>in</strong>g <strong>in</strong> the doma<strong>in</strong> is controlled by a paternally methylated impr<strong>in</strong>t<strong>in</strong>g center immediatelyupstream of H19, known as the H19 DMR. It is also referred to as impr<strong>in</strong>t<strong>in</strong>g center 1 orimpr<strong>in</strong>t<strong>in</strong>g control region 1 and is thought to act as a physical <strong>in</strong>sulator, controll<strong>in</strong>g access of(A)261(B)FIGURE 13.2Schematic diagram of the normal 11p15 GRR. (A) The maternal and paternal alleles at the normal 11p15 growth-regulatory region. The region is arranged <strong>in</strong>two impr<strong>in</strong>ted doma<strong>in</strong>s, the more telomeric impr<strong>in</strong>ted doma<strong>in</strong> 1 and the more centromeric impr<strong>in</strong>ted doma<strong>in</strong> 2. Each impr<strong>in</strong>ted doma<strong>in</strong> is controlled byan impr<strong>in</strong>t<strong>in</strong>g center conta<strong>in</strong><strong>in</strong>g a differentially methylated region (DMR). Impr<strong>in</strong>ted doma<strong>in</strong> 1 is controlled by the H19 DMR. This is methylated on the paternalallele (filled lollipops) and unmethylated on the paternal allele (open lollipops). Impr<strong>in</strong>ted doma<strong>in</strong> 2 is controlled by KvDMR1. This is methylated on the maternalallele and unmethylated on the paternal allele. At impr<strong>in</strong>ted doma<strong>in</strong> 1, IGF2 is expressed (solid outl<strong>in</strong>e and arrow at 5 0 end) from the paternal allele but silent(gray dashed outl<strong>in</strong>e and bar at 5 0 end) on the maternal allele while the non-cod<strong>in</strong>g H19 transcript is expressed from the maternal allele and silent onthe paternal allele. At impr<strong>in</strong>ted doma<strong>in</strong> 2, a number of genes <strong>in</strong>clud<strong>in</strong>g CDKN1C are expressed from the maternal allele but silent form the paternal allele, whileKCNQ1OT1 is expressed from the paternal allele but silent from the maternal allele. (B) Detailed view of the structure of the H19 DMR, which is arranged <strong>in</strong>two repeat blocks each conta<strong>in</strong><strong>in</strong>g one A- and three or four B-repeat elements. Six of the B-repeat elements conta<strong>in</strong><strong>in</strong>g target sites for the CTCF z<strong>in</strong>c-f<strong>in</strong>gerprote<strong>in</strong> (numbered above the repeat blocks) and a seventh CTCF target site lies between the repeat blocks and the H19 transcription start site.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>the compet<strong>in</strong>g H19 and IGF2 genes to telomeric enhancer elements [32]. On the normalmaternal allele, the H19 DMR is unmethylated allow<strong>in</strong>g H19 access to these enhancers. Thisresults <strong>in</strong> expression of H19 and silenc<strong>in</strong>g of IGF2. On the normal paternal allele, the H19DMR is methylated, allow<strong>in</strong>g IGF2 access to the enhancers. This results <strong>in</strong> expression of IGF2and silenc<strong>in</strong>g of H19.H19 DMR CONTROLS IMPRINTED DOMAIN 1In man, differential methylation at the H19 DMR extends for at least 5.5 kb upstream from the5 0 end of the H19 gene. At its core is a, 3.8-kb span of repeated sequence elements whichconta<strong>in</strong> multiple target sites for CTCF (Figure 13.2) [33,34]. These are arranged <strong>in</strong> two repeatblocks conta<strong>in</strong><strong>in</strong>g A- and B-repeat elements. Six CTCF target sites are present with<strong>in</strong> B-repeatelements and a seventh lies between the repeat blocks and the H19 transcription start site.B<strong>in</strong>d<strong>in</strong>g of CTCF at these sites occurs preferentially to the unmethylated maternal allele andmay protect it from abnormal methylation and/or mediate its function as a chromat<strong>in</strong> <strong>in</strong>sulator[35]. The functional importance of these CTCF target sites is supported by the H19hypermethylation and IGF2 loss of impr<strong>in</strong>t<strong>in</strong>g seen with their deletion or disruption of thematernal allele <strong>in</strong> model organisms and <strong>in</strong> man [31,34e37].262CHROMATIN LOOPING IN THE CONTROL OF IMPRINTED DOMAIN 1In the mouse, parent-of-orig<strong>in</strong>-specific chromat<strong>in</strong> loops have been found at impr<strong>in</strong>ted doma<strong>in</strong>1 and are thought to be important <strong>in</strong> the control of impr<strong>in</strong>t<strong>in</strong>g. These physical alterations <strong>in</strong>chromat<strong>in</strong> conformation mediate what is often referred to as the chromat<strong>in</strong> <strong>in</strong>sulator functionof the H19 DMR. The chromat<strong>in</strong> loops are formed by the physical <strong>in</strong>teraction of the H19 DMRwith differentially methylated regions at IGF2 and the different loops formed on each parentalallele probably result <strong>in</strong> impr<strong>in</strong>t<strong>in</strong>g at H19 and IGF2 by alter<strong>in</strong>g their access to enhancerelements [38].The unmethylated maternal H19 DMR is bound by CTCF and physically <strong>in</strong>teracts with IGF2DMR1. This creates two chromat<strong>in</strong> doma<strong>in</strong>s, with H19 <strong>in</strong> an active doma<strong>in</strong> with its enhancersand IGF2 <strong>in</strong> an <strong>in</strong>active doma<strong>in</strong> away from the enhancers. The methylated paternal H19 DMRis not bound by CTCF and <strong>in</strong>teracts with IGF2 DMR2. This results <strong>in</strong> IGF2 ly<strong>in</strong>g <strong>in</strong> the activedoma<strong>in</strong>, with access to the enhancers telomeric of H19. H19 is <strong>in</strong> the active doma<strong>in</strong> butsilenced, probably by the presence DNA methylation at its promoter (which is with<strong>in</strong> the H19DMR). It is presumed that a similar mechanism operates <strong>in</strong> man, though this is yet to bedemonstrated and the role of the human IGF2 DMRs is uncerta<strong>in</strong>.THE IGF2 DIFFERENTIALLY METHYLATED REGIONSThree differentially methylated regions at IGF2 have been identified <strong>in</strong> the mouse (IGF2DMR0, DMR1, and DMR2). As described above, IGF2 DMR1 and DMR2 have been demonstratedto be <strong>in</strong>volved <strong>in</strong> the formation of parent-of-orig<strong>in</strong>-specific chromat<strong>in</strong> loops. In man,only two IGF2 DMRs have been identified, the paternally methylated IGF2 DMR0 and IGF2DMR2 [38]. Their roles are not known and understand<strong>in</strong>g of the region was h<strong>in</strong>dered by <strong>in</strong>itialreports which <strong>in</strong>correctly assigned the parent-of-orig<strong>in</strong> of the methylated allele at IGF2 DMR0[39,40].13.5.6 Impr<strong>in</strong>ted Doma<strong>in</strong> 2Impr<strong>in</strong>ted doma<strong>in</strong> 2 conta<strong>in</strong>s the paternally expressed non-cod<strong>in</strong>g KCNQ1OT1 (KCNQ1-overlapp<strong>in</strong>g transcript 1) and a number of maternally expressed genes <strong>in</strong>clud<strong>in</strong>g the growthsuppressor CDKN1C (Cycl<strong>in</strong> dependent k<strong>in</strong>ase <strong>in</strong>hibitor 1C; Figure 13.2) [64]. KCNQ1OT1 andCDKN1C ma<strong>in</strong>ta<strong>in</strong> impr<strong>in</strong>ted expression <strong>in</strong> embryonic and many differentiated tissues. Bycontrast a number of maternally expressed genes <strong>in</strong> the region other than CDKN1C areimpr<strong>in</strong>ted only <strong>in</strong> the placenta or <strong>in</strong> a subset of embryonic or differentiated tissues [41].


CHAPTER 13Epigenetic Mechanisms of <strong>Human</strong> Impr<strong>in</strong>t<strong>in</strong>g DisordersImpr<strong>in</strong>t<strong>in</strong>g <strong>in</strong> the doma<strong>in</strong> is controlled <strong>in</strong> cis by an impr<strong>in</strong>t<strong>in</strong>g center at the promoter ofKCNQ1OT1 known as KvDMR1, which is also referred to as KCNQ1OT1, LIT1, impr<strong>in</strong>t<strong>in</strong>gcenter 2, or impr<strong>in</strong>t<strong>in</strong>g control region 2. On the normal maternal allele KvDMR1 is methylated,result<strong>in</strong>g <strong>in</strong> silenc<strong>in</strong>g of KCNQ1OT1 and expression of CDKN1C. On the normalpaternal allele KvDMR1 is unmethylated, result<strong>in</strong>g <strong>in</strong> expression of KCNQ1OT1 and silenc<strong>in</strong>gof CDKN1C.KVDMR1 CONTROLS IMPRINTING AT IMPRINTED DOMAIN 2KvDMR1 may control impr<strong>in</strong>t<strong>in</strong>g <strong>in</strong> doma<strong>in</strong> 2 through more than one mechanism. Two ma<strong>in</strong>mechanisms have been proposed: (1) a non-cod<strong>in</strong>g RNA mediated mechanism; and (2) anenhancer competition-mediated mechanism.On the paternal allele, many of the maternally expressed genes show impr<strong>in</strong>ted expression <strong>in</strong>the placenta <strong>in</strong> the absence of differential DNA methylation at their promoters [42]. This issimilar to that seen <strong>in</strong> the process of X <strong>in</strong>activation and it has been proposed that silenc<strong>in</strong>g ofthese genes on the paternal allele occurs by a similar process to that seen on the <strong>in</strong>active X:through repressive histone H3K27 methylation mediated by Polycomb group prote<strong>in</strong>s. Thisrepressive histone modification is observed <strong>in</strong> placental tissues across the doma<strong>in</strong> and may betargeted to the paternal allele by coat<strong>in</strong>g of the region <strong>in</strong> cis by the paternally expressed noncod<strong>in</strong>gRNA KCNQ1OT1, a mechanism parallel to that mediated by the Xist transcript on the<strong>in</strong>active X [43].The enhancer-competition mechanism similar to that seen at impr<strong>in</strong>ted doma<strong>in</strong> 1 has alsobeen proposed to expla<strong>in</strong> the ma<strong>in</strong>tenance of impr<strong>in</strong>t<strong>in</strong>g <strong>in</strong> the doma<strong>in</strong> that may be central tothe ma<strong>in</strong>tenance of impr<strong>in</strong>t<strong>in</strong>g at KCNQ1OT1, CDKN1C, and other genes that are impr<strong>in</strong>ted<strong>in</strong> embryonic tissues [44]. There is currently limited evidence to provide mechanistic understand<strong>in</strong>gof this model [45].13.5.7 Establishment of Impr<strong>in</strong>t<strong>in</strong>g at the 11p15 GRRDifferential methylation is observed <strong>in</strong> the germl<strong>in</strong>e at the impr<strong>in</strong>t<strong>in</strong>g centers <strong>in</strong> bothdoma<strong>in</strong>s: at the H19 DMR <strong>in</strong> impr<strong>in</strong>ted doma<strong>in</strong> 1 and at KvDMR1 <strong>in</strong> impr<strong>in</strong>ted doma<strong>in</strong> 2[42,44]. It is thought that this germl<strong>in</strong>e differential methylation is the driver of the establishmentof post-zygotic impr<strong>in</strong>t<strong>in</strong>g at each doma<strong>in</strong>. At impr<strong>in</strong>ted doma<strong>in</strong> 1 this modelpredicts that differential methylation at the H19 DMR is the first event <strong>in</strong> a cascade that<strong>in</strong>volves differential b<strong>in</strong>d<strong>in</strong>g of CTCF and possibly other factors, differential methylation at theIGF2 DMRs, establishment of parent-of-orig<strong>in</strong>-specific chromat<strong>in</strong> loops, and therefore thedifferential access to enhancers that mediate impr<strong>in</strong>t<strong>in</strong>g <strong>in</strong> the region. At impr<strong>in</strong>ted doma<strong>in</strong> 2,the model predicts that differential methylation at KvDMR1 is the first event <strong>in</strong> a cascade that<strong>in</strong>volves impr<strong>in</strong>ted expression of the non-cod<strong>in</strong>g KCNQ1OT1 transcript, which <strong>in</strong> turn iscrucial to the establishment of parent-of-orig<strong>in</strong>-specific histone modifications and otherepigenetic modifications that mediate impr<strong>in</strong>t<strong>in</strong>g at other genes.26313.5.8 Abnormalities at Impr<strong>in</strong>ted LociA variety of classes of molecular defects can result <strong>in</strong> disruption at impr<strong>in</strong>ted loci. These<strong>in</strong>clude both epigenetic and genetic defects.13.6 UNIPARENTAL DISOMYUniparental disomy (UPD) results when both chromosomes of a pair are <strong>in</strong>herited from thesame parent. When UPD encompasses an impr<strong>in</strong>ted locus, both alleles show the characteristicsof the reta<strong>in</strong>ed allele. For example, <strong>in</strong> a region of paternal UPD (pUPD), paternallyexpressed genes are expressed from both alleles and maternally expressed genes are silenced.Uniparental disomy can occur by a variety of mechanisms, either prezygotic (usually errors of


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>meiosis) or postzygotic (errors of mitosis) and can affect whole chromosomes or besegmental [46].13.7 EPIMUTATIONSEpimutations are isolated epigenetic defects that result <strong>in</strong> disruption of the normal pattern ofexpression. This can result <strong>in</strong> the silenc<strong>in</strong>g of the normally active allele or expression ofa normally silent allele. At impr<strong>in</strong>ted loci, the activation of a normally silent allele is termedloss of impr<strong>in</strong>t<strong>in</strong>g and results <strong>in</strong> biallelic expression of a normally monoallelically expressedgene. Disrupted expression is frequently associated with disruption of differential DNAmethylation, result<strong>in</strong>g <strong>in</strong> hypomethylation or hypermethylation at a DMR. The primary defectunderly<strong>in</strong>g epimutations is not known. One possibility is that they result from the stochasticloss or ga<strong>in</strong> of DNA methylation at key CpGs with<strong>in</strong> the relevant DMR.13.8 IMPRINTING CENTER MUTATIONSImpr<strong>in</strong>t<strong>in</strong>g center mutations are genetic mutations at impr<strong>in</strong>t<strong>in</strong>g control regions that result <strong>in</strong>epigenetic disruption of expression of the genes under their control (<strong>in</strong> cis). They are oftenassociated with disruption of methylation at DMRs and, as with other mutations at impr<strong>in</strong>tedloci, they show parent-of-orig<strong>in</strong>-specific pathogenicity. Impr<strong>in</strong>t<strong>in</strong>g center mutations identifiedto date have largely been microdeletions spann<strong>in</strong>g several kilobases and <strong>in</strong> some casesmegabases [34,47e51]. The 2.9-kb micro<strong>in</strong>sertion identified at the 11p15 H19 DMR representsa further class of impr<strong>in</strong>t<strong>in</strong>g center mutation and some balanced chromosome rearrangementswith breakpo<strong>in</strong>ts at impr<strong>in</strong>ted loci may be further examples. The mechanism ofpathogenicity of impr<strong>in</strong>t<strong>in</strong>g center mutations is often obscure.26413.9 MUTATIONS IN IMPRINTED GENESMutations <strong>in</strong> a number of impr<strong>in</strong>ted genes have been reported. They are typically only ofconsequence when <strong>in</strong>herited on the active allele, that is they show parent-of-orig<strong>in</strong>-dependentpathogenicity [52].13.10 COPY NUMBER ABNORMALITIES ENCOMPASSINGIMPRINTED GENESLarge copy number defects encompass<strong>in</strong>g impr<strong>in</strong>ted genes have been reported at a number ofloci, either as a result of <strong>in</strong>terstitial disruptions or unbalanced chromosome translocations. Aswith mutations <strong>in</strong> impr<strong>in</strong>ted genes, they have parent-of-orig<strong>in</strong>-specific effects: they would onlybe expected to alter the expression of genes which are active on the disrupted allele.13.11 MUTATIONS IN IMPRINTING ESTABLISHMENT ORMAINTENANCE MACHINERYRecently, biallelic mutations <strong>in</strong> ZFP57, a gene important <strong>in</strong> the establishment of DNAmethylation at maternally methylated DMRs, have been found to cause hypomethylation atmultiple impr<strong>in</strong>ted loci [56]. This represents an example of a dist<strong>in</strong>ct mechanism ofimpr<strong>in</strong>t<strong>in</strong>g disruption: mutation of a component of the mach<strong>in</strong>ery of the establishment orma<strong>in</strong>tenance of impr<strong>in</strong>t<strong>in</strong>g. Other examples <strong>in</strong>clude biallelic mutations <strong>in</strong> the genes NALP7and C6ORF221, which cause recurrent hydatidiform mole [53,54]. Interest<strong>in</strong>gly, mutations <strong>in</strong>the related gene NALP2 have been reported <strong>in</strong> one family with BeckwitheWiedemannsyndrome [55].


CHAPTER 13Epigenetic Mechanisms of <strong>Human</strong> Impr<strong>in</strong>t<strong>in</strong>g Disorders13.11.1 Phenotypes Associated with Constitutional Abnormalities atImpr<strong>in</strong>ted LociConstitutional abnormalities at impr<strong>in</strong>ted loci underlie a number of congenital syndromes <strong>in</strong>man (Table 13.3). The study of these disorders and the molecular abnormalities that underliethem resulted <strong>in</strong> the identification of many of the known impr<strong>in</strong>ted loci and has been centralto much of our understand<strong>in</strong>g of normal and abnormal impr<strong>in</strong>t<strong>in</strong>g. In a number of cases theseconstitutional molecular abnormalities, despite be<strong>in</strong>g present soma-wide, are mosaic.13.12 CHROMOSOME 6Q24Disruption of the impr<strong>in</strong>ted locus at 6q24 results <strong>in</strong> transient neonatal diabetes mellitus [57].In addition to diabetes mellitus from the neonatal period that can last until 18 months of age,other features of the condition <strong>in</strong>clude <strong>in</strong>trauter<strong>in</strong>e growth retardation, macroglossia, andumbilical hernia. Abnormalities at 6q24 account for approximately 90% of cases of transientneonatal diabetes mellitus and result <strong>in</strong> <strong>in</strong>creased expression of the paternally expressed genesPLAGL1 (Pleomorphic adenoma gene-like 1) and the HYMAI (Hydatidiform mole-associated andimpr<strong>in</strong>ted) transcript. Reported abnormalities <strong>in</strong>clude paternal UPD, epimutation at the DMRsat PLAGL1 and HYMAI (hypomethylation of the maternal allele) and paternal duplicationsencompass<strong>in</strong>g both genes [57e59].13.13 CHROMOSOME 7SilvereRussell syndrome is a growth-restriction disorder associated with pre- and postnatalgrowth restriction, relative macrocephaly, growth asymmetry, fifth f<strong>in</strong>ger cl<strong>in</strong>odactyly, anda characteristic facial appearance. Maternal uniparental disomy for chromosome 7 is found <strong>in</strong>approximately 10% of cases. Uniparental disomy usually affects the whole of chromosome 7but maternal segmental abnormalities have also been reported, provid<strong>in</strong>g <strong>in</strong>sights <strong>in</strong>to thelikely critical region [60,61]. Extensive work has identified a number of impr<strong>in</strong>ted genes onchromosome 7 (Table 13.2) [62,63]. As discussed below, abnormalities at the 11p15 growthregulatory region account for a further 25e40% of cases of SilvereRussell syndrome.26513.14 CHROMOSOME 11P15As set out earlier <strong>in</strong> this chapter, the 11p15 growth-regulatory region, is arranged <strong>in</strong> two adjacentbut <strong>in</strong>dependent impr<strong>in</strong>ted doma<strong>in</strong>s (Figure 13.2). Two oppos<strong>in</strong>g groups of abnormalities <strong>in</strong>the region result <strong>in</strong> overgrowth (most characteristically BeckwitheWiedemann syndrome) andgrowth restriction (most characteristically SilvereRussell syndrome) [64,65].Overgrowth is caused by abnormalities that result <strong>in</strong> <strong>in</strong>creased expression of paternallyexpressed growth promoters such as IGF2 or decreased expression of maternally expressedgrowth suppressors such as CDKN1C. Growth-promot<strong>in</strong>g abnormalities at 11p15 are found <strong>in</strong>approximately 80% of cases of BeckwitheWiedemann syndrome and <strong>in</strong>clude: paternaluniparental disomy 11p15; epimutations at impr<strong>in</strong>ted doma<strong>in</strong> 1 (hypermethylation of thematernal H19 DMR) or impr<strong>in</strong>ted doma<strong>in</strong> 2 (hypomethylation of the maternal KvDMR1);impr<strong>in</strong>t<strong>in</strong>g center mutations at impr<strong>in</strong>ted doma<strong>in</strong> 1 that result <strong>in</strong> H19 hypermethylation(microdeletion or micro<strong>in</strong>sertion with<strong>in</strong> the H19 impr<strong>in</strong>t<strong>in</strong>g control region); paternalduplications; maternal deletion of KCNQ1OT1 and KvDMR1; and maternal mutations <strong>in</strong>CDKN1C [64].Conversely, abnormalities that result <strong>in</strong> a net decrease <strong>in</strong> expression of growth promoters suchas IGF2 result <strong>in</strong> growth restriction. Growth-suppress<strong>in</strong>g abnormalities at 11p15 are found <strong>in</strong>25e40% of cases of SilvereRussell syndrome and <strong>in</strong>clude epimutations at impr<strong>in</strong>ted doma<strong>in</strong>1 (hypomethylation of the paternal H19 DMR) and maternal duplications of the region [65].


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>TABLE 13.3 Phenotypes Associated with Constitutional Abnormalities at Impr<strong>in</strong>tedLociLocus/Phenotype6q24Transient neonatal diabetesmellitusClass of Abnormality AbnormalityUPDEpimutationDuplication impr<strong>in</strong>tedgenesPaternal UPD 6q24Hypomethylation maternalPLAGL1 and HYMAI DMRsDuplication paternal 6q24266Chromosome 7Growth retardation/SilvereRussell UPDsyndrome11p15Overgrowth/Beckwithe UPDWiedemann syndromeEpimutationGrowth retardation/SilvereRussellsyndromeEpimutationImpr<strong>in</strong>t<strong>in</strong>g centremutationMutation impr<strong>in</strong>tedgenesDeletion impr<strong>in</strong>tedgenesDuplication impr<strong>in</strong>tedgenesEpimutationDuplication impr<strong>in</strong>tedgenesMaternal UPD 7, usually affect<strong>in</strong>gthe whole chromosomePaternal UPD 11p15Hypermethylation maternal H19DMRHypomethylation maternalKvDMR1 (referred to as KvDMR1loss of methylation)Microdeletion/micro<strong>in</strong>sertionmaternal H19 DMR caus<strong>in</strong>g H19hypermethylationMutation maternal CDKN1CDeletion encompass<strong>in</strong>g maternalKCNQ1OT1 and KvDMR1Duplication paternal 11p15Hypomethylation paternal H19DMRDuplication maternal 11p1514q32.2Maternal UPD 14-like phenotype UPD Maternal UPD 14q32.2Deletion impr<strong>in</strong>tedgenesDeletion encompass<strong>in</strong>g paternalDLK1Paternal UPD 14-like phenotype UPD Paternal UPD 14q32.2EpimutationHypermethylation maternalIG-DMR and MEG3-DMRDeletion impr<strong>in</strong>tedgenesDeletion encompass<strong>in</strong>g maternalMEG315q11-q13PradereWilli syndrome UPD Maternal UPD 15q11-q13EpimutationHypermethylation paternalSNRPN DMRImpr<strong>in</strong>t<strong>in</strong>g centermutationMicrodeletion paternal SNRPNregion caus<strong>in</strong>g hypermethylationDeletion impr<strong>in</strong>tedgenesDeletion paternal 15q11.2encompass<strong>in</strong>g snoRNAs <strong>in</strong>SNRPN region


CHAPTER 13Epigenetic Mechanisms of <strong>Human</strong> Impr<strong>in</strong>t<strong>in</strong>g DisordersTABLE 13.3Phenotypes Associated with Constitutional Abnormalities at Impr<strong>in</strong>tedLocidcont<strong>in</strong>uedLocus/PhenotypeClass of Abnormality AbnormalityAngelman syndrome UPD Paternal UPD 15q11-13EpimutationHypomethylation maternalSNRPN DMRImpr<strong>in</strong>t<strong>in</strong>g centremutationMicrodeletion/<strong>in</strong>version upstreamof maternal SNRPN caus<strong>in</strong>ghypomethylationMutation impr<strong>in</strong>ted Mutation maternal UBE3A20q13.32Pseudohypoparathyroidismtype 1aPseudohypoparathyroidsimtype 1bMultiple lociHypomethylation multipleimpr<strong>in</strong>ted locigeneDeletion impr<strong>in</strong>tedgenesMutation impr<strong>in</strong>tedgeneEpimutationImpr<strong>in</strong>t<strong>in</strong>g centermutationImpr<strong>in</strong>t<strong>in</strong>g centermutationEpimutation/unknownMutation impr<strong>in</strong>t<strong>in</strong>gmach<strong>in</strong>eryDeletion maternal 15q11.2encompass<strong>in</strong>g UBE3AMutation maternal GNASHypomethylation maternal GNASExon A/B DMR þ/e GNASXL andNESPAS DMRsDeletion maternal STX16 exon4e6 caus<strong>in</strong>g hypomethylationGNAS exonA/B DMRDeletion maternal NESP55 DMRcaus<strong>in</strong>g hypomethylation GNASexonA/B, GNASXL and NESPASDMRsHypomethylation maternallymethylated DMRs <strong>in</strong>clud<strong>in</strong>g:PLAGL1, GRB10, KvDMR1 andNESPASBiallelic mutations ZFP57 caus<strong>in</strong>ghypomethylation at multiplematernally methylated DMRs<strong>in</strong>clud<strong>in</strong>g PLAGL1, GRB10,KCNQ1OT1 and NESPAS267UPD, uniparental disomy; DMR, differentially methylated region.13.15 CHROMOSOME 14Q32.2Two oppos<strong>in</strong>g groups of abnormalities occur at 14q32.2 caus<strong>in</strong>g reciprocal abnormalities atimpr<strong>in</strong>ted genes <strong>in</strong> the region and result<strong>in</strong>g <strong>in</strong> two dist<strong>in</strong>ct phenotypes. The first, referred to asthe paternal UPD 14-like phenotype, is associated with developmental delay, a bell-shapedthorax, abdom<strong>in</strong>al wall defects, and dist<strong>in</strong>ctive facial appearance. Causative abnormalitiesresult <strong>in</strong> decreased expression of the maternally expressed genes such as MEG3 (Maternallyexpressed gene 3) and RTL1as (Retrotransposon-like gene 1 antisense) and/or <strong>in</strong>creased expressionof paternally expressed genes such as DLK1 (Delta, drosophila, homolog-like 1) and RTL1(Retrotransposon-like gene 1). Reported abnormalities <strong>in</strong>clude paternal UPD 14 and maternal14q32.2 deletions encompass<strong>in</strong>g the MEG3 DMR and/or the IG-DMR [50,51].The second phenotype, referred to as the maternal UPD 14-like phenotype, is associated withpre- and postnatal growth restriction, developmental delay, and early puberty. Causativeabnormalities result <strong>in</strong> <strong>in</strong>creased expression of the maternally expressed genes such as MEG3


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>and RTL1as and/or decreased expression of paternally expressed genes such as DLK1 and RTL1.Reported abnormalities <strong>in</strong>clude maternal UPD 14; epimutations at the IG-DMR and MEG3-DMR (hypomethylation at the maternal allele); and paternal 14q32.2 deletions encompass<strong>in</strong>gDLK1[50].13.16 CHROMOSOME 15Q11-Q13Similar to the 11p15 and 14q32.2 impr<strong>in</strong>ted loci, two oppos<strong>in</strong>g groups of abnormalities occurat 15q11.2 and cause dist<strong>in</strong>ct phenotypes. PradereWilli syndrome is characterized bymoderate developmental delay, neonatal hypotonia, hyperphagia, and hypogonadism. It iscaused by a variety of abnormalities at 15q11.2 that reduce expression of paternally expressedgenes <strong>in</strong>clud<strong>in</strong>g SNRPN (Small nucleolar ribonucleoprote<strong>in</strong> polypeptide N), the SNURF (SNRPNupstream read<strong>in</strong>g frame) and/or the nearby small nucleolar RNAs (snoRNAs). Reportedabnormalities <strong>in</strong>clude maternal uniparental disomy 15q11.2; epimutations at the SNRPNDMR (hypermethylation of the paternal allele); impr<strong>in</strong>t<strong>in</strong>g center mutations that result <strong>in</strong>SNRPN DMR hypermethylation (microdeletions affect<strong>in</strong>g a critical region that <strong>in</strong>cludesSNRPN exon 1); and paternal deletions that <strong>in</strong>clude the snoRNAs adjacent to SNRPN[66e68]. This last abnormality is the most frequent cause of the condition and oftenencompasses the whole of 15q11.2.268Angelman syndrome is characterized by developmental delay with absent or nearly absentspeech, an ataxic gait, seizures, and microcephaly. Causative abnormalities reduce expressionof the maternally expressed UBE3A (Ubiquit<strong>in</strong>-prote<strong>in</strong> ligase E3A) and <strong>in</strong>clude paternaluniparental disomy 15q11.2; epimutation at the SNRPN DMR (hypomethylation of thematernal allele); impr<strong>in</strong>t<strong>in</strong>g center mutations that result <strong>in</strong> SNRPN DMR hypomethylation(microdeletions or chromosomal <strong>in</strong>version affect<strong>in</strong>g a critical region upstream of SNRPN);maternal deletions at 15q11.2 encompass<strong>in</strong>g UBE3A; and maternal mutations <strong>in</strong> UBE3A[48,68,69].13.17 CHROMOSOME 20Q13.32Abnormalities at the impr<strong>in</strong>ted 20q13.32 locus that disturb expression of GNAS (Guan<strong>in</strong>enucleotide b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> alpha-stimulat<strong>in</strong>g activity polypeptide) and its surround<strong>in</strong>g transcriptsresult <strong>in</strong> a group of disorders associated with parathyroid hormone resistance (pseudohypoparathyroidism).Pseudoparathryoidism type 1a is characterized by Albright’s hereditaryosteodystrophy and resistance to numerous hormones typically <strong>in</strong>clud<strong>in</strong>g thyroid-stimulat<strong>in</strong>ghormone and gonadotroph<strong>in</strong>s <strong>in</strong> addition to parathyroid hormone. It is caused by mutationson the maternal GNAS allele [70,71].Pseudohypoparathroidism type 1b is characterized by resistance to parathyroid hormone and<strong>in</strong> some cases thyroid-stimulat<strong>in</strong>g hormone without features of Albright’s hereditary osteodystrophy.Causative epimutations are frequently found and result <strong>in</strong> hypomethylation of thematernal GNAS Exon A/B DMR and <strong>in</strong> some cases the GNASXL and NESPAS DMRs [71]. Inaddition, two dist<strong>in</strong>ct types of impr<strong>in</strong>t<strong>in</strong>g center mutations have been reported: deletions ofthe maternal STX16 (Syntax<strong>in</strong> 16) exons 4e6 that result <strong>in</strong> hypomethylation at the GNAS ExonA/B DMR; and deletions of the maternal NESP55 DMR that cause hypomethylation at thematernal GNAS Exon A/B, GNASXL, and NESPAS DMRs.13.18 HYPOMETHYLATION AT MULTIPLE IMPRINTED LOCIA number of <strong>in</strong>dividuals have recently been reported with hypomethylation at multiplematernally methylated loci <strong>in</strong>clud<strong>in</strong>g PLAGL1, GRB10, KvDMR1, and NESPAS. This pattern hasbeen identified <strong>in</strong> <strong>in</strong>dividuals orig<strong>in</strong>ally diagnosed with transient neonatal diabetes mellitusand BeckwitheWiedemann syndrome [72,73]. However, the spectrum of phenotypes withwhich hypomethylation at multiple impr<strong>in</strong>ted loci manifests and the determ<strong>in</strong>ants of the


CHAPTER 13Epigenetic Mechanisms of <strong>Human</strong> Impr<strong>in</strong>t<strong>in</strong>g Disordersphenotypic features rema<strong>in</strong> to be identified. Some cases present<strong>in</strong>g with features of transientneonatal diabetes mellitus are caused by biallelic mutations <strong>in</strong> ZFP57 [56]. The underly<strong>in</strong>gcause <strong>in</strong> others is unknown.13.19 CONCLUSIONA variety of molecular mechanisms can disrupt impr<strong>in</strong>ted loci and cause a number of humansyndromes. Their study has proved valuable <strong>in</strong> our understand<strong>in</strong>g of these disorders themselves.It has also led to important advances <strong>in</strong> our understand<strong>in</strong>g of the mechanisms by whichimpr<strong>in</strong>t<strong>in</strong>g is established and ma<strong>in</strong>ta<strong>in</strong>ed and by which it can be abrogated. Despite theconsiderable advances that have been made <strong>in</strong> these areas over the last few decades, muchrema<strong>in</strong>s to be understood.References[1] Bird A. Perceptions of epigenetics. Nature 2007;447:396e8.[2] Ledford H. Language: Disputed def<strong>in</strong>itions. Nature 2008;455:1023e8.[3] Wadd<strong>in</strong>gton CH. The Strategy of the Genes. London: Allen & Unw<strong>in</strong>; 1957.[4] Suzuki MM, Bird A. DNA methylation landscapes: provocative <strong>in</strong>sights from epigenomics. Nat Rev Genet2008;9:465e76.[5] Li E, Bestor TH, Jaenisch R. Targeted mutation of the DNA methyltransferase gene results <strong>in</strong> embryoniclethality. Cell 1992;69:915e26.[6] Walsh CP, Chaillet JR, Bestor TH. Transcription of IAP endogenous retroviruses is constra<strong>in</strong>ed by cytos<strong>in</strong>emethylation. Nat Genet 1998;20:116e7.[7] Okano M, Bell DW, Haber DA, Li E. DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novomethylation and mammalian development. Cell 1999;99:247e57.[8] Chen T, Li E. Structure and function of eukaryotic DNA methyltransferases. Curr Top Dev Biol 2004;60:55e89.[9] Karpf AR, Matsui S. Genetic disruption of cytos<strong>in</strong>e DNA methyltransferase enzymes <strong>in</strong>duces chromosomal<strong>in</strong>stability <strong>in</strong> human cancer cells. Cancer Res 2005;65:8635e9.[10] Dodge JE, Okano M, Dick F, Tsujimoto N, Chen T, Wang S, et al. Inactivation of Dnmt3b <strong>in</strong> mouse embryonicfibroblasts results <strong>in</strong> DNA hypomethylation, chromosomal <strong>in</strong>stability, and spontaneous immortalization.J Biol Chem 2005;280:17986e91.[11] Cedar H, Bergman Y. L<strong>in</strong>k<strong>in</strong>g DNA methylation and histone modification: patterns and paradigms. Nat RevGenet 2009;10:295e304.[12] Weber M, Hellmann I, Stadler MB, Ramos L, Paabo S, Rebhan M, et al. Distribution, silenc<strong>in</strong>g potentialand evolutionary impact of promoter DNA methylation <strong>in</strong> the human genome. Nat Genet2007;39:457e66.[13] Metivier R, Gallais R, Tiffoche C, Le PC, Jurkowska RZ, Carmouche RP, et al. Cyclical DNA methylation ofa transcriptionally active promoter. Nature 2008;452:45e50.[14] Gruenbaum Y, Cedar H, Raz<strong>in</strong> A. Substrate and sequence specificity of a eukaryotic DNA methylase. Nature1982;295:620e2.[15] Schaefer CB, Ooi SK, Bestor TH, Bourc’his D. Epigenetic decisions <strong>in</strong> mammalian germ cells. <strong>Science</strong>2007;316:398e9.[16] Reik W, Dean W, Walter J. Epigenetic reprogramm<strong>in</strong>g <strong>in</strong> mammalian development. <strong>Science</strong>2001;293:1089e93.[17] Shi Y. Histone lys<strong>in</strong>e demethylases: emerg<strong>in</strong>g roles <strong>in</strong> development, physiology and disease. Nat Rev Genet2007;8:829e33.[18] Haberland M, Montgomery RL, a ndOlson EN. The many roles of histone deacetylases <strong>in</strong> development andphysiology: implications for disease and therapy. Nat Rev Genet 2009;10:32e42.[19] Probst AV, Dunleavy E, Almouzni G. Epigenetic <strong>in</strong>heritance dur<strong>in</strong>g the cell cycle. Nat Rev Mol Cell Biol2009;10:192e206.[20] Wang J, Mager J, Chen Y, Schneider E, Cross JC, Nagy A, et al. Impr<strong>in</strong>ted X <strong>in</strong>activation ma<strong>in</strong>ta<strong>in</strong>ed by a mousePolycomb group gene. Nat Genet 2001;28:371e5.[21] Schwartz YB, Pirrotta V. Polycomb complexes and epigenetic states. Curr Op<strong>in</strong> Cell Biol 2008;20:266e73.[22] Francis NJ, Follmer NE, Simon MD, Aghia G, Butler JD. Polycomb prote<strong>in</strong>s rema<strong>in</strong> bound to chromat<strong>in</strong> andDNA dur<strong>in</strong>g DNA replication <strong>in</strong> vitro. Cell 2009;137:110e22.269


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>270[23] Herz<strong>in</strong>g LB, Romer JT, Horn JM, Ashworth A. Xist has properties of the X-chromosome <strong>in</strong>activation centre.Nature 1997;386:272e5.[24] Sleutels F, Zwart R, Barlow DP. The non-cod<strong>in</strong>g Air RNA is required for silenc<strong>in</strong>g autosomal impr<strong>in</strong>ted genes.Nature 2002;415:810e3.[25] Reik W. Stability and flexibility of epigenetic gene regulation <strong>in</strong> mammalian development. Nature2007;447:425e32.[26] Moore T, Haig D. Genomic impr<strong>in</strong>t<strong>in</strong>g <strong>in</strong> mammalian development: a parental tug-of-war. Trends Genet1991;7:45e9.[27] Haig D. Genomic impr<strong>in</strong>t<strong>in</strong>g and k<strong>in</strong>ship: how good is the evidence? Annu Rev Genet 2004;38:553e85.[28] Murrell A, Heeson S, Reik W. Interaction between differentially methylated regions partitions the impr<strong>in</strong>tedgenes Igf2 and H19 <strong>in</strong>to parent-specific chromat<strong>in</strong> loops. Nat Genet 2004;36:889e93.[29] Davis TL, Yang GJ, McCarrey JR, Bartolomei MS. The H19 methylation impr<strong>in</strong>t is erased and re-establisheddifferentially on the parental alleles dur<strong>in</strong>g male germ cell development. Hum Mol Genet 2000;9:2885e94.[30] Lucifero D, Mert<strong>in</strong>eit C, Clarke HJ, Bestor TH, Trasler JM. Methylation dynamics of impr<strong>in</strong>ted genes <strong>in</strong> mousegerm cells. Genomics 2002;79:530e8.[31] Scott RH, Douglas J, Baskcomb L, Huxter N, Barker K, Hanks S, et al. Constitutional 11p15 abnormalities, <strong>in</strong>clud<strong>in</strong>gheritable impr<strong>in</strong>t<strong>in</strong>g center mutations, cause nonsyndromic Wilms tumor. Nat Genet 2008a;40:1329e34.[32] Bell AC, Felsenfeld G. Methylation of a CTCF-dependent boundary controls impr<strong>in</strong>ted expression of the Igf2gene. Nature 2000;405:482e5.[33] Cui H, Niemitz EL, Ravenel JD, Onyango P, Brandenburg SA, Lobanenkov VV, et al. Loss of impr<strong>in</strong>t<strong>in</strong>g of<strong>in</strong>sul<strong>in</strong>-like growth factor-II <strong>in</strong> Wilms’ tumor commonly <strong>in</strong>volves altered methylation but not mutations ofCTCF or its b<strong>in</strong>d<strong>in</strong>g site. Cancer Res 2001;61:4947e50.[34] Sparago A, Cerrato F, Vernucci M, Ferrero GB, Silengo MC, Riccio A. Microdeletions <strong>in</strong> the human H19 DMRresult <strong>in</strong> loss of IGF2 impr<strong>in</strong>t<strong>in</strong>g and BeckwitheWiedemann syndrome. Nat Genet 2004;36:958e60.[35] Hark AT, Schoenherr CJ, Katz DJ, Ingram RS, Levorse JM, Tilghman SM. CTCF mediates methylation-sensitiveenhancer-block<strong>in</strong>g activity at the H19/Igf2 locus. Nature 2000;405:486e9.[36] Pant V, Kurukuti S, Pugacheva E, Shamsudd<strong>in</strong> S, Mariano P, Renkawitz R, et al. Mutation of a s<strong>in</strong>gle CTCFtarget site with<strong>in</strong> the H19 impr<strong>in</strong>t<strong>in</strong>g control region leads to loss of Igf2 impr<strong>in</strong>t<strong>in</strong>g and complex patterns of denovo methylation upon maternal <strong>in</strong>heritance. Mol Cell Biol 2004;24:3497e504.[37] Szabo PE, Tang SH, Silva FJ, Tsark WM, Mann JR. Role of CTCF b<strong>in</strong>d<strong>in</strong>g sites <strong>in</strong> the Igf2/H19 impr<strong>in</strong>t<strong>in</strong>gcontrol region. Mol Cell Biol 2004;24:4791e800.[38] Murrell A, Ito Y, Verde G, Huddleston J, Woodf<strong>in</strong>e K, Silengo MC, et al. Dist<strong>in</strong>ct methylation changes at theIGF2-H19 locus <strong>in</strong> congenital growth disorders and cancer. PLoS ONE 2008;3:e1849.[39] Cui H, Onyango P, Brandenburg S, Wu Y, Hsieh CL, Fe<strong>in</strong>berg AP. Loss of impr<strong>in</strong>t<strong>in</strong>g <strong>in</strong> colorectal cancer l<strong>in</strong>kedto hypomethylation of H19 and IGF2. Cancer Res 2002;62:6442e6.[40] Sullivan MJ, Taniguchi T, Jhee A, Kerr N, Reeve AE. Relaxation of IGF2 impr<strong>in</strong>t<strong>in</strong>g <strong>in</strong> Wilms tumours associatedwith specific changes <strong>in</strong> IGF2 methylation. Oncogene 1999;18:7527e34.[41] Apostolidou S, bu-Amero S, O’Donoghue K, Frost J, Olafsdottir O, Chavele KM, et al. Elevated placentalexpression of the impr<strong>in</strong>ted PHLDA2 gene is associated with low birth weight. J Mol Med 2007;85:379e87.[42] Lewis A, Mitsuya K, Umlauf D, Smith P, Dean W, Walter J, et al. Impr<strong>in</strong>t<strong>in</strong>g on distal chromosome 7 <strong>in</strong> theplacenta <strong>in</strong>volves repressive histone methylation <strong>in</strong>dependent of DNA methylation. Nat Genet 2004;36:1291e5.[43] Redrup L, Branco MR, Perdeaux ER, Krueger C, Lewis A, Santos F, et al. The long noncod<strong>in</strong>g RNA Kcnq1ot1organises a l<strong>in</strong>eage-specific nuclear doma<strong>in</strong> for epigenetic gene silenc<strong>in</strong>g. Development 2009;136:525e30.[44] Lewis A, Reik W. How impr<strong>in</strong>t<strong>in</strong>g centres work. Cytogenet Genome Res 2006;113:81e9.[45] Sh<strong>in</strong> JY, Fitzpatrick GV, Higg<strong>in</strong>s MJ. Two dist<strong>in</strong>ct mechanisms of silenc<strong>in</strong>g by the KvDMR1 impr<strong>in</strong>t<strong>in</strong>g controlregion. EMBO J 2008;27:168e78.[46] Kotzot D. Complex and segmental uniparental disomy updated. J Med Genet 2008;45:545e56.[47] Buit<strong>in</strong>g K, Saitoh S, Gross S, Dittrich B, Schwartz S, Nicholls RD, et al. Inherited microdeletions <strong>in</strong> theAngelman and PradereWilli syndromes def<strong>in</strong>e an impr<strong>in</strong>t<strong>in</strong>g centre on human chromosome 15. Nat Genet1995;9:395e400.[48] Buit<strong>in</strong>g K, Barnicoat A, Lich C, Pembrey M, Malcolm S, Horsthemke B. Disruption of the bipartite impr<strong>in</strong>t<strong>in</strong>gcenter <strong>in</strong> a family with Angelman syndrome. Am J Hum Genet 2001;68:1290e4.[49] Bastepe M, Frohlich LF, L<strong>in</strong>glart A, Abu-Zahra HS, Tojo K, Ward LM, et al. Deletion of the NESP55 differentiallymethylated region causes loss of maternal GNAS impr<strong>in</strong>ts and pseudohypoparathyroidism type Ib. NatGenet 2005;37:25e7.[50] Kagami M, Sekita Y, Nishimura G, Irie M, Kato F, Okada M, et al. Deletions and epimutations affect<strong>in</strong>g thehuman 14q32.2 impr<strong>in</strong>ted region <strong>in</strong> <strong>in</strong>dividuals with paternal and maternal upd(14)-like phenotypes. NatGenet 2008;40:237e42.


CHAPTER 13Epigenetic Mechanisms of <strong>Human</strong> Impr<strong>in</strong>t<strong>in</strong>g Disorders[51] Kagami M, O’Sullivan MJ, Green AJ, Watabe Y, Arisaka O, Masawa N, et al. The IG-DMR and the MEG3-DMR athuman chromosome 14q32.2: hierarchical <strong>in</strong>teraction and dist<strong>in</strong>ct functional properties as impr<strong>in</strong>t<strong>in</strong>g controlcenters. PLoS Genet 2010;6:e1000992.[52] Hatada I, Ohashi H, Fukushima Y, Kaneko Y, Inoue M, Komoto Y, et al. An impr<strong>in</strong>ted gene p57KIP2 is mutated<strong>in</strong> BeckwitheWiedemann syndrome. Nat Genet 1996;14:171e3.[53] Murdoch S, Djuric U, Mazhar B, Seoud M, Khan R, Kuick R, et al. Mutations <strong>in</strong> NALP7 cause recurrenthydatidiform moles and reproductive wastage <strong>in</strong> humans. Nat Genet 2006;38:300e2.[54] Parry DA, Logan CV, Hayward BE, Shires M, Landolsi H, Diggle C, et al. Mutations caus<strong>in</strong>g familial biparentalhydatidiform mole implicate c6orf221 as a possible regulator of genomic impr<strong>in</strong>t<strong>in</strong>g <strong>in</strong> the human oocyte. AmJ Hum Genet 2011;89:451e8.[55] Meyer E, Lim D, Pasha S, Tee LJ, Rahman F, Yates JR, et al. Germl<strong>in</strong>e mutation <strong>in</strong> NLRP2 (NALP2) <strong>in</strong> a familialimpr<strong>in</strong>t<strong>in</strong>g disorder (BeckwitheWiedemann Syndrome). PLoS Genet 2009;5:e1000423.[56] Mackay DJ, Callaway JL, Marks SM, White HE, Acer<strong>in</strong>i CL, Boonen SE, et al. Hypomethylation of multipleimpr<strong>in</strong>ted loci <strong>in</strong> <strong>in</strong>dividuals with transient neonatal diabetes is associated with mutations <strong>in</strong> ZFP57. NatGenet 2008;40:949e51.[57] Gardner RJ, Mackay DJ, Mungall AJ, Polychronakos C, Siebert R, Shield JP, et al. An impr<strong>in</strong>ted locus associatedwith transient neonatal diabetes mellitus. Hum Mol Genet 2000;9:589e96.[58] Temple IK, Gardner RJ, Mackay DJ, Barber JC, Rob<strong>in</strong>son DO, Shield JP. Transient neonatal diabetes: widen<strong>in</strong>gthe understand<strong>in</strong>g of the etiopathogenesis of diabetes. Diabetes 2000;49:1359e66.[59] Temple IK, Shrubb V, Lever M, Bullman H, Mackay DJ. Isolated impr<strong>in</strong>t<strong>in</strong>g mutation of the DLK1/GTL2 locusassociated with a cl<strong>in</strong>ical presentation of maternal uniparental disomy of chromosome 14. J.Med.Genet2007;44:637e40.[60] Monk D, Bentley L, Hitch<strong>in</strong>s M, Myler RA, Clayton-Smith J, Ismail S, et al. Chromosome 7p disruptions <strong>in</strong>SilvereRussell syndrome: del<strong>in</strong>eat<strong>in</strong>g an impr<strong>in</strong>ted candidate gene region. Hum Genet 2002;111:376e87.[61] Monk D, Wakel<strong>in</strong>g EL, Proud V, Hitch<strong>in</strong>s M, bu-Amero SN, Stanier P, et al. Duplication of 7p11.2-p13,<strong>in</strong>clud<strong>in</strong>g GRB10, <strong>in</strong> SilvereRussell syndrome. Am J Hum Genet 2000;66:36e46.[62] Hitch<strong>in</strong>s MP, Monk D, Bell GM, Ali Z, Preece MA, Stanier P, et al. Maternal repression of the human GRB10gene <strong>in</strong> the develop<strong>in</strong>g central nervous system; evaluation of the role for GRB10 <strong>in</strong> SilvereRussell syndrome.Eur J Hum Genet 2001;9:82e90.[63] Bentley L, Nakabayashi K, Monk D, Beechey C, Peters J, Birjandi Z, et al. The impr<strong>in</strong>ted region on humanchromosome 7q32 extends to the carboxypeptidase A gene cluster: an impr<strong>in</strong>ted candidate for SilvereRussellsyndrome. J Med Genet 2003;40:249e56.[64] Weksberg R, Shuman C, Smith AC. BeckwitheWiedemann syndrome. Am J Med Genet C.Sem<strong>in</strong>.Med Genet2005;137:12e23.[65] Abu-Amero S, Monk D, Frost J, Preece M, Stanier P, Moore GE. The genetic aetiology of SilvereRussellsyndrome. J Med Genet 2008;45:193e9.[66] Ohta T, Gray TA, Rogan PK, Buit<strong>in</strong>g K, Gabriel JM, Saitoh S, et al. Impr<strong>in</strong>t<strong>in</strong>g-mutation mechanisms <strong>in</strong>PradereWilli syndrome. Am J Hum Genet 1999;64:397e413.[67] Sahoo T, del GD, German JR, Sh<strong>in</strong>awi M, Peters SU, Person RE, et al. PradereWilli phenotype caused bypaternal deficiency for the HBII-85 C/D box small nucleolar RNA cluster. Nat Genet 2008;40:719e21.[68] Buit<strong>in</strong>g K, Gross S, Lich C, Gillessen-Kaesbach G, el-Maarri O, Horsthemke B. Epimutations <strong>in</strong> PradereWilliand Angelman syndromes: a molecular study of 136 patients with an impr<strong>in</strong>t<strong>in</strong>g defect. Am J Hum Genet2003;72:571e7.[69] Kish<strong>in</strong>o T, Lalande M, Wagstaff J. UBE3A/E6-AP mutations cause Angelman syndrome. Nat Genet1997;15:70e3.[70] Patten JL, Johns DR, Valle D, Eil C, Gruppuso PA, Steele G, et al. Mutation <strong>in</strong> the gene encod<strong>in</strong>g the stimulatory Gprote<strong>in</strong> of adenylate cyclase <strong>in</strong> Albright’s hereditary osteodystrophy. N Engl J Med 1990;322:1412e9.[71] Bastepe M, Juppner H. GNAS locus and pseudohypoparathyroidism. Horm Res 2005;63:65e74.[72] Mackay DJ, Hahnemann JM, Boonen SE, Poerksen S, Bunyan DJ, White HE, et al. Epimutation of the TNDMlocus and the BeckwitheWiedemann syndrome centromeric locus <strong>in</strong> <strong>in</strong>dividuals with transient neonataldiabetes mellitus. Hum Genet 2006;119:179e84.[73] Bliek J, Verde G, Callaway J, Maas SM, De CA, Sparago A, et al. Hypomethylation at multiple maternallymethylated impr<strong>in</strong>ted regions <strong>in</strong>clud<strong>in</strong>g PLAGL1 and GNAS loci <strong>in</strong> BeckwitheWiedemann syndrome. Eur JHum Genet 2009;17:611e9.[74] Impr<strong>in</strong>ted Gene Database. 2008. Available at http://www.geneimpr<strong>in</strong>t.com/site/genes-by-species.[75] Catalogue of Impr<strong>in</strong>ted Genes and Parent of Orig<strong>in</strong> Effects. 2009. Available at http://www.otago.ac.nz/IGC.271


This page <strong>in</strong>tentionally left blank


CHAPTER 14Epigenomic Factors <strong>in</strong><strong>Human</strong> ObesityChristopher G. BellUniversity College London, London, UKCHAPTER OUTLINE14.1 Introduction 27314.2 Epigenomic Marks 27514.3 A Role for Impr<strong>in</strong>t<strong>in</strong>gAbnormalities <strong>in</strong> Obesity 27514.4 Conflict Theory ofImpr<strong>in</strong>t<strong>in</strong>g 27614.5 Rare Impr<strong>in</strong>ted Abnormalitieswith Obesity-RelatedPhenotypes 27714.6 Dietary Influence on DNAMethylation <strong>in</strong> Mur<strong>in</strong>eModels 27914.7 Obesogenic EnvironmentEffects on Common <strong>Human</strong>Obesity 28014.8 Ag<strong>in</strong>g Effect on DNAMethylation 28114.9 Developmental EpigenomicDysregulation 28114.10 Fetal Plasticity 28214.11 Postnatal EnvironmentalMismatch 28314.12 Hypernutrition 28414.13 Epigenetic Analysis ofLept<strong>in</strong> 28514.14 Histone Epigenomic Modifiers eMaster MetabolicRegulators 28514.15 Metastable Alleles <strong>in</strong> <strong>Human</strong>Associated with Obesity 28614.16 Parent-of-Orig<strong>in</strong> GeneticEffects 28714.17 Epigenomic-Wide AssociationStudies (EWAS) <strong>in</strong> <strong>Human</strong>Obesity 28814.18 Future Prospects 289References 28927314.1 INTRODUCTIONThe detrimental effects of obesity on health can be starkly brought <strong>in</strong>to focus by a recentpublication from the United States, stat<strong>in</strong>g that the escalat<strong>in</strong>g rates of this condition will nownegate all the positive public health ga<strong>in</strong>s made by the reduction <strong>in</strong> tobacco consumption overthe past 50 years [1]. This will led, after the steady rise <strong>in</strong> life expectancy dur<strong>in</strong>g the last century,to a decl<strong>in</strong>e <strong>in</strong> lifespan for those children born today [2]. The high prevalence of obesity isa result of the current “obesogenic” environment widespread throughout the Western world;a coupl<strong>in</strong>g of reduced energy expenditure both at work and leisure, with <strong>in</strong>creas<strong>in</strong>gly easyaccess to high-calorific foods [3]. A major driver <strong>in</strong> the energy <strong>in</strong>take overload has beendocumented as simply the <strong>in</strong>crease <strong>in</strong> both portion sizes and eat<strong>in</strong>g opportunities [4]. Additionallythis high obesity rate is now be<strong>in</strong>g swiftly caught up to by those <strong>in</strong> develop<strong>in</strong>gcountries, as they are <strong>in</strong>creas<strong>in</strong>gly removed from a rural existence and rapidly adopt modernT. Tollefsbol (Ed): <strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>. DOI: 10.1016/B978-0-12-388415-2.00014-7Copyright Ó 2012 Elsevier Inc. All rights reserved.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>urban life [5,6]. Obesity <strong>in</strong>creases the risk of type 2 diabetes (T2D), coronary vascular disease,hypertension, and some forms of cancer [7]. This obesity-driven <strong>in</strong>crease <strong>in</strong> T2D alone isputt<strong>in</strong>g a considerable stra<strong>in</strong> on health care provision because of its chronic nature andmultisystemic complications [8]. Furthermore, estimates placed obesity as causative <strong>in</strong> z14%and z20% of all cancer deaths <strong>in</strong> the US <strong>in</strong> men and women, respectively [9].Although on a population-wide scale this environmental and nutritional <strong>in</strong>fluence isextremely pervasive, it does not affect all <strong>in</strong>dividuals equally. There is considerable variancebetween those most susceptible to weight ga<strong>in</strong> to those least at risk. Obesity, as a complexpolygenic trait, is the result of environmental and genomic effects and there is substantialgenetic variation <strong>in</strong> <strong>in</strong>dividual response to this “obesogenic” pressure to put on weight [3].Due to this genetic susceptibility certa<strong>in</strong> ethnic groups have been found to be at even greaterrisk when they encounter this environment [10]. However, the emergence of this dramatically<strong>in</strong>creas<strong>in</strong>g rate over the time span of a few generations is too fast for the appearance of newobesity-promot<strong>in</strong>g mutant alleles, therefore cannot be attributed to a pure genetic effect alone.The discovery of Mendelian childhood-onset extreme obesity syndromes, caused by s<strong>in</strong>glegene mutations, revealed the critical role of neurons with<strong>in</strong> the hypothalamus, particularlythose resid<strong>in</strong>g <strong>in</strong> the arcuate nucleus [11,12] (see Figure 14.1). These neurons are <strong>in</strong>volved <strong>in</strong>274FIGURE 14.1Arcuate nucleus control of central energybalance between food <strong>in</strong>take and energyexpenditure. Peripheral tissue hormonalsignals are summated by theneuropeptide Y (NPY)/agouti-relatedprote<strong>in</strong> (AGRP) neurons (orexigenicpromot<strong>in</strong>gfood <strong>in</strong>take), and the proopiomelanocort<strong>in</strong>(POMC)/coca<strong>in</strong>e andamphetam<strong>in</strong>e-related transcript (CART)neurons (with opposite anorexigeniceffect) <strong>in</strong> the arcuate nucleus of thehypothalamus. These hormones <strong>in</strong>cludePYY 3-36 from the distal gastro<strong>in</strong>test<strong>in</strong>altract via Y2 receptors (Y2R), Lept<strong>in</strong> fromadipose tissue via the lept<strong>in</strong> receptor(LEPR), and Ghrel<strong>in</strong> from the stomachand duodenum via the growth hormonesecretagog receptors (GHSRs) and <strong>in</strong>sul<strong>in</strong>from the pancreas. The NPY/AGRPneurons also have a direct <strong>in</strong>hibitoryeffect on the POMC/CART neuronsthrough gamma-am<strong>in</strong>obutyric acid(GABA) release. Second-orderdownstream effector neurons are<strong>in</strong>fluenced by the NPY/AGRP and POMC/CART neurons, and also receivemodify<strong>in</strong>g <strong>in</strong>puts from dopam<strong>in</strong>e,seroton<strong>in</strong>, and endocannab<strong>in</strong>oid signals,with receptors that <strong>in</strong>clude the Y1receptor (Y1R) and the melanocort<strong>in</strong> 4receptor (MC4R). This figure isreproduced <strong>in</strong> the color plate section.Source: From [56], Ó Nature Publish<strong>in</strong>gGroup.


CHAPTER 14Epigenomic Factors <strong>in</strong> <strong>Human</strong> Obesitycentral appetite control and energy balance and have highlighted the importance of thelept<strong>in</strong>emelanocort<strong>in</strong> pathway <strong>in</strong> this peripheral tissue to bra<strong>in</strong> control feedback loop [19].However these monogenic causes are rare, the most frequent, MC4R mutations, account<strong>in</strong>g foronly ~6% of extreme obesity <strong>in</strong> children [14].Considerable success has come <strong>in</strong> gene discovery for all common diseases <strong>in</strong> the last 5 years,with the advent of high-throughput s<strong>in</strong>gle nucleotide polymorphism (SNP)-chips facilitat<strong>in</strong>ggenome-wide association studies (GWAS). These population-based case-control studies led tothe identification of common genetic variants associated with common obesity, which currentlystands at 32 loci <strong>in</strong>fluenc<strong>in</strong>g body mass <strong>in</strong>dex (BMI) [15,16]. Further, rarer but stronger causalvariants have also begun to be found [17,18] and more will be able to be unearthed by wholegenomesequenc<strong>in</strong>g studies currently underway. These common variants have modest effectsize, the largest with<strong>in</strong> the FTO locus lead<strong>in</strong>g to a 1.67-fold higher rate of obesity [19], althoughthe potential to ga<strong>in</strong> new <strong>in</strong>sights to the pathophysiology from these replicated associations isconsiderable. Work, current and on-go<strong>in</strong>g <strong>in</strong> the com<strong>in</strong>g years, will enable the unravell<strong>in</strong>g of theetiological significance of these genomic loci, with likely therapeutic advances.The comb<strong>in</strong>ed effect of these common SNP variants known to date is only 2e3% of theestimated 40e70% <strong>in</strong>herited fraction of obesity risk [20]. Additional power to detect genetic<strong>in</strong>volvement, however, may come by detect<strong>in</strong>g: the actual causal variant that the associationSNP may only be an adequate proxy for; further common pathogenic variants with<strong>in</strong> theseloci; rare variants with<strong>in</strong> these [21] or yet to be discovered loci; and f<strong>in</strong>ally the possible role ofnon-sequence-based or epigenetic factors <strong>in</strong> susceptibility [22]. The ability to detect theseepigenetic alterations through genetic means alone though, will range from obligatory tostochastic, depend<strong>in</strong>g on their direct association with the underly<strong>in</strong>g genome [23]. Furthermore,epigenetic elements are <strong>in</strong>fluenced by the environment, as well as the genetic sequence,so therefore act at this crucial <strong>in</strong>terface with the genome [24], hence be<strong>in</strong>g a plausiblemechanism for the obesogenic environment. These marks can lead to longstand<strong>in</strong>g metabolicchanges [25]; consequently epigenetic factors may make up some of the “miss<strong>in</strong>g heritability”<strong>in</strong> the complex disease of obesity [26e28]. However this obviously can be <strong>in</strong> only thoseepigenomic marks that bear some obligatory or facultative relationship with genome sequenceor they will not contribute to heritability measures.27514.2 EPIGENOMIC MARKSEpigenetic marks <strong>in</strong>clude modifications of DNA, such as DNA methylation or hydroxymethylation,post-translational modifications of histone tails, <strong>in</strong>clud<strong>in</strong>g acetylation, phosphorylation,sumoylation, ubiquit<strong>in</strong>ation, and methylation, and some non-cod<strong>in</strong>g RNAspecies. The ability to self-propagate and be conserved through mitotic division limits thisdef<strong>in</strong>ition currently to DNA methylation, and perhaps a few histone modifications [29]. Thisma<strong>in</strong>tenance permits l<strong>in</strong>eage-specific epigenomes to be preserved, to allow tissue-specific rolesto be performed, <strong>in</strong> genome-identical somatic cells [23].DNA methylation is the addition of a methyl group to the 5 0 carbon of cytos<strong>in</strong>e, pr<strong>in</strong>cipally <strong>in</strong>the context of the CpG d<strong>in</strong>ucleotide, a C followed by G <strong>in</strong> the genomic sequence, <strong>in</strong> differentiatedcells. This stable mark is also the most well-studied, so will be the major factordiscussed here<strong>in</strong>. As was the case with the genetic causes of human obesity, the <strong>in</strong>itial <strong>in</strong>sightsto the epigenetic <strong>in</strong>fluence <strong>in</strong> this disorder of adiposity have come from rare childhood-onsethuman syndromes, <strong>in</strong> this <strong>in</strong>stance the impr<strong>in</strong>t<strong>in</strong>g disorders such as PradereWilli syndrome(PWS), and mur<strong>in</strong>e models, e.g. the Agouti-viable mouse (A vy /a).14.3 A ROLE FOR IMPRINTING ABNORMALITIES IN OBESITYGenomic impr<strong>in</strong>t<strong>in</strong>g allows only one of the allelic pair of genes to be expressed, dependenton whether it is of paternal, or maternal, orig<strong>in</strong> [30]. This parent-of-orig<strong>in</strong> conditional


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>TABLE 14.1 Impr<strong>in</strong>t<strong>in</strong>g Disorders with an Obesity-Associated PhenotypeSyndrome OMIM Locus Parental Impr<strong>in</strong>t ObesityPradereWilli #176270 15q11.2 Maternal SeverePseudohypoparathyroidism 1a #103580 20q13.3 Maternal ModerateMaternal Uniparental Disomy 14 d 14 d TruncalAngelman #105830 15q11.2 Paternal Late-onsetSource: Impr<strong>in</strong>t<strong>in</strong>g syndromes with an obesity-associated phenotype [53,191].276allele-specific expression (ASE) is driven by parent-of-orig<strong>in</strong> conditional allele-specific DNAmethylation (ASM) [31]. The attribution of which parental allele, paternal or maternal, isimpr<strong>in</strong>ted is by convention referred to the allele on which the precise DNA methylation occurs,therefore the allele silenced not that which is expressed. Impr<strong>in</strong>ted genes commonly are found<strong>in</strong> clusters with reciprocally impr<strong>in</strong>ted genes, i.e. both maternal and paternal impr<strong>in</strong>ted genesare colocated [32]. Impr<strong>in</strong>ted loci are coord<strong>in</strong>ately regulated, via impr<strong>in</strong>t<strong>in</strong>g control regions,various species of non-cod<strong>in</strong>g RNA and methylation-sensitive boundary elements, lead<strong>in</strong>g toconsiderable regulatory complexity [33,34]. Furthermore, the impr<strong>in</strong>t<strong>in</strong>g of <strong>in</strong>dividual genesmay not occur body-wide, but may be present, or escape this mark<strong>in</strong>g, <strong>in</strong> only one or moreparticular organs. This tissue-specific impr<strong>in</strong>t<strong>in</strong>g is very prevalent <strong>in</strong> the cells of the placentaand bra<strong>in</strong> [30]. Placental impr<strong>in</strong>t<strong>in</strong>g is key <strong>in</strong> <strong>in</strong>fluenc<strong>in</strong>g this vital <strong>in</strong>terface of resourceallocation between the fetus and mother, and this has been proposed as a major driver <strong>in</strong> itsevolution <strong>in</strong> eutherian (placental) mammals [35]. Genomic impr<strong>in</strong>t<strong>in</strong>g and placentationevolved around a similar time period <strong>in</strong> mammals [36]. With regards to bra<strong>in</strong> impr<strong>in</strong>t<strong>in</strong>g, thepotential complexity of orchestrated impr<strong>in</strong>t<strong>in</strong>g variation amongst the numerous regions ofthe central nervous system and furthermore throughout the process of bra<strong>in</strong> development hasonly just begun to be explored [37]. However, <strong>in</strong>trigu<strong>in</strong>g evidence for the <strong>in</strong>tricate <strong>in</strong>terplay ofimpr<strong>in</strong>t<strong>in</strong>g and impr<strong>in</strong>t<strong>in</strong>g loss for correct neurogenesis <strong>in</strong> the mouse has recently beenpublished [38].Obesity is often observed as part of the spectrum of an impr<strong>in</strong>t<strong>in</strong>g abnormality phenotype(see Table 14.1). Speculation that dysregulation of the impr<strong>in</strong>ted genes <strong>in</strong> placental resourceallocation pathways, or impr<strong>in</strong>ted control of bra<strong>in</strong> development genes <strong>in</strong>volved <strong>in</strong> thelept<strong>in</strong>emelanocort<strong>in</strong> hypothalamic pathway, has been proposed <strong>in</strong> metabolic disease [39,40].Understand<strong>in</strong>g why these impr<strong>in</strong>t<strong>in</strong>g mechanisms have evolved may aid <strong>in</strong> unravell<strong>in</strong>g howthey may be causative <strong>in</strong> obesity.14.4 CONFLICT THEORY OF IMPRINTINGEutherian mammals facilitate nutrient uptake via a placenta dur<strong>in</strong>g fetal growth. The “k<strong>in</strong>ship”or “gene conflict” theorem proposes that this evolved to control the allocation of resourcesfrom mother to offspr<strong>in</strong>g e <strong>in</strong> terms of the “<strong>in</strong>terest” of the genes from the two parents.Maternal genes see equal benefit <strong>in</strong> all of their progeny, due to an equal genetic contribution toeach, so therefore will hope to provide for all <strong>in</strong> an equal fashion. However paternal genes aredivided between offspr<strong>in</strong>g sired by possibly differ<strong>in</strong>g fathers [41]. Genetic <strong>in</strong>fluences <strong>in</strong> highermammals that help them acquire maternal resources, as well as those with<strong>in</strong> parents that aidthis process, have an evolutionary advantage <strong>in</strong> that they <strong>in</strong>crease the survival and propagationof those genes [36,42]. Therefore the parental conflict theory of gene impr<strong>in</strong>t<strong>in</strong>g proposes thatgrowth-promot<strong>in</strong>g genes, that <strong>in</strong>crease direct <strong>in</strong>vestment <strong>in</strong> the currently develop<strong>in</strong>g fetus, willbe under paternal control, whereas the opposite effect will be maternally controlled, as themother tries to equally distribute resources between all of her offspr<strong>in</strong>g [36,43]. The extrememanifestation of this drive can be seen <strong>in</strong> the pathological case of the hydatidiform mole,formed by the fusion of two paternal gametes, which therefore lacks any maternal restra<strong>in</strong><strong>in</strong>gimpr<strong>in</strong>ts and leads to an extreme, tumor-like growth, which can even <strong>in</strong>vade <strong>in</strong> rare cases [44].


CHAPTER 14Epigenomic Factors <strong>in</strong> <strong>Human</strong> ObesityKnockout mouse models show that impr<strong>in</strong>ted genes play a role <strong>in</strong> the blood nutrient transfer<strong>in</strong>terface of the placenta [45]. Impr<strong>in</strong>ted placental genes control nutrition supply and fetalimpr<strong>in</strong>ted genes control demand by growth regulation [36]. The paternally expressed Igf2 isshown to be critical <strong>in</strong> placental growth [46]. Deletion of the placenta-specific promoter of thisgene leads to decreased permeability, by reduc<strong>in</strong>g the exchange barrier and <strong>in</strong>creas<strong>in</strong>g thethickness of the placenta, thereby <strong>in</strong>fluenc<strong>in</strong>g nutrient supply with subsequent effects on thegrowth of the develop<strong>in</strong>g fetus <strong>in</strong> early gestation [45]. However, the placenta is capable offunctionally adapt<strong>in</strong>g to this <strong>in</strong>terference, enabl<strong>in</strong>g it to respond to fetal need requirements, bythe up-regulation of gene pathways of the placental supply genes <strong>in</strong> the system A am<strong>in</strong>o acidtransport systems, <strong>in</strong>clud<strong>in</strong>g Slc38a4, which is also paternally expressed [47]. This counterbalanceis not sufficient to ma<strong>in</strong>ta<strong>in</strong> this correction <strong>in</strong> the late gestation period though. It canbe hypothesized that fetal and maternal blood flow and the transportation of nutrients mayalso be under impr<strong>in</strong>ted genetic regulation [36]. It is possible this “conflict” may also swaymaternal postnatal resource allocation via breast milk and the control of suckl<strong>in</strong>g. Indeeddisruption of the paternally expressed transcript of the impr<strong>in</strong>ted Gnas locus, isoform XLaS,<strong>in</strong>a mouse model led to reduced growth and survival due to effects <strong>in</strong>clud<strong>in</strong>g a poor suckl<strong>in</strong>gresponse [48]. Impr<strong>in</strong>ted genes additionally <strong>in</strong>fluence cognitive and social behavior whichmay also be used to ga<strong>in</strong> resources [49].14.5 RARE IMPRINTED ABNORMALITIES WITH OBESITY-RELATEDPHENOTYPESPradereWilli syndrome (PWS) is the classic example of an impr<strong>in</strong>t<strong>in</strong>g-related syndrome andthis was first recognized as be<strong>in</strong>g the pathogenic mechanism <strong>in</strong> 1989 [50]. The phenotype<strong>in</strong>cludes dysmorphic features, short stature, low lean body mass, muscular hypotonia, mildmental retardation, and behavioral abnormalities [51]. The region of the complex locusassociated with PWS is maternally impr<strong>in</strong>ted, so only the paternal copy is normally expressed.The <strong>in</strong>verse paternally impr<strong>in</strong>ted disorder leds to the non-obese Angelman syndrome (AS)child [51]. However, AS <strong>in</strong>dividuals are at risk of moderate later-onset adiposity [52]. Theobesity observed <strong>in</strong> PWS is so severe that it is life-threaten<strong>in</strong>g and develops with<strong>in</strong> the first 6years of childhood. PWS is estimated to occur approximately <strong>in</strong> 0.5e1 <strong>in</strong>dividuals per 10 000births with no racial bias, although it is cl<strong>in</strong>ically diagnosed and reported more frequently <strong>in</strong>Caucasians, and affects approximately 350 000e400 000 <strong>in</strong>dividuals worldwide [53]. Diagnosisis via abnormal DNA methylation <strong>in</strong> 99% of PWS patients, enabl<strong>in</strong>g the detection ofdeletions, uniparental disomy (UPD) of chromosome 15, or an impr<strong>in</strong>t<strong>in</strong>g control region(ICR) defect [54].277The behavioral and cognitive impairment <strong>in</strong> these <strong>in</strong>dividuals leads to an extreme anduncontrolled appetite, due to a proposed <strong>in</strong>ability to experience the normal satiety responseafter eat<strong>in</strong>g [55]. As mentioned above, energy balance is controlled and regulated with<strong>in</strong> thehypothalamus, with the arcuate nucleus perform<strong>in</strong>g a major role <strong>in</strong> this coord<strong>in</strong>ation(Figure 14.1). With<strong>in</strong> the arcuate there is crosstalk and <strong>in</strong>terchange between the orexigenic orappetite-promot<strong>in</strong>g NPY/AGRP neurons and the oppos<strong>in</strong>g anorexigenic POMC/CARTneurons [56]. Therefore a mechanism that disrupts this f<strong>in</strong>ely tuned apparatus has beensuggested to expla<strong>in</strong> this extreme behavior [55]. The stomach-secreted hormone Ghrel<strong>in</strong>, isassociated with <strong>in</strong>duc<strong>in</strong>g satiation follow<strong>in</strong>g feed<strong>in</strong>g, and is found to be <strong>in</strong>creased <strong>in</strong> PWS<strong>in</strong>dividuals [57]. Ghrel<strong>in</strong> stimulates the NPY/AGRP neurons via growth hormone secretagogreceptors, though a def<strong>in</strong>ite role <strong>in</strong> the pathogenesis of PWS is not yet confirmed. SNRPN hadlong been f<strong>in</strong>gered as causative <strong>in</strong> PWS, but patients with smaller microdeletions have enabledthe critical region to be narrowed to a locus conta<strong>in</strong><strong>in</strong>g non-cod<strong>in</strong>g HBII-85 snoRNAs [58].PWS babies are often born small-for-gestation-age with poor feed<strong>in</strong>g responses [59]. Thiscould be <strong>in</strong>terpreted <strong>in</strong> a reductionist way by the “conflict theory” as <strong>in</strong>dicative of the lack offetal paternal gene expression, with the later onset of extreme appetite driv<strong>in</strong>g the response due


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>to abnormal neurotrophic central pathway formation <strong>in</strong> the bra<strong>in</strong> dur<strong>in</strong>g this restricteddevelopment.The GNAS locus is another complex impr<strong>in</strong>ted region with an associated abnormal phenotypethat <strong>in</strong>cludes obesity. Isoforms of the GNAS (guan<strong>in</strong>e nucleotide b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>, a-stimulat<strong>in</strong>g)gene, the a-stimulatory subunit of the G-prote<strong>in</strong>, are created from multiple promotersand splic<strong>in</strong>g variations and an antisense non-cod<strong>in</strong>g RNA GNAS-AS1 (GNAS antisense RNA 1(non-prote<strong>in</strong> cod<strong>in</strong>g)) is also transcribed [60]. Biallelic, paternally and maternally expressedgenes are positioned with<strong>in</strong> this locus. GNAS itself is derived predom<strong>in</strong>ately from both alleles,with the exception of maternal-only expression <strong>in</strong> the tissues of the renal proximal tubules,thyroid, gonads, and pituitary. Loss of the maternal allele is associated with greater weight ga<strong>in</strong>[48,61]. XLAS is a large isoform variant of GNAS that is paternally expressed with<strong>in</strong> the nervoussystem and neuroendocr<strong>in</strong>e tissues. With<strong>in</strong> this cluster another GNAS isoform, previouslytermed Nesp <strong>in</strong> mouse, is maternally expressed [62]. Whilst maternal nonsense geneticmutations of GNAS are causative of pseudohypoparathyroidism type 1A (MIM #103580, alsoknown as Albright’s hereditary osteodystrophy), the loss of maternal-specific exon 1A DNAmethylation leads to pseudohypoparathyroidism type 1B (MIM #603233) [63]. The associatedmultiple hormone resistance is proposed to be due to these molecules utiliz<strong>in</strong>g signal<strong>in</strong>gpathways through G-prote<strong>in</strong>-coupled receptors and the associated obesity is thought similarlyto be due to abnormalities of these G-prote<strong>in</strong> receptors centrally [64]. Allelic variation <strong>in</strong> theHistone H3K4 state of the GNAS exon 1A promoter region with<strong>in</strong> proximal renal tubules hasalso been identified, illustrat<strong>in</strong>g the coord<strong>in</strong>ated nature of these post-translational histonemodifications with DNA methylation [65].278The human overgrowth disorder, BeckwitheWiedemann syndrome (BWS) (MIM #130650), iscaused by mutation or deletion of impr<strong>in</strong>ted genes with<strong>in</strong> the chromosome 11p15.5 region.Growth is of an <strong>in</strong>creased rate dur<strong>in</strong>g the latter half of pregnancy and <strong>in</strong> the first few years oflife, and whilst not lead<strong>in</strong>g to obesity per se, also displays the phenotypic potential ofimpr<strong>in</strong>t<strong>in</strong>g <strong>in</strong> terms of DNA methylation abnormalities and growth control throughoutdevelopment. It has also been proposed that <strong>in</strong> vitro assisted reproductive technology mayaffect the impr<strong>in</strong>t<strong>in</strong>g process and with some <strong>in</strong>creased levels of BWS <strong>in</strong> those conceived via thistechnique and confirmatory molecular studies recogniz<strong>in</strong>g BWS-specific epigenetic alterations[66]. This manipulation of germ cells and embryos that occurs <strong>in</strong> vitro, especially at such anearly crucial po<strong>in</strong>t <strong>in</strong> development, therefore demonstrates the fragility of the epigenomecompared to the genome, exemplified by this specific abnormality at an impr<strong>in</strong>ted locus.There is considerable variation <strong>in</strong> estimates of the actual level of impr<strong>in</strong>ted regions <strong>in</strong> themammalian, <strong>in</strong>clud<strong>in</strong>g the human, genome. Recent evidence has h<strong>in</strong>ted at the possibility ofhigh levels of particular bra<strong>in</strong> tissue-specific impr<strong>in</strong>t<strong>in</strong>g <strong>in</strong> a mouse model [67]. Soitisplausible that there is still an underestimation of impr<strong>in</strong>ted loci, particularly with the <strong>in</strong>clusionof tissue-specific and developmental-stage-specific variation. Current confirmed estimatesstand at around 50 genes <strong>in</strong> human [68,69]. DNA sequence characteristics and recomb<strong>in</strong>ationrates at impr<strong>in</strong>ted loci have led to predictions of 150 human genes, however without confirmationof differentially methylated regions (DMRs) [70] and further studies have also<strong>in</strong>dicated there are other undiscovered impr<strong>in</strong>ted loci [71,72]. Moreover whilst there is as yetno def<strong>in</strong>itive set of human or other eutherian mammalian impr<strong>in</strong>ted genes, there does appearto be significant dissimilarities between the species. These differences could be reconciled withthe “paternal conflict” theory, for <strong>in</strong>stance, as be<strong>in</strong>g driven by variation <strong>in</strong> litter size betweenmouse and human [73].Mouse models have <strong>in</strong>dicated a role of impr<strong>in</strong>ted genes <strong>in</strong> adipocyte metabolism. This<strong>in</strong>cludes the paternally expressed genes Dlk1 (delta-like 1 homolog, Drosophila), Mest(mesoderm specific transcript) (also known as Peg1) and Ndn (Necd<strong>in</strong>) [74]. Cheverud et al.calculated the effects that impr<strong>in</strong>t<strong>in</strong>g may have on complex traits <strong>in</strong> mice by estimat<strong>in</strong>g thevariation body size between reciprocal heterozygote, i.e. Aa compared to aA [75]. There was


CHAPTER 14Epigenomic Factors <strong>in</strong> <strong>Human</strong> Obesitya small but discernible difference of 0.25 standard deviation units or approximately 1e4% ofthe variance, which <strong>in</strong>cluded body weight, reproductive fat pad, and various organ weights.Further mur<strong>in</strong>e work look<strong>in</strong>g specifically at obesity quantitative trait loci (QTL) estimated thepossible effect of impr<strong>in</strong>t<strong>in</strong>g on diet-dependent obesity was high, with 61% of the QTLsidentified with a strong parental <strong>in</strong>fluence [76]. These sites were predom<strong>in</strong>ately not <strong>in</strong> knownimpr<strong>in</strong>ted regions.Two further paternally expressed genes Mest(Peg1) and Peg3 are <strong>in</strong>volved not only <strong>in</strong> fetal andpostnatal growth, but also can affect maternal nurtur<strong>in</strong>g success [77,78]. These impr<strong>in</strong>tedgenes are strongly expressed <strong>in</strong> hypothalamus, preoptic area, and septum, therefore they areexcellent candidates for neuronal programm<strong>in</strong>g [39].14.6 DIETARY INFLUENCE ON DNA METHYLATION IN MURINEMODELSThe epigenetic modulation of the expression of the non-impr<strong>in</strong>ted Agouti gene <strong>in</strong> the mouseviable yellow (A vy /a) allele is the most extensively studied “metastable epiallele” and hasbecome the archetypical model of dietary modulation of DNA methylation with subsequentphenotypic effects (see Figure 14.2). Metastable epialleles are so termed as these loci ofepigenetically variability are established very early <strong>in</strong> embryogenesis and subsequently rema<strong>in</strong>stable whilst permeat<strong>in</strong>g through all ensu<strong>in</strong>g developmental stages and germ layers [79]. In thewild-type mouse the Agouti gene encodes a signal<strong>in</strong>g molecule that produces either blackeumelan<strong>in</strong> (a) or yellow phaeomelan<strong>in</strong> (A). Transcription is normally <strong>in</strong>itiated from a hairspecificpromoter <strong>in</strong> exon 2, with transient expression of the A allele lead<strong>in</strong>g to the mottledbrown fur. In the agouti mutant mouse the <strong>in</strong>sertion upstream of the agouti gene of a s<strong>in</strong>gle<strong>in</strong>tracisternal A particle sequence (IAP) (endogenous retrovirus-like element), creates a crypticpromoter lead<strong>in</strong>g to an <strong>in</strong>crease of pheomela<strong>in</strong><strong>in</strong> over eumelan<strong>in</strong> [80]. This overaction results<strong>in</strong> a lighten<strong>in</strong>g of the coat color as ectopic expression of the <strong>in</strong>verse agonist at melanocort<strong>in</strong>receptors, agouti, antagonizes the action of melan<strong>in</strong> [79]. The “viable yellow” heterozygote(A vy /a) mouse has a shortened live span with yellow fur, obesity, and an <strong>in</strong>creased susceptibilityto neoplasia [81]. The associated obesity phenotype is due to the ability of the agoutiprote<strong>in</strong> to centrally mimic the Agouti-Related Prote<strong>in</strong> (AGRP) and to stimulate appetite <strong>in</strong> thehypothalamus via the NPY/AGRP orexigenic neurons.279FIGURE 14.2Agouti mouse show<strong>in</strong>g epigenetic dietary effects with graduation from obese Agouti to normal-sized Pseudoagouti (from [192].Reproduced with permission from Environmental Health Perspectives.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>There is a spectrum of expression of agouti depend<strong>in</strong>g on the epigenetic state of the IAPelement lead<strong>in</strong>g to a cont<strong>in</strong>uum from the agouti yellow, mottled to the brown “pseudoagouti”.The methylation of the IAP results <strong>in</strong> epigenetic suppression of this novel promoter and thepseudoagouti mouse is by comparison phenotypically lean with a normal lifespan. The crucialfunctional methylation variability occurs at six CpG sites that reside <strong>in</strong> the 5 0 long term<strong>in</strong>alrepeat (LTR) of the IAP element [82]. A dietary <strong>in</strong>fluence on the methylation of this upstreamIAP element was identified, whereby the use of methyl-supplemented diet or other nutritionalmodulations, <strong>in</strong>clud<strong>in</strong>g Geniste<strong>in</strong>, <strong>in</strong> the pregnant black (a/a) mother led to an alteration <strong>in</strong>the portion of heterozygote (A vy /a) sired and resultant pups with a skew towards the darkenedpseudoagouti phenotype [83,84] (see Figure 14.2). Histone modifications accompany thehypomethylation of the IAP element, with H3K and H4K diacetylation, whilst with DNAmethylation of the IAP, the repressive histone H4K20me3 mark is displayed (82). Thisphenotypic modulation of an <strong>in</strong>dividual locus by maternal food <strong>in</strong>take has led to hypotheses<strong>in</strong> regards to how the diet affects the develop<strong>in</strong>g embryo particularly by direct DNA methylationepiallele variation across the entire genome [79]. Additionally, this visible dietaryphenotypic effect is at a non-impr<strong>in</strong>ted locus, further expand<strong>in</strong>g the nutritional possibilities ofepigenomic modulation, particularly <strong>in</strong> a developmental sett<strong>in</strong>g, as an example of modulationof gene expression with potential <strong>in</strong>fluence on disease susceptibility.Dietary impact on impr<strong>in</strong>ted genes nevertheless has been documented, <strong>in</strong> the impr<strong>in</strong>ted Igf2locus <strong>in</strong> a mouse model [85]. Instalment of a methyl-donor-deficient diet post-wean<strong>in</strong>g led toloss of impr<strong>in</strong>t<strong>in</strong>g at this locus with subsequent modification of expression.280The <strong>in</strong>bred mouse stra<strong>in</strong> C57Bl/6J is documented as be<strong>in</strong>g highly susceptible to diet-<strong>in</strong>ducedobesity, but furthermore has also been observed to show a wide range of variability <strong>in</strong> this weightga<strong>in</strong> when fed a high-calorie diet [86]. Phenotype divergence <strong>in</strong>to those who would becomehigh weight-ga<strong>in</strong>ers versus low was even evident <strong>in</strong> measures before commenc<strong>in</strong>g an obesitypromot<strong>in</strong>gdiet. After this regimen was <strong>in</strong>troduced this led to a four-fold difference <strong>in</strong> obesity.Additionally these dissimilarities persisted even when the mice were switched back to a calorierestricteddiet. Koza et al. also detected gene expression differences <strong>in</strong> these isogenic mice,between the high- and low-weight ga<strong>in</strong>ers, prior to the high-calorie diet <strong>in</strong>troduction, lead<strong>in</strong>g tothe hypothesis that pre-exist<strong>in</strong>g epigenetic factors were <strong>in</strong>volved <strong>in</strong> these observations [86].Overfeed<strong>in</strong>g <strong>in</strong> rats, <strong>in</strong>duced by limit<strong>in</strong>g the litter size, led to an obese phenotype [87].Investigation of hypothalamic tissue showed hypermethylation of the promoter of POMC, theanorexigenic neurohormone, proopiomelanocort<strong>in</strong>, <strong>in</strong>volved <strong>in</strong> appetite repression [87].Lept<strong>in</strong> and <strong>in</strong>sul<strong>in</strong> stimulate this pathway via two Sp1-related b<strong>in</strong>d<strong>in</strong>g sequences with<strong>in</strong> thispromoter. The hyperlept<strong>in</strong>emia and hyper<strong>in</strong>sul<strong>in</strong>emia present <strong>in</strong> these rats therefore did notlead to an up-regulation of POMC expression. The DNA methylation levels with<strong>in</strong> theseb<strong>in</strong>d<strong>in</strong>g sites were <strong>in</strong>versely correlated to the quotients of POMC expression/lept<strong>in</strong> and POMCexpression/<strong>in</strong>sul<strong>in</strong>, demonstrat<strong>in</strong>g the function of these acquired epigenomic alterations <strong>in</strong>modify<strong>in</strong>g the “set po<strong>in</strong>t” parameters of this promoter critical for bodyweight regulation byoverfeed<strong>in</strong>g: an example of obesity epigenetic reprogramm<strong>in</strong>g.Recently <strong>in</strong> humans, obese women, who were separated <strong>in</strong>to high and low responders todietary restriction, were found to have measurable differences <strong>in</strong> subcutaneous adipose tissueDNA methylation [88]. In another <strong>in</strong>vestigation of human muscle cells, a reversible effect onDNA methylation <strong>in</strong> the key metabolic regulator Peroxisome Proliferator-Activated Receptor g,coactivator 1 (PPARGC1A) with overfeed<strong>in</strong>g was seen [89].14.7 OBESOGENIC ENVIRONMENT EFFECTS ON COMMON HUMANOBESITYOver and above the cumulative genetic susceptibility risks recently identified from GWAS, arethe major environmental elements that are the product of urban and socioeconomic


CHAPTER 14Epigenomic Factors <strong>in</strong> <strong>Human</strong> Obesitytransitions over the last 60 years and the formation of the aforementioned “obesogenic”environment. This can be perceptibly displayed by the observation that the common geneticsusceptibilities towards the trait are not acted upon, unless certa<strong>in</strong> compound<strong>in</strong>g causes areencountered. Lifestyle contributors such as diet and exercise are central <strong>in</strong> liability to metabolicdisease and also have a substantial aggregate effect [90]. It has been proposed that the etiologyof common diseases are under both genetic and epigenetic <strong>in</strong>fluence and these disease-relatedepigenetic factors could be environmentally <strong>in</strong>duced with subsequent modulatory effects ongenetic susceptibility [91]. Epigenetic marks such as DNA methylation provide a mechanismby which these environmental <strong>in</strong>fluences can modulate genetic susceptibility to disease, asevidence from mur<strong>in</strong>e models and humans has alluded to. Nutrition, behavior, stress, tox<strong>in</strong>s,and stochastic action can all have an impact on DNA methylation lead<strong>in</strong>g to variation <strong>in</strong>epialleles [92]. Epigenetic effect on gene expression by the modification of the target cellepigenome [93], thereby changes metabolic risk [79,94]. Comb<strong>in</strong>ed gene by environmental<strong>in</strong>teractions have been observed with known genetic susceptibility factors for obesity, as seenwith accentuated adiposity effect of the FTO susceptibility variant <strong>in</strong> <strong>in</strong>dividuals through lowphysical activity [95]. The significant role of epigenetics <strong>in</strong> the pathogenesis of cancer is wellestablished [96] but has also been seen <strong>in</strong> other diseases such as <strong>in</strong> the etiology of atheroscleroticplaques [97], and evidence is accumulat<strong>in</strong>g <strong>in</strong> the metabolic syndrome.14.8 AGING EFFECT ON DNA METHYLATIONIn demonstrat<strong>in</strong>g the non-rigid state of DNA methylation, alteration over time has been an<strong>in</strong>itial target. An accumulation of DNA variation over a lifetime was found by compar<strong>in</strong>gdivergence between monozygotic tw<strong>in</strong>s by age [98]. This may be caused by accrued environmentaleffect and/or “epigenetic drift” due to defective transmission through multiple mitoticreplications. DNA methylation over time devolves from its fixed bimodal extremes; withhypomethylated CpGs with<strong>in</strong> islands ga<strong>in</strong><strong>in</strong>g methylation and hypermethylated CpGs los<strong>in</strong>gmethylation [99]. Therefore it has been speculated these shifts may modify metabolic pathways,becom<strong>in</strong>g gradually suboptimal, lead<strong>in</strong>g to slow late-onset weight ga<strong>in</strong> [100]. Now thathigher-resolution DNA methylome analysis has become available more subtle signatures ofag<strong>in</strong>g have begun to be identified, such as hypermethylation at the promoters of the targetgenes of the repressive Polycomb Group prote<strong>in</strong>s [101].28114.9 DEVELOPMENTAL EPIGENOMIC DYSREGULATIONWhilst DNA methylation is a comparatively stable repressive mark, it is however required to beremoved from specific genomic regions, as well as genome-wide, through the process ofdevelopment. The major stages are at postfertilization and at germ cell differentiation <strong>in</strong> malesand females. Two global waves of demethylation are undertaken with the embryo be<strong>in</strong>g mostsusceptible to environmental perturbation dur<strong>in</strong>g the re-establishment of DNA methylationpostfertilization but preimplantation [92]. Experimental evidence <strong>in</strong> mice shows that thepreimplantation embryo is sensitive to epigenetic modifications [102]. Furthermore, the entireprocess of embryogenesis <strong>in</strong>volves exact DNA methylation configuration <strong>in</strong> order that normaltissue development occurs and so is a w<strong>in</strong>dow of possible <strong>in</strong>fluence [92].Therefore, these adverse environmental effects with<strong>in</strong> specific critical junctures, such as whenthe tissue-specific epigenome is be<strong>in</strong>g cast <strong>in</strong> develop<strong>in</strong>g cells, have been proposed to havesignificant last<strong>in</strong>g effects on metabolism <strong>in</strong>to adult life with a concurrent risk of chronicdiseases [92,103,104]. Direct evidence of dietary modulation dur<strong>in</strong>g these time-po<strong>in</strong>ts, andthe latent ability to affect long-term risk of chronic metabolic disease health has beenattempted us<strong>in</strong>g mur<strong>in</strong>e models, through from the periconceptual period to postwean<strong>in</strong>g[105]. The epigenetic state of the transcription factor Hnf4a was <strong>in</strong>vestigated <strong>in</strong> the pancreasof rats that had been subjected to poor maternal diet and controls [106]. HNF4A is implicated<strong>in</strong> T2D pathogenesis and mutants are associated with a monogenic form of T2D


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>(MODY, type I, MIM #125850). Poor maternal diet dur<strong>in</strong>g critical periods of development, aswell as ag<strong>in</strong>g, was shown to down-regulate an islet-specific promoter and the <strong>in</strong>teractionbetween the promoter and an enhancer was also down-regulated. A small study <strong>in</strong> humansalso identified methylation changes <strong>in</strong> HNF4A <strong>in</strong> cord blood follow<strong>in</strong>g <strong>in</strong>trauter<strong>in</strong>e growthretardation (IUGR) [107].Further rat models of IUGR <strong>in</strong>vestigated the DNA methylation and gene expression ofpancreatic islets [108]. Cellular memory <strong>in</strong> the pancreatic cells of the developmental <strong>in</strong>trauter<strong>in</strong>eenvironment was sought by the <strong>in</strong>vestigation of approximately 1 million CpG sites <strong>in</strong>the rat methylome of these cells at the later age of 7 weeks. This study p<strong>in</strong>po<strong>in</strong>ted changes <strong>in</strong>methylation state of approximately 1400 CpGs consistent with undergo<strong>in</strong>g IUGR. Thesealterations were preferably found <strong>in</strong> highly conserved non-cod<strong>in</strong>g <strong>in</strong>tergenic genomic loci,which may be cis-regulatory sites, and frequently near genes <strong>in</strong>volved <strong>in</strong> the deregulation ofislets follow<strong>in</strong>g IUGR, <strong>in</strong>clud<strong>in</strong>g <strong>in</strong>sul<strong>in</strong> secretion, vascularization, cell proliferation, and celldeath. Also they were additionally associated with mRNA expression levels preced<strong>in</strong>g thedevelopment of diabetes.28214.10 FETAL PLASTICITYThe dramatic and startl<strong>in</strong>g <strong>in</strong>crease <strong>in</strong> obesity over such a short historical period has led to theconcept that fetal programm<strong>in</strong>g or plasticity may be <strong>in</strong>volved. This proposes that the periconceptual,<strong>in</strong> utero, and postnatal developmental environment can impact on long-term riskfor adult-onset disease by set po<strong>in</strong>t adaptive changes [109e111]. This suggests that “poor”nutrition at critical growth stages <strong>in</strong>creases the chance of develop<strong>in</strong>g the metabolic syndrome(<strong>in</strong>sul<strong>in</strong> resistance, obesity, dyslipidemia, and hypertension) <strong>in</strong> later life and these developmentalorig<strong>in</strong>s of adult disease put forward epigenetic <strong>in</strong>heritance as the possible mechanism<strong>in</strong> this programm<strong>in</strong>g [92].Although ischemic heart disease <strong>in</strong>creases <strong>in</strong> a population as it becomes wealthier, it was notedthat those <strong>in</strong> the poorest regions suffered disproportionately [112]. An early epidemiologicalstudy <strong>in</strong> England and Wales identified a connection between poor nutrition <strong>in</strong> early life and<strong>in</strong>creased susceptibility to ischemic heart disease, that was suggested to be due to a moredetrimental effect of an adiposity-promot<strong>in</strong>g diet <strong>in</strong> these <strong>in</strong>dividuals [112]. Furthermore,impaired glucose tolerance <strong>in</strong> men aged 64 was correlated to low weight at birth and at 1 year[113]. The “thrifty phenotype” hypothesis proposed by Barker encapsulates this prospectiveeffect of fetal programm<strong>in</strong>g and the reprogramm<strong>in</strong>g of the lept<strong>in</strong>emelanocort<strong>in</strong> hypothalamicaxis, pituitary, adrenal, or islet development, or <strong>in</strong>sul<strong>in</strong>-signal<strong>in</strong>g pathways may be mechanisms<strong>in</strong> these long-term effects [114].Documented historical fam<strong>in</strong>es furthermore allow these questions to be proposed to agecohorts, and the Dutch Hunger W<strong>in</strong>ter of November 1944 to May 1945 has been a classicexample <strong>in</strong> the literature. Upon reach<strong>in</strong>g the army draft age of 19 years the obesity rates of300 000 men were compared [115]. The outcomes were dependent on when dur<strong>in</strong>g theirdevelopment the severe wartime fam<strong>in</strong>e had occurred. Those exposed <strong>in</strong> the last trimester orthe first months of life were less likely to be obese, whilst those whose mothers had been <strong>in</strong>early or mid-pregnancy dur<strong>in</strong>g this severe wartime fam<strong>in</strong>e were twice as likely to be obese atarmy draft. This would imply early changes may differ <strong>in</strong> programm<strong>in</strong>g from later changes,fitt<strong>in</strong>g with hypotheses of developmental programm<strong>in</strong>g and critical epigenetic w<strong>in</strong>dows.Epigenetic exam<strong>in</strong>ation of DNA methylation <strong>in</strong> these Dutch Hunger W<strong>in</strong>ter <strong>in</strong>dividuals 60years after this fam<strong>in</strong>e displayed variation at the impr<strong>in</strong>ted DMR of IGF2 that was found to beless methylated among periconceptionally exposed <strong>in</strong>dividuals [116]. Individuals conceiveddur<strong>in</strong>g the fam<strong>in</strong>e were compared aga<strong>in</strong>st a sibl<strong>in</strong>g of the same sex to attempt to reduceenvironmental and genetic modulators. Follow-up work <strong>in</strong>vestigated a further 15 loci withknown <strong>in</strong>volvement <strong>in</strong> growth and metabolism [117]. In those with periconceptual fam<strong>in</strong>e,


CHAPTER 14Epigenomic Factors <strong>in</strong> <strong>Human</strong> ObesityDNA methylation was <strong>in</strong>creased <strong>in</strong> the promoter and impr<strong>in</strong>ted DMRs of GNAS-AS1 andMEG3 (maternally expressed 3 (non-prote<strong>in</strong> cod<strong>in</strong>g)), as well as proximal promoters of IL10(<strong>in</strong>terleuk<strong>in</strong> 10), ABCA1 (ATP-b<strong>in</strong>d<strong>in</strong>g cassette, sub-family A (ABC1), member 1), and LEP(Lept<strong>in</strong>). Comparative decreased DNA methylation was also recognized <strong>in</strong> the promoter ofimpr<strong>in</strong>ted INS-IGF2 (INS-IGF2 read-through) which is also part of the INS (<strong>in</strong>sul<strong>in</strong>) promoter.These studies have highlighted the periconceptual period as a volatile spatiotemporal w<strong>in</strong>dow<strong>in</strong> epigenomic development with additional support for this com<strong>in</strong>g from animal models[118]. Manipulation of dietary vitam<strong>in</strong> B and methion<strong>in</strong>e dur<strong>in</strong>g this periconceptual period <strong>in</strong>sheep led to heavier, fatter, and <strong>in</strong>sul<strong>in</strong>-resistant animals [119].In a large population-based obesity study, the maternal impact on this trait was shown to begreater [120], possibly due to maternally impr<strong>in</strong>ted genes. As these impr<strong>in</strong>ted loci are strongly<strong>in</strong>terconnected with resource allocation, energy balance, and feed<strong>in</strong>g behavior, early environmentaleffects may have long-reach<strong>in</strong>g consequences. This makes these parental-specificmarks obviously strong <strong>in</strong>itial candidates <strong>in</strong> any “fetal programm<strong>in</strong>g” or “plasticity” <strong>in</strong>fluenceon chronic disease. It may be that these impr<strong>in</strong>t<strong>in</strong>g resource allocation tools are subverted bythe “fetal programm<strong>in</strong>g” mechanism, enabl<strong>in</strong>g a shift of resource regulation over the course ofa lifetime and subsequent risk for adult-onset disease [39]. This metabolic plasticity enablesa non-changed genome to produce a range of phenotypes <strong>in</strong> response to variation <strong>in</strong> environment,specifically early nutritional status [121,122]. Subtle modulation of impr<strong>in</strong>t<strong>in</strong>gpathways could be a mechanism, or it may be that they are too critical for an adaptive processand changes are only seen <strong>in</strong> severe disease manifestations [123].Two <strong>in</strong>dependent, but sometimes overlapp<strong>in</strong>g, pathways show<strong>in</strong>g how the nutritional statethrough development can lead to <strong>in</strong>creased susceptibility to later-onset obesity have beenproposed [123]. First, this may be a “mismatch” pathway, which can be either severe orpredictive. These are developmental plasticity modifications of the genetically driven phenotypecued by prenatal undernutrition or possibly stress, which then may not be correctlygeared, if the postnatal environment is obesogenic. Second, a pathway due to the risks causedby maternal obesity and the “hypernutrition” experienced by the fetus <strong>in</strong> this situation.28314.11 POSTNATAL ENVIRONMENTAL MISMATCHSurvival-<strong>in</strong>duced modifications are due to severe maternal ill health or placental abnormalityand lead to growth reduction <strong>in</strong> order to survive. Fetal growth retardation leads to changes <strong>in</strong>gene expression driven by epigenetic changes [114]. Recent work has shown that via theimpr<strong>in</strong>ted Peg3 placental sacrifice will occur <strong>in</strong> order to protect bra<strong>in</strong> development [124].Optimization for survival favors some organs aga<strong>in</strong>st overall growth, but may lead to persistentmetabolic and endocr<strong>in</strong>ological changes that over time become detrimental when subsequentlyencounter<strong>in</strong>g an obesogenic environment [114]. For <strong>in</strong>stance the development of<strong>in</strong>sul<strong>in</strong> resistance <strong>in</strong> order to survive <strong>in</strong> utero undernutrition [103].Significant correlations between low birth weight and adult-onset disease <strong>in</strong> studies acrossEurope and <strong>in</strong> the USA have been identified, even after corrections for possible confounderssuch as socioeconomic status, physical activity, smok<strong>in</strong>g, etc. [114]. There is a disproportionateloss of subcutaneous compared to visceral fat <strong>in</strong> undernourished babies, which leads toa relative visceral adiposity and additionally evidence of specific further ga<strong>in</strong> <strong>in</strong> visceral fat <strong>in</strong>IUGR children [125]. Those born with the smallest birth weight (


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>Restricted <strong>in</strong>fant growth and fast weight ga<strong>in</strong> <strong>in</strong> childhood <strong>in</strong>tensify the effects of impairedprenatal growth [128]. Evidence from many animals po<strong>in</strong>ts to the additive effect of prenataldiet restriction and then subsequent hypercalorific diet <strong>in</strong> the develop<strong>in</strong>g <strong>in</strong>fant lead<strong>in</strong>g tostronger risk of weight ga<strong>in</strong> than the latter alone [129]. Mouse evidence shows high levels of“catch-up” growth after <strong>in</strong> utero growth restriction leads to even further <strong>in</strong>creased susceptibilityto the adverse effects on lifespan of a postwean<strong>in</strong>g obesogenic diet [130]. Furtherevidence for probable programm<strong>in</strong>g of chronic disease by impaired fetal nutrition wasestablished<strong>in</strong>anIndianpopulationstudy,where small-for-gestational-age babies witha high “catch up” growth and therefore high fat mass at ages 2 through to 12, later had thehighest levels of <strong>in</strong>sul<strong>in</strong> resistance [5]. Some studies have, however, found <strong>in</strong>conclusiveevidence of early undernutrition <strong>in</strong>creas<strong>in</strong>g risk of later obesity, but did support a role forovernutrition [131].Predictive responses via phenotypic plasticity mechanisms are <strong>in</strong> order to improve or optimizefitness at a later stage of development [132]. These are an evolutionary beneficial ability toenable adaptation to probable future environmental likelihoods, but are not advantageous atthe time [133]. These modifications allow subtle modification where this is possible. Thisresponse is made with<strong>in</strong> the variation of normal range <strong>in</strong> development and is a nonpathologicalenvironmental prediction adjustment.284Hypothalamic neuroendocr<strong>in</strong>e gene expression effects were identified <strong>in</strong> a rat model of thisresponse [134]. Prenatal undernutrition and postnatal high-fat diet with subsequent assessmentat 24 weeks revealed specific alterations <strong>in</strong> the hypothalamic gene expression of POMC,NPY, AGRP, and the Ob-Rb isoform of LEPR, as well as the observation of circulat<strong>in</strong>g hyper<strong>in</strong>sul<strong>in</strong>emiaand hyperlept<strong>in</strong>emia. These f<strong>in</strong>d<strong>in</strong>g were supportive therefore of a predictiveresponse hav<strong>in</strong>g <strong>in</strong>fluenced the neurogenesis of the hypothalamic pathways that had subsequentlybeen <strong>in</strong>correct for the later environment encountered.Exploration <strong>in</strong> humans for epigenetic evidence <strong>in</strong>vestigated 68 CpGs 5 0 of five candidate genesfrom neonatal umbilical cord tissue DNA and ascerta<strong>in</strong>ed an epiallele association withmaternal pregnancy diet and childhood fat mass at age 9 years <strong>in</strong>clud<strong>in</strong>g an <strong>in</strong>dividual CpG 5 0of RXRA (Ret<strong>in</strong>oid X receptor-a) and NOS3 (nitric oxide synthase 3 (endothelial cell))previously known as eNOS [135]. RXRA also associated with lower maternal carbohydrate<strong>in</strong>put and additionally only the RXRA adiposity result replicated <strong>in</strong> a second group. If thisresult can be corroborated by other researchers this may represent likely “low-hang<strong>in</strong>g fruit” orchanges that are strong enough to be exposed by candidate studies. If so this will bode well formore powerful metabolic epigenome-wide association studies (EWAS) currently be<strong>in</strong>gproposed. Changes <strong>in</strong>clude <strong>in</strong>creases as well as decreases <strong>in</strong> methylation, therefore dietrestriction of methyl donors is a too simplistic explanation of this observation, rather anadaptive response is hypothesized [103,110].14.12 HYPERNUTRITIONSecondly, the direct effect on the <strong>in</strong>trauter<strong>in</strong>e environment of maternal diabetes and obesity, aswell as <strong>in</strong>fant overfeed<strong>in</strong>g of high-calorific foods, lead to greater risk of adult obesity.Increas<strong>in</strong>g numbers of women now are overweight when pregnant [39]. Hyper<strong>in</strong>sul<strong>in</strong>emia iscausative of macrosomia <strong>in</strong> diabetic mothers with the greatest effect on fat mass and subsequentlygreater risk of childhood obesity [136].In a Danish study the adult children of women who suffered from gestational diabetes dur<strong>in</strong>gpregnancy had a 2.5 times <strong>in</strong>creased rate of metabolic syndrome and a twofold risk of overweightcompared to a selected control population [137]. Central appetite regulatory circuitry<strong>in</strong> the hypothalamus is active with<strong>in</strong> the fetus and affected by the nutritional state <strong>in</strong> developmentalstages, therefore it may lead to long-term effects <strong>in</strong> the gear<strong>in</strong>g of this system due tothis chronic hypernutrition [138]. This neural programm<strong>in</strong>g may be variable with malleability


CHAPTER 14Epigenomic Factors <strong>in</strong> <strong>Human</strong> Obesityof this system at critical species-specific w<strong>in</strong>dows dependent on restricted or excess diet [138].High levels of white fat <strong>in</strong> human <strong>in</strong>fants compared to other mammals is proposed as anenergy store buffer to protect bra<strong>in</strong> development post-wean<strong>in</strong>g [139], so this level ofpreload<strong>in</strong>g will not be drawn upon and reduce with excess feed<strong>in</strong>g. Increas<strong>in</strong>g affluence <strong>in</strong>develop<strong>in</strong>g countries leads to an <strong>in</strong>crease <strong>in</strong> availability of ref<strong>in</strong>ed foods which are cheaper buthave less nutritional value than fresh foods [6]. Also, early feed<strong>in</strong>g of differ<strong>in</strong>g diet, breast orformula, may affect absorption and gut microbiotic changes have been found to have a role <strong>in</strong>obesity [140]. One study on <strong>in</strong>trauter<strong>in</strong>e evidence for effect on obesity, showed an associationwith maternal and paternal BMI, imply<strong>in</strong>g that shared environmental <strong>in</strong>fluence also cannot bediscounted [141].14.13 EPIGENETIC ANALYSIS OF LEPTINAdipose tissue is an endocr<strong>in</strong>e organ, key <strong>in</strong> energy homeostasis, which secretes adipok<strong>in</strong>es,the major player be<strong>in</strong>g lept<strong>in</strong>. This supplies vital feedback to the hypothalamus about fat mass.The promoter of lept<strong>in</strong> moves from a highly methylated to a low methylation state <strong>in</strong> thedifferentiation from pre-adipocyte to adipocyte, thus facilitat<strong>in</strong>g expression of this endocr<strong>in</strong>ehormone <strong>in</strong> mature fat cells [142]. Lept<strong>in</strong> additionally has been shown to have a role <strong>in</strong>neurogenesis and specifically with<strong>in</strong> the critical hypothalamic pathways [143,144]. Visualizationof NPY and POMC neurons, by expression of fluorescent prote<strong>in</strong>s, displayed a lept<strong>in</strong>mediatedmodulation of synaptic density onto these cells. Lept<strong>in</strong>’s neurotrophic role <strong>in</strong> thehypothalamus is furthermore illustrated by the lack of neuronal projection pathways from thearcuate nucleus <strong>in</strong> the lept<strong>in</strong>-deficient mouse model. This neuroanatomical deficiency cannotbe reversed with adulthood lept<strong>in</strong> adm<strong>in</strong>istration, but can, if delivered dur<strong>in</strong>g the neonatalperiod [145].Rats fed a high-fat diet become obese and hyperlept<strong>in</strong>emic, but also <strong>in</strong>crease methylation <strong>in</strong>the promoter of lept<strong>in</strong> and this was collated with a comparative reduction <strong>in</strong> expression oflept<strong>in</strong> [146]. Further <strong>in</strong> vivo analysis of the lept<strong>in</strong> promoter <strong>in</strong> human and mouse identified itstissue-specific variability <strong>in</strong> methylation (t-DMR). A critical C/EBPa (CCAAT/enhancerb<strong>in</strong>d<strong>in</strong>gprote<strong>in</strong> alpha) [147] transcription b<strong>in</strong>d<strong>in</strong>g site is located with<strong>in</strong> the CpG Island andgenerally has overall lower methylation <strong>in</strong> adipose compared to peripheral blood tissue [148].Interest<strong>in</strong>gly the mouse CpG Island possessed a higher level of <strong>in</strong>termediate methylation andis smaller ( 1 / 3 size) and a less dense CpG Island, therefore it is speculated to have lost CpGs viadeam<strong>in</strong>ation, suggest<strong>in</strong>g it may have higher levels of germl<strong>in</strong>e methylation [148]. No DNAmethylation differences <strong>in</strong> the comparison of T2D cases versus controls <strong>in</strong> the lept<strong>in</strong> promotervia MeDIP-chip of peripheral blood were however identified [149] [see Figure 14.3] and nochanges were found <strong>in</strong> a study post weigh loss via bariatric surgery [150].28514.14 HISTONE EPIGENOMIC MODIFIERS e MASTER METABOLICREGULATORSRecent work <strong>in</strong> mur<strong>in</strong>e models has illum<strong>in</strong>ated the potential role of modifiers of the epigenetichistone state, such as methyltransferases and demethylases, <strong>in</strong> metabolism and energyhomeostasis gene pathways. Particularly this has highlight<strong>in</strong>g the critical role of Kdm3a (lys<strong>in</strong>e(k)-specific demethylase 3a), also known as Jhdm2a, <strong>in</strong> metabolic regulation [151]. This acts asa H3K9-specific demethylase, catalyz<strong>in</strong>g the removal of mono- and dimethylation from H3K9,therefore has a de-repressive function. Abrogation of action was shown to lead to significantmultigenic effects on metabolic pathways result<strong>in</strong>g <strong>in</strong> a maturity-onset obesity, hypercholesterolemia,hyper<strong>in</strong>sul<strong>in</strong>emia, hypertriglycerolemia, and hyperlipidemia phenotype. This<strong>in</strong>volved reduction with<strong>in</strong> the skeletal muscle of fat oxidation and the release of glycerol and <strong>in</strong>brown fat cell disruption of oxygen consumption and the b-adrenergic stimulated secretion ofthe glycerol. The expression of this demethylase is stimulated by the b-adrenergic mechanismsand it plays a significant role <strong>in</strong> expression of the transcription factor Ppara (peroxisome


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>FIGURE 14.3Methylation DNA immunoprecipitation (MeDIP) graphical result for the Lept<strong>in</strong> Promoter CpG Island. Slid<strong>in</strong>g scale for methylation level: yellow ¼ low, green ¼mid, blue ¼ high. Hypomethylation seen over the CpG Island and no significant difference seen between T2D cases (f<strong>in</strong>al_avg_diabetes) versus controls(f<strong>in</strong>al_ave_control). This figure is reproduced <strong>in</strong> the color plate section.286proliferator-activated receptor alpha), essential <strong>in</strong> fatty acid metabolism, and Ucp1 (Uncoupl<strong>in</strong>gprote<strong>in</strong> 1) <strong>in</strong>volved <strong>in</strong> decoupl<strong>in</strong>g energy creation to generate heat <strong>in</strong> the mitochondriaof brown adipose tissue. These abnormalities <strong>in</strong> adipose storage and energy balance were alsodisplayed <strong>in</strong> the knockout mouse by hypothermia and reduced energy production via fatutilization [152].14.15 METASTABLE ALLELES IN HUMAN ASSOCIATED WITHOBESITYGenetic <strong>in</strong>fluence on the methylation state, or allele-specific variation <strong>in</strong> methylation levels[153,154] by polymorphism such as CpGeSNPs, has been acknowledged as hav<strong>in</strong>ga considerable bear<strong>in</strong>g on region methylation levels [155,156]. Furthermore, DNA methylationlevels driven by CpG ga<strong>in</strong> and loss lead<strong>in</strong>g to higher variance <strong>in</strong> local methylation havebeen proposed as a major driver <strong>in</strong> evolution as well as common disease susceptibility [157]and epigenetic and other concepts of non-DNA <strong>in</strong>heritance are now beg<strong>in</strong>n<strong>in</strong>g to be <strong>in</strong>corporated<strong>in</strong>to evolutionary theory [158]. Transgenerational transmission possibilities are stillunclear with current evidence, with some reports of <strong>in</strong>complete epigenetic erasure [159],however it is very difficult to completely exclude genetic effects (for review see [160]). Paternal<strong>in</strong>heritance effects have been proposed <strong>in</strong> two recent papers [159,160].Epigenetic variability, or epialleles, can vary purely without genetic <strong>in</strong>fluence, or play a facilitativeor obligatory role conferred by genetic variants. Furthermore, they are generally <strong>in</strong>tr<strong>in</strong>sicallytissue-specific, however a subset of these epialleles is determ<strong>in</strong>ed very early <strong>in</strong>development and subsequently propagate through all differentiat<strong>in</strong>g cell l<strong>in</strong>eages as illustratedby the Agouti mouse. <strong>Human</strong> “metastable epialleles” with correlations across tissues represent<strong>in</strong>gall three develop<strong>in</strong>g germ layers have also been observed [162]. These loci were shownto have highly different values, between monogenic tw<strong>in</strong>s, <strong>in</strong>dicat<strong>in</strong>g that they are likely set <strong>in</strong>


CHAPTER 14Epigenomic Factors <strong>in</strong> <strong>Human</strong> Obesitya stochastic fashion. Furthermore, DNA methylation at these epialleles was shown to besignificantly associated with the experienced periconceptual nutritional environment <strong>in</strong> a setof rural Gambians. A higher level of methylation was identified <strong>in</strong> those who had beenconceived dur<strong>in</strong>g the nutrition-reduced ra<strong>in</strong>y season [162]. Metastable epialleles of impr<strong>in</strong>tedgenes have also been hypothesized to play a major part <strong>in</strong> adaption and evolution [163].Fe<strong>in</strong>berg et al. catalogued 227 genomic regions that showed a wide level of DNA methylationvariability between 638 Icelandic <strong>in</strong>dividuals [164]. These regions were enriched for developmentaland morphogenesis genes. The <strong>in</strong>dividuals were sampled at two time periodsapproximately 11 years apart. Approximately half of these, or 119 regions, termed variablemethylation regions (VMRs), were found to be stable over this time course with<strong>in</strong> <strong>in</strong>dividuals[164]. These VMRs therefore could create an <strong>in</strong>dividual epigenomic signature or f<strong>in</strong>gerpr<strong>in</strong>t.Four of these stable VMRs correlated with BMI at both timepo<strong>in</strong>t visits: these loci resided <strong>in</strong> ornear the genes PM20D1, MMP9, PRKG1, and RFC5. Up-regulation of MMP9 has been seen <strong>in</strong>obese <strong>in</strong>dividuals [165], and PRKG1 has been associated with energy balance and foodbehavior <strong>in</strong> a number of organisms [166]. These highly variable regions were proposed toreside with<strong>in</strong> loci that could possibly be highly susceptible to environmental modulation andcould be <strong>in</strong>vestigated for strong environmental <strong>in</strong>fluences such as tox<strong>in</strong>s, smok<strong>in</strong>g, dietaryvariation, etc. However, a caveat stated that this variability may be contributed to by the mixedcell type of peripheral blood that was exam<strong>in</strong>ed. The <strong>in</strong>tra<strong>in</strong>dividual time-stable VMRs couldbe expected to have a strong genetic component, such as possible CpG-SNP or haplotypespecificmethylation effect [167].14.16 PARENT-OF-ORIGIN GENETIC EFFECTSGenome-wide l<strong>in</strong>kage has been used to try to f<strong>in</strong>d impr<strong>in</strong>ted obesity-related genomic loci[168]. Whilst highly successful <strong>in</strong> discover<strong>in</strong>g monogenic disease genes, this technique has hadlow success <strong>in</strong> polygenic traits. GWAS have achieved far more <strong>in</strong> identify<strong>in</strong>g variants stronglyassociated with these common diseases, <strong>in</strong>clud<strong>in</strong>g obesity risk and BMI modulation [15,169].Furthermore, success has been found <strong>in</strong> look<strong>in</strong>g for parent-of-orig<strong>in</strong> effects <strong>in</strong> GWAS associationSNPs, with significant results <strong>in</strong> other complex disease traits, such as type 1 diabetes[170].287Analysis from deCODE Genetics, also of GWAS SNP data, revealed a number of parent-oforig<strong>in</strong>effects [69]. By utiliz<strong>in</strong>g the extensive known genealogies of these Icelandic <strong>in</strong>dividuals,as well as long-range phas<strong>in</strong>g of haplotypes, parental state of <strong>in</strong>herited alleles was determ<strong>in</strong>ed.They focused on those disease-associated SNPs with<strong>in</strong> 500 kb of known impr<strong>in</strong>ted regions(approximately 1% of the genome), which <strong>in</strong>formed that SNPs with<strong>in</strong> two known clusters onchromosome 11p15 (conta<strong>in</strong><strong>in</strong>g H19/IGF2 and KCNQ1 impr<strong>in</strong>ted clusters) and 7q32(<strong>in</strong>clud<strong>in</strong>g the maternally expressed genes, CPA4 and KLF14 surround<strong>in</strong>g paternally expressedgenes MEST (PEG1) and MESTIT1) were present. Whilst not obesity SNP associations, T2Ddisease-associated traits were located here. Five parent-of-orig<strong>in</strong> SNP associations were able tobe made, <strong>in</strong>clud<strong>in</strong>g three with T2D. The maternally expressed KCNQ1 conta<strong>in</strong>ed two SNPs,rs2237892 and rs231362, each with allele C significantly associated when transmittedmaternally. Both these SNPs are at CpG-SNP sites, with the C variant of the polymorphismfacilitat<strong>in</strong>g methylation by the creation of a CpG.A third SNP, allele C of rs4731702, aga<strong>in</strong> via maternal <strong>in</strong>heritance at 7q32, was significantlyassociated and additionally this risk allele correlated with lower expression of KLF14 <strong>in</strong>adipose but not blood tissue and aga<strong>in</strong> only when <strong>in</strong>herited from the maternal side. Subsequentwork by Small et al. discovered this cis-modulatory SNP rs4731702 of this impr<strong>in</strong>tedKLF14 transcription factor gene to be a significant trans regulator <strong>in</strong> gene expression studies <strong>in</strong>subcutaneous adipose tissue biopsies from a cohort of 776 healthy female tw<strong>in</strong>s [171]. The<strong>in</strong>fluence of this factor was enriched for metabolic syndrome phenotypes, <strong>in</strong>clud<strong>in</strong>g BMI, with


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>evidence from APH1B, ARSD, C8orf82, GNB1, SLC7A10, and TPMT expression levels andassociated the control of this trans expression network by KLF14 with risk of metabolic disease.Kong et al. also established a novel association at SNP rs2334499, with susceptibility via thepaternal, and a protective effect via maternal l<strong>in</strong>eage, and there was correlation witha decreased methylation state of a nearby CTCF-b<strong>in</strong>d<strong>in</strong>g region, which are known genomic<strong>in</strong>sulators, and this susceptibility variant [69].These parent-of-orig<strong>in</strong> associations with metabolic syndrome traits h<strong>in</strong>t at the role of subtlegenetic variation, and potentially epiallele variation, <strong>in</strong> these impr<strong>in</strong>ted loci <strong>in</strong>fluenc<strong>in</strong>g thesediseases [172]. This is particularly the case when these genetic variants may be either directlyimpact<strong>in</strong>g on methylation ability, <strong>in</strong> the case of CpG-SNPs, or additionally <strong>in</strong>fluenc<strong>in</strong>gneighbor<strong>in</strong>g CpGs methylation state, or the density of CpGs, or are perhaps <strong>in</strong> l<strong>in</strong>kagedisequilibrium with such variants. These disparities <strong>in</strong> monoallelic impr<strong>in</strong>t<strong>in</strong>g may be viaslight allele-specific expression balance changes through to complete loss of impr<strong>in</strong>t<strong>in</strong>g.28814.17 EPIGENOMIC-WIDE ASSOCIATION STUDIES (EWAS) INHUMAN OBESITYThe potential to <strong>in</strong>vestigate the “methylome” or “chromat<strong>in</strong>ome” with the same power as thatof GWAS may enable commonly perturbed epigenomic pathways <strong>in</strong> common disease to bediscovered. As the genome is controlled by epigenetic mechanisms that <strong>in</strong>form development,but also respond and are affected by the environment, <strong>in</strong>vestigation of this “epigenome” <strong>in</strong>disease promises major <strong>in</strong>sights <strong>in</strong>to both cause and effect [24,173]. DNA methylation, be<strong>in</strong>gthe most stable of these marks, is currently the most amenable to the high-throughput analysisrequired <strong>in</strong> order to exam<strong>in</strong>e enough <strong>in</strong>dividuals to make these studies sufficiently powered toidentify replicable associations. Recent technological advances have now made it possible forthese DNA methylation epigenome-wide association studies (EWAS) to be considered[174,175]. By utiliz<strong>in</strong>g a chemical modification of the genome, bisulfite conversion, to createa pseudo-SNP at variably methylated cytos<strong>in</strong>e sites, SNP array technology has been adapted forthis task. The recent availability of the next generation of array platform (Illum<strong>in</strong>a 450k eInf<strong>in</strong>ium <strong>Human</strong>Methylation450 BeadChip performed on the iScan), supersed<strong>in</strong>g previous27-k arrays, allows greater than a magnitude <strong>in</strong>crease <strong>in</strong> coverage and importantly <strong>in</strong>cludes<strong>in</strong>vestigation of more dynamic regions of DNA methylation change, such as CpG Island shores[176].EWAS design needs to carefully consider the pr<strong>in</strong>cipal facets that differentiate the epigenomefrom the genome. First, that the epigenome is tissue-specific; second, that epigenetic marks canbe <strong>in</strong>fluenced to vary<strong>in</strong>g degrees by the underly<strong>in</strong>g genome; and third, that it can be modifiedby environmental factors. Regard<strong>in</strong>g tissue-specificity when <strong>in</strong>vestigat<strong>in</strong>g the obesity phenotype,adipose, muscle and more difficult to acquire hypothalamic tissue are <strong>in</strong>itial strongetiological candidate epigenomes. Mixture of cell type may impede signal detection, thereforepure cell isolation is a major advantage. Inflammatory processes <strong>in</strong> obesity may also make<strong>in</strong>flammatory cells an <strong>in</strong>terest<strong>in</strong>g target [177,178]. A population-based design will encountersignificant genetic heterogeneity with subsequent impact on DNA methylation. This will leadto a loss of power <strong>in</strong> comparison to a disease-discordant monozygotic tw<strong>in</strong> design analysis,although these cohorts are very difficult to resource for any, but particularly this, phenotype.Furthermore the genetically <strong>in</strong>duced variation may be more complex than can be dissectedfrom array genotype data alone. F<strong>in</strong>ally, cause and effect cannot be separated with regards toany DNA methylation changes seen. In obesity therefore any epigenetic association may be<strong>in</strong>duced by the adiposity state itself or confound<strong>in</strong>g factors related to obesity such as diet orphysical activity [104].There is obviously added complexity <strong>in</strong>terpret<strong>in</strong>g the functional significance of small changes<strong>in</strong> <strong>in</strong>dividual CpG, versus a consistent result over an entire CpG island or shore region


CHAPTER 14Epigenomic Factors <strong>in</strong> <strong>Human</strong> Obesitycompris<strong>in</strong>g a number of CpGs commonly seen <strong>in</strong> cancer methylation results. Caution will berequired <strong>in</strong> the <strong>in</strong>terpretation of EWAS and the lessons from genetic association studies need tobe remembered. Without proper correction for multiple hypothesis test<strong>in</strong>g all EWAS will be <strong>in</strong>danger of over<strong>in</strong>terpretation due to the high type 1 error [104]. This will make validation byreplication <strong>in</strong> multiple subject groups vital, as is required <strong>in</strong> step-wise GWAS.It is currently unclear whether GWAS-type population studies will be adequately powered andhighly successful <strong>in</strong> the EWAS sett<strong>in</strong>g, though the <strong>in</strong>crease <strong>in</strong> coverage of the newer arrays mayhelp to answer this question. Other study designs however may be more amenable, such as thelongitud<strong>in</strong>al. The extreme phenotype has been a powerful and successful strategy <strong>in</strong> the searchfor obesity susceptibility genetic alleles [179] and may also be useful <strong>in</strong> epiallelic hunt<strong>in</strong>g.Longitud<strong>in</strong>al studies <strong>in</strong>vestigat<strong>in</strong>g epigenomic tissue-specific changes <strong>in</strong> adipose or muscletissue, before and after dramatic physiological adjustment, such as gastric bypass operation, <strong>in</strong>severe adult morbid obesity cases, would remove genetic variability. This EWAS temporalanalysis of extreme <strong>in</strong>tervention, lead<strong>in</strong>g to dramatic reduction <strong>in</strong> BMI and glucose controlimprovement, may reveal epigenetic deviations that can be attempted to be validated <strong>in</strong> lessextreme lifestyle or weight-loss program subjects. Follow-up validation <strong>in</strong> larger sample sets byfocus<strong>in</strong>g on a smaller number of CpG, us<strong>in</strong>g for <strong>in</strong>stance quantitative methylation analysis byQiagen Pyromark Bisulphite conversion Pyrosequenc<strong>in</strong>g (Pyro Q-CpG) would facilitate rapidtarget validation. Subsequent correlation with expression <strong>in</strong> these tissues of these genes withtheir methylation changes would also be strongly supportive. Successful EWAS epialleleassociations to environmental <strong>in</strong>fluences have recently begun to be published, with a s<strong>in</strong>gleCpG, cg03636183, be<strong>in</strong>g associated and replicated with lower methylation <strong>in</strong> DNA derivedfrom peripheral blood <strong>in</strong> smokers [180].14.18 FUTURE PROSPECTSLarge amounts of genomic data are now available for common complex diseases, <strong>in</strong>clud<strong>in</strong>gobesity, therefore an <strong>in</strong>tegrative appropriate utiliz<strong>in</strong>g this gathered <strong>in</strong>formation would becomplementary and also theoretically more powerful [181]. Initial pilot work <strong>in</strong>to the <strong>in</strong>tegrationof DNA methylation data with GWAS T2D loci has shown the ability to detect DNAmethylation variation with respect to risk haplotype status [149]. This method was used toshow higher levels of methylation were present on the FTO obesity-associated risk haplotype[167]. Chromat<strong>in</strong> state <strong>in</strong>tegration recognized allelic dissimilarity <strong>in</strong> the open/closed chromat<strong>in</strong>structure <strong>in</strong> the pancreas, dependent on the T2D-associated SNP <strong>in</strong> TCF7L2 [182] andthese approaches can aid formulation of further novel functional hypotheses.289Full <strong>in</strong>tegration of genomic, epigenomic, and transcriptomic data, or an “Omni-seq”approach, will have the potential to m<strong>in</strong>e out subtle disease association variation that is notpossible by one modality only [183,184]. Furthermore, the advent of third-generationsequenc<strong>in</strong>g [185,196] will remove the necessity of chemical alteration enabl<strong>in</strong>g direct read<strong>in</strong>gof DNA modifications and will greatly improve the ability to <strong>in</strong>terpret disease significance ofDNA methylation as well as newer modifications, such as hydroxymethylation [187],formylcytos<strong>in</strong>e, and carboxylcytos<strong>in</strong>e [188]. This will also be useful <strong>in</strong> the extremely complexarea of the multitude of histone tail modifications and variants, and non-cod<strong>in</strong>g RNAs and thepossiblity of RNA epigenetic change. The ability for environmental <strong>in</strong>fluences to modulate theepigenetic state make sequence-specific epidrugs to correct these epimutations an attractiveconcept, thereby <strong>in</strong>clud<strong>in</strong>g pharmacoepigenomics <strong>in</strong> the future of personalized medic<strong>in</strong>e forthe metabolic disease [189,190].References[1] Stewart ST, Cutler DM, Rosen AB. Forecast<strong>in</strong>g the effects of obesity and smok<strong>in</strong>g on U.S. life expectancy. NEngl J Med 2009;361:2252e60.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>290[2] Olshansky SJ, Passaro DJ, Hershow RC, Layden J, Carnes BA, Brody J, et al. A potential decl<strong>in</strong>e <strong>in</strong> lifeexpectancy <strong>in</strong> the United States <strong>in</strong> the 21st century. N Engl J Med 2005;352:1138e45.[3] Friedman JM. Obesity: Causes and control of excess body fat. Nature 2009;459:340e2.[4] Duffey KJ, Popk<strong>in</strong> BM. Energy density, portion size, and eat<strong>in</strong>g occasions: contributions to <strong>in</strong>creased energy<strong>in</strong>take <strong>in</strong> the United States, 1977-2006. PLoS medic<strong>in</strong>e 2011;8:e1001050.[5] Bhargava SK, Sachdev HS, Fall CH, Osmond C, Lakshmy R, Barker DJ, et al. Relation of serial changes <strong>in</strong>childhood body-mass <strong>in</strong>dex to impaired glucose tolerance <strong>in</strong> young adulthood. N Engl J Med2004;350:865e75.[6] Gluckman PD, Hanson M, Zimmet P, Forrester T. Los<strong>in</strong>g the war aga<strong>in</strong>st obesity: the need for a developmentalperspective. Sci Trans Med 2011;3:93cm19.[7] Kopelman PG. Obesity as a medical problem. Nature 2000;404:635e43.[8] Zimmet P, Alberti KG, Shaw J. Global and societal implications of the diabetes epidemic. Nature2001;414:782e7.[9] Calle EE, Rodriguez C, Walker-Thurmond K, Thun MJ. Overweight, obesity, and mortality from cancer <strong>in</strong>a prospectively studied cohort of U.S. adults. N Engl J Med 2003;348:1625e38.[10] Ogden CL, Carroll MD, Curt<strong>in</strong> LR, McDowell MA, Tabak CJ, Flegal KM. Prevalence of overweight andobesity <strong>in</strong> the United States, 1999e2004. JAMA: the journal of the American Medical Association2006;295:1549e55.[11] Choquet H, Meyre D. Genomic <strong>in</strong>sights <strong>in</strong>to early-onset obesity. Genome medic<strong>in</strong>e 2010;2:36.[12] Blakemore AI, Froguel P. Investigation of Mendelian forms of obesity holds out the prospect of personalizedmedic<strong>in</strong>e. Ann NY Acad Sci 2010;1214:180e9.[13] O’Rahilly S. <strong>Human</strong> genetics illum<strong>in</strong>ates the paths to metabolic disease. Nature 2009;462:307e14.[14] Farooqi IS, Keogh JM, Yeo GS, Lank EJ, Cheetham T, O’Rahilly S. Cl<strong>in</strong>ical spectrum of obesity and mutations<strong>in</strong> the melanocort<strong>in</strong> 4 receptor gene. N Engl J Med 2003;348:1085e95.[15] Walley AJ, Asher JE, Froguel P. The genetic contribution to non-syndromic human obesity. Nat Rev Genet2009;10:431e42.[16] Kilpela<strong>in</strong>en TO, Zillikens MC, Stancakova A, F<strong>in</strong>ucane FM, Ried, et al. Genetic variation near IRS1 associateswith reduced adiposity and an impaired metabolic profile. Nat Genet 2011;43:753e60.[17] Walters RG, Jacquemont S, Valsesia A, de Smith AJ, Mart<strong>in</strong>et D, et al. A new highly penetrant form of obesitydue to deletions on chromosome 16p11.2. Nature 2010;463:671e5.[18] Bochukova EG, Huang N, Keogh J, Henn<strong>in</strong>g E, Purmann C, Blaszczyk K, et al. Large, rare chromosomaldeletions associated with severe early-onset obesity. Nature 2010;463:666e70.[19] Frayl<strong>in</strong>g TM, Timpson NJ, Weedon MN, Zegg<strong>in</strong>i E, Freathy RM, L<strong>in</strong>dgren CM, et al. A common variant <strong>in</strong> theFTO gene is associated with body mass <strong>in</strong>dex and predisposes to childhood and adult obesity. <strong>Science</strong>2007;316:889e94.[20] Marti A, Ordovas J. <strong>Epigenetics</strong> lights up the obesity field. Obesity facts 2011;4:187e90.[21] Nejentsev S, Walker N, Riches D, Egholm M, Todd JA. Rare variants of IFIH1, a gene implicated <strong>in</strong> antiviralresponses, protect aga<strong>in</strong>st type 1 diabetes. <strong>Science</strong> 2009;324:387e9.[22] McCarthy MI, Hirschhorn JN. Genome-wide association studies: potential next steps on a genetic journey.Hum Mol Genet 2008;17:R156e65.[23] Richards EJ. Inherited epigenetic variationerevisit<strong>in</strong>g soft <strong>in</strong>heritance. Nat Rev Genet 2006;7:395e401.[24] Bell CG, Beck S. The epigenomic <strong>in</strong>terface between genome and environment <strong>in</strong> common complex diseases.Brief<strong>in</strong>gs funct genomics 2010;9:477e85.[25] Barres R, Zierath JR. DNA methylation <strong>in</strong> metabolic disorders. Am J Cl<strong>in</strong> Nutr 2011;93:897Se900.[26] Bjornsson HT, Fall<strong>in</strong> MD, Fe<strong>in</strong>berg AP. An <strong>in</strong>tegrated epigenetic and genetic approach to common humandisease. Trends Genet 2004;20:350e8.[27] Manolio TA, Coll<strong>in</strong>s FS, Cox NJ, Goldste<strong>in</strong> DB, H<strong>in</strong>dorff LA, Hunter DJ, et al. F<strong>in</strong>d<strong>in</strong>g the miss<strong>in</strong>g heritabilityof complex diseases. Nature 2009;461:747e53.[28] Herrera BM, Keildson S, L<strong>in</strong>dgren CM. Genetics and epigenetics of obesity. Maturitas 2011;69:41e9.[29] Berger SL, Kouzarides T, Shiekhattar R, Shilatifard A. An operational def<strong>in</strong>ition of epigenetics. Genes Dev2009;23:781e3.[30] Ferguson-Smith AC. Genomic impr<strong>in</strong>t<strong>in</strong>g: the emergence of an epigenetic paradigm. Nat Rev Genet2011;12:565e75.[31] Reik W, Walter J. Genomic impr<strong>in</strong>t<strong>in</strong>g: parental <strong>in</strong>fluence on the genome. Nat Rev Genet 2001;2:21e32.[32] Edwards CA, Ferguson-Smith AC. Mechanisms regulat<strong>in</strong>g impr<strong>in</strong>ted genes <strong>in</strong> clusters. Curr Op<strong>in</strong> Cell Biol2007;19:281e9.


CHAPTER 14Epigenomic Factors <strong>in</strong> <strong>Human</strong> Obesity[33] Sleutels F, Zwart R, Barlow DP. The non-cod<strong>in</strong>g Air RNA is required for silenc<strong>in</strong>g autosomal impr<strong>in</strong>ted genes.Nature 2002;415:810e3.[34] Pont<strong>in</strong>g CP, Oliver PL, Reik W. Evolution and functions of long noncod<strong>in</strong>g RNAs. Cell 2009;136:629e41.[35] Reik W, Lewis A. Co-evolution of X-chromosome <strong>in</strong>activation and impr<strong>in</strong>t<strong>in</strong>g <strong>in</strong> mammals. Nat Rev Genet2005;6:403e10.[36] Constancia M, Kelsey G, Reik W. Resourceful impr<strong>in</strong>t<strong>in</strong>g. Nature 2004;432:53e7.[37] Wilk<strong>in</strong>son LS, Davies W, Isles AR. Genomic impr<strong>in</strong>t<strong>in</strong>g effects on bra<strong>in</strong> development and function. Nat RevNeurosci 2007;8:832e43.[38] Ferron SR, Charalambous M, Radford E, McEwen K, Wildner H, H<strong>in</strong>d E, et al. Postnatal loss of Dlk1impr<strong>in</strong>t<strong>in</strong>g <strong>in</strong> stem cells and niche astrocytes regulates neurogenesis. Nature 2011;475:381e5.[39] Gallou-Kabani C, Junien C. Nutritional epigenomics of metabolic syndrome: new perspective aga<strong>in</strong>st theepidemic. Diabetes 2005;54:1899e906.[40] Waterland RA, Garza C. Potential mechanisms of metabolic impr<strong>in</strong>t<strong>in</strong>g that lead to chronic disease. Am J Cl<strong>in</strong>Nutr 1999;69:179e97.[41] Moore T, Haig D. Genomic impr<strong>in</strong>t<strong>in</strong>g <strong>in</strong> mammalian development: a parental tug-of-war. Trends Genet1991;7:45e9.[42] Dawk<strong>in</strong>s R. The selfish gene. Oxford: Oxford University Press; 1976.[43] Wilk<strong>in</strong>s JF, Haig D. What good is genomic impr<strong>in</strong>t<strong>in</strong>g: the function of parent-specific gene expression. NatRev Genet 2003;4:359e68.[44] Berkowitz RS, Goldste<strong>in</strong> DP. Cl<strong>in</strong>ical practice. Molar pregnancy. N Engl J Med 2009;360:1639e45.[45] Sibley CP, Coan PM, Ferguson-Smith AC, Dean W, Hughes J, Smith P, et al. Placental-specific <strong>in</strong>sul<strong>in</strong>-likegrowth factor 2 (Igf2) regulates the diffusional exchange characteristics of the mouse placenta. Proc Natl AcadSci USA 2004;101:8204e8.[46] Constancia M, Hemberger M, Hughes J, Dean W, Ferguson-Smith A, Fundele R, et al. Placental-specific IGF-IIis a major modulator of placental and fetal growth. Nature 2002;417:945e8.[47] Constancia M, Angiol<strong>in</strong>i E, Sandovici I, Smith P, Smith R, Kelsey G, et al. Adaptation of nutrient supply tofetal demand <strong>in</strong> the mouse <strong>in</strong>volves <strong>in</strong>teraction between the Igf2 gene and placental transporter systems. ProcNatl Acad Sci USA 2005;102:19219e24.[48] Plagge A, Gordon E, Dean W, Boiani R, C<strong>in</strong>ti S, Peters J, et al. The impr<strong>in</strong>ted signal<strong>in</strong>g prote<strong>in</strong> XL alpha s isrequired for postnatal adaptation to feed<strong>in</strong>g. Nat Genet 2004;36:818e26.[49] Isles AR, Davies W, Wilk<strong>in</strong>son LS. Genomic impr<strong>in</strong>t<strong>in</strong>g and the social bra<strong>in</strong>. Philosophical transactions of theRoyal Society of London. B Biol Sci 2006;361:2229e37.[50] Nicholls RD, Knoll JH, Butler MG, Karam S, Lalande M. Genetic impr<strong>in</strong>t<strong>in</strong>g suggested by maternal heterodisomy<strong>in</strong> nondeletion PradereWilli syndrome. Nature 1989;342:281e5.[51] Horsthemke B, Wagstaff J. Mechanisms of impr<strong>in</strong>t<strong>in</strong>g of the PradereWilli/Angelman region. Americanjournal of medical genetics. A 2008;146A:2041e52.[52] Williams CA, Beaudet AL, Clayton-Smith J, Knoll JH, Kyllerman M, Laan LA, et al. Angelman syndrome 2005:updated consensus for diagnostic criteria. Am J Med Genet A 2006;140:413e8.[53] Butler MG. Genomic impr<strong>in</strong>t<strong>in</strong>g disorders <strong>in</strong> humans: a m<strong>in</strong>i-review. J Assist Reprod Genet 2009;26:477e86.[54] Boyle J, Hawk<strong>in</strong>s M, Barton DE, Meaney K, Guitart M, O’Grady A, et al. Establishment of the first WHO<strong>in</strong>ternational genetic reference panel for Prader Willi and Angelman syndromes. Eur J Hum Genet2011;19:857e64.[55] Stoger R. <strong>Epigenetics</strong> and obesity. Pharmacogenomics 2008;9:1851e60.[56] Bell CG, Walley AJ, Froguel P. The genetics of human obesity. Nature reviews. Genetics 2005;6:221e34.[57] Cumm<strong>in</strong>gs DE, Clement K, Purnell JQ, Vaisse C, Foster KE, Frayo RS, et al. Elevated plasma ghrel<strong>in</strong> levels <strong>in</strong>Prader Willi syndrome. Nat Med 2002;8:643e4.[58] Sahoo T, del Gaudio D, German JR, Sh<strong>in</strong>awi M, Peters SU, Person RE, et al. PradereWilli phenotype causedby paternal deficiency for the HBII-85 C/D box small nucleolar RNA cluster. Nat Genet 2008;40:719e21.[59] Goldstone AP. PradereWilli syndrome: advances <strong>in</strong> genetics, pathophysiology and treatment. TrendsEndocr<strong>in</strong>ol Metab 2004;15:12e20.[60] Williamson CM, Turner MD, Ball ST, Nott<strong>in</strong>gham WT, Glenister P, Fray M, et al. Identification of animpr<strong>in</strong>t<strong>in</strong>g control region affect<strong>in</strong>g the expression of all transcripts <strong>in</strong> the Gnas cluster. Nat Genet2006;38:350e5.[61] Chen M, Gavrilova O, Liu J, Xie T, Deng C, Nguyen AT, et al. Alternative Gnas gene products have oppositeeffects on glucose and lipid metabolism. Proc Natl Acad Sci USA 2005;102:7386e91.291


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>292[62] Bastepe M, Frohlich LF, L<strong>in</strong>glart A, Abu-Zahra HS, Tojo K, Ward LM, et al. Deletion of the NESP55 differentiallymethylated region causes loss of maternal GNAS impr<strong>in</strong>ts and pseudohypoparathyroidism type Ib.Nat Genet 2005;37:25e7.[63] Mariot V, Maupetit-Mehouas S, S<strong>in</strong>d<strong>in</strong>g C, Kottler ML, L<strong>in</strong>glart A. A maternal epimutation of GNAS leads toAlbright osteodystrophy and parathyroid hormone resistance. J Cl<strong>in</strong> Endocr<strong>in</strong>ol Metab 2008;93:661e5.[64] Spiegel AM, We<strong>in</strong>ste<strong>in</strong> LS. Inherited diseases <strong>in</strong>volv<strong>in</strong>g g prote<strong>in</strong>s and g prote<strong>in</strong>-coupled receptors. Annu RevMed 2004;55:27e39.[65] Sakamoto A, Liu J, Greene A, Chen M, We<strong>in</strong>ste<strong>in</strong> LS. Tissue-specific impr<strong>in</strong>t<strong>in</strong>g of the G prote<strong>in</strong> Gsalpha isassociated with tissue-specific differences <strong>in</strong> histone methylation. Hum Mol Genet 2004;13:819e28.[66] DeBaun MR, Niemitz EL, Fe<strong>in</strong>berg AP. Association of <strong>in</strong> vitro fertilization with BeckwitheWiedemannsyndrome and epigenetic alterations of LIT1 and H19. Am J Hum Genet 2003;72:156e60.[67] Gregg C, Zhang J, Weissbourd B, Luo S, Schroth GP, Haig D, et al. High-resolution analysis of parent-of-orig<strong>in</strong>allelic expression <strong>in</strong> the mouse bra<strong>in</strong>. <strong>Science</strong> 2010;329:643e8.[68] Morison IM, Ramsay JP, Spencer HG. A census of mammalian impr<strong>in</strong>t<strong>in</strong>g. Trends Genet 2005;21:457e65.[69] Kong A, Ste<strong>in</strong>thorsdottir V, Masson G, Thorleifsson G, Sulem P, Besenbacher S, et al. Parental orig<strong>in</strong> ofsequence variants associated with complex diseases. Nature 2009;462:868e74.[70] Luedi PP, Dietrich FS, Weidman JR, Bosko JM, Jirtle RL, Hartem<strong>in</strong>k AJ. Computational and experimentalidentification of novel human impr<strong>in</strong>ted genes. Genome Res 2007;17:1723e30.[71] Wen B, Wu H, Bjornsson H, Green RD, Irizarry R, Fe<strong>in</strong>berg AP. Overlapp<strong>in</strong>g euchromat<strong>in</strong>/heterochromat<strong>in</strong>associatedmarks are enriched <strong>in</strong> impr<strong>in</strong>ted gene regions and predict allele-specific modification. Genome Res2008;18:1806e13.[72] Choufani S, Shapiro JS, Susiarjo M, Butcher DT, Grafodatskaya D, Lou Y, et al. A novel approach identifiesnew differentially methylated regions (DMRs) associated with impr<strong>in</strong>ted genes. Genome Res2011;21:465e76.[73] Monk D, Arnaud P, Apostolidou S, Hills FA, Kelsey G, Stanier P, et al. Limited evolutionary conservation ofimpr<strong>in</strong>t<strong>in</strong>g <strong>in</strong> the human placenta. Proc Natl Acad Sci USA 2006;103:6623e8.[74] Frontera M, Dick<strong>in</strong>s B, Plagge A, Kelsey G. Impr<strong>in</strong>ted genes, postnatal adaptations and endur<strong>in</strong>g effects onenergy homeostasis. Adv Exp Med Biol 2008;626:41e61.[75] Cheverud JM, Hager R, Roseman C, Fawcett G, Wang B, Wolf JB. Genomic impr<strong>in</strong>t<strong>in</strong>g effects on adult bodycomposition <strong>in</strong> mice. Proc Natl Acad Sci USA 2008;105:4253e8.[76] Cheverud JM, Lawson HA, Fawcett GL, Wang B, Pletscher LS, A, R. F., et al. Diet-dependent genetic andgenomic impr<strong>in</strong>t<strong>in</strong>g effects on obesity <strong>in</strong> mice. Obesity 2011;19:160e70.[77] Curley JP, Barton S, Surani A, Keverne EB. Coadaptation <strong>in</strong> mother and <strong>in</strong>fant regulated by a paternallyexpressed impr<strong>in</strong>ted gene. Proc Bio Sci Royal Soc 2004;271:1303e9.[78] Lefebvre L, Viville S, Barton SC, Ish<strong>in</strong>o F, Keverne EB, Surani MA. Abnormal maternal behaviour and growthretardation associated with loss of the impr<strong>in</strong>ted gene Mest. Nat Genet 1998;20:163e9.[79] Jirtle RL, Sk<strong>in</strong>ner MK. Environmental epigenomics and disease susceptibility. Nat Rev Genet 2007;8:253e62.[80] Morgan HD, Sutherland HG, Mart<strong>in</strong> DI, Whitelaw E. Epigenetic <strong>in</strong>heritance at the agouti locus <strong>in</strong> the mouse.Nat Genet 1999;23:314e8.[81] Duhl DM, Vriel<strong>in</strong>g H, Miller KA, Wolff GL, Barsh GS. Neomorphic agouti mutations <strong>in</strong> obese yellow mice.Nat Genet 1994;8:59e65.[82] Dol<strong>in</strong>oy DC, We<strong>in</strong>house C, Jones TR, Rozek LS, Jirtle RL. Variable histone modifications at the A(vy)metastable epiallele. Epig Official J DNA Methy Soc 2010;5:637e44.[83] Wolff GL, Kodell RL, Moore SR, Cooney CA. Maternal epigenetics and methyl supplements affect agouti geneexpression <strong>in</strong> Avy/a mice. FASEB J Official Publ Fed Am Soc for Exper Bio 1998;12:949e57.[84] Waterland RA, Jirtle RL. Transposable elements: targets for early nutritional effects on epigenetic generegulation. Mol Cell Bio 2003;23:5293e300.[85] Waterland RA, L<strong>in</strong> JR, Smith CA, Jirtle RL. Post-wean<strong>in</strong>g diet affects genomic impr<strong>in</strong>t<strong>in</strong>g at the <strong>in</strong>sul<strong>in</strong>-likegrowth factor 2 (Igf2) locus. Hum Mol Genet 2006;15:705e16.[86] Koza RA, Nikonova L, Hogan J, Rim JS, Mendoza T, Faulk C, et al. Changes <strong>in</strong> gene expression foreshadowdiet-<strong>in</strong>duced obesity <strong>in</strong> genetically identical mice. PLoS genetics 2006;2:e81.[87] Plagemann A, Harder T, Brunn M, Harder A, Roepke K, Wittrock-Staar M, et al. Hypothalamic proopiomelanocort<strong>in</strong>promoter methylation becomes altered by early overfeed<strong>in</strong>g: an epigenetic model of obesity andthe metabolic syndrome. J Physiol 2009;587:4963e76.[88] Bouchard L, Rabasa-Lhoret R, Faraj M, Lavoie ME, Mill J, Perusse L, et al. Differential epigenomic andtranscriptomic responses <strong>in</strong> subcutaneous adipose tissue between low and high responders to caloricrestriction. Am J Cl<strong>in</strong> Nut 2010;91:309e20.


CHAPTER 14Epigenomic Factors <strong>in</strong> <strong>Human</strong> Obesity[89] Brons C, Jacobsen S, Nilsson E, Ronn T, Jensen CB, Storgaard H, et al. Deoxyribonucleic acid methylationand gene expression of PPARGC1A <strong>in</strong> human muscle is <strong>in</strong>fluenced by high-fat overfeed<strong>in</strong>g <strong>in</strong>a birth-weight-dependent manner. J Cl<strong>in</strong> Endocr<strong>in</strong>ol Metab 2010;95:3048e56.[90] Mozaffarian D, Hao T, Rimm EB, Willett WC, Hu FB. Changes <strong>in</strong> diet and lifestyle and long-term weight ga<strong>in</strong><strong>in</strong> women and men. N Engl J Med 2011;364:2392e404.[91] Fe<strong>in</strong>berg AP. Phenotypic plasticity and the epigenetics of human disease. Nature 2007;447:433e40.[92] Faulk C, Dol<strong>in</strong>oy DC. Tim<strong>in</strong>g is everyth<strong>in</strong>g: The when and how of environmentally <strong>in</strong>duced changes <strong>in</strong> theepigenome of animals. <strong>Epigenetics</strong> 2011;6.[93] Esau C, Kang X, Peralta E, Hanson E, Marcusson EG, Ravichandran LV, et al. MicroRNA-143 regulatesadipocyte differentiation. J Bio Chem 2004;279:52361e5.[94] Jaenisch R, Bird A. Epigenetic regulation of gene expression: how the genome <strong>in</strong>tegrates <strong>in</strong>tr<strong>in</strong>sic and environmentalsignals. Nat Genet 2003;33(Suppl):245e54.[95] Andreasen CH, Stender-Petersen KL, Mogensen MS, Torekov SS, Wegner L, Andersen G, et al. Low physicalactivity accentuates the effect of the FTO rs9939609 polymorphism on body fat accumulation. Diabetes2008;57:95e101.[96] Esteller M. Aberrant DNA methylation as a cancer-<strong>in</strong>duc<strong>in</strong>g mechanism. Annu Rev Pharmacol Toxicol2005;45:629e56.[97] Y<strong>in</strong>g AK, Hassana<strong>in</strong> HH, Roos CM, Smiraglia DJ, Issa JJ, Michler RE, et al. Methylation of the estrogenreceptor-alpha gene promoter is selectively <strong>in</strong>creased <strong>in</strong> proliferat<strong>in</strong>g human aortic smooth muscle cells.Cardio Res 2000;46:172e9.[98] Fraga MF, Ballestar E, Paz MF, Ropero S, Setien F, Ballestar ML, et al. Epigenetic differences arise dur<strong>in</strong>g thelifetime of monozygotic tw<strong>in</strong>s. Proc Natl Acad Sci USA 2005;102:10604e9.[99] Christensen BC, Houseman EA, Marsit CJ, Zheng S, Wrensch MR, Wiemels JL, et al. Ag<strong>in</strong>g and environmentalexposures alter tissue-specific DNA methylation dependent upon CpG island context. PLoS Genet2009;5:e1000602.[100] Issa JP. Epigenetic variation and human disease. J Nut 2002;132:2388Se92S.[101] Teschendorff AE, Menon U, Gentry-Maharaj A, Ramus SJ, Weisenberger DJ, Shen H, et al. Age-dependentDNA methylation of genes that are suppressed <strong>in</strong> stem cells is a hallmark of cancer. Genome Res2010;20:440e6.[102] Reik W, Romer I, Barton SC, Surani MA, Howlett SK, Klose J. Adult phenotype <strong>in</strong> the mouse can be affectedby epigenetic events <strong>in</strong> the early embryo. Develop 1993;119:933e42.[103] Hales CN, Barker DJ. The thrifty phenotype hypothesis. Br Med Bull 2001;60:5e20.[104] Franks PW, L<strong>in</strong>g C. <strong>Epigenetics</strong> and obesity: the devil is <strong>in</strong> the details. BMC Med 2010;8:88.[105] McMillen IC, Rob<strong>in</strong>son JS. Developmental orig<strong>in</strong>s of the metabolic syndrome: prediction, plasticity, andprogramm<strong>in</strong>g. Physiol Rev 2005;85:571e633.[106] Sandovici I, Smith NH, Nitert MD, Ackers-Johnson M, Uribe-Lewis S, Ito Y, et al. Maternal diet and ag<strong>in</strong>g alterthe epigenetic control of a promoter-enhancer <strong>in</strong>teraction at the Hnf4a gene <strong>in</strong> rat pancreatic islets. Proc NatlAcad Sci USA 2011;108:5449e54.[107] E<strong>in</strong>ste<strong>in</strong> F, Thompson RF, Bhagat TD, Fazzari MJ, Verma A, Barzilai N, et al. Cytos<strong>in</strong>e methylation dysregulation<strong>in</strong> neonates follow<strong>in</strong>g <strong>in</strong>trauter<strong>in</strong>e growth restriction. PLoS One 2010;5:e8887.[108] Thompson RF, Fazzari MJ, Niu H, Barzilai N, Simmons RA, Greally JM. Experimental <strong>in</strong>trauter<strong>in</strong>e growthrestriction <strong>in</strong>duces alterations <strong>in</strong> DNA methylation and gene expression <strong>in</strong> pancreatic islets of rats. J BiolChem 2010;285:15111e8.[109] Barker DJ, Eriksson JG, Forsen T, Osmond C. Fetal orig<strong>in</strong>s of adult disease: strength of effects and biologicalbasis. Int J Epidemiol 2002;31:1235e9.[110] Gluckman PD, Hanson MA, Cooper C, Thornburg KL. Effect of <strong>in</strong> utero and early-life conditions on adulthealth and disease. N Engl J Med 2008;359:61e73.[111] Godfrey KM, Gluckman PD, Hanson MA. Developmental orig<strong>in</strong>s of metabolic disease: life course and<strong>in</strong>tergenerational perspectives. Trends Endocr<strong>in</strong>ol Metab 2010;21:199e205.[112] Barker DJ, Osmond C. Infant mortality, childhood nutrition, and ischaemic heart disease <strong>in</strong> England andWales. Lancet 1986;1:1077e81.[113] Hales CN, Barker DJ, Clark PM, Cox LJ, Fall C, Osmond C, et al. Fetal and <strong>in</strong>fant growth and impaired glucosetolerance at age 64. BMJ 1991;303:1019e22.[114] Simmons R. Developmental orig<strong>in</strong>s of adult metabolic disease: concepts and controversies. Trends <strong>in</strong>endocr<strong>in</strong>ology and metabolism: TEM 2005;16:390e4.[115] Ravelli GP, Ste<strong>in</strong> ZA, Susser MW. Obesity <strong>in</strong> young men after fam<strong>in</strong>e exposure <strong>in</strong> utero and early <strong>in</strong>fancy.N Engl J Med 1976;295:349e53.293


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>294[116] Heijmans BT, Tobi EW, Ste<strong>in</strong> AD, Putter H, Blauw GJ, Susser ES, et al. Persistent epigenetic differencesassociated with prenatal exposure to fam<strong>in</strong>e <strong>in</strong> humans. Proc Natl Acad Sci USA 2008;105:17046e9.[117] Tobi EW, Lumey LH, Talens RP, Kremer D, Putter H, Ste<strong>in</strong> AD, et al. DNA methylation differences afterexposure to prenatal fam<strong>in</strong>e are common and tim<strong>in</strong>g- and sex-specific. Hum Mol Genet 2009;18:4046e53.[118] Reik W. Stability and flexibility of epigenetic gene regulation <strong>in</strong> mammalian development. Nature2007;447:425e32.[119] S<strong>in</strong>clair KD, Allegrucci C, S<strong>in</strong>gh R, Gardner DS, Sebastian S, Bispham J, et al. DNA methylation, <strong>in</strong>sul<strong>in</strong>resistance, and blood pressure <strong>in</strong> offspr<strong>in</strong>g determ<strong>in</strong>ed by maternal periconceptional B vitam<strong>in</strong> and methion<strong>in</strong>estatus. Proc Natl Acad Sci USA 2007;104:19351e6.[120] Whitaker KL, Jarvis MJ, Beeken RJ, Boniface D, Wardle J. Compar<strong>in</strong>g maternal and paternal <strong>in</strong>tergenerationaltransmission of obesity risk <strong>in</strong> a large population-based sample. Am J Cl<strong>in</strong> Nutr 2010;91:1560e7.[121] Radford EJ, Ferron SR, Ferguson-Smith AC. Genomic impr<strong>in</strong>t<strong>in</strong>g as an adaptative model of developmentalplasticity. FEBS letters 2011;585:2059e66.[122] Gluckman PD, Lillycrop KA, Vickers MH, Pleasants AB, Phillips ES, Beedle AS, et al. Metabolic plasticitydur<strong>in</strong>g mammalian development is directionally dependent on early nutritional status. Proc Natl Acad SciUSA 2007;104:12796e800.[123] Gluckman PD, Hanson MA. Developmental and epigenetic pathways to obesity: an evolutionarydevelopmentalperspective. International journal of obesity 2008;32(Suppl. 7):S62e71.[124] Broad KD, Keverne EB. Placental protection of the fetal bra<strong>in</strong> dur<strong>in</strong>g short-term food deprivation. Proc NatlAcad Sci USA 2011;108:15237e41.[125] Ibanez L, Suarez L, Lopez-Bermejo A, Diaz M, Valls C, de Zegher F. Early development of visceral fat excessafter spontaneous catch-up growth <strong>in</strong> children with low birth weight. J Cl<strong>in</strong> Endocr<strong>in</strong>ol Metab2008;93:925e8.[126] Jaquet D, Gaboriau A, Czernichow P, Levy-Marchal C. Insul<strong>in</strong> resistance early <strong>in</strong> adulthood <strong>in</strong> subjects bornwith <strong>in</strong>trauter<strong>in</strong>e growth retardation. J Cl<strong>in</strong> Endocr<strong>in</strong>ol Metab 2000;85:1401e6.[127] Hattersley AT, Tooke JE. The fetal <strong>in</strong>sul<strong>in</strong> hypothesis: an alternative explanation of the association of lowbirthweight with diabetes and vascular disease. Lancet 1999;353:1789e92.[128] Barker DJ. The developmental orig<strong>in</strong>s of adult disease. J Am Coll Nutr 2004;23:588Se95S.[129] Vickers MH, Breier BH, Cutfield WS, Hofman PL, Gluckman PD. Fetal orig<strong>in</strong>s of hyperphagia, obesity, andhypertension and postnatal amplification by hypercaloric nutrition. Am J Physiol Endocr<strong>in</strong>al Metab2000;279:E83e7.[130] Ozanne SE, Hales CN. Lifespan: catch-up growth and obesity <strong>in</strong> male mice. Nature 2004;427:411e2.[131] Martorell R, Ste<strong>in</strong> AD, Schroeder DG. Early nutrition and later adiposity. J Nutr 2001;131:874Se80S.[132] Gluckman PD, Hanson MA, Spencer HG. Predictive adaptive responses and human evolution. Trends EcoEvol 2005;20:527e33.[133] Bateson P, Barker D, Clutton-Brock T, Deb D, D’Ud<strong>in</strong>e B, Foley RA, et al. Developmental plasticity andhuman health. Nature 2004;430:419e21.[134] Ikenasio-Thorpe BA, Breier BH, Vickers MH, Fraser M. Prenatal <strong>in</strong>fluences on susceptibility to diet-<strong>in</strong>ducedobesity are mediated by altered neuroendocr<strong>in</strong>e gene expression. J Endocr<strong>in</strong>ol 2007;193:31e7.[135] Godfrey KM, Sheppard A, Gluckman PD, Lillycrop KA, Burdge GC, McLean C, et al. Epigenetic GenePromoter Methylation at Birth Is Associated With Child’s Later Adiposity. Diabetes 2011;60:1528e34.[136] Hillier TA, Pedula KL, Schmidt MM, Mullen JA, Charles MA, Pettitt DJ. Childhood obesity and metabolicimpr<strong>in</strong>t<strong>in</strong>g: the ongo<strong>in</strong>g effects of maternal hyperglycemia. Diabetes care 2007;30:2287e92.[137] Clausen TD, Mathiesen ER, Hansen T, Pedersen O, Jensen DM, Lauenborg J, et al. Overweight and themetabolic syndrome <strong>in</strong> adult offspr<strong>in</strong>g of women with diet-treated gestational diabetes mellitus or type 1diabetes. J Cl<strong>in</strong> Endocr<strong>in</strong>ol Metab 2009;94:2464e70.[138] Muhlhausler BS. Programm<strong>in</strong>g of the appetite-regulat<strong>in</strong>g neural network: a l<strong>in</strong>k between maternal overnutritionand the programm<strong>in</strong>g of obesity? J neuroendocr<strong>in</strong>ol 2007;19:67e72.[139] Kuzawa CW. Adipose tissue <strong>in</strong> human <strong>in</strong>fancy and childhood: an evolutionary perspective. Am J PhysAnthropol 1998;(Suppl 27):177e209.[140] Ley RE, Turnbaugh PJ, Kle<strong>in</strong> S, Gordon JI. Microbial ecology: human gut microbes associated with obesity.Nature 2006;444:1022e3.[141] Davey Smith G, Steer C, Leary S, Ness A. Is there an <strong>in</strong>trauter<strong>in</strong>e <strong>in</strong>fluence on obesity? Evidence from parentchild associations <strong>in</strong> the Avon Longitud<strong>in</strong>al Study of Parents and Children (ALSPAC). Arch Dis Child2007;92:876e80.[142] Melzner I, Scott V, Dorsch K, Fischer P, Wabitsch M, Bruderle<strong>in</strong> S, et al. Lept<strong>in</strong> gene expression <strong>in</strong> humanpreadipocytes is switched on by maturation-<strong>in</strong>duced demethylation of dist<strong>in</strong>ct CpGs <strong>in</strong> its proximalpromoter. J Bio Chem 2002;277:45420e7.


CHAPTER 14Epigenomic Factors <strong>in</strong> <strong>Human</strong> Obesity[143] P<strong>in</strong>to S, Roseberry AG, Liu H, Diano S, Shanabrough M, Cai X, et al. Rapid rewir<strong>in</strong>g of arcuate nucleusfeed<strong>in</strong>g circuits by lept<strong>in</strong>. <strong>Science</strong> 2004;304:110e5.[144] Bouret SG, Simerly RB. M<strong>in</strong>ireview: Lept<strong>in</strong> and development of hypothalamic feed<strong>in</strong>g circuits. Endocr<strong>in</strong>ol2004;145:2621e6.[145] Bouret SG, Draper SJ, Simerly RB. Trophic action of lept<strong>in</strong> on hypothalamic neurons that regulate feed<strong>in</strong>g.<strong>Science</strong> 2004;304:108e10.[146] Milagro FI, Campion J, Garcia-Diaz DF, Goyenechea E, Paterna<strong>in</strong> L, Mart<strong>in</strong>ez JA. High fat diet-<strong>in</strong>ducedobesity modifies the methylation pattern of lept<strong>in</strong> promoter <strong>in</strong> rats. J Phys Biochem 2009;65:1e9.[147] Rosen ED, Hsu CH, Wang X, Sakai S, Freeman MW, Gonzalez FJ, et al. C/EBPalpha <strong>in</strong>duces adipogenesisthrough PPARgamma: a unified pathway. Genes Dev 2002;16:22e6.[148] Stoger R. In vivo methylation patterns of the lept<strong>in</strong> promoter <strong>in</strong> human and mouse. <strong>Epigenetics</strong> 2006;1:155e62.[149] Bell CG, F<strong>in</strong>er S, L<strong>in</strong>dgren CM, Wilson GA, Rakyan VK, Teschendorff AE, et al. Integrated Genetic andEpigenetic Analysis Identifies Haplotype-Specific Methylation <strong>in</strong> the FTO Type 2 Diabetes and ObesitySusceptibility Locus. PLoS One 2010;5:e14040.[150] Marchi M, Lisi S, Curcio M, Barbuti S, Piaggi P, Ceccar<strong>in</strong>i G, et al. <strong>Human</strong> lept<strong>in</strong> tissue distribution, but notweight loss-dependent change <strong>in</strong> expression, is associated with methylation of its promoter. Epi official JDNA Methy Soc 2011;6.[151] Inagaki T, Tachibana M, Magoori K, Kudo H, Tanaka T, Okamura M, et al. Obesity and metabolic syndrome <strong>in</strong>histone demethylase JHDM2a-deficient mice. Genes cells 2009;14:991e1001.[152] Tateishi K, Okada Y, Kall<strong>in</strong> EM, Zhang Y. Role of Jhdm2a <strong>in</strong> regulat<strong>in</strong>g metabolic gene expression and obesityresistance. Nature 2009;458:757e61.[153] Schalkwyk LC, Meaburn EL, Smith R, Dempster EL, Jeffries AR, Davies MN, et al. Allelic skew<strong>in</strong>g of DNAmethylation is widespread across the genome. Am J Hum Genet 2010;86:196e212.[154] Kerkel K, Spadola A, Yuan E, Kosek J, Jiang L, Hod E, et al. Genomic surveys by methylation-sensitive SNPanalysis identify sequence-dependent allele-specific DNA methylation. Nat Genet 2008;40:904e8.[155] Shoemaker R, Deng J, Wang W, Zhang K. Allele-specific methylation is prevalent and is contributed byCpG-SNPs <strong>in</strong> the human genome. Genome Res 2010;20:883e9.[156] Gertz J, Varley KE, Reddy TE, Bowl<strong>in</strong>g KM, Pauli F, Parker SL, et al. Analysis of DNA Methylation <strong>in</strong>a Three-Generation Family Reveals Widespread Genetic Influence on Epigenetic Regulation. PLoS Genet2011;7:e1002228.[157] Fe<strong>in</strong>berg AP, Irizarry RA. Evolution <strong>in</strong> health and medic<strong>in</strong>e Sackler colloquium: Stochastic epigenetic variationas a driv<strong>in</strong>g force of development, evolutionary adaptation, and disease. Proc Natl Acad Sci USA2010;107(Suppl. 1):1757e64.[158] Danch<strong>in</strong> E, Charmantier A, Champagne FA, Mesoudi A, Pujol B, Blanchet S. Beyond DNA: <strong>in</strong>tegrat<strong>in</strong>g<strong>in</strong>clusive <strong>in</strong>heritance <strong>in</strong>to an extended theory of evolution. Nature reviews. Genetics 2011;12:475e86.[159] Carone BR, Fauquier L, Habib N, Shea JM, Hart CE, Li R, et al. Paternally <strong>in</strong>duced transgenerational environmentalreprogramm<strong>in</strong>g of metabolic gene expression <strong>in</strong> mammals. Cell 2010;143:1084e96.[160] Dax<strong>in</strong>ger L, Whitelaw E. Transgenerational epigenetic <strong>in</strong>heritance: more questions than answers. Genome Res2010;20:1623e8.[161] Ng SF, L<strong>in</strong> RC, Laybutt DR, Barres R, Owens JA, Morris MJ. Chronic high-fat diet <strong>in</strong> fathers programs beta-celldysfunction <strong>in</strong> female rat offspr<strong>in</strong>g. Nature 2010;467:963e6.[162] Waterland RA, Kellermayer R, Laritsky E, Rayco-Solon P, Harris RA, Travisano M, et al. Season of Conception <strong>in</strong>Rural Gambia Affects DNA Methylation at Putative <strong>Human</strong> Metastable Epialleles. PLoS Genet 2010;6:e1001252.[163] Dol<strong>in</strong>oy DC, Das R, Weidman JR, Jirtle RL. Metastable epialleles, impr<strong>in</strong>t<strong>in</strong>g, and the fetal orig<strong>in</strong>s of adultdiseases. Pediatric Res 2007;61:30Re7R.[164] Fe<strong>in</strong>berg AP, Irizarry RA, Frad<strong>in</strong> D, Aryee MJ, Murakami P, Aspelund T, et al. Personalized epigenomicsignatures that are stable over time and covary with body mass <strong>in</strong>dex. Sci Transl Med 2010;2:49ra67.[165] Nair S, Lee YH, Rousseau E, Cam M, Tataranni PA, Baier LJ, et al. Increased expression of <strong>in</strong>flammationrelatedgenes <strong>in</strong> cultured preadipocytes/stromal vascular cells from obese compared with non-obese PimaIndians. Diabetologia 2005;48:1784e8.[166] Kaun KR, Sokolowski MB. cGMP-dependent prote<strong>in</strong> k<strong>in</strong>ase: l<strong>in</strong>k<strong>in</strong>g forag<strong>in</strong>g to energy homeostasis.Genome 2009;52:1e7.[167] Bell CG. Integration of genomic and epigenomic DNA methylation data <strong>in</strong> common complex diseases byhaplotype-specific methylation analysis. Personalized Medic<strong>in</strong>e 2011;8:243.[168] Dong C, Li WD, Geller F, Lei L, Li D, Gorlova OY, et al. Possible genomic impr<strong>in</strong>t<strong>in</strong>g of three human obesityrelatedgenetic loci. Am J Hum Genet 2005;76:427e37.295


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>296[169] H<strong>in</strong>dorff LA, Sethupathy P, Junk<strong>in</strong>s HA, Ramos EM, Mehta JP, Coll<strong>in</strong>s FS, et al. Potential etiologic andfunctional implications of genome-wide association loci for human diseases and traits. Proc Natl Acad SciUSA 2009;106:9362e7.[170] Wallace C, Smyth DJ, Maisuria-Armer M, Walker NM, Todd JA, Clayton DG. The impr<strong>in</strong>ted DLK1-MEG3 generegion on chromosome 14q32.2 alters susceptibility to type 1 diabetes. Nat Genet 2010;42:68e71.[171] Small KS, Hedman AK, Grundberg E, Nica AC, Thorleifsson G, Kong A, et al. Identification of an impr<strong>in</strong>tedmaster trans regulator at the KLF14 locus related to multiple metabolic phenotypes. Nat Genet2011;43:561e4.[172] Heijmans BT, Kremer D, Tobi EW, Boomsma DI, Slagboom PE. Heritable rather than age-related environmentaland stochastic factors dom<strong>in</strong>ate variation <strong>in</strong> DNA methylation of the human IGF2/H19 locus. HumMol Genet 2007;16:547e54.[173] Relton CL, Davey Smith G. Epigenetic epidemiology of common complex disease: prospects for prediction,prevention, and treatment. PLoS Med 2010;7:e1000356.[174] Bell CG, Teschendorff AE, Rakyan VK, Maxwell AP, Beck S, Savage DA. Genome-wide DNA methylationanalysis for diabetic nephropathy <strong>in</strong> type 1 diabetes mellitus. BMC Med Genomics 2010;3:33.[175] Rakyan VK, Down TA, Bald<strong>in</strong>g DJ, Beck S. Epigenome-wide association studies for common human diseases.Nat Rev Genet 2011;12:529e41.[176] Irizarry RA, Ladd-Acosta C, Wen B, Wu Z, Montano C, Onyango P, et al. The human colon cancer methylomeshows similar hypo- and hypermethylation at conserved tissue-specific CpG island shores. Nat Genet2009;41:178e86.[177] Vandanmagsar B, Youm YH, Ravuss<strong>in</strong> A, Galgani JE, Stadler K, Mynatt RL, et al. The NLRP3 <strong>in</strong>flammasome<strong>in</strong>stigates obesity-<strong>in</strong>duced <strong>in</strong>flammation and <strong>in</strong>sul<strong>in</strong> resistance. Nat Med 2011.[178] Pietila<strong>in</strong>en KH, Rog T, Seppanen-Laakso T, Virtue S, Gopalacharyulu P, Tang J, et al. Association oflipidome remodel<strong>in</strong>g <strong>in</strong> the adipocyte membrane with acquired obesity <strong>in</strong> humans. PLoS biology 2011;9: e1000623.[179] Froguel P, Blakemore AI. The power of the extreme <strong>in</strong> elucidat<strong>in</strong>g obesity. N Engl J Med 2008;359:891e3.[180] Breitl<strong>in</strong>g LP, Yang R, Korn B, Burw<strong>in</strong>kel B, Brenner H. Tobacco-smok<strong>in</strong>g-related differential DNA methylation:27K discovery and replication. Am J Hum Genet 2011;88:450e7.[181] Birney E. Chromat<strong>in</strong> and heritability: how epigenetic studies can complement genetic approaches. TrendsGenet 2011;27:172e6.[182] Gaulton KJ, Nammo T, Pasquali L, Simon JM, Giresi PG, Fogarty MP, et al. A map of open chromat<strong>in</strong> <strong>in</strong>human pancreatic islets. Nat Genet 2010;42:255e9.[183] Bell CG, Beck S. Advances <strong>in</strong> the identification and analysis of allele-specific expression. Genome Med 2009;1:56.[184] Cedar H, Bergman Y. L<strong>in</strong>k<strong>in</strong>g DNA methylation and histone modification: patterns and paradigms. Nat RevGenet 2009;10:295e304.[185] Flusberg BA, Webster DR, Lee JH, Travers KJ, Olivares EC, Clark TA, et al. Direct detection of DNA methylationdur<strong>in</strong>g s<strong>in</strong>gle-molecule, real-time sequenc<strong>in</strong>g. Nat Methods 2010;7:461e5.[186] Clarke J, Wu HC, Jayas<strong>in</strong>ghe L, Patel A, Reid S, Bayley H. Cont<strong>in</strong>uous base identification for s<strong>in</strong>gle-moleculenanopore DNA sequenc<strong>in</strong>g. Nat Nanotechnol 2009;4:265e70.[187] Kriaucionis S, He<strong>in</strong>tz N. The nuclear DNA base 5-hydroxymethylcytos<strong>in</strong>e is present <strong>in</strong> Purk<strong>in</strong>je neurons andthe bra<strong>in</strong>. <strong>Science</strong> 2009;324:929e30.[188] Ito S, Shen L, Dai Q, Wu SC, Coll<strong>in</strong>s LB, Swenberg JA, et al. Tet Prote<strong>in</strong>s Can Convert 5-Methylcytos<strong>in</strong>e to5-Formylcytos<strong>in</strong>e and 5-Carboxylcytos<strong>in</strong>e. <strong>Science</strong> 2011.[189] Ingelman-Sundberg M, Gomez A. The past, present and future of pharmacoepigenomics. Pharmacogenomics2010;11:625e7.[190] Egger G, Liang G, Aparicio A, Jones PA. <strong>Epigenetics</strong> <strong>in</strong> human disease and prospects for epigenetic therapy.Nature 2004;429:457e63.[191] Delrue MA, Michaud JL. Fat chance: genetic syndromes with obesity. Cl<strong>in</strong> Genet 2004;66:83e93.[192] Dol<strong>in</strong>oy DC, Weidman JR, Waterland RA, Jirtle RL. Maternal geniste<strong>in</strong> alters coat color and protects Avymouse offspr<strong>in</strong>g from obesity by modify<strong>in</strong>g the fetal epigenome. Environ Health perspect 2006;114:567e72.


CHAPTER 15Epigenetic Approaches toControl ObesityAbigail S. Lapham 1 , Karen A. Lillycrop 1 , Graham C. Burdge 1 , Peter D. Gluckman 2 ,Mark A. Hanson 1 , Keith M. Godfrey 1,3,41 University of Southampton, Southampton, UK2 University of Auckland, Auckland, New Zealand3 MRC Lifecourse Epidemiology Unit, Southampton, UK4 Southampton NIHR Nutrition Biomedical Research Centre, Southampton, UKCHAPTER OUTLINE15.1 The Chang<strong>in</strong>g Epidemiology ofObesity 29715.2 Developmental Orig<strong>in</strong>s ofObesity 29815.3 Animal Studies of EarlyDevelopment and MetabolicProgramm<strong>in</strong>g 30315.3.1 The Maternal Prote<strong>in</strong>-Restricted Diet 30315.3.2 Global DietaryRestriction 30415.3.3 High-Fat Diet Dur<strong>in</strong>gPregnancy 30415.4 Developmental Plasticity 30515.5 <strong>Epigenetics</strong> and DevelopmentalProgramm<strong>in</strong>g by the Early LifeEnvironment 30615.6 <strong>Epigenetics</strong> and Early-LifeNutrition 30715.6.1 TransgenerationalEffects 30915.6.2 Mechanism of EpigeneticChange 31015.6.3 Animal Models of MaternalNutrition and EpigeneticAlterations 31015.6.4 <strong>Human</strong> Studies of MaternalNutrition and EpigeneticAlterations 31215.7 Identification of PredictiveEpigenetic Markers of FutureObesity 31215.8 Conclusions 314References 31429715.1 THE CHANGING EPIDEMIOLOGY OF OBESITYIn the developed world, obesity, diabetes, cardiovascular disease, and non-alcoholic fatty liverdisease (NAFLD) are all <strong>in</strong>creas<strong>in</strong>g at alarm<strong>in</strong>g rates. These non-communicable diseases(NCDs) now account for 60% of deaths globally [1]. It is predicted that by 2030 there will be2.16 billion overweight and 1.12 billion obese adults worldwide [2]. Obesity <strong>in</strong> childhood isof particular concern, with recent estimates that as many as 10% of school-aged children areeither overweight or obese, although the prevalence is higher <strong>in</strong> economically developedregions [3]. A recent statement released by the World Watch Institute revealed that for the firsttime <strong>in</strong> human history the number of overweight people rivals the number of underweight [4].T. Tollefsbol (Ed): <strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>. DOI: 10.1016/B978-0-12-388415-2.00015-9Copyright Ó 2012 Elsevier Inc. All rights reserved.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>They found that while the world’s underfed population has decl<strong>in</strong>ed slightly s<strong>in</strong>ce 1980 to1.1 billion, the number of overweight has surged to <strong>in</strong> excess of this figure.In the develop<strong>in</strong>g world, obesity is also <strong>in</strong>creas<strong>in</strong>gly becom<strong>in</strong>g as significant a problem asunderfeed<strong>in</strong>g. The number of overweight people <strong>in</strong> Ch<strong>in</strong>a has risen from less than 10% toover 15% <strong>in</strong> a period of 3 years. In Brazil and Colombia the numbers of overweight <strong>in</strong>dividualsare comparable to those seen <strong>in</strong> a number of European countries, at around 40% ofadults. Even <strong>in</strong> sub-Saharan Africa, a region home to the largest proportion of the world’shungry, an <strong>in</strong>crease <strong>in</strong> obesity has been observed. This <strong>in</strong>crease has been most marked <strong>in</strong>women liv<strong>in</strong>g <strong>in</strong> urban areas [5].The large and <strong>in</strong>creas<strong>in</strong>g numbers of overweight and obese people presents a huge cl<strong>in</strong>ical andpublic health burden. For example, <strong>in</strong> the UK alone, annual direct costs are estimated to be£4.2 billion and Foresight (an <strong>in</strong>-depth study carried out by the Department of Bus<strong>in</strong>ess andInnovation Skills), which is a UK m<strong>in</strong>isterial department, have predicted that this will morethan double by 2050 if we cont<strong>in</strong>ue as we are. There are also costs to society and the economymore broadly e for example, sickness absence reduces productivity. Foresight estimated thatweight problems already cost the wider UK economy <strong>in</strong> the region of £16 billion, and that thiswill rise to £50 billion per year by 2050 if left unchecked [6].298The number of overweight children is <strong>in</strong>creas<strong>in</strong>g so rapidly that there is an urgent need toidentify risk factors for obesity <strong>in</strong> order to prevent further <strong>in</strong>creases and to identify possible<strong>in</strong>tervention strategies. Apart from the likelihood that these children will rema<strong>in</strong> overweightthroughout adolescence and their entire adult life, the consequences of childhood obesity arenow beg<strong>in</strong>n<strong>in</strong>g to be fully understood. Be<strong>in</strong>g overweight has a negative effect on the psychologicalwellbe<strong>in</strong>g of the child and studies have shown that overweight children have a lowerhealth-related quality of life [7], as well as poorer educational and social outcomes as comparedto children of normal weight [8]. Direct health consequences of be<strong>in</strong>g an overweight child<strong>in</strong>clude an <strong>in</strong>creased risk of type 2 diabetes, which is now be<strong>in</strong>g seen <strong>in</strong> adolescents due to thepediatric obesity epidemic [9]. Studies have also l<strong>in</strong>ked be<strong>in</strong>g overweight <strong>in</strong> childhood with<strong>in</strong>creased risk of impaired glucose tolerance and cardiovascular disease <strong>in</strong> later life [10].Although it is well established that the risk of an <strong>in</strong>dividual develop<strong>in</strong>g obesity is dependentupon the <strong>in</strong>teraction between their genotype and lifestyle factors such as an energy-rich dietand sedentary behavior, it is becom<strong>in</strong>g clear that these are not the sole causes of the obesityepidemic. Whilst there is a genetic component related to the ways that genes can favor fataccumulation <strong>in</strong> a given environment (Table 15.1 shows a list of 54 genes associated withobesity phenotypes), there is now substantial evidence that the fetal and early postnatalenvironment strongly <strong>in</strong>fluences the risk of develop<strong>in</strong>g obesity and that altered epigeneticregulation is central to this process.15.2 DEVELOPMENTAL ORIGINS OF OBESITYThe association between the quality of the early life environment and subsequent risk ofcardio-metabolic disease has been described <strong>in</strong> a series of epidemiological studies. Theseshowed a strong geographical relationship between <strong>in</strong>fant mortality and risk of cardiovasculardisease (CVD) disease 50e60 years later [11]. Subsequent retrospective studies <strong>in</strong> cohorts <strong>in</strong>developed and develop<strong>in</strong>g nations <strong>in</strong>clud<strong>in</strong>g the UK, North America, India, and Ch<strong>in</strong>a haveshown consistently that lower birth weight with<strong>in</strong> the normal range is associated with an<strong>in</strong>creased risk <strong>in</strong> later life of CVD and the metabolic syndrome (hypertension, <strong>in</strong>sul<strong>in</strong> resistance,type 2 diabetes, dyslipidemia, and obesity).The Dutch Hunger W<strong>in</strong>ter provides an example of how the tim<strong>in</strong>g of nutritional constra<strong>in</strong>tdur<strong>in</strong>g pregnancy is important <strong>in</strong> determ<strong>in</strong><strong>in</strong>g the future risk of disease. This short-termfam<strong>in</strong>e dur<strong>in</strong>g the w<strong>in</strong>ter of 1944e1945 resulted <strong>in</strong> 18 000 deaths with adult rations <strong>in</strong> cities


TABLE 15.1 The 54 Loci Associated with Anthropomorphic Obesity PhenotypesClosest Gene(s)ChromosomalLocationPhenotypeAssociatedLead SNP(s)Proposed Molecular or CellularFunctionAdditionalPhenotypesTBX15eWARS2 1p12 WHR rs984222 Transcription factor <strong>in</strong>volved <strong>in</strong> adipocyteand specific adipose depot developmentImplicated <strong>in</strong> Cous<strong>in</strong>syndromePTBP2 1p21.3 BMI rs1555543 eNEGR1 1p31 BMI rs2815752, Neuronal outgrowthrs3101336,rs2568958TNNI3K 1p31.1 BMI rs1514175 eDNM3ePIGC 1q24.3 WHR rs1011731 Dom<strong>in</strong>ant, negative mutations <strong>in</strong> DNM enzymes promote GLUT6 andGLUT8 transporters to adipocyte cell surface <strong>in</strong> rats.SEC16B, RASAL2 1q25 BMI rs10913469 eLYPLAL1; ZC3H11B 1q41 WHR rs2605100 Encodes prote<strong>in</strong> thought to act as triglyceride lipase and isupregulated <strong>in</strong> subcutaneous adipose tissue <strong>in</strong> obese patientsSDCCAG8 1q43eq44 BMI rs12145833 eFANCL 2p16.1 BMI rs887912 eRBJeADCY3ePOMC 2p23.3 BMI rs713586 e Rare POMC mutationscause human obesityTMEM18 2p25 BMI rs6548238, Neural developmentAssociated with T2Drs2867125,rs4854344,rs7561317,rs11127485ZNRF3eKREMEN1 2q12.1 WHR rs4823006 e Kremen1 prote<strong>in</strong>forms a complex withLDL receptor-relatedprote<strong>in</strong> 6LRP1B 2q22.2 BMI rs2890652 e LRP1B deletions seen<strong>in</strong> several types ofhuman cancersGRB14 2q24.3 WHR rs10195252 e Associated withtriglyceride and <strong>in</strong>sul<strong>in</strong>levels. GRB14-deficient mice exhibit<strong>in</strong>creased bodyADAMTS9 3p14.1 WHR rs6795735 Important for spatial distribution of cells <strong>in</strong> Associated with T2Dembryonic developmentNISCHeSTAB1 3p21.1 WHR rs6784615 Interacts with <strong>in</strong>sul<strong>in</strong> receptor substrateCADM2 3p21.1 BMI rs13078807 eETV5 (locus with threegenes, strongestassociation <strong>in</strong> ETV5)3q27 BMI rs7647305 eCont<strong>in</strong>uedCHAPTER 15Epigenetic Approaches to Control Obesity299


300TABLE 15.1Closest Gene(s)The 54 Loci Associated with Anthropomorphic Obesity Phenotypesdcont<strong>in</strong>uedChromosomalLocationPhenotypeAssociatedLead SNP(s)Proposed Molecular or CellularFunctionAdditionalPhenotypesGene desert; GNPDA2 4p13 BMI rs10938397 e Associated with T2Dis one of three genesnearbySLC39A8 4q24 BMI rs13107325 eFLJ35779 5q13.3 BMI rs2112347 eZNF608 5q23.2 BMI rs4836133 eCPEB4 5q35.2 WHR rs6861681 Regulates polyadenylation elongationTFAP2B 6p12 WC, BMI rs987237 eLocus conta<strong>in</strong><strong>in</strong>g 6p21 BMI rs2844479, eNCR3, AIF1 and BAT2rs2260000,rs1077393VEGFA 6p21.1 WHR rs6905288 Involved <strong>in</strong> vascular development. Keymediator of adipogenesisNUDT3eHMGA1 6p21.31 BMI rs206936 ePRL 6p22.2ep21.3 BMI rs4712652 eLY86 6p25.1 WHR rs1294421 Plays a role <strong>in</strong> recognition oflipopolysaccharideRSPOS 6q22.33 WHR rs9491696 Promotes angiogenesis and vasculardevelopmentNFE2L3 7p15.2 WHR rs1055144 eMSRA 8p23.1 WC, BMI rs7826222, ers17150703LRRN6C 9p21.3 BMI rs10968576 ePTER 10p12 BMI rs10508503 eMTCH2 (locus with 11p11.2 BMI rs10838738 Cellular apoptosis14 genes)BDNF (locus with fourgenes, strongestassociation nearBDNF)11p14 BMI rs4074134,rs4923461,rs925946,rs10501087,BDNF expression is regulated bynutritional state and MC4R signall<strong>in</strong>gAssociated withweight, not BMIVEGFA variantsnom<strong>in</strong>ally associatedwith T2DAssociated withasthmaOncogene <strong>in</strong> mousemammary epithelialcellsAssociated with T2D.Individuals with WAGRsyndrome with BDNFdeletion have BMI>95thrs6265RPL27A 11p15.4 BMI rs4929949 eITPR2eSSPN 12p21.1 WHR rs718314 e Mice lack<strong>in</strong>g ITPR2and ITPR3 exhibitedhypoglycaemia andlean body typeHOXC13 12q13.13 WHR rs1443512 Transcription factor important <strong>in</strong> cell spatialdistribution <strong>in</strong> embryonic development<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>


Closest Gene(s)ChromosomalLocationPhenotypeAssociatedLead SNP(s)Proposed Molecular or CellularFunctionFAIM2 (locus also 12q13 BMI rs7138803 Adipocyte apoptosisconta<strong>in</strong>s BCDIN3D)C12orf51 12q24 WHR rs2074356 eMTIF3eGTF3A 13q12.2 BMI rs4771122 ePRKD1 14q12 BMI rs11847697 eNRXN3 14q31 WC, BMI rs10146997 eMAP2K5 15q23 BMI rs2241423 eSH2B1 (locus with19e25 genes)16p11.2 BMI rs7498665,rs8049439,rs4788102,rs7498665Neuronal role <strong>in</strong> energy homeostasisGPRC5B 16p12.3 BMI rs12444979 eMAF 16q22eq23 BMI rs1424233 Transcription factor <strong>in</strong>volved <strong>in</strong>adipogenesis and <strong>in</strong>sul<strong>in</strong>eglucagonregulationFTO 16q22.2 BMI rs9939609,rs6499640,rs8050136,rs3751812,rs7190492,rs8044769,rs1558902Neuronal function associated with controlof appetiteAdditionalPhenotypesSh2b1-null mice areobese and diabeticAssociated with T2DNPC1 18q11.2 BMI rs1805081 Intracellular lipid transport NPC1-null mice showlate-onset weight lossand poor food <strong>in</strong>take.NPC1 <strong>in</strong>terferes withfunctionMC4R 18q22 BMI rs17782313,rs12970134,rs17700144Hypothalamic signall<strong>in</strong>gHaplo-<strong>in</strong>sufficiency <strong>in</strong>humans is associatedwith morbid obesity.MC4R-deficient miceshowKCTD15 19q13.11 BMI rs11084753, ers29941QPTCL-GIPR 19q13.32 BMI rs2287019 Encodes <strong>in</strong>cret<strong>in</strong> receptor Associated withfast<strong>in</strong>g and 2-hglucoseTMEM160 19q13.32 BMI rs3810291 e(adapted from Herrera (143)CHAPTER 15Epigenetic Approaches to Control Obesity301


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>such as Amsterdam dropp<strong>in</strong>g to below 1000 kilocalories (4200 kilojoules) a day by the end ofNovember 1944 and to 580 kilocalories <strong>in</strong> the west by the end of February 1945. The DutchFam<strong>in</strong>e Birth Cohort Study found that the children of pregnant women exposed to fam<strong>in</strong>e <strong>in</strong>early gestation were more susceptible to CVD and obesity while those exposed <strong>in</strong> later pregnancywere more susceptible to hypertension and <strong>in</strong>sul<strong>in</strong> resistance, diabetes, obesity,microalbum<strong>in</strong>uria, CVD, and other health problems [12].Suboptimal <strong>in</strong>trauter<strong>in</strong>e environments, such as <strong>in</strong> <strong>in</strong>trauter<strong>in</strong>e growth restriction (IUGR), maylead to fetal adaptations to assist short-term survival but may be detrimental <strong>in</strong> the long term[13]. Small babies who were born at term and undergo early catch-up growth, characterized bya greater accumulation of fat mass relative to lean body mass, have a particularly <strong>in</strong>creased riskof becom<strong>in</strong>g obese <strong>in</strong> later life compared to those born at higher birth weights [14]. Earlycatch-up growth <strong>in</strong> <strong>in</strong>fants born preterm and who were fed formula milk is also associatedwith an <strong>in</strong>creased cardio-metabolic risk <strong>in</strong> later life [15], <strong>in</strong>clud<strong>in</strong>g obesity. A number ofstudies have shown a greater <strong>in</strong>cidence of obesity <strong>in</strong> adults who were formula-fed as opposedto breast-fed dur<strong>in</strong>g <strong>in</strong>fancy. These f<strong>in</strong>d<strong>in</strong>gs highlight the likely role of postnatal feed<strong>in</strong>g.302Overnutrition <strong>in</strong> early life also <strong>in</strong>creases susceptibility to future obesity. It is believed that highmaternal plasma concentrations of glucose, free fatty acids, and am<strong>in</strong>o acids can result <strong>in</strong> lifelongchanges <strong>in</strong> appetite control, neuroendocr<strong>in</strong>e function, or energy metabolism <strong>in</strong> the develop<strong>in</strong>gfetus which lead to obesity later <strong>in</strong> life. Fetal overnutrition is more likely <strong>in</strong> mothers who havea greater BMI dur<strong>in</strong>g pregnancy [16] as factors such as <strong>in</strong>sul<strong>in</strong> resistance, glucose <strong>in</strong>tolerancelead<strong>in</strong>g to higher plasma concentrations of glucose, and free fatty acids correlate positively withBMI. Dorner and Plagemann [17] have reported that children of obese women are themselvesmore likely to become overweight and develop <strong>in</strong>sul<strong>in</strong> resistance <strong>in</strong> later life. Gestational weightga<strong>in</strong> irrespective of prepregnancy weight is positively associated with greater childhood adiposity[18] and even moderate weight ga<strong>in</strong> between successive pregnancies has been shown to result <strong>in</strong>an <strong>in</strong>crease <strong>in</strong> large-for-gestational-age births [19]. However, maternal weight loss throughbariatric surgery prevents transmission of obesity to children compared with the offspr<strong>in</strong>g ofmothers who did not undergo the surgery and rema<strong>in</strong>ed obese [20]. European and Indian studieshave shown that high maternal weight and adiposity are associated with coronary heart disease,<strong>in</strong>sul<strong>in</strong> deficiency, and type II diabetes <strong>in</strong> the offspr<strong>in</strong>g. These data suggest that even with<strong>in</strong>a relatively normal dietary range, modest alterations can affect the development of the fetus [21].However, it is possible that these correlations may not be due to an <strong>in</strong>trauter<strong>in</strong>e effect butresult from shared socioeconomic lifestyle factors between the mother and offspr<strong>in</strong>g or thetransmission of genetic factors. Studies tak<strong>in</strong>g <strong>in</strong>to account paternal BMI have been <strong>in</strong>consistent[22e24], with some f<strong>in</strong>d<strong>in</strong>g a strong maternal effect, some f<strong>in</strong>d<strong>in</strong>g a stronger paternaleffect, and others f<strong>in</strong>d<strong>in</strong>g similar paternal and maternal relations. However, these studies wereall relatively small and may have lacked sufficient power.The thrifty phenotype hypothesis proposes that reduced fetal growth is associated witha number of chronic conditions <strong>in</strong> later life [25]. These conditions <strong>in</strong>clude coronary heartdisease, stroke, diabetes, and hypertension. This <strong>in</strong>creased susceptibility is proposed to resultfrom adaptations made by the fetus <strong>in</strong> utero due to its limited supply of nutrients. Thehypothesis is that poor nutrient supply <strong>in</strong> utero results <strong>in</strong> fetal adaptations such that the <strong>in</strong>fantwill be prepared for survival <strong>in</strong> an environment <strong>in</strong> which resources are likely to be limited,result<strong>in</strong>g <strong>in</strong> a thrifty phenotype.Those with a thrifty phenotype who actually develop <strong>in</strong> an affluent environment may be moreprone to metabolic disorders, such as obesity and type 2 diabetes, whereas those who havereceived a good nutrient supply <strong>in</strong> utero will be adapted to good conditions and thereforebetter able to cope with rich diets. This idea is now widely accepted and is a source of concernfor societies such as those <strong>in</strong> the develop<strong>in</strong>g world where rapid socioeconomic improvement isunderway result<strong>in</strong>g <strong>in</strong> a transition from sparse to adequate or good nutrition [26].


CHAPTER 15Epigenetic Approaches to Control ObesityA study of IUGR <strong>in</strong>fants born at full term [27] showed that although these <strong>in</strong>fants were bornwith reduced subcutaneous fat, visceral fat depots were preserved creat<strong>in</strong>g an imbalancebetween central and peripheral fat deposits. In IUGR <strong>in</strong>fants accelerated postnatal growth isknown to exacerbate the negative effects of be<strong>in</strong>g born small-for-gestational-age, <strong>in</strong>creas<strong>in</strong>g therisk of develop<strong>in</strong>g impaired glucose tolerance [28], <strong>in</strong>sul<strong>in</strong> resistance [29], obesity [30] andtype 2 diabetes [31].15.3 ANIMAL STUDIES OF EARLY DEVELOPMENT ANDMETABOLIC PROGRAMMINGThe processes by which environmental cues <strong>in</strong>duce altered adult phenotypes and an <strong>in</strong>creasedrisk of obesity <strong>in</strong> the offspr<strong>in</strong>g are not yet fully understood, but <strong>in</strong>creas<strong>in</strong>g evidence po<strong>in</strong>ts tothe importance of epigenetic processes. Animal models have been useful <strong>in</strong> understand<strong>in</strong>g theeffects on adult phenotypes result<strong>in</strong>g from perturbations <strong>in</strong> the developmental environment.15.3.1 The Maternal Prote<strong>in</strong>-Restricted DietThe best-studied and most-characterized animal model of nutritional <strong>in</strong>duction of an alteredphenotype is feed<strong>in</strong>g pregnant rodents a prote<strong>in</strong>-restricted (PR) diet. Feed<strong>in</strong>g the PR dietdur<strong>in</strong>g pregnancy results <strong>in</strong> impaired glucose homeostasis [32], vascular dysfunction [33],impaired immunity [34], <strong>in</strong>creased susceptibility to oxidative stress [35], <strong>in</strong>creased fat deposition,and altered feed<strong>in</strong>g behavior [36,37]. The <strong>in</strong>duction dur<strong>in</strong>g early life of persistentchanges to the phenotype of the offspr<strong>in</strong>g by perturbations <strong>in</strong> maternal diet implies stablealteration of gene transcription which, <strong>in</strong> turn, results <strong>in</strong> the altered activities of metabolicpathways and homeostatic control processes. Initially us<strong>in</strong>g a candidate gene approach manygroups reported long-term changes <strong>in</strong> the expression of key metabolic genes <strong>in</strong> response tovariations <strong>in</strong> maternal diet. For example, feed<strong>in</strong>g a PR diet to pregnant rats <strong>in</strong>creased glucocorticoidreceptor (GR) expression and reduced expression of 11b-hydroxysteroid dehydrogenasetype 2 (11bHSD)-2, the enzyme which <strong>in</strong>activates corticosteroids, <strong>in</strong> liver, lung, kidney,and bra<strong>in</strong> <strong>in</strong> the offspr<strong>in</strong>g [38]. In the liver, <strong>in</strong>creased GR activity up-regulates phosphoenolpyruvatecarboxyk<strong>in</strong>ase (PEPCK) expression and activity and so <strong>in</strong>creases capacity forgluconeogenesis. This may contribute to the <strong>in</strong>duction of <strong>in</strong>sul<strong>in</strong> resistance <strong>in</strong> this model [39].Altered expression of GR has also been reported <strong>in</strong> the lung, liver, adrenals, and kidneys of theoffspr<strong>in</strong>g of sheep fed a restricted diet dur<strong>in</strong>g pregnancy [40e42]. Feed<strong>in</strong>g a PR diet to pregnantrats up-regulates glucok<strong>in</strong>ase expression <strong>in</strong> the liver of the offspr<strong>in</strong>g, which implies<strong>in</strong>creased capacity for glucose uptake [43]. The expression of genes <strong>in</strong>volved <strong>in</strong> lipidhomeostasis is also altered by maternal PR. Peroxisomal proliferator-activated receptor(PPAR)-a expression was <strong>in</strong>creased <strong>in</strong> the liver of the offspr<strong>in</strong>g of rats fed a PR diet dur<strong>in</strong>gpregnancy and was accompanied by up-regulation of its target gene acyl-CoA oxidase (AOX)[44]. The expression of acetyl-CoA carboxylase and fatty acid synthase have been reported to be<strong>in</strong>creased <strong>in</strong> the liver of the offspr<strong>in</strong>g of rats fed a PR diet dur<strong>in</strong>g pregnancy and lactation [45].303More recently genome-wide approaches have been used to determ<strong>in</strong>e which genes are altered<strong>in</strong> response to diet. Transcriptome-wide analysis of adult liver from PR offspr<strong>in</strong>g revealedapproximately 1.3% of genes with<strong>in</strong> the genome are changed <strong>in</strong> response to maternal prote<strong>in</strong>restriction. This change <strong>in</strong> a relatively small subset of genes suggests that these may representan orchestrated response to the nutritional challenge and be part of an adaptive response [46].The pathways changed <strong>in</strong> the liver <strong>in</strong> response to maternal PR were those <strong>in</strong>volved <strong>in</strong> developmentalprocesses, ion transport, and hormonal and stress responses, which is consistentwith the phenotypic changes observed <strong>in</strong> PR offspr<strong>in</strong>g.The alterations <strong>in</strong> offspr<strong>in</strong>g metabolism and physiology <strong>in</strong>duced by maternal prote<strong>in</strong> restrictionare dependent upon the tim<strong>in</strong>g of the nutritional challenge. Bertram et al. has shown <strong>in</strong>the gu<strong>in</strong>ea pig model that female offspr<strong>in</strong>g born to dams fed a PR diet <strong>in</strong> the first half of


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>pregnancy (1e35 days) have raised mean arterial blood pressure which was associated with an<strong>in</strong>creased <strong>in</strong>traventricular septum and anterior left ventricle wall thickness. They did notexhibit growth restriction at any time; <strong>in</strong> contrast, the offspr<strong>in</strong>g from dams fed a PR diet <strong>in</strong> lategestation (36e70 days) were growth restricted but did not display alterations <strong>in</strong> blood pressureor left ventricular structure [47].Animal studies have also shown a clear <strong>in</strong>teraction between the pre- and postnatal environments[48,49], with variations <strong>in</strong> the diet fed after wean<strong>in</strong>g exacerbat<strong>in</strong>g the effects of maternalundernutrition on the phenotype of the offspr<strong>in</strong>g. For example, dyslipidemia and impairedglucose homeostasis <strong>in</strong>duced by feed<strong>in</strong>g dams a PR diet dur<strong>in</strong>g pregnancy were exacerbated <strong>in</strong>adult male and female rats fed a diet conta<strong>in</strong><strong>in</strong>g 10% (w/w) fat after wean<strong>in</strong>g compared toa 4% (w/w) fat postwean<strong>in</strong>g diet [50].30415.3.2 Global Dietary RestrictionA number of groups have also used global dietary restriction dur<strong>in</strong>g pregnancy to <strong>in</strong>vestigatehow maternal diet can <strong>in</strong>fluence disease susceptibility <strong>in</strong> later life. Woodall et al. used globalnutrient restriction, feed<strong>in</strong>g rats 30% of an ad libitum <strong>in</strong>take throughout gestation, whichresults <strong>in</strong> a model of IUGR [51]. Offspr<strong>in</strong>g born to dams fed this diet dur<strong>in</strong>g pregnancy aresignificantly smaller at birth than control offspr<strong>in</strong>g. They also exhibit higher systolic bloodpressure, hyper<strong>in</strong>sul<strong>in</strong>emia, hyperlept<strong>in</strong>emia, hyperphagia, reduced locomotion, and obesity.These metabolic alterations are all augmented by feed<strong>in</strong>g a high-fat postnatal diet [52].However, even modest global nutrient restriction dur<strong>in</strong>g pregnancy <strong>in</strong>duces alterations <strong>in</strong>metabolism and the HPA axis. In gu<strong>in</strong>ea pigs fed 85% of an ad libitum diet throughoutgestation, alterations <strong>in</strong> postnatal cholesterol homeostasis were observed <strong>in</strong> the male offspr<strong>in</strong>g[53]. In the sheep, a 15% global nutrient restriction dur<strong>in</strong>g the first half of pregnancy led toreduced adrenocorticotroph<strong>in</strong> hormone (ACTH) and cortisol responses to exogenouscorticotrop<strong>in</strong>-releas<strong>in</strong>g hormone and arg<strong>in</strong><strong>in</strong>e vasopress<strong>in</strong> adm<strong>in</strong>istration, and a bluntedcortisol response to ACTH [54]. Long-term changes <strong>in</strong> gene expression have also been reported<strong>in</strong> adult offspr<strong>in</strong>g of dams fed a global undernutrition diet dur<strong>in</strong>g pregnancy. Gluckman et al.,have showed that expression of PPARaand GR <strong>in</strong> the rat liver are both down-regulated <strong>in</strong> adultoffspr<strong>in</strong>g born to dams fed a global nutrient-restricted diet of 30% ad libitum dur<strong>in</strong>g pregnancy[55].15.3.3 High-Fat Diet Dur<strong>in</strong>g PregnancyWith recent concerns about the levels of obesity <strong>in</strong> the Western world, a number of new animalmodels of overnutrition dur<strong>in</strong>g pregnancy have also been developed. Feed<strong>in</strong>g an obesogenicdiet to female rats from before mat<strong>in</strong>g and through lactation leads to maternal obesity as wellas hyperphagia, <strong>in</strong>creased adiposity, decreased muscle mass, reduced locomotion, and acceleratedpuberty <strong>in</strong> the offspr<strong>in</strong>g [56]. Samuelsson et al. have also shown that offspr<strong>in</strong>g frompregnant rats fed a “junk food diet” of 16% fat, 33% sugar throughout pregnancy and lactationexhibited higher blood pressure, greater adiposity, and <strong>in</strong>sul<strong>in</strong> resistance <strong>in</strong> comparison tocontrol offspr<strong>in</strong>g [57]. The type of fat may also be important as when dams were fed diets withdifferent ratios of n-6/n-3 fatty acids <strong>in</strong>sul<strong>in</strong> sensitivity and weight ga<strong>in</strong> varied accord<strong>in</strong>g to therelative amounts of these fatty acids <strong>in</strong> the maternal diet [58]. Maternal carbohydrate <strong>in</strong>takehas also been shown to have an effect with high-carbohydrate diets found to result <strong>in</strong> offspr<strong>in</strong>gthat rema<strong>in</strong>ed lighter <strong>in</strong> weight and had <strong>in</strong>creased responsiveness to the neuromodulator,neuropeptide Y (NPY) [59]. Persistent alterations <strong>in</strong> the expression of PPAR g2, 11bHSD-1 andthe b2 and b3 adrenoreceptors <strong>in</strong> adipose tissue [57] were seen, which may lead to <strong>in</strong>creasedadipogenesis and decreased lipolysis <strong>in</strong> these rats. Interest<strong>in</strong>gly, however, studies by Ng et al.have shown that not only can variations <strong>in</strong> maternal diet affect subsequent phenotype but thatpaternal diet is also important <strong>in</strong> determ<strong>in</strong><strong>in</strong>g future disease risk. Paternal high-fat-diet (HFD)exposure <strong>in</strong>duces <strong>in</strong>creased body weight, adiposity, impaired glucose tolerance, and <strong>in</strong>sul<strong>in</strong>sensitivity <strong>in</strong> female offspr<strong>in</strong>g [60]. Paternal HFD altered the expression of 642 pancreatic islet


CHAPTER 15Epigenetic Approaches to Control Obesitygenes <strong>in</strong> adult female offspr<strong>in</strong>g; these genes <strong>in</strong>cluded those <strong>in</strong>volved <strong>in</strong> cation and ATPb<strong>in</strong>d<strong>in</strong>g, cytoskeleton, and <strong>in</strong>tracellular transport.In rodents there is <strong>in</strong>creas<strong>in</strong>g evidence that the period of susceptibility extends <strong>in</strong>to postnatallife as the suckl<strong>in</strong>g period has been shown to be critical <strong>in</strong> the developmental <strong>in</strong>duction ofmetabolic disease. Studies of rats <strong>in</strong> cross-foster<strong>in</strong>g experiments show that high-fat feed<strong>in</strong>g <strong>in</strong>the suckl<strong>in</strong>g period leads to an <strong>in</strong>crease <strong>in</strong> adiposity, hyperlept<strong>in</strong>emia, and hypertension <strong>in</strong> theadult offspr<strong>in</strong>g fed a normal diet after wean<strong>in</strong>g [61e63]. Schmidt et al. have also shown thatoverfeed<strong>in</strong>g rats dur<strong>in</strong>g the suckl<strong>in</strong>g period by rear<strong>in</strong>g them <strong>in</strong> small litters produces hyperphagiaand obesity <strong>in</strong> adulthood [64]. There is grow<strong>in</strong>g evidence that overnutrition dur<strong>in</strong>gprenatal and/or early postnatal life alters the maturation of the appetite and energy-regulat<strong>in</strong>gneural network <strong>in</strong> the hypothalamus. Overfeed<strong>in</strong>g rat pups by rear<strong>in</strong>g them <strong>in</strong> small littersleads to an <strong>in</strong>creased food <strong>in</strong>take <strong>in</strong> the per<strong>in</strong>atal period and this was also associated witha persistent <strong>in</strong>crease <strong>in</strong> appetite drive <strong>in</strong> later life [65,66]. In rodents, exposure to a diabeticenvironment before birth or exposure <strong>in</strong> early postnatal life results <strong>in</strong> significant changes <strong>in</strong> thearchitecture of the hypothalamus, a reduced sensitivity of central hypothalamic neuropeptidesto signals of <strong>in</strong>creased nutrition, and a central resistance to peripheral signals of satiety signals[62,67e69]. The effect of overnutrition on hypothalamic function has been observed not only<strong>in</strong> rodents where appetite circuits are not fully mature until postnatal day 16 [67] but also <strong>in</strong>sheep where the neural network is relatively mature at birth as <strong>in</strong> humans [70].15.4 DEVELOPMENTAL PLASTICITYDevelopment is a period of rapid change <strong>in</strong> which environmental cues may <strong>in</strong>duce persistentchanges <strong>in</strong> phenotype <strong>in</strong> order to prepare the offspr<strong>in</strong>g for the predicted future environment;allow<strong>in</strong>g the organism to adapt much more rapidly than could be achieved by mutation. Forexample, the duration of daylight to which meadow voles (Microtus pennsylvanicus) are exposedto prior to conception <strong>in</strong>fluences the thickness of their coat <strong>in</strong> anticipation of either w<strong>in</strong>ter orsummer temperatures [71]. Work by Gluckman and Hanson argues that the developmentalenvironment can produce a range of effects with both immediate and later-life consequences.These effects do not confer any immediate advantage to the offspr<strong>in</strong>g but give a later fitnessadvantage <strong>in</strong> later life when <strong>in</strong> an environment as predicted by the developmental experience.This type of response has been termed a predictive adaptive response (PAR) [72].305PARs rely on the environment rema<strong>in</strong><strong>in</strong>g relatively constant throughout the life of theoffspr<strong>in</strong>g. For these PARSs to confer a fitness advantage it is not necessary for the fidelity ofpredictions to be particularly high. If the predicted environment does not fluctuate significantlyover many generations then the favorable trait may become assimilated, whereby it isfixed or genetically encoded [73]. It is also important to consider that we now live much longerthan our ancestors. Therefore, mechanisms that enhanced fitness <strong>in</strong> early evolution may nolonger have an advantage, or may be advantageous for the young only. Epigenetic/nongenomic<strong>in</strong>heritance that may have previously conferred a survival advantage may nowexacerbate a risk for successive generations. This may play a role <strong>in</strong> the current epidemic ofobesity and CVD.PARs are only adaptive when the developmental environment of the offspr<strong>in</strong>g is with<strong>in</strong> thepredicted range. If the environment differs significantly from that which was predicted the<strong>in</strong>dividual is said to be “mismatched”, that is hav<strong>in</strong>g a phenotype that is not appropriate forthe environment [72]. This mismatch does not have to be as a result of an extreme pre- orpostnatal environment, simply a phenotype be<strong>in</strong>g <strong>in</strong>duced dur<strong>in</strong>g development which is notsuitable for respond<strong>in</strong>g to the postnatal environment. This mismatch can affect the offspr<strong>in</strong>g<strong>in</strong> a range of ways, <strong>in</strong>clud<strong>in</strong>g abdom<strong>in</strong>al fat deposition [74]. Mismatch can be due to a range ofcircumstances such as poorer environmental conditions dur<strong>in</strong>g development followed byricher conditions later <strong>in</strong> life or vice versa, or due to exposure to a postnatal environment,


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>which is evolutionarily novel and as such outside of the predictive capabilities of the fetus.Maternal disease, unbalanced diet or body composition can lead to mismatch even if theoffspr<strong>in</strong>g goes on to have a balanced healthy diet; conversely an <strong>in</strong>crease <strong>in</strong> energy-dense foodsand limited physical activity <strong>in</strong> the offspr<strong>in</strong>g (the Western lifestyle) will <strong>in</strong>crease the degree ofmismatch if the <strong>in</strong>trauter<strong>in</strong>e environment was poor. Changes <strong>in</strong> lifestyle factors betweengenerations are of particular significance for countries <strong>in</strong> which rapid socioeconomic transitionis underway as contemporary westernized diets and lifestyles constitute novel environments,thus compound<strong>in</strong>g the mismatch [75]. The elevated risk of obesity is due to the degreeof mismatch between the pre- and postnatal environment rather than any absolute levels <strong>in</strong>the postnatal environment. This concept is supported by a number of animal studies <strong>in</strong> whichthe maternal pre- and postnatal diets were manipulated, as described later <strong>in</strong> this chapter.Both fetal and neonatal life are characterized by a high degree of plasticity (the potential of anorganism to alter it phenotype) which provides the potential for organisms to respond rapidlyand effectively to environmental change. Phenotypic plasticity is usually def<strong>in</strong>ed as a propertyof <strong>in</strong>dividual genotypes to produce different phenotypes when exposed to different environmentalconditions [76]. This plasticity can be expressed at a number of levels <strong>in</strong>clud<strong>in</strong>gbehavioral, biochemical, physiological, or developmental. Not all phenotypic plasticity isadaptive and it does not necessarily always serve to improve the <strong>in</strong>dividual’s survival. Sometraits are plastic due to unavoidable constra<strong>in</strong>ts <strong>in</strong> the biochemistry or physiology of theorganism. Developmental plasticity forms a component of phenotypic plasticity and <strong>in</strong>contrast to biochemical and physiological responses, which can be reversed over short timescales,developmental plasticity tends to be irreversible or take longer to be reversed.306Recently there have been advances <strong>in</strong> understand<strong>in</strong>g of epigenetic effects dur<strong>in</strong>g developmentand the key role which they can play <strong>in</strong> plastic processes. However, studies <strong>in</strong> animal modelshave proved to be key <strong>in</strong> show<strong>in</strong>g how the developmental environment, <strong>in</strong>clud<strong>in</strong>g themother’s diet, alters epigenetic processes [44,77,78]. In animal studies the effects of epigeneticchanges <strong>in</strong>duced experimentally dur<strong>in</strong>g development have been shown to produce lifelongphysiological changes of relevance to human disease such as metabolic alterations known to<strong>in</strong>fluence obesity [79].15.5 EPIGENETICS AND DEVELOPMENTAL PROGRAMMING BYTHE EARLY LIFE ENVIRONMENTMost studies exam<strong>in</strong><strong>in</strong>g the effect of early-life nutrition and its role <strong>in</strong> gene regulation havefocused on DNA methylation, rather than other epigenetic processes such as histonemodification and non-cod<strong>in</strong>g RNAs. DNA methylation can <strong>in</strong>duce transcriptional silenc<strong>in</strong>gby block<strong>in</strong>g the b<strong>in</strong>d<strong>in</strong>g of transcription factors and/or through promot<strong>in</strong>g the b<strong>in</strong>d<strong>in</strong>g ofthe methyl CpG-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> (MeCP2). The latter b<strong>in</strong>ds to methylated cytos<strong>in</strong>es and, <strong>in</strong>turn, recruits histone-modify<strong>in</strong>g complexes to the DNA. MeCP2 recruits both histonedeacetylases (HDACs), which remove acetyl groups from the histones, a signal of transcriptionallyactive chromat<strong>in</strong>, and histone methyl transferases (HMTs), such as Suv39H,which methylates lys<strong>in</strong>e 9 on H3, result<strong>in</strong>g <strong>in</strong> a closed chromat<strong>in</strong> structure and transcriptionalsilenc<strong>in</strong>g. Methylation of CpG d<strong>in</strong>ucleotides de novo is catalyzed by DNA methyltransferases(Dnmt) 3a and 3b, and is ma<strong>in</strong>ta<strong>in</strong>ed through mitosis by gene-specificmethylation of hemimethylated DNA by Dnmt1. DNA methylation is important for asymmetricalsilenc<strong>in</strong>g of impr<strong>in</strong>ted genes, X chromosome <strong>in</strong>activation, and silenc<strong>in</strong>g of retrotransposons.DNA methylation is also critical for cell differentiation by silenc<strong>in</strong>g theexpression of specific genes dur<strong>in</strong>g the development and differentiation of <strong>in</strong>dividual tissues[80,81]. Methylation of CpGs is largely established dur<strong>in</strong>g embryogenesis or <strong>in</strong> early postnatallife. Follow<strong>in</strong>g fertilization, maternal and paternal genomes undergo extensive demethylationfollowed by global methylation de novo just prior to blastocyst implantation[82], dur<strong>in</strong>g which 70% of CpGs are methylated, ma<strong>in</strong>ly <strong>in</strong> repressive heterochromat<strong>in</strong>


CHAPTER 15Epigenetic Approaches to Control Obesityregions and <strong>in</strong> repetitive sequences such as retrotransposable elements [83]. L<strong>in</strong>eage-specificmethylation of tissue-specific genes occurs throughout prenatal development and earlypostnatal life and determ<strong>in</strong>es developmental fates of differentiat<strong>in</strong>g cells. Epigenetic marksare essentially ma<strong>in</strong>ta<strong>in</strong>ed throughout life. However, environmental perturbations dur<strong>in</strong>gperiods when methylation patterns are <strong>in</strong>duced may impair the program of gene silenc<strong>in</strong>g oractivation with potential long-term adverse consequences.Epigenetic marks <strong>in</strong>duced dur<strong>in</strong>g development were thought to persist <strong>in</strong>to adulthood. However,there is now much evidence that ag<strong>in</strong>g is associated with tissue-specific epigenetic drift. The firstlarge-scale tw<strong>in</strong> study exam<strong>in</strong>ed 20 3-year-old and 50-year-old Spanish MZ tw<strong>in</strong> pairs. Thesestudies revealed that while the tw<strong>in</strong>s had very similar epigenetic profiles, <strong>in</strong>dicative of a high levelof epigenetic heritability, there was a degree of epigenetic variability which <strong>in</strong>creased with ageacross a range of tissues. It is of <strong>in</strong>terest that the greatest differences were <strong>in</strong> tw<strong>in</strong>s who differedmost <strong>in</strong> lifestyle [84]. This study was cross-sectional rather than longitud<strong>in</strong>al and for this reason itwas not possible to <strong>in</strong>vestigate observed <strong>in</strong>dividual variability. A more recent study [85] measuredthe DNA methylation status of three genes (DRD4-Dopam<strong>in</strong>e receptor 4 gene/seroton<strong>in</strong> transportergene-SERT/X-l<strong>in</strong>ked monoam<strong>in</strong>e oxidase A gene) <strong>in</strong> 46 monozygotic and 45 dizygotictw<strong>in</strong>s sampled at 5 and 10 years of age. This study revealed that DNA methylation differences wereapparent even at 5 years <strong>in</strong> genetically identical <strong>in</strong>dividuals. Analysis at the later timepo<strong>in</strong>tsuggested that these differences were not stable over time. The study suggests that environmental<strong>in</strong>fluences are important for determ<strong>in</strong><strong>in</strong>g methylation patterns found <strong>in</strong> the <strong>in</strong>dividual andhighlights the importance of longitud<strong>in</strong>al research designs for epigenetic studies.Studies on isolated embryos first supported the hypothesis that variations <strong>in</strong> nutrient availabilitycan alter the methylation of genes with<strong>in</strong> the embryo [86]. In these studies decreased expressionof both H19 and IGF2 was observed, coupled with <strong>in</strong>creased DNA methylation at the H19impr<strong>in</strong>t<strong>in</strong>g control region <strong>in</strong> embryos cultured <strong>in</strong> the presence of fetal calf serum as compared tocontrols. Data from these studies demonstrate that early nutrition can cause epigenetic changeswhich are ma<strong>in</strong>ta<strong>in</strong>ed <strong>in</strong> later developmental stages, at least <strong>in</strong> the case of impr<strong>in</strong>ted genes.307Manipulation of human embryos <strong>in</strong> vitro can <strong>in</strong>duce similar impr<strong>in</strong>t<strong>in</strong>g alterations to those seen<strong>in</strong> mice. Angelman’s syndrome is a human neurogenetic disorder caused by loss of function ofthe maternal allelle (paternally impr<strong>in</strong>ted and therefore maternally expressed) of UBE3A. This isusually as the result of a genetic mutation or rarely as a result of a sporadic impr<strong>in</strong>t<strong>in</strong>g error [87].However, there have been cases reported where Angelman’s syndrome has been found <strong>in</strong>children conceived us<strong>in</strong>g <strong>in</strong>tracytoplasmic sperm <strong>in</strong>jection [87,88]. In vitro fertilization (IVF)has also been l<strong>in</strong>ked to the <strong>in</strong>creased <strong>in</strong>cidence of another impr<strong>in</strong>t<strong>in</strong>g disorder,BeckwitheWiedemann syndrome. This congenital disorder is caused by a loss of impr<strong>in</strong>t<strong>in</strong>g ofa group of genes (which <strong>in</strong>cludes H19 and IGF2) on human chromosome 11p15 [89]. Thesestudies provide evidence that the early environment can cause epigenetic alterations atimpr<strong>in</strong>ted loci, lead<strong>in</strong>g to human disorders that <strong>in</strong>clude obesity as a cl<strong>in</strong>ical characteristic.A number of factors dur<strong>in</strong>g early life alter the epigenome of the fetus, produc<strong>in</strong>g long-termchanges <strong>in</strong> gene expression. In an elegant study of the effect of maternal behavior dur<strong>in</strong>gsuckl<strong>in</strong>g on the development of stress response <strong>in</strong> the offspr<strong>in</strong>g, Weaver et al. showed that pupsraised by rat dams which showed poorer nurtur<strong>in</strong>g had an <strong>in</strong>creased stress response [90]. Theeffect was due to hypermethylation of specific CpG d<strong>in</strong>ucleotides with<strong>in</strong> the promoter of theGR gene <strong>in</strong> the hippocampus of the offspr<strong>in</strong>g. These changes were reversed <strong>in</strong> the bra<strong>in</strong>s of theadults by <strong>in</strong>tracranial adm<strong>in</strong>istration of the histone deacetylase <strong>in</strong>hibitor Trichostat<strong>in</strong> A andL-methion<strong>in</strong>e [91].15.6 EPIGENETICS AND EARLY-LIFE NUTRITIONDifferences <strong>in</strong> the maternal <strong>in</strong>take of nutrients have been shown to alter the methylation ofnon-impr<strong>in</strong>ted genes, result<strong>in</strong>g <strong>in</strong> subtle effects on fetal and offspr<strong>in</strong>g development. In the


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>agouti mouse variations <strong>in</strong> the maternal <strong>in</strong>take dur<strong>in</strong>g pregnancy of nutrients <strong>in</strong>volved <strong>in</strong>1-carbon metabolism <strong>in</strong>duces differences <strong>in</strong> the coat color of the offspr<strong>in</strong>g. In agouti mice A vythere is an <strong>in</strong>sertion of an IAP retrotransposon <strong>in</strong> the 5 0 end of the Agouti gene which acts asa cryptic promoter direct<strong>in</strong>g expression of the agouti gene encod<strong>in</strong>g a paracr<strong>in</strong>e signal<strong>in</strong>gmolecule which produces black eumelan<strong>in</strong> or yellow phaeomelan<strong>in</strong>. The methylation status ofthe IAP element produces a range of coat colors between yellow (unmethylated) and brown(methylated) [77,92]. Supplementation of the mother’s diet with methyl donors such asbeta<strong>in</strong>e, chol<strong>in</strong>e, folic acid, and vitam<strong>in</strong> B 12 shifted the distribution of coat color of theoffspr<strong>in</strong>g from yellow (agouti) to brown (pseudo-agouti) [93]. This shift is due to <strong>in</strong>creasedmethylation of the IAP element [77]. Thus maternal <strong>in</strong>take of nutrients <strong>in</strong>volved <strong>in</strong> 1-carbonmetabolism can <strong>in</strong>duce graded changes to DNA methylation and gene expression <strong>in</strong> theoffspr<strong>in</strong>g which persist <strong>in</strong>to adulthood.308Feed<strong>in</strong>g pregnant rats a PR diet <strong>in</strong>duced hypomethylation of the GR and PPARa promoters <strong>in</strong>the livers of juvenile and adult offspr<strong>in</strong>g. Hypomethylation of the GR promoter was associatedwith histone modifications which facilitate transcription; acetylation of histones H3 andH4 and methylation of histone H3 at lys<strong>in</strong>e K4, while those that suppress gene expressionwere reduced or unchanged [94]. Although functionally consistent, the mechanistic relationshipbetween GR hypomethylation and the associated histone changes is not known.These studies showed for the first time that, <strong>in</strong> contrast to modify<strong>in</strong>g the maternal <strong>in</strong>take ofnutrients directly <strong>in</strong>volved 1-carbon metabolism [44], stable changes to the epigeneticregulation of the expression of transcription factors can be <strong>in</strong>duced <strong>in</strong> the offspr<strong>in</strong>g by modestchanges to maternal macronutrient balance dur<strong>in</strong>g pregnancy. Expression of PPARa and GR,and of their respective target genes, acyl-CoA oxidase and carnit<strong>in</strong>e palmitoyl-transferase-1,and PEPCK was <strong>in</strong>creased <strong>in</strong> juvenile and adult offspr<strong>in</strong>g [44,94e96]. This is consistent withraised plasma b-hydroxybutyrate and glucose concentrations <strong>in</strong> the fast<strong>in</strong>g offspr<strong>in</strong>g [97].Sequenc<strong>in</strong>g analysis of the PPARa promoter showed that four specific CpGs were hypomethylated,and that two CpGs located with<strong>in</strong> transcription factor response elementspredicted the level of the transcript [95]. Thus the effects of the maternal PR diet on theoffspr<strong>in</strong>g are targeted to specific CpGs. The mechanisms <strong>in</strong>volved are not known but byregulat<strong>in</strong>g effects of transcription factors on expression they may have important effects onphenotype.Together, these results <strong>in</strong>dicate that modest dietary prote<strong>in</strong> restriction dur<strong>in</strong>g pregnancy<strong>in</strong>duces an altered phenotype through epigenetic changes <strong>in</strong> specific genes. Methylation of theGR and PPARa promoters was also reduced <strong>in</strong> the heart of the offspr<strong>in</strong>g [98] and the PPARapromoter was hypomethylated <strong>in</strong> the whole umbilical cord [99]. These f<strong>in</strong>d<strong>in</strong>gs are consistentwith <strong>in</strong>creased GR mRNA expression <strong>in</strong> a range of tissues from the offspr<strong>in</strong>g of rats fed a PRdiet dur<strong>in</strong>g pregnancy [100]. However, PPARa methylation does not differ between controland PR offspr<strong>in</strong>g <strong>in</strong> skeletal muscle, spleen, and adipose tissue, <strong>in</strong>dicat<strong>in</strong>g that the effects of thematernal diet are tissue specific [Lillycrop and Burdge, unpublished]. Hypomethylation of theGR promoter has also been found <strong>in</strong> the offspr<strong>in</strong>g of mice fed a PR diet dur<strong>in</strong>g pregnancy [99],which suggests that the effect of the PR diet may not be specific to one species.The fundamental role of changes <strong>in</strong> the epigenetic regulation of transcription factor expression<strong>in</strong> alter<strong>in</strong>g the activity of pathways controlled by their target genes is underl<strong>in</strong>ed by theobservation that although glucok<strong>in</strong>ase expression was <strong>in</strong>creased <strong>in</strong> the liver of the PR offspr<strong>in</strong>g,this was not accompanied by changes <strong>in</strong> the methylation status of the glucok<strong>in</strong>ase promoter[43]. S<strong>in</strong>ce GR activity <strong>in</strong>creases glucok<strong>in</strong>ase expression through enhancement of <strong>in</strong>sul<strong>in</strong>action [101], greater glucok<strong>in</strong>ase expression <strong>in</strong> the PR offspr<strong>in</strong>g may have been due to<strong>in</strong>creased GR activity as a result of hypomethylation of the GR promoter rather than a directeffect of prenatal undernutrition on glucok<strong>in</strong>ase.In contrast to the effects of maternal PR diet on the epigenetic regulation of hepatic genes <strong>in</strong>the offspr<strong>in</strong>g, a 70% reduction of total food <strong>in</strong>take dur<strong>in</strong>g pregnancy <strong>in</strong> rats <strong>in</strong>duced


CHAPTER 15Epigenetic Approaches to Control Obesityhypermethylation and lower PPARa and GR expression <strong>in</strong> the liver of 170-day-old offspr<strong>in</strong>g[55]. One explanation may lie <strong>in</strong> the differences <strong>in</strong> severity of nutritional restriction betweenthese two dietary regimens. If the <strong>in</strong>duction of altered phenotypes is predictive, then it may beanticipated that <strong>in</strong>duced changes <strong>in</strong> the epigenome would differ accord<strong>in</strong>g to dietary regimen,<strong>in</strong> order to match the phenotype to the predicted future environment. Thus the maternal PRdiet could be regarded as a moderate nutrient constra<strong>in</strong>t which <strong>in</strong>duces <strong>in</strong> the offspr<strong>in</strong>g<strong>in</strong>creased capacity for us<strong>in</strong>g nutrient reserves for energy production. In contrast more severeglobal undernutrition <strong>in</strong>duces conservation of energy substrates. These <strong>in</strong>terpretations areconsistent with the phenotypes <strong>in</strong>duced <strong>in</strong> the offspr<strong>in</strong>g [52,55,95].There is also evidence that an excessive early nutritional environment can alter the epigeneticregulation of genes. In the hypothalamus lept<strong>in</strong> triggers specific neuronal subpopulations suchas pro-opiomelanocort<strong>in</strong> (POMC) and neuropeptide Y (NPY), and activates several <strong>in</strong>tracellularsignal<strong>in</strong>g events which result <strong>in</strong> decreased food <strong>in</strong>take and/or <strong>in</strong>creased energy expenditure.The importance of POMC <strong>in</strong> food <strong>in</strong>take and energy balance can be demonstrated bymutations <strong>in</strong> the POMC gene which result <strong>in</strong> an obese phenotype [102]. Targeted disruption ofthe POMC gene <strong>in</strong> a mouse model was found to result <strong>in</strong> hyperphagia and lower oxygenconsumption lead<strong>in</strong>g to <strong>in</strong>creased fat mass and obesity [103].Plagemann et al. [104] showed that neonatal overfeed<strong>in</strong>g <strong>in</strong>duced by rais<strong>in</strong>g rat pups <strong>in</strong> smalllitters <strong>in</strong>duces the hypermethylation of two CpG d<strong>in</strong>ucleotides with<strong>in</strong> the POMC promoterwhich are essential for POMC <strong>in</strong>duction by lept<strong>in</strong> and <strong>in</strong>sul<strong>in</strong>. Consequently POMC expressionis not up-regulated <strong>in</strong> these rats despite hyper<strong>in</strong>sul<strong>in</strong>emia and hyperlept<strong>in</strong>emia [104].This suggests that overfeed<strong>in</strong>g dur<strong>in</strong>g early postnatal life when the appetite circuitry with<strong>in</strong> thehypothalamus is still develop<strong>in</strong>g can alter the methylation of genes critical for bodyweightregulation, result<strong>in</strong>g <strong>in</strong> the altered programm<strong>in</strong>g of this system and an <strong>in</strong>creased tendencytowards obesity <strong>in</strong> later life. Ng et al. have also shown that Il13ra2 was hypomethylated <strong>in</strong>female offspr<strong>in</strong>g after high-fat feed<strong>in</strong>g of the fathers [60,60].30915.6.1 Transgenerational EffectsEmerg<strong>in</strong>g evidence from small animal models suggests that <strong>in</strong>duced phenotypes can pass tomore than one generation by a non-genomic mechanism. In rats, feed<strong>in</strong>g a PR diet to the F 0generation dur<strong>in</strong>g pregnancy results <strong>in</strong> elevated blood pressure, endothelial dysfunction, and<strong>in</strong>sul<strong>in</strong> resistance <strong>in</strong> the F 1 and F 2 generations [105e107] despite adequate nutrition dur<strong>in</strong>gpregnancy <strong>in</strong> the F 1 generation. The adverse effects on glucose homeostasis of feed<strong>in</strong>g a PRdur<strong>in</strong>g pregnancy <strong>in</strong> the F 0 generation have been found <strong>in</strong> the offspr<strong>in</strong>g up to F 3 generation[108]. The adm<strong>in</strong>istration of dexamethasone to dams <strong>in</strong> late pregnancy <strong>in</strong>duced <strong>in</strong>creasedexpression of the glucocorticoid receptor (GR) and its target gene phosphoenolpyruvatecarboxyk<strong>in</strong>ase (PEPCK) <strong>in</strong> the liver of the F 1 and F 2 , but not F 3 , offspr<strong>in</strong>g [109]. These f<strong>in</strong>d<strong>in</strong>gsraise the important issue that assessment of true non-genomic transmission between generationsrequires studies which cont<strong>in</strong>ue to at least the F 3 generation [110].There is substantial evidence for transgenerational epigenetic <strong>in</strong>heritance <strong>in</strong> non-mammalianspecies and its role <strong>in</strong> evolutionary biology has been reviewed [111,112]. Although epidemiologicaland experimental studies have shown transmission of <strong>in</strong>duced phenotypes betweengenerations, to date only one study has reported transmission of nutritionally <strong>in</strong>ducedepigenetic marks between generations [96]. The tendency towards obesity <strong>in</strong> A vy mice isexacerbated thorough successive generations [113]. Transmission of the obese phenotype wasprevented by supplementation of females with a methyl donors and cofactors, although thiswas not associated with a change <strong>in</strong> the methylation status of the A vy locus.The mechanism by which <strong>in</strong>duced epigenetic marks are transmitted to subsequent generationsis not known, although studies have begun to unpick the mechanisms <strong>in</strong>volved [114]. Whenthe transmission is only to the F 2 generation, a direct effect of the diet fed to the F 0 dams on


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>germ cells which gave rise to the F 2 offspr<strong>in</strong>g cannot be ruled out. Sequential transmissionfrom F 1 to F 2 , and possibly beyond, would <strong>in</strong>volve <strong>in</strong>duction <strong>in</strong> the germl<strong>in</strong>e of alteredepigenetic marks and such changes <strong>in</strong> DNA methylation would have to be preserved dur<strong>in</strong>ggenome-wide demethylation dur<strong>in</strong>g fertilization, possibly by a similar mechanism to thatwhich preserves the methylation of impr<strong>in</strong>ted genes and/or by targeted preservation ofnucleosome structure as occurs for specific developmental genes dur<strong>in</strong>g spermatogenesis[115]. An alternative possibility is that prenatal nutritional constra<strong>in</strong>t <strong>in</strong>duces physical orphysiological changes <strong>in</strong> the female which, <strong>in</strong> turn, restrict the <strong>in</strong>trauter<strong>in</strong>e environment <strong>in</strong>which her offspr<strong>in</strong>g develop. In this case, transmission of an altered phenotype betweengenerations would <strong>in</strong>volve <strong>in</strong>duction of changes <strong>in</strong> gene methylation de novo <strong>in</strong> eachgeneration. If so, the magnitude of the <strong>in</strong>duced effect, epigenetic or phenotypic, might differbetween generations.31015.6.2 Mechanism of Epigenetic ChangeThe processes by which environmental cues <strong>in</strong>duce altered epigenetic regulation <strong>in</strong> the embryorema<strong>in</strong> unknown. Studies <strong>in</strong> liver from juvenile offspr<strong>in</strong>g have, however, provided some<strong>in</strong>sights. Feed<strong>in</strong>g a PR diet to pregnant rats <strong>in</strong>duced lower Dnmt1 expression and reducedb<strong>in</strong>d<strong>in</strong>g of Dnmt1 at the GR promoter [94]. However, the expression of Dnmt3a, Dnmt3b, andMBD-2, and b<strong>in</strong>d<strong>in</strong>g of Dnmt3a at the GR promoter were unaltered [94]. This suggests thathypomethylation of the hepatic GR promoter <strong>in</strong> the offspr<strong>in</strong>g, and probably other genes<strong>in</strong>clud<strong>in</strong>g PPARa, is <strong>in</strong>duced by reduced capacity to ma<strong>in</strong>ta<strong>in</strong> patterns of cytos<strong>in</strong>e methylationdur<strong>in</strong>g mitosis rather than failure of methylation de novo or active demethylation [94,99].This is consistent with lower MeCP2 b<strong>in</strong>d<strong>in</strong>g and <strong>in</strong>creased histone modifications whichfacilitate transcription at the GR promoter. Reduced Dnmt1 activity might be expected to result<strong>in</strong> global demethylation. However, studies <strong>in</strong> vitro show loss of Dnmt1-<strong>in</strong>duced demethylationof only a subset of genes [116,117]. This <strong>in</strong>dicates that Dnmt1 is targeted to specific genes,consistent with selective hypomethylation <strong>in</strong> the liver <strong>in</strong> the PR offspr<strong>in</strong>g [44]. Dnmt1 activityis also required for progression through mitosis [118] and its expression is substantiallyreduced <strong>in</strong> non-proliferat<strong>in</strong>g cells [119]. Thus, suppression of Dnmt1 activity <strong>in</strong> the preimplantationperiod could also account for the changes <strong>in</strong> the number of cell types dur<strong>in</strong>g earlyembryonic development <strong>in</strong> this model [120]. However, recent studies have shown that 5-methylcytos<strong>in</strong>e (5mC) is not the only epigenetic mark of DNA itself, and the role of TET (Teneleven-translocation)prote<strong>in</strong>s must also be considered. Tet1, is an enzyme which catalyzes theconversion of 5-methylcytos<strong>in</strong>e (5mC) to 5-hydroxymethylcytos<strong>in</strong>e [121,122] and hastherefore been considered as a promis<strong>in</strong>g candidate for demethylation. The discovery of 5hmC<strong>in</strong> the mouse cerebellum [123] and <strong>in</strong> embryonic stem cells (ESCs) [124] has led to thehypothesis that 5hmC may be an <strong>in</strong>termediate <strong>in</strong> the removal of 5mC . Studies have shownthat 5hmC levels across the genome are low, consistent with the hypothesis that these may beshort-lived. Alternatively, 5hmC may be an epigenetic modification <strong>in</strong> its own right, attract<strong>in</strong>gits own chromat<strong>in</strong> or transcriptional modifications. The mark is significantly enriched <strong>in</strong> CpGd<strong>in</strong>ucleotides with<strong>in</strong> genes, particularly at exons and this has been found to be associated withgene expression as well as polycomb-mediated silenc<strong>in</strong>g [125]. Genome-wide profil<strong>in</strong>gmethods have also shown that the distribution of 5hmC is dist<strong>in</strong>ct to that of 5mC [125]. InESCs Tet1 is the primary Tet enzyme and is repressed follow<strong>in</strong>g differentiation <strong>in</strong>to embryoidbodies, which correlates with a reduction <strong>in</strong> 5hmC levels [124]. High levels of Tet1 <strong>in</strong>primordial germ cells have also been observed [126] suggest<strong>in</strong>g that Tet1 is associated with thepluripotent state.15.6.3 Animal Models of Maternal Nutrition and Epigenetic AlterationsIt is clear that some <strong>in</strong>dividuals are more predisposed than others to obesity-associateddiseases. It is difficult to identify those <strong>in</strong>dividuals most at risk and those who would mostbenefit from <strong>in</strong>dividualized monitor<strong>in</strong>g and care. It is important to remember that all fat is not


CHAPTER 15Epigenetic Approaches to Control Obesityequal and that the site of fat accumulation can have important implications [127]. In the worst<strong>in</strong>stances preferential accumulation of fat occurs <strong>in</strong> visceral adipose tissue and ectopic fatdeposition <strong>in</strong> <strong>in</strong>sul<strong>in</strong>-sensitive tissues such as muscle, liver, and pancreas, which correlatesstrongly with severe generalized <strong>in</strong>sul<strong>in</strong> resistance due to the development of a chronic<strong>in</strong>flammatory state partly due to <strong>in</strong>filtration of adipose tissue by macrophages.Supplementation of the maternal PR diet with glyc<strong>in</strong>e [128] or folic acid [33] has been shownto prevent the <strong>in</strong>duction of hypertension and endothelial dysfunction <strong>in</strong> the offspr<strong>in</strong>g. Folicacid supplementation of the PR diet was also shown [50] to prevent dyslipidemia <strong>in</strong> the adultoffspr<strong>in</strong>g, <strong>in</strong> contrast to the same supplementation of the control diet, which <strong>in</strong>ducedimpaired endothelial dysfunction and dyslipidemia <strong>in</strong> the offspr<strong>in</strong>g [33,50]. Earlier studieshave shown that this supplementation prevents hypomethylation of the PPARa and GRpromoters <strong>in</strong> the liver of offspr<strong>in</strong>g. A more detailed analysis of the promoters of these genesshowed that an <strong>in</strong>crease <strong>in</strong> maternal folic acid <strong>in</strong>take <strong>in</strong>duced subtle changes <strong>in</strong> gene regulationand altered the methylation of <strong>in</strong>dividual CpGs dependent on the supplementation given[95]. Animal studies have also identified puberty as a key time for <strong>in</strong>tervention. Folic acidsupplementation of the diet of rats dur<strong>in</strong>g their juvenile-pubertal period [129] was found to<strong>in</strong>duce impaired lipid homeostasis <strong>in</strong> addition to <strong>in</strong>creased weight ga<strong>in</strong>. These effects wereseen irrespective of the maternal diet given and were associated with altered methylation statusof specific genes <strong>in</strong> the liver.These observations are supportive of the view that puberty is a time of <strong>in</strong>creased <strong>in</strong>stability ofthe epigenome. In this particular <strong>in</strong>stance the phenotypic changes were undesirable. However,this study highlights the ability to alter effects of prenatal nutrition with <strong>in</strong>terventions dur<strong>in</strong>gpuberty. Studies carried out by Waterland and colleagues on a mouse model of obesity [113]were also able to demonstrate that obesity <strong>in</strong> offspr<strong>in</strong>g could be prevented by appropriatesupplementation of the maternal diet. The mouse A vy allele results from a transposition ofa mur<strong>in</strong>e <strong>in</strong>tracisternal A particle retrotransposon upstream of the agouti gene. The agoutisignal<strong>in</strong>g molecule <strong>in</strong>duces yellow pigmentation <strong>in</strong> the hair follicles as well as antagoniz<strong>in</strong>gsatiety signal<strong>in</strong>g at the melanocort<strong>in</strong> 4 receptor <strong>in</strong> the hypothalamus; as a result the mice haveyellow coats and are prone to hyperphagic obesity. In these studies the altered A v/y allele waspassed through three successive generations of A vy /a females and a cumulative effect on coatcolor and obesity was observed. This study was carried out on two populations of mice <strong>in</strong>parallel; one fed a standard diet, the other a methyl-supplemented diet that <strong>in</strong>duces DNAhypermethylation dur<strong>in</strong>g development. The work found that maternal obesity could causetransgenerational amplification of <strong>in</strong>creased body weight and that a methyl-supplementeddiet was able to prevent this effect. This confirms that epigenetic mechanisms such as methylationplay a role <strong>in</strong> the transgenerational <strong>in</strong>creases <strong>in</strong> mammalian obesity, but also providesevidence that dietary <strong>in</strong>tervention dur<strong>in</strong>g pregnancy to prevent obesity is possible. These <strong>in</strong>itialstudies po<strong>in</strong>t to the need for further work to determ<strong>in</strong>e whether <strong>in</strong>creased adiposity occurs asa result of <strong>in</strong>creased energy <strong>in</strong>take, decreased energy expenditure, or both. For this <strong>in</strong>formationto be of cl<strong>in</strong>ical value further studies will be required to elucidate a causal relationship betweenDNA methylation and obesity. Hav<strong>in</strong>g an understand<strong>in</strong>g of the epigenetic mechanisms whichunderlie the observed <strong>in</strong>crease <strong>in</strong> obesity presents the opportunity to prevent or reverse further<strong>in</strong>creases <strong>in</strong> obesity.311Among the best-characterized of the animal models of <strong>in</strong>tervention is neonatal lept<strong>in</strong> treatment.Lept<strong>in</strong> is produced by white adipose tissue and plays a key role <strong>in</strong> ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g bodyweight homeostasis [130]. Lept<strong>in</strong> was <strong>in</strong>itially proposed as an antiobesity therapy to reducefood <strong>in</strong>take. However, measurement of serum lept<strong>in</strong> levels <strong>in</strong> obese subjects showed thatcirculat<strong>in</strong>g lept<strong>in</strong> levels were <strong>in</strong> most cases elevated, <strong>in</strong> keep<strong>in</strong>g with a state of lept<strong>in</strong> resistance.More recent studies have shown that lept<strong>in</strong> has a broader range of functions than first thoughtand that it is particularly important dur<strong>in</strong>g growth and development. Lept<strong>in</strong> measurements <strong>in</strong>the serum of mice show that lept<strong>in</strong> levels drop dur<strong>in</strong>g <strong>in</strong>trauter<strong>in</strong>e and early postnatal life


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>before <strong>in</strong>creas<strong>in</strong>g 5e10-fold at postnatal days 5e10. Breast milk conta<strong>in</strong>s lept<strong>in</strong> and it isthought that this may contribute to the circulat<strong>in</strong>g levels <strong>in</strong> the neonate. However, the sourceof this lept<strong>in</strong> surge is controversial with work <strong>in</strong> rodents suggest<strong>in</strong>g that it is derived entirelyfrom the develop<strong>in</strong>g neonate [131]. Cord blood lept<strong>in</strong> levels reflect neonatal fat mass and lowcord blood lept<strong>in</strong> levels are associated with rapid postnatal weight ga<strong>in</strong> <strong>in</strong> small-forgestational-age<strong>in</strong>fants. Studies carried out by Vickers et al. <strong>in</strong> New Zealand [132] exam<strong>in</strong>edneonatal lept<strong>in</strong> treatment of rats <strong>in</strong> whom maternal undernutrition dur<strong>in</strong>g pregnancy results<strong>in</strong> offspr<strong>in</strong>g obesity, hyper<strong>in</strong>sul<strong>in</strong>emia, and hyperlept<strong>in</strong>emia, especially <strong>in</strong> the presence ofa high-fat postnatal diet. Treatment with lept<strong>in</strong> from postnatal days 3e13 resulted <strong>in</strong> a slow<strong>in</strong>gof neonatal weight ga<strong>in</strong>, particularly <strong>in</strong> programmed offspr<strong>in</strong>g, and normalized calorie <strong>in</strong>take,locomoter activity, body weight, fat mass, <strong>in</strong>sul<strong>in</strong> and lept<strong>in</strong> concentrations. This was <strong>in</strong>contrast to the control animals that were given a sal<strong>in</strong>e substitute, which were observed todevelop all of the features listed above. This study was able to demonstrate that the effects ofdevelopmental programm<strong>in</strong>g are potentially reversible if <strong>in</strong>tervention is made dur<strong>in</strong>g a periodof developmental plasticity, <strong>in</strong> this <strong>in</strong>stance the neonatal period.31215.6.4 <strong>Human</strong> Studies of Maternal Nutrition and Epigenetic AlterationsAt present there are sparse data l<strong>in</strong>k<strong>in</strong>g maternal nutrition to epigenetic changes <strong>in</strong> humanoffspr<strong>in</strong>g. A study of genomic DNA prepared from the blood of adults who were <strong>in</strong> uterodur<strong>in</strong>g the Dutch Hunger W<strong>in</strong>ter has revealed differences <strong>in</strong> the methylation of the differentiallymethylated region of the impr<strong>in</strong>ted <strong>in</strong>sul<strong>in</strong>-like growth factor-2 gene (IGF2 DMR). Thisstudy has revealed that adults who were <strong>in</strong> utero dur<strong>in</strong>g the fam<strong>in</strong>e have this region of the genehypomethylated. Comparisons made us<strong>in</strong>g same-sex sibl<strong>in</strong>gs whose gestation was unaffectedby the fam<strong>in</strong>e reveal that the mean level of methylation of this region was 52% <strong>in</strong> exposed<strong>in</strong>dividuals as compared to 49% <strong>in</strong> those who were unexposed [133]. Further studies [134] ofpeople exposed to the Dutch Hunger W<strong>in</strong>ter fam<strong>in</strong>e have provided evidence that periconceptionalfam<strong>in</strong>e exposure may alter methylation of the promoter region of impr<strong>in</strong>ted andnon-impr<strong>in</strong>ted genes which are implicated <strong>in</strong> growth and metabolic disease such as theimpr<strong>in</strong>ted genes INSIGN and MEG3 (which were hypo- and hypermethylated, respectively).Some evidence of altered methylation <strong>in</strong> non-impr<strong>in</strong>ted genes was observed. However,differences between unexposed and exposed subjects were very small and with<strong>in</strong> the range oferror for the technique used to measure methylation. A recent study look<strong>in</strong>g at the wholeblood methylation levels of the IGF2 DMR <strong>in</strong> the children of mothers who took a 400 mgsupplement of folic acid dur<strong>in</strong>g pregnancy was found to be 49.5% as compared to 47.4% <strong>in</strong>those mothers not tak<strong>in</strong>g the supplement. This study provides further evidence that <strong>in</strong>humans, maternal nutrition can have an effect on the epigenetic process and levels ofmethylation <strong>in</strong> the fetus [135]. Studies of patients with hyperhomocyste<strong>in</strong>emia have also beensupportive of the notion that folate therapy can alter methylation status of specific genes.Hyperhomocyste<strong>in</strong>emia (def<strong>in</strong>ed as a blood homocyste<strong>in</strong>e concentration above 15 mmol/l) isassociated with <strong>in</strong>creased risk of thrombosis, myocardial <strong>in</strong>farction, and stroke and is knownto occur <strong>in</strong> patients with several genetically determ<strong>in</strong>ed disorders as well as be<strong>in</strong>g highlyprevalent <strong>in</strong> patients with uremia. Comparison of methylation levels <strong>in</strong> leukocytes of normalversus diseased patients revealed that total DNA hypomethylation was <strong>in</strong>creased <strong>in</strong> hyperhomocyste<strong>in</strong>emia;however, treatment of patients us<strong>in</strong>g folate therapy was found to restoremethylation to normal levels <strong>in</strong> addition to correct<strong>in</strong>g patterns of gene expression [136].15.7 IDENTIFICATION OF PREDICTIVE EPIGENETIC MARKERS OFFUTURE OBESITYThe evidence to date <strong>in</strong> both animal models and <strong>in</strong> humans suggests that the early-life environment,particularly variations <strong>in</strong> nutrition, can <strong>in</strong>duce epigenetic alterations <strong>in</strong> the fetuswhich then persist throughout the lifecourse, lead<strong>in</strong>g to long-term changes <strong>in</strong> gene expression


CHAPTER 15Epigenetic Approaches to Control ObesityFIGURE 15.1Non-communicable diseases (NCDs) do not fit the medicalmodel <strong>in</strong> which an <strong>in</strong>dividual is healthy until they contract thedisease. Risk <strong>in</strong>creases throughout the lifecourse as a result ofdecl<strong>in</strong><strong>in</strong>g plasticity (green triangle) and the result<strong>in</strong>gaccumulative effects of <strong>in</strong>adequate responses to new challenges(brown triangle). However, although the greatest <strong>in</strong>crease occurs<strong>in</strong> adult life, the trajectory is set much earlier, be<strong>in</strong>g <strong>in</strong>fluencedby factors such as the mother’s diet and body compositionbefore and dur<strong>in</strong>g pregnancy, and fetal, <strong>in</strong>fant, and childhoodnutrition and development. Adopt<strong>in</strong>g a lifecourse perspectiveallows identification of phenotype and markers of risk early, withthe possibility of nutritional and other lifestyle <strong>in</strong>terventions.Timely, relatively modest <strong>in</strong>terventions <strong>in</strong> early life (red area) canhave a large effect on disease risk later (red arrow), while later<strong>in</strong>tervention (p<strong>in</strong>k area) can rema<strong>in</strong> impactful for vulnerablegroups (p<strong>in</strong>k arrow). Early-life preventive measures requirea long-term <strong>in</strong>vestment, but are more likely to be effective thanpopulation screen<strong>in</strong>g programs that identify the early stages ofdisease or treatments <strong>in</strong>itiated after the disease is manifest. Thisfigure is reproduced <strong>in</strong> the color plate section.and phenotype and an altered susceptibility to disease. Characterization of such alteredepigenetic marks <strong>in</strong> early life may allow the identification of <strong>in</strong>dividuals at risk of later obesity,enabl<strong>in</strong>g early <strong>in</strong>tervention and the development of new therapies. Such concepts arefundamental to current lifecourse strategies to the prevention and treatment of NCDs,<strong>in</strong>clud<strong>in</strong>g obesity (Figure 15.1).313Proof of concept for a potential role of epigenetic biomarkers <strong>in</strong> such a lifecourse approach hasrecently been published. Measurements of the epigenetic profile of a number of genes <strong>in</strong>umbilical cord tissue at birth were found [137] to predict phenotypic outcomes <strong>in</strong> childhood<strong>in</strong>dependent of birthweight. Greater methylation of a s<strong>in</strong>gle CpG with<strong>in</strong> the ret<strong>in</strong>oid Xreceptor alpha (RXRA) promoter measured <strong>in</strong> umbilical cord was strongly associated withgreater adiposity <strong>in</strong> later childhood. Per<strong>in</strong>atal measurements of DNA methylation expla<strong>in</strong>ed>25% of the variance <strong>in</strong> childhood adiposity. These f<strong>in</strong>d<strong>in</strong>gs were replicated <strong>in</strong> a second<strong>in</strong>dependent cohort. Given the complexities of ret<strong>in</strong>oid receptor biology, <strong>in</strong>creased RXRAmethylation might be act<strong>in</strong>g through a variety of pathways, but an association between<strong>in</strong>creased RXRA methylation and adiposity is consistent with the observation of stronglydim<strong>in</strong>ished RXRA expression <strong>in</strong> visceral white adipose tissue from obese mice [138]. Moreover,a role for ret<strong>in</strong>oid receptor methylation <strong>in</strong> developmental <strong>in</strong>fluences on later adiposity issupported by recent experimental data show<strong>in</strong>g an <strong>in</strong>fluence of maternal diet dur<strong>in</strong>g pregnancyon methylation of LXRA, a heterodimeric partner of RXRA [139].In the above human studies, associations were also observed between levels of RXRA methylationand the mothers’ carbohydrate <strong>in</strong>take <strong>in</strong> early pregnancy, supportive of the concept thatnutritional conditions <strong>in</strong> early pregnancy can affect a child’s adiposity <strong>in</strong> later life. The work hasprovided novel evidence for the importance of the developmental contribution to later adiposityand was able to clearly show that specific components of the epigenetic state at birth could beused to predict adiposity <strong>in</strong> later childhood. The associations between the methylation of thisCpG and both maternal diet and child’s phenotype are supportive of the notion that epigeneticprocesses are able to exert a f<strong>in</strong>e control on developmental outcomes and therefore theseepigenetic measurements taken at birth could have prognostic value. It is not known whethermethylation <strong>in</strong> readily available tissue such as blood, buccal, or <strong>in</strong> this case umbilical cord


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>reflects the levels of methylation <strong>in</strong> other tissue. In a recent study by Heijmans and collegues <strong>in</strong>the Netherlands [140] comparison of methylation of candidate loci <strong>in</strong> blood and buccal cellsfound that <strong>in</strong> half of the loci tested DNA methylation measured <strong>in</strong> blood was a marker for that<strong>in</strong> buccal cells, despite the fact that these cell types stem from different germ layers (mesodermand ectoderm, respectively). In other studies DNA methylation <strong>in</strong> blood has been shown toserve as a marker for methylation measured <strong>in</strong> colon tissue [141,142]. However there are clearlywell-documented tissue-specific differences <strong>in</strong> gene methylation observed and much moreresearch is required to determ<strong>in</strong>e whether methylation levels <strong>in</strong> blood, buccal, or cord mayprovide useful proxy markers of methylation <strong>in</strong> more metabolically relevant tissues and whethersuch marks can then be used as predictive markers of future disease risk.31415.8 CONCLUSIONSThere is now a considerable body of evidence to suggest that variations <strong>in</strong> the quality of theearly life environment <strong>in</strong>duce a differential risk of obesity and subsequent noncommunicablediseases <strong>in</strong> later life. Furthermore, elements of the heritable or familialcomponent of disease susceptibility may be transmitted by non-genomic means. Studieshave shown that the mechanisms <strong>in</strong>clude altered methylation of DNA and covalent modificationof histones. This non-genomic tun<strong>in</strong>g of the phenotype through developmentalplasticity has adaptive value because it attempts to match the <strong>in</strong>dividual’s responses to thepredicted future environment based on cues received dur<strong>in</strong>g development. When theresponses are mismatched the risk of subsequent obesity rises. Epigenetic processes such asthose <strong>in</strong>duced by the mother’s diet and body composition before and dur<strong>in</strong>g pregnancy anddur<strong>in</strong>g the early life of the child set the trajectory for obesity dur<strong>in</strong>g the lifecourse of the<strong>in</strong>dividual. The demonstration of a role for altered epigenetic regulation of genes <strong>in</strong> thedevelopmental <strong>in</strong>duction of obesity <strong>in</strong> early life and the identification of obesity biomarkerspo<strong>in</strong>ts to the possibility of nutritional or lifestyle <strong>in</strong>terventions or perhaps pharmacological<strong>in</strong>terventions which could modify long-term obesity risk and reverse the current epidemic ofobesity.References[1] World Health Organization. 2008e2013 Action Plan for the Global Strategy for the Prevention and Controlof Noncommunicable <strong>Disease</strong>s. World Health Organ 2011:2008.[2] Kelly T, Yang W, Chen CS, Reynolds K, He J. Global burden of obesity <strong>in</strong> 2005 and projections to 2030. Int JObes (Lond) 2008;32:1431e7.[3] Lobste<strong>in</strong> T, Baur L, Uauy R. Obesity <strong>in</strong> children and young people: a crisis <strong>in</strong> public health. Obes Rev2004;5(Suppl 1):4e104.[4] Garder G, Halweil B. Overfed and Underfed. The Global Epidemic of Malnutrition 2011.[5] International Obesity Taskforce. http, www.iaso.org/iotf/obesity; 2011.[6] Foresight Project, via the Department for Bus<strong>in</strong>ess, Innovation and Skills. http://www.bis.gov.uk/foresight;2011.[7] Williams J, Wake M, Hesketh K, Maher E, Waters E. Health-related quality of life of overweight and obesechildren. JAMA 2005;293:70e6.[8] Gortmaker SL, Must A, Perr<strong>in</strong> JM, Sobol AM, Dietz WH. Social and economic consequences of overweight <strong>in</strong>adolescence and young adulthood. N Engl J Med 1993;329:1008e12.[9] Ebbel<strong>in</strong>g CB, Pawlak DB, Ludwig DS. Childhood obesity: public-health crisis, common sense cure. Lancet2002;360:473e82.[10] Bhargava SK, Sachdev HS, Fall CH, Osmond C, Lakshmy R, Barker DJ, et al. Relation of serial changes <strong>in</strong>childhood body-mass <strong>in</strong>dex to impaired glucose tolerance <strong>in</strong> young adulthood. N Engl J Med2004;350:865e75.[11] Barker DJ. Intrauter<strong>in</strong>e programm<strong>in</strong>g of coronary heart disease and stroke. Acta Paediatr Suppl1997;423:178e82.[12] Schulz LC. The Dutch Hunger W<strong>in</strong>ter and the developmental orig<strong>in</strong>s of health and disease. Proc Natl Acad SciUSA 2010;107:16757e8.


CHAPTER 15Epigenetic Approaches to Control Obesity[13] Barker DJ, Bull AR, Osmond C, Simmonds SJ. Fetal and placental size and risk of hypertension <strong>in</strong> adult life.BMJ 1990;301:259e62.[14] Ong KK, Ahmed ML, Emmett PM, Preece MA, Dunger DB. Association between postnatal catch-up growthand obesity <strong>in</strong> childhood: prospective cohort study. BMJ 2000;320:967e71.[15] Fall CH, Barker DJ, Osmond C, W<strong>in</strong>ter PD, Clark PM, Hales CN. Relation of <strong>in</strong>fant feed<strong>in</strong>g to adult serumcholesterol concentration and death from ischaemic heart disease. BMJ 1992;304:801e5.[16] Oken E, Gillman MW. Fetal orig<strong>in</strong>s of obesity. Obes Res 2003;11:496e506.[17] Dorner G, Plagemann A. Per<strong>in</strong>atal hyper<strong>in</strong>sul<strong>in</strong>ism as possible predispos<strong>in</strong>g factor for diabetes mellitus,obesity and enhanced cardiovascular risk <strong>in</strong> later life. Horm Metab Res 1994;26:213e21.[18] Fraser A, Till<strong>in</strong>g K, Macdonald-Wallis C, Sattar N, Brion MJ, Benfield L, et al. Association of maternal weightga<strong>in</strong> <strong>in</strong> pregnancy with offspr<strong>in</strong>g obesity and metabolic and vascular traits <strong>in</strong> childhood. Circulation2010;121:2557e64.[19] Villamor E, Cnatt<strong>in</strong>gius S. Interpregnancy weight change and risk of adverse pregnancy outcomes: a population-basedstudy. Lancet 2006;368:1164e70.[20] Smith J, Cianflone K, Biron S, Hould FS, Lebel S, Marceau S, et al. Effects of maternal surgical weight loss <strong>in</strong>mothers on <strong>in</strong>tergenerational transmission of obesity. J Cl<strong>in</strong> Endocr<strong>in</strong>ol Metab 2009;94:4275e83.[21] Guilloteau P, Zabielski R, Hammon HM, Metges CC. Adverse effects of nutritional programm<strong>in</strong>g dur<strong>in</strong>gprenatal and early postnatal life, some aspects of regulation and potential prevention and treatments. JPhysiol Pharmacol 2009;60(Suppl 3):17e35.[22] Davey SG, Steer C, Leary S, Ness A. Is there an <strong>in</strong>trauter<strong>in</strong>e <strong>in</strong>fluence on obesity? Evidence from parent childassociations <strong>in</strong> the Avon Longitud<strong>in</strong>al Study of Parents and Children (ALSPAC). Arch Dis Child2007;92:876e80.[23] Lawlor DA, Timpson NJ, Harbord RM, Leary S, Ness A, McCarthy MI, et al. Explor<strong>in</strong>g the developmentalovernutrition hypothesis us<strong>in</strong>g parental-offspr<strong>in</strong>g associations and FTO as an <strong>in</strong>strumental variable. PLoSMed 2008;5:e33.[24] Li L, Law C, Lo CR, Power C. Intergenerational <strong>in</strong>fluences on childhood body mass <strong>in</strong>dex: the effect ofparental body mass <strong>in</strong>dex trajectories. Am J Cl<strong>in</strong> Nutr 2009;89:551e7.[25] Hales CN, Barker DJ. Type 2 (non-<strong>in</strong>sul<strong>in</strong>-dependent) diabetes mellitus: the thrifty phenotype hypothesis.Diabetologia 1992;35:595e601.[26] Rob<strong>in</strong>son R. The fetal orig<strong>in</strong>s of adult disease. BMJ 2001;322:375e6.[27] Modi N, Thomas EL, Harr<strong>in</strong>gton TA, Uthaya S, Dore CJ, Bell JD. Determ<strong>in</strong>ants of adiposity dur<strong>in</strong>gprewean<strong>in</strong>g postnatal growth <strong>in</strong> appropriately grown and growth-restricted term <strong>in</strong>fants. Pediatr Res2006;60:345e8.[28] Crowther NJ, Cameron N, Trusler J, Gray IP. Association between poor glucose tolerance and rapid post natalweight ga<strong>in</strong> <strong>in</strong> seven-year-old children. Diabetologia 1998;41:1163e7.[29] Iniguez G, Ong K, Bazaes R, Avila A, Salazar T, Dunger D, et al. Longitud<strong>in</strong>al changes <strong>in</strong> <strong>in</strong>sul<strong>in</strong>-like growthfactor-I, <strong>in</strong>sul<strong>in</strong> sensitivity, and secretion from birth to age three years <strong>in</strong> small-for-gestational-age children.J Cl<strong>in</strong> Endocr<strong>in</strong>ol Metab 2006;91:4645e9.[30] Ong KK, Loos RJ. Rapid <strong>in</strong>fancy weight ga<strong>in</strong> and subsequent obesity: systematic reviews and hopefulsuggestions. Acta Paediatr 2006;95:904e8.[31] Yajnik CS. Early life orig<strong>in</strong>s of <strong>in</strong>sul<strong>in</strong> resistance and type 2 diabetes <strong>in</strong> India and other Asian countries. J Nutr2004;134:205e10.[32] Fernandez-Tw<strong>in</strong>n DS, Wayman A, Ekizoglou S, Mart<strong>in</strong> MS, Hales CN, Ozanne SE. Maternal prote<strong>in</strong> restrictionleads to hyper<strong>in</strong>sul<strong>in</strong>emia and reduced <strong>in</strong>sul<strong>in</strong>-signal<strong>in</strong>g prote<strong>in</strong> expression <strong>in</strong> 21-mo-old female ratoffspr<strong>in</strong>g. Am J Physiol Regul Integr Comp Physiol 2005;288:R368e73.[33] Torrens C, Brawley L, Anthony FW, Dance CS, Dunn R, Jackson AA, et al. Folate supplementation dur<strong>in</strong>gpregnancy improves offspr<strong>in</strong>g cardiovascular dysfunction <strong>in</strong>duced by prote<strong>in</strong> restriction. Hypertension2006;47:982e7.[34] Calder PC, Yaqoob P. The level of prote<strong>in</strong> and type of fat <strong>in</strong> the diet of pregnant rats both affect lymphocytefunction <strong>in</strong> the offspr<strong>in</strong>g. Nutr Res 2000;20:995e1005.[35] Langley-Evans SC, Sculley DV. Programm<strong>in</strong>g of hepatic antioxidant capacity and oxidative <strong>in</strong>jury <strong>in</strong> theage<strong>in</strong>g rat. Mech Age<strong>in</strong>g Dev 2005;126:804e12.[36] Bell<strong>in</strong>ger L, Lilley C, Langley-Evans SC. Prenatal exposure to a maternal low-prote<strong>in</strong> diet programmesa preference for high-fat foods <strong>in</strong> the young adult rat. Br J Nutr 2004;92:513e20.[37] Bell<strong>in</strong>ger L, Sculley DV, Langley-Evans SC. Exposure to undernutrition <strong>in</strong> fetal life determ<strong>in</strong>es fat distribution,locomotor activity and food <strong>in</strong>take <strong>in</strong> age<strong>in</strong>g rats. Int J Obes (Lond) 2006;30:729e38.[38] Bertram CE, Hanson MA. Animal models and programm<strong>in</strong>g of the metabolic syndrome. Br Med Bull2001;60:103e21.315


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>316[39] Burns SP, Desai M, Cohen RD, Hales CN, Iles RA, Germa<strong>in</strong> JP, et al. Gluconeogenesis, glucose handl<strong>in</strong>g, andstructural changes <strong>in</strong> livers of the adult offspr<strong>in</strong>g of rats partially deprived of prote<strong>in</strong> dur<strong>in</strong>g pregnancy andlactation. J Cl<strong>in</strong> Invest 1997;100:1768e74.[40] Whorwood CB, Firth KM, Budge H, Symonds ME. Maternal undernutrition dur<strong>in</strong>g early to midgestationprograms tissue-specific alterations <strong>in</strong> the expression of the glucocorticoid receptor, 11beta-hydroxysteroiddehydrogenase isoforms, and type 1 angiotens<strong>in</strong> ii receptor <strong>in</strong> neonatal sheep. Endocr<strong>in</strong>ol2001;142:2854e64.[41] Brennan KA, Gopalakrishnan GS, Kurlak L, Rh<strong>in</strong>d SM, Kyle CE, Brooks AN, et al. Impact of maternalundernutrition and fetal number on glucocorticoid, growth hormone and <strong>in</strong>sul<strong>in</strong>-like growth factor receptormRNA abundance <strong>in</strong> the ov<strong>in</strong>e fetal kidney. Reprod 2005;129:151e9.[42] Gnanal<strong>in</strong>gham MG, Mostyn A, Dandrea J, Yakubu DP, Symonds ME, Stephenson T. Ontogeny and nutritionalprogramm<strong>in</strong>g of uncoupl<strong>in</strong>g prote<strong>in</strong>-2 and glucocorticoid receptor mRNA <strong>in</strong> the ov<strong>in</strong>e lung. J Physiol2005;565:159e69.[43] Bogdar<strong>in</strong>a I, Murphy HC, Burns SP, Clark AJ. Investigation of the role of epigenetic modification of the ratglucok<strong>in</strong>ase gene <strong>in</strong> fetal programm<strong>in</strong>g. Life Sci 2004;74:1407e15.[44] Lillycrop KA, Phillips ES, Jackson AA, Hanson MA, Burdge GC. Dietary prote<strong>in</strong> restriction of pregnant rats<strong>in</strong>duces and folic acid supplementation prevents epigenetic modification of hepatic gene expression <strong>in</strong> theoffspr<strong>in</strong>g. J Nutr 2005;135:1382e6.[45] Maloney CA, Gosby AK, Phuyal JL, Denyer GS, Bryson JM, Caterson ID. Site-specific changes <strong>in</strong> the expressionof fat-partition<strong>in</strong>g genes <strong>in</strong> weanl<strong>in</strong>g rats exposed to a low-prote<strong>in</strong> diet <strong>in</strong> utero. Obes Res 2003;11:461e8.[46] Lillycrop KA, Rodford J, Garratt ES, Slater-Jefferies JL, Godfrey KM, Gluckman PD, et al. Maternal prote<strong>in</strong>restriction with or without folic acid supplementation dur<strong>in</strong>g pregnancy alters the hepatic transcriptome <strong>in</strong>adult male rats. Br J Nutr 2010;103:1711e9.[47] Bertram C, Khan O, Ohri S, Phillips DI, Matthews SG, Hanson MA. Transgenerational effects of prenatalnutrient restriction on cardiovascular and hypothalamic-pituitary-adrenal function. J Physiol2008;586:2217e29.[48] Ozanne SE, Hales CN. Lifespan: catch-up growth and obesity <strong>in</strong> male mice. Nature 2004;427:411e2.[49] Zambrano E, Bautista CJ, Deas M, Mart<strong>in</strong>ez-Samayoa PM, Gonzalez-Zamorano M, Ledesma H, et al. A lowmaternal prote<strong>in</strong> diet dur<strong>in</strong>g pregnancy and lactation has sex- and w<strong>in</strong>dow of exposure-specific effects onoffspr<strong>in</strong>g growth and food <strong>in</strong>take, glucose metabolism and serum lept<strong>in</strong> <strong>in</strong> the rat. J Physiol2006;571:221e30.[50] Burdge GC, Lillycrop KA, Jackson AA, Gluckman PD, Hanson MA. The nature of the growth pattern and ofthe metabolic response to fast<strong>in</strong>g <strong>in</strong> the rat are dependent upon the dietary prote<strong>in</strong> and folic acid <strong>in</strong>takes oftheir pregnant dams and post-wean<strong>in</strong>g fat consumption. Br J Nutr 2008;99:540e9.[51] Woodall SM, Johnston BM, Breier BH, Gluckman PD. Chronic maternal undernutrition <strong>in</strong> the rat leads todelayed postnatal growth and elevated blood pressure of offspr<strong>in</strong>g. Pediatr Res 1996;40:438e43.[52] Vickers MH, Breier BH, Cutfield WS, Hofman PL, Gluckman PD. Fetal orig<strong>in</strong>s of hyperphagia, obesity, andhypertension and postnatal amplification by hypercaloric nutrition. Am J Physiol Endocr<strong>in</strong>ol Metab2000;279:E83e7.[53] K<strong>in</strong>d KL, Clifton PM, Katsman AI, Tsiounis M, Rob<strong>in</strong>son JS, Owens JA. Restricted fetal growth and theresponse to dietary cholesterol <strong>in</strong> the gu<strong>in</strong>ea pig. Am J Physiol 1999;277:R1675e82.[54] Hawk<strong>in</strong>s P, Steyn C, McGarrigle HH, Saito T, Ozaki T, Stratford LL, et al. Effect of maternal nutrient restriction<strong>in</strong> early gestation on responses of the hypothalamic-pituitary-adrenal axis to acute isocapnic hypoxaemia <strong>in</strong>late gestation fetal sheep. Exp Physiol 2000;85:85e96.[55] Gluckman PD, Lillycrop KA, Vickers MH, Pleasants AB, Phillips ES, Beedle AS, et al. Metabolic plasticitydur<strong>in</strong>g mammalian development is directionally dependent on early nutritional status. Proc Natl Acad SciUSA 2007;104:12796e800.[56] Guo F, Jen KL. High-fat feed<strong>in</strong>g dur<strong>in</strong>g pregnancy and lactation affects offspr<strong>in</strong>g metabolism <strong>in</strong> rats. PhysiolBehav 1995;57:681e6.[57] Samuelsson AM, Matthews PA, Argenton M, Christie MR, McConnell JM, Jansen EH, et al. Diet-<strong>in</strong>ducedobesity <strong>in</strong> female mice leads to offspr<strong>in</strong>g hyperphagia, adiposity, hypertension, and <strong>in</strong>sul<strong>in</strong> resistance: a novelmur<strong>in</strong>e model of developmental programm<strong>in</strong>g. Hypertension 2008;51:383e92.[58] Korotkova M, Gabrielsson BG, Holmang A, Larsson BM, Hanson LA, Strandvik B. Gender-related long-termeffects <strong>in</strong> adult rats by per<strong>in</strong>atal dietary ratio of n-6/n-3 fatty acids. Am J Physiol Regul Integr Comp Physiol2005;288:R575e9.[59] Kozak R, Burlet A, Burlet C, Beck B. Dietary composition dur<strong>in</strong>g fetal and neonatal life affects neuropeptideY function<strong>in</strong>g <strong>in</strong> adult offspr<strong>in</strong>g. Bra<strong>in</strong> Res Dev Bra<strong>in</strong> Res 2000;125:75e82.[60] Ng SF, L<strong>in</strong> RC, Laybutt DR, Barres R, Owens JA, Morris MJ. Chronic high-fat diet <strong>in</strong> fathers programs b-celldysfunction <strong>in</strong> female rat offspr<strong>in</strong>g. Nature 2010;467:963e6.


CHAPTER 15Epigenetic Approaches to Control Obesity[61] Khan IY, Dekou V, Douglas G, Jensen R, Hanson MA, Poston L, et al. A high-fat diet dur<strong>in</strong>g rat pregnancy orsuckl<strong>in</strong>g <strong>in</strong>duces cardiovascular dysfunction <strong>in</strong> adult offspr<strong>in</strong>g. Am J Physiol Regul Integr Comp Physiol2005;288:R127e33.[62] Plagemann A, Harder T, Rake A, Melchior K, Rittel F, Rohde W, et al. Hypothalamic <strong>in</strong>sul<strong>in</strong> and neuropeptideY <strong>in</strong> the offspr<strong>in</strong>g of gestational diabetic mother rats. Neuroreport 1998;9:4069e73.[63] Plagemann A, Heidrich I, Gotz F, Rohde W, Dorner G. Obesity and enhanced diabetes and cardiovascular risk<strong>in</strong> adult rats due to early postnatal overfeed<strong>in</strong>g. Exp Cl<strong>in</strong> Endocr<strong>in</strong>ol 1992;99:154e8.[64] Schmidt I, Fritz A, Scholch C, Schneider D, Simon E, Plagemann A. The effect of lept<strong>in</strong> treatment onthe development of obesity <strong>in</strong> overfed suckl<strong>in</strong>g Wistar rats. Int J Obes Relat Metab Disord2001;25:1168e74.[65] Davidowa H, Plagemann A. Decreased <strong>in</strong>hibition by lept<strong>in</strong> of hypothalamic arcuate neurons <strong>in</strong> neonatallyoverfed young rats. Neuroreport 2000;11:2795e8.[66] Plagemann A, Harder T, Melchior K, Rake A, Rohde W, Dorner G. Elevation of hypothalamic neuropeptideY-neurons <strong>in</strong> adult offspr<strong>in</strong>g of diabetic mother rats. Neuroreport 1999;10:3211e6.[67] Plagemann A, Harder T, Rake A, Melchior K, Rohde W, Dorner G. Increased number of galan<strong>in</strong>-neurons <strong>in</strong> theparaventricular hypothalamic nucleus of neonatally overfed weanl<strong>in</strong>g rats. Bra<strong>in</strong> Res 1999;818:160e3.[68] Plagemann A, Harder T, Janert U, Rake A, Rittel F, Rohde W, et al. Malformations of hypothalamic nuclei <strong>in</strong>hyper<strong>in</strong>sul<strong>in</strong>emic offspr<strong>in</strong>g of rats with gestational diabetes. Dev Neurosci 1999;21:58e67.[69] Plagemann A, Harder T, Rake A, Melchior K, Rittel F, Rohde W, et al. Hypothalamic <strong>in</strong>sul<strong>in</strong> and neuropeptideY <strong>in</strong> the offspr<strong>in</strong>g of gestational diabetic mother rats. Neuroreport 1998;9:4069e73.[70] Muhlhausler BS. Programm<strong>in</strong>g of the appetite-regulat<strong>in</strong>g neural network: a l<strong>in</strong>k between maternal overnutritionand the programm<strong>in</strong>g of obesity? J Neuroendocr<strong>in</strong>ol 2007;19:67e72.[71] Lee TM, Zucker I. Vole <strong>in</strong>fant development is <strong>in</strong>fluenced per<strong>in</strong>atally by maternal photoperiodic history. Am JPhysiol 1988;255:R831e8.[72] Gluckman PD, Hanson MA, Spencer HG. Predictive adaptive responses and human evolution. Trends EcolEvol 2005;20:527e33.[73] West-Eberhard MJ. Alternative adaptations, speciation, and phylogeny (A Review). Proc Natl Acad Sci USA1986;83:1388e92.[74] Gluckman PD, Hanson MA. The Fetal Matrix: Evolution, Developmental and <strong>Disease</strong>. Cambridge, UK:Cambridge University Press; 2005.[75] Godfrey KM, Inskip HM, Hanson MA. The long-term effects of prenatal development on growth andmetabolism. Sem<strong>in</strong> Reprod Med 2011;29:257e65.[76] Pigliucci M. Evolution of phenotypic plasticity: where are we go<strong>in</strong>g now? Trends Ecol Evol 2005;20:481e6.[77] Waterland RA, Jirtle RL. Transposable elements: targets for early nutritional effects on epigenetic generegulation. Mol Cell Biol 2003;23:5293e300.[78] Gluckman PD, Hanson MA, Beedle AS. Early life events and their consequences for later disease: a life historyand evolutionary perspective. Am J Hum Biol 2007;19:1e19.[79] Gluckman PD, Hanson MA, Beedle AS, Spencer HG. Predictive adaptive responses <strong>in</strong> perspective. TrendsEndocr<strong>in</strong>ol Metab 2008;19:109e10.[80] Bird A. DNA methylation patterns and epigenetic memory. Genes Dev 2002;16:6e21.[81] Bird A. Molecular biology: Methylation talk between histones and DNA. <strong>Science</strong> 2001;294:2113e5.[82] Morgan HD, Santos F, Green K, Dean W, Reik W. Epigenetic reprogramm<strong>in</strong>g <strong>in</strong> mammals. Hum Mol Genet2005;14 Spec No 1:R47e58.[83] Reik W, Dean W, Walter J. Epigenetic reprogramm<strong>in</strong>g <strong>in</strong> mammalian development. <strong>Science</strong> 2001;293:1089e93.[84] Fraga MF, Ballestar E, Paz MF, Ropero S, Setien F, Ballestar ML, et al. Epigenetic differences arise dur<strong>in</strong>g thelifetime of monozygotic tw<strong>in</strong>s. Proc Natl Acad Sci USA 2005;102:10604e9.[85] Wong CC, Caspi A, Williams B, Craig IW, Houts R, Ambler A, et al. A longitud<strong>in</strong>al study of epigeneticvariation <strong>in</strong> tw<strong>in</strong>s. <strong>Epigenetics</strong> 2010;5:516e26.[86] Khosla S, Dean W, Reik W, Feil R. Culture of preimplantation embryos and its long-term effects on geneexpression and phenotype. Hum Reprod Update 2001;7:419e27.[87] Cox GF, Burger J, Lip V, Mau UA, Sperl<strong>in</strong>g K, Wu BL, et al. Intracytoplasmic sperm <strong>in</strong>jection may <strong>in</strong>crease therisk of impr<strong>in</strong>t<strong>in</strong>g defects. Am J Hum Genet 2002;71:162e4.[88] Orstavik KH, Eiklid K, van der Hagen CB, Spetalen S, Kierulf K, Skjeldal O, et al. Another case of impr<strong>in</strong>t<strong>in</strong>gdefect <strong>in</strong> a girl with Angelman syndrome who was conceived by <strong>in</strong>tracytoplasmic semen <strong>in</strong>jection. Am J HumGenet 2003;72:218e9.[89] DeBaun MR, Niemitz EL, Fe<strong>in</strong>berg AP. Association of <strong>in</strong> vitro fertilization with BeckwitheWiedemannsyndrome and epigenetic alterations of LIT1 and H19. Am J Hum Genet 2003;72:156e60.317


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>318[90] Weaver IC, Cervoni N, Champagne FA, D’Alessio AC, Sharma S, Seckl JR, et al. Epigenetic programm<strong>in</strong>g bymaternal behavior. Nat Neurosci 2004;7:847e54.[91] Weaver IC, Champagne FA, Brown SE, Dymov S, Sharma S, Meaney MJ, et al. Reversal of maternalprogramm<strong>in</strong>g of stress responses <strong>in</strong> adult offspr<strong>in</strong>g through methyl supplementation: alter<strong>in</strong>g epigeneticmark<strong>in</strong>g later <strong>in</strong> life. J Neurosci 2005;25:11045e54.[92] Wolff GL, Kodell RL, Moore SR, Cooney CA. Maternal epigenetics and methyl supplements affect agouti geneexpression <strong>in</strong> Avy/a mice. FASEB J 1998;12:949e57.[93] Bird AP, Wolffe AP. Methylation-<strong>in</strong>duced repressionebelts, braces, and chromat<strong>in</strong>. Cell 1999;99:451e4.[94] Lillycrop KA, Slater-Jefferies JL, Hanson MA, Godfrey KM, Jackson AA, Burdge GC. Induction of alteredepigenetic regulation of the hepatic glucocorticoid receptor <strong>in</strong> the offspr<strong>in</strong>g of rats fed a prote<strong>in</strong>-restricted dietdur<strong>in</strong>g pregnancy suggests that reduced DNA methyltransferase-1 expression is <strong>in</strong>volved <strong>in</strong> impaired DNAmethylation and changes <strong>in</strong> histone modifications. Br J Nutr 2007;97:1064e73.[95] Lillycrop KA, Phillips ES, Torrens C, Hanson MA, Jackson AA, Burdge GC. Feed<strong>in</strong>g pregnant rats a prote<strong>in</strong>restricteddiet persistently alters the methylation of specific cytos<strong>in</strong>es <strong>in</strong> the hepatic PPAR alpha promoter ofthe offspr<strong>in</strong>g. Br J Nutr 2008;100:278e82.[96] Burdge GC, Slater-Jefferies J, Torrens C, Phillips ES, Hanson MA, Lillycrop KA. Dietary prote<strong>in</strong> restriction ofpregnant rats <strong>in</strong> the F0 generation <strong>in</strong>duces altered methylation of hepatic gene promoters <strong>in</strong> the adult maleoffspr<strong>in</strong>g <strong>in</strong> the F1 and F2 generations. Br J Nutr 2007;97:435e9.[97] Burdge GC, Slater-Jefferies JL, Grant RA, Chung WS, West AL, Lillycrop KA, et al. Sex, but not maternal prote<strong>in</strong>or folic acid <strong>in</strong>take, determ<strong>in</strong>es the fatty acid composition of hepatic phospholipids, but not of triacylglycerol,<strong>in</strong> adult rats. Prostagland<strong>in</strong>s Leukot Essent Fatty Acids 2008;78:73e9.[98] Lillycrop KA, Phillips ES, Jackson AA, Hanson MA, Burdge GC. Dietary prote<strong>in</strong> restriction <strong>in</strong> the pregnant rat<strong>in</strong>duces altered epigenetic regulation of the glucocorticoid receptor and peroxisomal proliferator-activatedreceptor alpha <strong>in</strong> the heart of the offspr<strong>in</strong>g which is prevented by folic acid. Proc Nutr Soc 2006;65:65A.[99] Burdge GC, Hanson MA, Slater-Jefferies JL, Lillycrop KA. Epigenetic regulation of transcription: a mechanismfor <strong>in</strong>duc<strong>in</strong>g variations <strong>in</strong> phenotype (fetal programm<strong>in</strong>g) by differences <strong>in</strong> nutrition dur<strong>in</strong>g early life? Br JNutr 2007;97:1036e46.[100] Bertram C, Trowern AR, Cop<strong>in</strong> N, Jackson AA, Whorwood CB. The maternal diet dur<strong>in</strong>g pregnancy programsaltered expression of the glucocorticoid receptor and type 2 11beta-hydroxysteroid dehydrogenase: potentialmolecular mechanisms underly<strong>in</strong>g the programm<strong>in</strong>g of hypertension <strong>in</strong> utero. Endocr<strong>in</strong>ol2001;142:2841e53.[101] Pr<strong>in</strong>tz MP, Jirout M, Jaworski R, Alemayehu A, Kren V. Genetic Models <strong>in</strong> Applied Physiology. HXB/BXH ratrecomb<strong>in</strong>ant <strong>in</strong>bred stra<strong>in</strong> platform: a newly enhanced tool for cardiovascular, behavioral, and developmentalgenetics and genomics. J Appl Physiol 2003;94:2510e22.[102] Krude H, Biebermann H, Gruters A. Mutations <strong>in</strong> the human proopiomelanocort<strong>in</strong> gene. Ann N Y Acad Sci2003;994:233e9.[103] Challis BG, Coll AP, Yeo GS, P<strong>in</strong>nock SB, Dickson SL, Thresher RR, et al. Mice lack<strong>in</strong>g pro-opiomelanocort<strong>in</strong>are sensitive to high-fat feed<strong>in</strong>g but respond normally to the acute anorectic effects of peptide-YY(3-36). ProcNatl Acad Sci USA 2004;101:4695e700.[104] Plagemann A, Harder T, Brunn M, Harder A, Roepke K, Wittrock-Staar M, et al. Hypothalamic proopiomelanocort<strong>in</strong>promoter methylation becomes altered by early overfeed<strong>in</strong>g: an epigenetic model of obesity andthe metabolic syndrome. J Physiol 2009;587:4963e76.[105] Torrens C, Poston L, Hanson MA. Transmission of raised blood pressure and endothelial dysfunction to theF2 generation <strong>in</strong>duced by maternal prote<strong>in</strong> restriction <strong>in</strong> the F0, <strong>in</strong> the absence of dietary challenge <strong>in</strong> the F1generation. Br J Nutr 2008;100:760e6.[106] Mart<strong>in</strong> JF, Johnston CS, Han CT, Benyshek DC. Nutritional orig<strong>in</strong>s of <strong>in</strong>sul<strong>in</strong> resistance: a rat model fordiabetes-prone human populations. J Nutr 2000;130:741e4.[107] Zambrano E, Mart<strong>in</strong>ez-Samayoa PM, Bautista CJ, Deas M, Guillen L, Rodriguez-Gonzalez GL, et al. Sex differences<strong>in</strong> transgenerational alterations of growth and metabolism <strong>in</strong> progeny (F2) of female offspr<strong>in</strong>g (F1) of rats feda low prote<strong>in</strong> diet dur<strong>in</strong>g pregnancy and lactation. J Physiol (Lond) 2005;566:225e36.[108] Benyshek DC, Johnston CS, Mart<strong>in</strong> JF. Glucose metabolism is altered <strong>in</strong> the adequately-nourished grandoffspr<strong>in</strong>g(F3 generation) of rats malnourished dur<strong>in</strong>g gestation and per<strong>in</strong>atal life. Diabetologia 2006;49:1117e9.[109] Drake AJ, Walker BR, Seckl JR. Intergenerational consequences of fetal programm<strong>in</strong>g by <strong>in</strong> utero exposure toglucocorticoids <strong>in</strong> rats. Am J Physiol Regul.Integr. Comp Physiol 2005;288:R34e8.[110] Sk<strong>in</strong>ner MK. What is an epigenetic transgenerational phenotype? F3 or F2. Reprod Toxicol 2008;25:2e6.[111] Anway MD, Sk<strong>in</strong>ner MK. Epigenetic programm<strong>in</strong>g of the germ l<strong>in</strong>e: effects of endocr<strong>in</strong>e disruptors on thedevelopment of transgenerational disease. Reprod Biomed Onl<strong>in</strong>e 2008;16:23e5.[112] Jablonka E. Inheritance systems and the evolution of new levels of <strong>in</strong>dividuality. J Theor Biol1994;170:301e9.


CHAPTER 15Epigenetic Approaches to Control Obesity[113] Waterland RA, Travisano M, Tahiliani KG, Rached MT, Mirza S. Methyl donor supplementation preventstransgenerational amplification of obesity. Int J Obes (Lond) 2008;32:1373e9.[114] Hoile SP, Lillycrop KA, Thomas NA, Hanson MA, Burdge GC. Dietary prote<strong>in</strong> restriction dur<strong>in</strong>g F0 pregnancy<strong>in</strong> rats <strong>in</strong>duces transgenerational changes <strong>in</strong> the hepatic transcriptome <strong>in</strong> female offspr<strong>in</strong>g. PLoS ONE 2011;6.e21668.[115] Hammoud SS, Nix DA, Zhang H, Purwar J, Carrell DT, Cairns BR. Dist<strong>in</strong>ctive chromat<strong>in</strong> <strong>in</strong> human spermpackages genes for embryo development. Nature 2009;460:473e8.[116] Jackson-Grusby L, Beard C, Possemato R, Tudor M, Fambrough D, Csankovszki G, et al. Loss of genomicmethylation causes p53-dependent apoptosis and epigenetic deregulation. Nat Genet 2001;27:31e9.[117] Rhee I, Jair KW, Yen RWC, Lengauer C, Herman JG, K<strong>in</strong>zler KW, et al. CpG methylation is ma<strong>in</strong>ta<strong>in</strong>ed <strong>in</strong>human cancer cells lack<strong>in</strong>g DNMT1. Nature 2000;404:1003e7.[118] Milut<strong>in</strong>ovic S, Zhuang Q, Niveleau A, Szyf M. Epigenomic stress response. Knockdown of DNA methyltransferase1 triggers an <strong>in</strong>tra-S-phase arrest of DNA replication and <strong>in</strong>duction of stress response genes. J BiolChem 2003;278:14985e95.[119] Suetake I, Shi LH, Watanabe D, Nakamura M, Tajima S. Proliferation stage-dependent expression of DNAmethyltransferase (Dnmt1) <strong>in</strong> mouse small <strong>in</strong>test<strong>in</strong>e. Cell Struct Funct 2001;26:79e86.[120] Kwong WY, Wild AE, Roberts P, Willis AC, Flem<strong>in</strong>g TP. Maternal undernutrition dur<strong>in</strong>g the preimplantationperiod of rat development causes blastocyst abnormalities and programm<strong>in</strong>g of postnatal hypertension.Development 2000;127:4195e202.[121] Ito S, D’Alessio AC, Taranova OV, Hong K, Sowers LC, Zhang Y. Role of Tet prote<strong>in</strong>s <strong>in</strong> 5mC to 5hmCconversion, ES-cell self-renewal and <strong>in</strong>ner cell mass specification. Nature 2010;466:1129e33.[122] Wu H, D’Alessio AC, Ito S, Xia K, Wang Z, Cui K, et al. Dual functions of Tet1 <strong>in</strong> transcriptional regulation <strong>in</strong>mouse embryonic stem cells. Nature 2011;473:389e93.[123] Kriaucionis S, He<strong>in</strong>tz N. The nuclear DNA base 5-hydroxymethylcytos<strong>in</strong>e is present <strong>in</strong> Purk<strong>in</strong>je neurons andthe bra<strong>in</strong>. <strong>Science</strong> 2009;324:929e30.[124] Tahiliani M, Koh KP, Shen Y, Pastor WA, Bandukwala H, Brudno Y, et al. Conversion of 5-methylcytos<strong>in</strong>e to5-hydroxymethylcytos<strong>in</strong>e <strong>in</strong> mammalian DNA by MLL partner TET1. <strong>Science</strong> 2009;324:930e5.[125] Wu H, Zhang Y. Tet1 and 5-hydroxymethylation: a genome-wide view <strong>in</strong> mouse embryonic stem cells. CellCycle 2011;10:2428e36.[126] Hajkova P, Jeffries SJ, Lee C, Miller N, Jackson SP, Surani MA. Genome-wide reprogramm<strong>in</strong>g <strong>in</strong> the mousegerm l<strong>in</strong>e entails the base excision repair pathway. <strong>Science</strong> 2010;329:78e82.[127] Walley AJ, Blakemore AI, Froguel P. Genetics of obesity and the prediction of risk for health. Hum Mol Genet2006;15 Spec No 2:R124e30.[128] Jackson AA, Dunn RL, Marchand MC, Langley-Evans SC. Increased systolic blood pressure <strong>in</strong> rats <strong>in</strong>duced bya maternal low-prote<strong>in</strong> diet is reversed by dietary supplementation with glyc<strong>in</strong>e. Cl<strong>in</strong> Sci (Lond)2002;103:633e9.[129] Burdge GC, Lillycrop KA, Phillips ES, Slater-Jefferies JL, Jackson AA, Hanson MA. Folic acid supplementationdur<strong>in</strong>g the juvenile-pubertal period <strong>in</strong> rats modifies the phenotype and epigenotype <strong>in</strong>duced by prenatalnutrition. J Nutr 2009;139:1054e60.[130] Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM. Positional clon<strong>in</strong>g of the mouse obesegene and its human homologue. Nature 1994;372:425e32.[131] Cottrell EC, Mercer JG, Ozanne SE. Postnatal development of hypothalamic lept<strong>in</strong> receptors. Vitam Horm2010;82:201e17.[132] Vickers MH, Gluckman PD, Coveny AH, Hofman PL, Cutfield WS, Gertler A, et al. Neonatal lept<strong>in</strong> treatmentreverses developmental programm<strong>in</strong>g. Endocr<strong>in</strong>ol 2005;146:4211e6.[133] Heijmans BT, Tobi EW, Ste<strong>in</strong> AD, Putter H, Blauw GJ, Susser ES, et al. Persistent epigenetic differencesassociated with prenatal exposure to fam<strong>in</strong>e <strong>in</strong> humans. Proc Natl Acad Sci USA 2008;105:17046e9.[134] Tobi EW, Lumey LH, Talens RP, Kremer D, Putter H, Ste<strong>in</strong> AD, et al. DNA methylation differencesafter exposure to prenatal fam<strong>in</strong>e are common and tim<strong>in</strong>g- and sex-specific. Hum Mol Genet2009;18:4046e53.[135] Steegers-Theunissen RP, Obermann-Borst SA, Kremer D, L<strong>in</strong>demans J, Siebel C, Steegers EA, et al. Periconceptionalmaternal folic acid use of 400 microg per day is related to <strong>in</strong>creased methylation of the IGF2gene <strong>in</strong> the very young child. PLoS ONE 2009;4. e7845.[136] Ingrosso D, Cimm<strong>in</strong>o A, Perna AF, Masella L, De Santo NG, et al. Folate treatment and unbalancedmethylation and changes of allelic expression <strong>in</strong>duced by hyperhomocyste<strong>in</strong>aemia <strong>in</strong> patients with uraemia.Lancet 2003;361:1693e9.[137] Godfrey KM, Sheppard A, Gluckman PD, Lillycrop KA, Burdge GC, McLean C, et al. Epigenetic gene promotermethylation at birth is associated with child’s later adiposity. Diabetes 2011;60:1528e34.319


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>[138] Lefebvre B, Benomar Y, Gued<strong>in</strong> A, Langlois A, Hennuyer N, Dumont J, et al. Proteasomal degradation ofret<strong>in</strong>oid X receptor alpha reprograms transcriptional activity of PPARgamma <strong>in</strong> obese mice and humans.J Cl<strong>in</strong> Invest 2010;120:1454e68.[139] van Straten EM, Bloks VW, Huijkman NC, Baller JF, van MH, et al. The liver X-receptor gene promoter ishypermethylated <strong>in</strong> a mouse model of prenatal prote<strong>in</strong> restriction. Am J Physiol Regul Integr Comp Physiol2010;298:R275e82.[140] Talens RP, Boomsma DI, Tobi EW, Kremer D, Jukema JW, Willemsen G, et al. Variation, patterns, andtemporal stability of DNA methylation: considerations for epigenetic epidemiology. FASEB J2010;24:3135e44.[141] Ally MS, Al-Ghnaniem R, Pufulete M. The relationship between gene-specific DNA methylation <strong>in</strong> leukocytesand normal colorectal mucosa <strong>in</strong> subjects with and without colorectal tumors. Cancer Epidemiol BiomarkersPrev 2009;18:922e8.[142] Cui H, Cruz-Correa M, Giardiello FM, Hutcheon DF, Kafonek DR, Brandenburg S, et al. Loss of IGF2impr<strong>in</strong>t<strong>in</strong>g: a potential marker of colorectal cancer risk. <strong>Science</strong> 2003;299:1753e5.[143] Herrera BM, Keildson S, L<strong>in</strong>dgren CM. Genetics and epigenetics of obesity. Maturitas 2011;69:41e9.320


CHAPTER 16<strong>Epigenetics</strong> of Diabetes<strong>in</strong> <strong>Human</strong>sCharlotte L<strong>in</strong>g, T<strong>in</strong>a RönnLund University, Malmö, SwedenCHAPTER OUTLINE16.1 Introduction 32116.2 Epigenetic Mechanisms 32216.3 <strong>Epigenetics</strong>, Insul<strong>in</strong> Secretion,and Diabetes 32216.4 <strong>Epigenetics</strong>, Insul<strong>in</strong> Resistance,and Diabetes 32516.5 Prospective 326References 32616.1 INTRODUCTIONDiabetes is characterized by chronic hyperglycemia and, accord<strong>in</strong>g to the World HealthOrganization (WHO), the def<strong>in</strong>ition of chronic hyperglycemia is a fast<strong>in</strong>g plasma glucoseconcentration 7.0 mmol/l and/or plasma glucose 11.1 mmol/l 2 hours after a 75-g oralglucose load measured on two occasions [1]. There are different subtypes of diabetes, the mostwell-characterized be<strong>in</strong>g type 1 diabetes, which is an autoimmune disease and type 2 diabetes,which is a polygenic multifactorial disease that develops due to impaired <strong>in</strong>sul<strong>in</strong> secretionfrom pancreatic beta cells <strong>in</strong> comb<strong>in</strong>ation with impaired <strong>in</strong>sul<strong>in</strong> action <strong>in</strong> target tissues,maturity-onset diabetes of the young (MODY), which is a group of monogenic forms ofdiabetes, latent autoimmune diabetes <strong>in</strong> adults (LADA) and maternally <strong>in</strong>herited diabetes anddeafness (MIDD). The prevalence of diabetes is <strong>in</strong>creas<strong>in</strong>g worldwide, particularly <strong>in</strong> develop<strong>in</strong>gcountries, and the disease has reached epidemic proportions [2]. Due to a sedentarylifestyle and an <strong>in</strong>creas<strong>in</strong>g age of many populations, type 2 diabetes is not only the mostcommon form of diabetes but also the subtype <strong>in</strong>creas<strong>in</strong>g the most. This chapter will ma<strong>in</strong>lyfocus on epigenetic mechanisms <strong>in</strong>fluenc<strong>in</strong>g the development of type 2 diabetes.321It is well established that comb<strong>in</strong>ations of non-genetic and genetic risk factors <strong>in</strong>fluence thesusceptibility for type 2 diabetes. A sedentary lifestyle, <strong>in</strong>clud<strong>in</strong>g high-calorie food <strong>in</strong>take,obesity, and physical <strong>in</strong>activity, as well as ag<strong>in</strong>g, represent non-genetic risk factors for type 2diabetes. The genetic contribution has been confirmed by family and tw<strong>in</strong> studies [3]. Moreover,recent genome-wide association studies have identified more than 40 polymorphismsassociated with an <strong>in</strong>creased risk for the disease [4e13]. Although most of the <strong>in</strong>itial genomewideanalyses were performed <strong>in</strong> Europeans, many of the identified polymorphisms do alsoconfer risk of type 2 diabetes <strong>in</strong> other populations [14,15]. There is further a grow<strong>in</strong>g body ofresearch suggest<strong>in</strong>g that epigenetic mechanisms may affect the pathogenesis of type 2 diabetesand this chapter will provide some <strong>in</strong>sights <strong>in</strong>to the role of epigenetics <strong>in</strong> type 2 diabetes.T. Tollefsbol (Ed): <strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>. DOI: 10.1016/B978-0-12-388415-2.00016-0Copyright Ó 2012 Elsevier Inc. All rights reserved.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>32216.2 EPIGENETIC MECHANISMSThe epigenome, <strong>in</strong>clud<strong>in</strong>g DNA methylation and histone modifications, plays a key role <strong>in</strong>controll<strong>in</strong>g the function of the genome. Even though different mammalian cell types <strong>in</strong> a bodyconta<strong>in</strong> the same genomic DNA, they have a cell-specific gene expression pattern <strong>in</strong>fluenc<strong>in</strong>gtheir phenotype. Epigenetic modifications <strong>in</strong>clude chemical modifications of both the DNAsequence itself and of the prote<strong>in</strong>s it is wrapped around, the histones, and are mechanismsused to control this cell-specific gene expression [16]. Cells do also use epigenetic modificationsfor parental impr<strong>in</strong>t<strong>in</strong>g, X-chromosome <strong>in</strong>activation, to regulate cell differentiationand to silence non-cod<strong>in</strong>g DNA regions. <strong>Epigenetics</strong> can be described as heritable changes <strong>in</strong>gene function that occur without a change <strong>in</strong> the nucleotide sequence and epigenetic modificationscan be transferred between one cell generation and the next (mitotic <strong>in</strong>heritance) andbetween generations (meiotic <strong>in</strong>heritance) [17]. However, although it is well-established thatepigenetic modifications can be <strong>in</strong>herited between generations <strong>in</strong> plants, there are onlya limited number of studies suggest<strong>in</strong>g that this is also the case <strong>in</strong> mammals [18e21].Indifferentiated mammalian cells, DNA methylation ma<strong>in</strong>ly takes place on cytos<strong>in</strong>e residues <strong>in</strong>CG d<strong>in</strong>ucleotides [22]. An <strong>in</strong>creased degree of DNA methylation has been associated withtranscriptional silenc<strong>in</strong>g through either repress<strong>in</strong>g the b<strong>in</strong>d<strong>in</strong>g of transcription factors to thepromoter regions or by the recruitment of prote<strong>in</strong>s that specifically b<strong>in</strong>d to methylated CGs,methyl-CG-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s, which can further recruit prote<strong>in</strong>s that close down the chromat<strong>in</strong>structure. Two groups of methyltransferases are responsible for DNA methylation: DNMT1,which ma<strong>in</strong>ta<strong>in</strong>s methylation dur<strong>in</strong>g replication through copy<strong>in</strong>g the DNA methylationpattern between cell generations, and DNMT3a and DNMT3b, which are responsible for denovo methylation. Histone modifications can be associated with either an active or repressivestate, depend<strong>in</strong>g on where the modifications take place [23]. Moreover, numerous differentenzymes are responsible for generat<strong>in</strong>g these histone modifications [24]. While emerg<strong>in</strong>g datademonstrate that the epigenome is dynamic and may change <strong>in</strong> response to environmentalexposures, <strong>in</strong>clud<strong>in</strong>g risk factors for type 2 diabetes, it is further possible that the epigeneticchanges <strong>in</strong>duced by today’s sedentary lifestyle may be <strong>in</strong>herited by future generations [25].16.3 EPIGENETICS, INSULIN SECRETION, AND DIABETESInsul<strong>in</strong> is a key hormone regulat<strong>in</strong>g metabolic homeostasis. The secretion of <strong>in</strong>sul<strong>in</strong> iscontrolled by fuel metabolism <strong>in</strong> pancreatic beta cells. When blood glucose levels rise, glucoseis transported <strong>in</strong>to the beta cells where it is metabolized to generate elevated ATP/ADP levels,which triggers exocytosis of <strong>in</strong>sul<strong>in</strong> [26]. Pancreatic <strong>in</strong>sul<strong>in</strong> secretion is determ<strong>in</strong>ed both bythe total beta cell mass and the function of each <strong>in</strong>dividual cell. Type 2 diabetes develops whenthe <strong>in</strong>sul<strong>in</strong> secretion is not sufficient to ma<strong>in</strong>ta<strong>in</strong> normoglycemia. Both genetic and nongeneticrisk factors are known to affect <strong>in</strong>sul<strong>in</strong> secretion [11e13,27e30]. Moreover, recentstudies from our group and others have exam<strong>in</strong>ed whether changes <strong>in</strong> DNA methylation andhistone modification may play <strong>in</strong> role <strong>in</strong> the regulation of <strong>in</strong>sul<strong>in</strong> secretion. Kuroda et al.hypothesized that s<strong>in</strong>ce the DNA region surround<strong>in</strong>g the <strong>in</strong>sul<strong>in</strong> gene is impr<strong>in</strong>ted, the geneencod<strong>in</strong>g <strong>in</strong>sul<strong>in</strong> may be regulated by DNA methylation [31]. They demonstrated that the<strong>in</strong>sul<strong>in</strong> promoter is demethylated <strong>in</strong> the <strong>in</strong>sul<strong>in</strong>-produc<strong>in</strong>g beta cells compared with other celltypes not express<strong>in</strong>g <strong>in</strong>sul<strong>in</strong>. Moreover, us<strong>in</strong>g a reporter gene construct they showed that<strong>in</strong>creased DNA methylation reduces the expression of the <strong>in</strong>sul<strong>in</strong> gene. In particular, DNAmethylation of a CpG site 182 bp upstream of the <strong>in</strong>sul<strong>in</strong> promoter, which is part of a cyclicadenos<strong>in</strong>e monophosphate (cAMP) responsive element (CRE), <strong>in</strong>fluences the expression ofthis gene. Mutskov et al. have further shown that the <strong>in</strong>sul<strong>in</strong> gene is part of a large openchromat<strong>in</strong> doma<strong>in</strong> <strong>in</strong> human islets and it displays high levels of histone modifications(hyperacetylation of H4 and dimethylation of H3 lys<strong>in</strong>e 4) typical of active genes [32,33]. Toexam<strong>in</strong>e if the epigenetic regulation of the <strong>in</strong>sul<strong>in</strong> gene plays a role <strong>in</strong> patients with type 2diabetes, we studied pancreatic islets from diabetic and non-diabetic donors as well as FACSsortedbeta and alpha cells from human islets [34]. We found that four CpG sites, located 234,


CHAPTER 16<strong>Epigenetics</strong> of Diabetes <strong>in</strong> <strong>Human</strong>s180, and 102 bp upstream and 63 bp downstream of the transcription start site respectively,showed <strong>in</strong>creased DNA methylation <strong>in</strong> islets from type 2 diabetic compared with non-diabetic<strong>in</strong>dividuals. Moreover glucose-stimulated <strong>in</strong>sul<strong>in</strong> secretion, <strong>in</strong>sul<strong>in</strong> content and <strong>in</strong>sul<strong>in</strong>expression were reduced <strong>in</strong> pancreatic islets from the patients with type 2 diabetes [34]. Wefurther showed that DNA methylation of the <strong>in</strong>sul<strong>in</strong> promoter was <strong>in</strong>creased <strong>in</strong> alphacompared with beta cells from human donors, propos<strong>in</strong>g that DNA methylation may play animportant role <strong>in</strong> regulat<strong>in</strong>g cell-specific <strong>in</strong>sul<strong>in</strong> gene expression. In agreement with thefunctional luciferase experiments performed by Kuroda et al., demonstrat<strong>in</strong>g a negative effectof DNA methylation on <strong>in</strong>sul<strong>in</strong> expression, we found a strong negative correlation between thelevel of methylation and <strong>in</strong>sul<strong>in</strong> gene expression <strong>in</strong> human pancreatic islets. As type 2 diabetesis characterized by hyperglycemia and patients with the disease often have elevated HbA1clevels, we also <strong>in</strong>vestigated the relation between <strong>in</strong>sul<strong>in</strong> DNA methylation and HbA1c. Indeed,the level of <strong>in</strong>sul<strong>in</strong> promoter DNA methylation <strong>in</strong> the human islets correlated positively withHbA1c levels, suggest<strong>in</strong>g that high levels of glucose may <strong>in</strong>crease methylation of the <strong>in</strong>sul<strong>in</strong>gene. This hypothesis was further confirmed <strong>in</strong> clonal beta cells, where cells exposed to highlevels of glucose for 48 hours showed <strong>in</strong>creased <strong>in</strong>sul<strong>in</strong> promoter DNA methylation comparedwith cells cultured <strong>in</strong> normal levels of glucose [34].Insul<strong>in</strong> secretion is <strong>in</strong>creased <strong>in</strong> response to nutrient stimulation and metabolism, which leadsto elevated ATP/ADP levels <strong>in</strong> the pancreatic beta cells. S<strong>in</strong>ce this process requires a sufficientmitochondrial metabolism and oxidative phosphorylation, it is of no surprise that mitochondrialdysfunctions have been implicated <strong>in</strong> impaired <strong>in</strong>sul<strong>in</strong> secretion and type 2 diabetes[11,26,29,35e37]. While common genetic variation near genes <strong>in</strong>fluenc<strong>in</strong>g oxidative phosphorylationis associated with impaired <strong>in</strong>sul<strong>in</strong> secretion, we have also shown that epigeneticvariation may reduce the expression of genes <strong>in</strong>volved <strong>in</strong> oxidative phosphorylation(OXPHOS genes) <strong>in</strong> human pancreatic islets [11,29,36,37]. The transcriptional coactivatorPGC-1a (encoded by PPARGC1A) regulates the expression of multiple genes with key functionswith<strong>in</strong> the mitochondria and it may hence affect ATP production and possibly <strong>in</strong>sul<strong>in</strong>secretion <strong>in</strong> pancreatic beta cells. Indeed, while the expression of PGC-1a correlates positivelywith <strong>in</strong>sul<strong>in</strong> secretion, it is reduced <strong>in</strong> pancreatic islets from patients with type 2 diabetes [37].We further found that DNA methylation of the PPARGC1A gene is <strong>in</strong>creased <strong>in</strong> pancreatic isletsfrom diabetic patients, propos<strong>in</strong>g that epigenetic modifications may affect gene expression andsubsequently <strong>in</strong>sul<strong>in</strong> secretion. In a different study, it was shown that the expression ofOXPHOS genes was reduced <strong>in</strong> islets from diabetic patients [36]. DNA methylation wasanalyzed <strong>in</strong> the promoter region of a subset of the OXPHOS genes show<strong>in</strong>g reduced expression,however, there were no differences <strong>in</strong> methylation of the studied genes due to diabetes.Nevertheless, a negative correlation was found between the level of expression and DNAmethylation for one OXPHOS gene, COX11. Future studies are needed to further dissect therole for DNA methylation <strong>in</strong> the regulation of gene expression and <strong>in</strong>sul<strong>in</strong> secretion <strong>in</strong> isletsfrom patients with type 2 diabetes.323Two recent studies have performed genome-wide analyses of histone modifications <strong>in</strong> humanpancreatic islets [38,39]. Bhandare et al. analyzed two histone marks associated with geneactivation (H3K4me1 and H3K4me2) and one histone mark associated with gene repression(H3K27me3) <strong>in</strong> human islets and identified relationships between these histone modificationsand gene expression. They further identified 18 polymorphisms, previously associatedwith type 2 diabetes, located with<strong>in</strong> 500 bp of an H3K4me1 GLITR region and suggest thats<strong>in</strong>ce H3K4me1 is frequently associated with enhancer regions, these SNPs have the potentialto affect gene expression [38]. Stitzel et al. did also analyze two histone marks associated withgene activation (H3K4me1 and H3K4me3) and one histone mark associated with generepression (H3K79me2) <strong>in</strong> human pancreatic islets and they propose to use their data asa snapshot of the epigenome <strong>in</strong> human islets [39]. A different method to analyze the chromat<strong>in</strong>structure is through the use of a formaldehyde-assisted isolation of regulatory elements(FAIRE) assay [40]. Us<strong>in</strong>g this approach, approximately 80 000 open chromat<strong>in</strong> sites <strong>in</strong>


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>human pancreatic islets have been identified. Interest<strong>in</strong>gly, one polymorphism previouslyassociated with type 2 diabetes and located close to the TCF7L2 gene was found to be located<strong>in</strong> one of these islet-selective open chromat<strong>in</strong> regions.324In humans, an adverse <strong>in</strong>trauter<strong>in</strong>e environment has been associated with an <strong>in</strong>creased risk fordiabetes and metabolic disease <strong>in</strong> postnatal life [41e48]. Inadequate nutrition may lead tochronic alterations <strong>in</strong> the body’s ability to ma<strong>in</strong>ta<strong>in</strong> metabolism, hormone levels, and the cellnumber of important organs [49]. Intrauter<strong>in</strong>e growth restriction could be due to maternal,placental, or genetic factors, and this perturbed environment <strong>in</strong> early life is thought to affectphysiological and cellular adaptive responses <strong>in</strong> key organs, as summarized <strong>in</strong> Figure 16.1. Theassociation with susceptibility of metabolic disease <strong>in</strong> adult life suggests permanent alterations,a cell memory, potentially mediated by epigenetic mechanisms tak<strong>in</strong>g place <strong>in</strong> utero [50,51].Supportively, rodents exposed to an adverse <strong>in</strong>trauter<strong>in</strong>e environment show impaired <strong>in</strong>sul<strong>in</strong>secretion and develop diabetes <strong>in</strong> adult life due to epigenetic modifications that take placedur<strong>in</strong>g embryonic development [52e55]. Pancreatic duodenal homeobox 1 (PDX-1) isa homeodoma<strong>in</strong>-conta<strong>in</strong><strong>in</strong>g transcription factor that plays a key role <strong>in</strong> pancreas developmentand function and patients with certa<strong>in</strong> mutations <strong>in</strong> PDX-1 develop a monogenic form ofdiabetes (MODY4) [56]. Knockout animals lack<strong>in</strong>g Pdx-1 expression <strong>in</strong> beta cells developdiabetes due to impaired <strong>in</strong>sul<strong>in</strong> secretion [57]. Moreover, <strong>in</strong>trauter<strong>in</strong>e growth retardation <strong>in</strong>rodents results <strong>in</strong> a decl<strong>in</strong>e <strong>in</strong> islet Pdx-1 expression and diabetes of the offspr<strong>in</strong>g [52]. Thisdecl<strong>in</strong>e <strong>in</strong> Pdx-1 expression is also associated with progressive changes <strong>in</strong> epigenetic modifications<strong>in</strong>clud<strong>in</strong>g histone modification and DNA methylation at the Pdx-1 locus. A recent studyshowed that when these animals were treated with Exend<strong>in</strong>-4, a long-act<strong>in</strong>g glucagon-likepeptide-1 (GLP-1) analog, <strong>in</strong> the newborn period, the development of diabetes was preventeddue to <strong>in</strong>creased islet Pdx-1 expression and epigenetic changes [55]. A genome-wide analysis ofDNA methylation <strong>in</strong> islets from rodents exposed to an <strong>in</strong>trauter<strong>in</strong>e growth retardation didfurther show epigenetic changes of genes regulat<strong>in</strong>g beta cell proliferation, <strong>in</strong>sul<strong>in</strong> secretion, andvascularization [54]. An additional candidate gene for type 2 diabetes and MODY is HNF4A. Amaternal low-prote<strong>in</strong> diet was associated with reduced Hnf4a expression and epigenetic changes<strong>in</strong> islets of rodent offspr<strong>in</strong>g [53]. This phenotype was further associated with impaired <strong>in</strong>sul<strong>in</strong>secretion and diabetes. In addition, this study showed that epigenetic modifications of Hnf4acontrolled the expression from tissue-specific promoters <strong>in</strong> both rodent and human islets.Overall, these studies demonstrate that epigenetic changes <strong>in</strong> pancreatic islets and beta cellsmay affect the expression of candidate genes for type 2 diabetes and hence <strong>in</strong>sul<strong>in</strong> secretionand risk for disease.FIGURE 16.1Intrauter<strong>in</strong>e environment, epigenetic programm<strong>in</strong>g, and thedevelopment of type 2 diabetes.


CHAPTER 16<strong>Epigenetics</strong> of Diabetes <strong>in</strong> <strong>Human</strong>s16.4 EPIGENETICS, INSULIN RESISTANCE, AND DIABETESInsul<strong>in</strong> resistance is a condition when cells <strong>in</strong> the body become unable to respond to normalamounts of <strong>in</strong>sul<strong>in</strong>. This results <strong>in</strong> impaired glucose clearance from the blood and failure tosuppress hepatic glucose production, both of which contribute to hyperglycemia. The majortarget organs for <strong>in</strong>sul<strong>in</strong> are skeletal muscle and the liver, as these are the sites where the majorglucose uptake occurs. Adipose tissue only accounts for a small proportion of glucose clearance,but is still important <strong>in</strong> ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g normoglycemia as <strong>in</strong>sul<strong>in</strong> resistance <strong>in</strong> fat cellsresults <strong>in</strong> <strong>in</strong>creased hydrolysis of triglycerides, which may further <strong>in</strong>crease <strong>in</strong>sul<strong>in</strong> resistance.Reduced oxidative capacity of the mitochondria <strong>in</strong> skeletal muscle has been suggested tocontribute to <strong>in</strong>sul<strong>in</strong> resistance and type 2 diabetes [58]. Moreover, the expression of genes<strong>in</strong>fluenc<strong>in</strong>g oxidative phosphorylation (OXPHOS genes) has been shown to be downregulated<strong>in</strong> skeletal muscle from patients with type 2 diabetes [59e62]. One of theseOXPHOS genes, COX7A1, was selected as a candidate to <strong>in</strong>vestigate the impact of genetic,epigenetic, and non-genetic factors on the gene expression <strong>in</strong> human skeletal muscle [63]. Thisstudy observed an age-related <strong>in</strong>crease <strong>in</strong> DNA methylation of the COX7A1 promoter as well asa decrease <strong>in</strong> mRNA expression of this gene <strong>in</strong> human skeletal muscle. Additionally, there wasa positive correlation between COX7A1 mRNA expression and <strong>in</strong>sul<strong>in</strong>-stimulated glucoseuptake and also a genetic component <strong>in</strong>fluenc<strong>in</strong>g gene expression. This proposes a mechanismfor regulation of COX7A1 <strong>in</strong> human skeletal muscle <strong>in</strong>volv<strong>in</strong>g age, epigenetic, and geneticvariation, which <strong>in</strong> turn could affect <strong>in</strong> vivo metabolism [63].NDUFB6 is another example of a gene, which encodes for a prote<strong>in</strong> of the respiratory cha<strong>in</strong>,with decreased gene expression <strong>in</strong> muscle from diabetic patients [59]. To determ<strong>in</strong>e whetherthis defect is <strong>in</strong>herited or acquired, our group <strong>in</strong>vestigated the association of genetic, epigenetic,and non-genetic factors with the mRNA expression of NDUFB6 [64]. We founda polymorphism (rs629566) <strong>in</strong> the NDUFB6 promoter region that was associated witha decl<strong>in</strong>e <strong>in</strong> muscle NDUFB6 expression with age. Young subjects carry<strong>in</strong>g the rs629566 G/Ggenotype exhibited higher muscle NDUFB6 expression, but this genotype was associated withreduced expression <strong>in</strong> elderly subjects. This was expla<strong>in</strong>ed by the f<strong>in</strong>d<strong>in</strong>g of <strong>in</strong>creased DNAmethylation <strong>in</strong> the promoter of elderly, but not young subjects, carry<strong>in</strong>g the G/G genotype.Moreover, this polymorphism <strong>in</strong>troduced a CG d<strong>in</strong>ucleotide <strong>in</strong>troduc<strong>in</strong>g a possible DNAmethylation site. The degree of DNA methylation correlated negatively with muscle NDUFB6expression, which <strong>in</strong> turn was associated with <strong>in</strong>sul<strong>in</strong> sensitivity. This provides an example ofhow genetic and epigenetic factors may <strong>in</strong>teract to <strong>in</strong>crease age-dependent susceptibility to<strong>in</strong>sul<strong>in</strong> resistance [64].325PPARGC1A is a key regulator of the OXPHOS genes, and it seems to be differentiallymethylated not only <strong>in</strong> pancreatic islets, hav<strong>in</strong>g a role <strong>in</strong> <strong>in</strong>sul<strong>in</strong> secretion [37], but also <strong>in</strong>human skeletal muscle, possibly contribut<strong>in</strong>g to <strong>in</strong>sul<strong>in</strong> resistance [65,66]. The study by Bronset al. showed that when <strong>in</strong>dividuals born with a low birth weight are challenged with a high-fatdiet they develop <strong>in</strong>sul<strong>in</strong> resistance and reduced expression of both PPARGC1A and downstreamOXPHOS genes. A novel f<strong>in</strong>d<strong>in</strong>g <strong>in</strong> this study was that <strong>in</strong>dividuals with a low birthweight had a constitutive <strong>in</strong>crease <strong>in</strong> PPARGC1A promoter DNA methylation, whereas <strong>in</strong>contrast, those born with a normal birth weight were able to reverse the changes <strong>in</strong> DNAmethylation <strong>in</strong>duced by the high-fat diet. Supportively, a genome-wide promoter screen<strong>in</strong>gof DNA methylation demonstrated epigenetic modifications of the PPARGC1A promoter <strong>in</strong>skeletal muscle from patients with type 2 diabetes [65]. Their methylation levels correlatednegatively with PPARGC1A mRNA and mitochondrial DNA, and <strong>in</strong>terest<strong>in</strong>gly the highestproportion of cytos<strong>in</strong>e methylation was found <strong>in</strong> non-CpG nucleotides.An adverse <strong>in</strong>trauter<strong>in</strong>e environment could affect not only <strong>in</strong>sul<strong>in</strong> secretion, but also <strong>in</strong>sul<strong>in</strong>resistance (Figure 16.1). One potential explanation for this is that epigenetic mechanisms maydrive a state that is beneficial for the fetus, for example <strong>in</strong>sul<strong>in</strong> resistance, which <strong>in</strong> adult life


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>facilitates the development of type 2 diabetes and the metabolic syndrome when exposed to anobesogenic environment [67]. <strong>Human</strong> studies <strong>in</strong> this area are still sparse, but recent animalstudies show promis<strong>in</strong>g support of this idea. For example Lillycrop et al. have shown a model<strong>in</strong> rats where a maternal low-prote<strong>in</strong> diet <strong>in</strong>duces persistent, gene-specific epigenetic changesthat alter mRNA expression <strong>in</strong> the liver. These changes can be reversed by folic acid supplementationdur<strong>in</strong>g pregnancy, suggest<strong>in</strong>g that changes <strong>in</strong> DNA methylation may reflect animpaired supply of folic acid from the mother [68e70]. Dietary prote<strong>in</strong> restriction of pregnantrats <strong>in</strong>duces, and folic acid supplementation prevents, epigenetic modification of hepatic geneexpression <strong>in</strong> the offspr<strong>in</strong>g. In humans, the Dutch Hunger W<strong>in</strong>ter provides an example wherethe offspr<strong>in</strong>g to pregnant women exposed to fam<strong>in</strong>e show <strong>in</strong>creases <strong>in</strong> <strong>in</strong>sul<strong>in</strong> levels,suggest<strong>in</strong>g an association with <strong>in</strong>sul<strong>in</strong> resistance [41]. Furthermore, <strong>in</strong> those <strong>in</strong>dividualsexposed to fam<strong>in</strong>e, changes <strong>in</strong> DNA methylation were present six decades later, <strong>in</strong>clud<strong>in</strong>gdecreased DNA methylation <strong>in</strong> the promoters of IGF2 and INSIGF, and <strong>in</strong>creased DNAmethylation <strong>in</strong> the promoters of IL10, LEP, ABCA1, GNASAS, and MEG3 [50,51].32616.5 PROSPECTIVEAlthough there has been substantial progress <strong>in</strong> the field of epigenetics <strong>in</strong> human diabetes,additional genome-wide and functional studies are needed to fully dissect andunderstand the impact of epigenetic modifications <strong>in</strong> the pathogenesis of the disease.Future studies need to be carried out <strong>in</strong> several organs under different environmentalconditions, s<strong>in</strong>ce there are multiple environmental risk factors for type 2 diabetes thattarget different organs. It is possible that these studies will generate <strong>in</strong>formation that canbe used <strong>in</strong> the prediction and prevention of type 2 diabetes. Moreover, <strong>in</strong> the future it ispossible that new drugs target<strong>in</strong>g epigenetic factors can be developed for patients withtype 2 diabetes.References[1] Alberti KG, Zimmet PZ. Def<strong>in</strong>ition, diagnosis and classification of diabetes mellitus and its complications. Part1: diagnosis and classification of diabetes mellitus provisional report of a WHO consultation. Diabet Med1998;15:539e53.[2] Zimmet P, Alberti KG, Shaw J. Global and societal implications of the diabetes epidemic. Nature2001;414:782e7.[3] Kobberl<strong>in</strong>g J, Tillil H. Genetic and nutritional factors <strong>in</strong> the etiology and pathogenesis of diabetes mellitus.World Rev Nutr Diet 1990;63:102e15.[4] McCarthy M. Genomics, Type 2 Diabetes, and Obesity. The New England Journal of Medic<strong>in</strong>e2010;363:2339e50.[5] Saxena R, Voight BF, Lyssenko V, Burtt NP, de Bakker PI, Chen H, et al. Genome-wide association analysisidentifies loci for type 2 diabetes and triglyceride levels. <strong>Science</strong> 2007;316:1331e6.[6] Scott LJ, Mohlke KL, Bonnycastle LL, Willer CJ, Li Y, Duren WL, et al. A genome-wide association study of type2 diabetes <strong>in</strong> F<strong>in</strong>ns detects multiple susceptibility variants. <strong>Science</strong> 2007;316:1341e5.[7] Sladek R, Rocheleau G, Rung J, D<strong>in</strong>a C, Shen L, Serre D, et al. A genome-wide association study identifies novelrisk loci for type 2 diabetes. Nature 2007;445:881e5.[8] Zegg<strong>in</strong>i E, Weedon MN, L<strong>in</strong>dgren CM, Frayl<strong>in</strong>g TM, Elliott KS, Lango H, et al. Replication of genome-wideassociation signals <strong>in</strong> UK samples reveals risk loci for type 2 diabetes. <strong>Science</strong> 2007;316:1336e41.[9] Zegg<strong>in</strong>i E, Scott LJ, Saxena R, Voight BF, March<strong>in</strong>i JL, Hu T, et al. Meta-analysis of genome-wide association dataand large-scale replication identifies additional susceptibility loci for type 2 diabetes. Nat Genet2008;40:638e45.[10] Voight BF, Scott LJ, Ste<strong>in</strong>thorsdottir V, Morris AP, D<strong>in</strong>a C, Welch RP, et al. Twelve type 2 diabetes susceptibilityloci identified through large-scale association analysis. Nat Genet 2010;42:579e89.[11] Koeck T, Olsson AH, Nitert MD, Sharoyko VV, Ladenvall C, Kotova O, et al. A common variant <strong>in</strong> TFB1M isassociated with reduced <strong>in</strong>sul<strong>in</strong> secretion and <strong>in</strong>creased future risk of type 2 diabetes. Cell Metab2011;13:80e91.


CHAPTER 16<strong>Epigenetics</strong> of Diabetes <strong>in</strong> <strong>Human</strong>s[12] Lyssenko V, Nagorny CL, Erdos MR, Wierup N, Jonsson A, Spegel P, et al. Common variant <strong>in</strong> MTNR1Bassociated with <strong>in</strong>creased risk of type 2 diabetes and impaired early <strong>in</strong>sul<strong>in</strong> secretion. Nat Genet2009;41:82e8.[13] Lyssenko V, Jonsson A, Almgren P, Pulizzi N, Isomaa B, Tuomi T, et al. Cl<strong>in</strong>ical risk factors, DNA variants, andthe development of type 2 diabetes. N Engl J Med 2008;359:2220e32.[14] Ronn T, Wen J, Yang Z, Lu B, Du Y, Groop L, et al. A common variant <strong>in</strong> MTNR1B, encod<strong>in</strong>g melaton<strong>in</strong>receptor 1B, is associated with type 2 diabetes and fast<strong>in</strong>g plasma glucose <strong>in</strong> Han Ch<strong>in</strong>ese <strong>in</strong>dividuals.Diabetologia 2009.[15] Wen J, Ronn T, Olsson A, Yang Z, Lu B, Du Y, et al. Investigation of type 2 diabetes risk alleles supportCDKN2A/B, CDKAL1, and TCF7L2 as susceptibility genes <strong>in</strong> a Han Ch<strong>in</strong>ese cohort. PLoS ONE 2010;5:e9153.[16] Gibney ER, Nolan CM. <strong>Epigenetics</strong> and gene expression. Heredity 2010;105:4e13.[17] Bird A. Perceptions of epigenetics. Nature 2007;447:396e8.[18] Cubas P, V<strong>in</strong>cent C, Coen E. An epigenetic mutation responsible for natural variation <strong>in</strong> floral symmetry.Nature 1999;401:157e61.[19] Chong S, Whitelaw E. Epigenetic germl<strong>in</strong>e <strong>in</strong>heritance. Curr Op<strong>in</strong> Genet Dev 2004;14:692e6.[20] Anway MD, Cupp AS, Uzumcu M, Sk<strong>in</strong>ner MK. Epigenetic transgenerational actions of endocr<strong>in</strong>e disruptorsand male fertility. <strong>Science</strong> 2005;308:1466e9.[21] Ng SF, L<strong>in</strong> RC, Laybutt DR, Barres R, Owens JA, Morris MJ. Chronic high-fat diet <strong>in</strong> fathers programs beta-celldysfunction <strong>in</strong> female rat offspr<strong>in</strong>g. Nature 2010;467:963e6.[22] Lister R, Pelizzola M, Dowen RH, Hawk<strong>in</strong>s RD, Hon G, Tonti-Filipp<strong>in</strong>i J, et al. <strong>Human</strong> DNA methylomes atbase resolution show widespread epigenomic differences. Nature 2009;462(7271):315e22.[23] Lennartsson A, Ekwall K. Histone modification patterns and epigenetic codes. Biochimica et Biophysica Acta2009;1790(9):863e8.[24] Turner B M. Environmental sens<strong>in</strong>g by chromat<strong>in</strong>: an epigenetic contribution to evolutionary change. FEBSLett 585, 2032-2040[25] L<strong>in</strong>g C, Groop L. <strong>Epigenetics</strong>: a molecular l<strong>in</strong>k between environmental factors and type 2 diabetes. Diabetes2009;58:2718e25.[26] Mulder H, L<strong>in</strong>g C. Mitochondrial dysfunction <strong>in</strong> pancreatic beta-cells <strong>in</strong> Type 2 diabetes. Mol Cell Endocr<strong>in</strong>ol2009;297:34e40.[27] Lyssenko V, Lupi R, Marchetti P, Del Guerra S, Orho-Melander M, Almgren P, et al. Mechanisms bywhich common variants <strong>in</strong> the TCF7L2 gene <strong>in</strong>crease risk of type 2 diabetes. J Cl<strong>in</strong> Invest 2007;117:2155e63.[28] Lyssenko V, Almgren P, Anevski D, Perfekt R, Lahti K, Nissen M, et al. Predictors of and longitud<strong>in</strong>al changes <strong>in</strong><strong>in</strong>sul<strong>in</strong> sensitivity and secretion preced<strong>in</strong>g onset of type 2 diabetes. Diabetes 2005;54:166e74.[29] Olsson AH, Ronn T, Ladenvall C, Parikh H, Isomaa B, Groop L, et al. Two common genetic variants nearnuclear encoded OXPHOS genes are associated with <strong>in</strong>sul<strong>in</strong> secretion <strong>in</strong> vivo. Eur J Endocr<strong>in</strong>ol 2011.[30] L<strong>in</strong>g C, Groop L, Guerra SD, Lupi R. Calpa<strong>in</strong>-10 expression is elevated <strong>in</strong> pancreatic islets from patients withtype 2 diabetes. PLoS ONE 2009;4: e6558.[31] Kuroda A, Rauch TA, Todorov I, Ku HT, Al-Abdullah IH, Kandeel F, et al. Insul<strong>in</strong> gene expression is regulated byDNA methylation. PLoS ONE 2009;4: e6953.[32] Mutskov V, Raaka BM, Felsenfeld G, Gershengorn MC. The human <strong>in</strong>sul<strong>in</strong> gene displays transcriptionallyactive epigenetic marks <strong>in</strong> islet-derived mesenchymal precursor cells <strong>in</strong> the absence of <strong>in</strong>sul<strong>in</strong> expression. Stemcells (Dayton, Ohio) 2007;25:3223e33.[33] Mutskov V, Felsenfeld G. The human <strong>in</strong>sul<strong>in</strong> gene is part of a large open chromat<strong>in</strong> doma<strong>in</strong> specific for humanislets. Proc Natl Acad Sci USA 2009;106:17419e24.[34] Cimen H, Han MJ, Yang Y, Tong Q, Koc H, Koc EC. Regulation of succ<strong>in</strong>ate dehydrogenase activity by SIRT3 <strong>in</strong>mammalian mitochondria. Biochem 2010;49:304e11.[35] Henqu<strong>in</strong> JC. Regulation of <strong>in</strong>sul<strong>in</strong> secretion: a matter of phase control and amplitude modulation. Diabetologia2009;52:739e51.[36] Olsson AH, Yang BT, Hall E, Taneera J, Salehi A, Dekker Nitert M, et al. Decreased expression of genes <strong>in</strong>volved<strong>in</strong> oxidative phosphorylation <strong>in</strong> human pancreatic islets from patients with type 2 diabetes. Eur J Endocr<strong>in</strong>ol2011.[37] L<strong>in</strong>g C, Del Guerra S, Lupi R, Ronn T, Granhall C, Luthman H, et al. Epigenetic regulation of PPARGC1A <strong>in</strong>human type 2 diabetic islets and effect on <strong>in</strong>sul<strong>in</strong> secretion. Diabetologia 2008;51:615e22.[38] Bhandare R, Schug J, Le Lay J, Fox A, Smirnova O, Liu C, et al. Genome-wide analysis of histone modifications<strong>in</strong> human pancreatic islets. Genome Res 2010;20:428e33.327


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>328[39] Stitzel ML, Sethupathy P, Pearson DS, Ch<strong>in</strong>es PS, Song L, Erdos MR, et al. Global epigenomic analysis ofprimary human pancreatic islets provides <strong>in</strong>sights <strong>in</strong>to type 2 diabetes susceptibility loci. Cell Metabol2010;12:443e55.[40] Gaulton KJ, Nammo T, Pasquali L, Simon JM, Giresi PG, Fogarty MP, et al. A map of open chromat<strong>in</strong> <strong>in</strong>human pancreatic islets. Nat Genet 2010;42:255e9.[41] Ravelli AC, van der Meulen JH, Michels RP, Osmond C, Barker DJ, Hales CN, et al. Glucose tolerance <strong>in</strong> adultsafter prenatal exposure to fam<strong>in</strong>e. Lancet 1998;351:173e7.[42] Jensen CB, Storgaard H, Dela F, Holst JJ, Madsbad S, Vaag AA. Early differential defects of <strong>in</strong>sul<strong>in</strong> secretion andaction <strong>in</strong> 19-year-old caucasian men who had low birth weight. Diabetes 2002;51:1271e80.[43] Jensen CB, Storgaard H, Madsbad S, Richter EA, Vaag AA. Altered skeletal muscle fiber composition and sizeprecede whole-body <strong>in</strong>sul<strong>in</strong> resistance <strong>in</strong> young men with low birth weight. J Cl<strong>in</strong> Endocr<strong>in</strong>ol Metab2007;92:1530e4.[44] Barker DJ. The <strong>in</strong>trauter<strong>in</strong>e environment and adult cardiovascular disease. Ciba Foundation symposium1991;156:3e10. discussion 10e16.[45] Ozanne SE, Jensen CB, T<strong>in</strong>gey KJ, Storgaard H, Madsbad S, Vaag AA. Low birthweight is associated withspecific changes <strong>in</strong> muscle <strong>in</strong>sul<strong>in</strong>-signall<strong>in</strong>g prote<strong>in</strong> expression. Diabetologia 2005;48:547e52.[46] Ozanne SE, Jensen CB, T<strong>in</strong>gey KJ, Mart<strong>in</strong>-Gronert MS, Grunnet L, Brons C, et al. Decreased prote<strong>in</strong> levels ofkey <strong>in</strong>sul<strong>in</strong> signall<strong>in</strong>g molecules <strong>in</strong> adipose tissue from young men with a low birthweight: potential l<strong>in</strong>k to<strong>in</strong>creased risk of diabetes? Diabetologia 2006;49:2993e9.[47] Poulsen P, Vaag AA, Kyvik KO, Moller Jensen D, Beck-Nielsen H. Low birth weight is associated with NIDDM<strong>in</strong> discordant monozygotic and dizygotic tw<strong>in</strong> pairs. Diabetologia 1997;40:439e46.[48] Poulsen P, Vaag A. The <strong>in</strong>trauter<strong>in</strong>e environment as reflected by birth size and tw<strong>in</strong> and zygosity status<strong>in</strong>fluences <strong>in</strong>sul<strong>in</strong> action and <strong>in</strong>tracellular glucose metabolism <strong>in</strong> an age- or time-dependent manner. Diabetes2006;55:1819e25.[49] Barker DJ. The developmental orig<strong>in</strong>s of <strong>in</strong>sul<strong>in</strong> resistance. Hormone Res 2005;64(Suppl. 3):2e7.[50] Tobi EW, Lumey LH, Talens RP, Kremer D, Putter H, Ste<strong>in</strong> AD, et al. DNA Methylation differences afterexposure to prenatal fam<strong>in</strong>e are common and tim<strong>in</strong>g- and sex-specific. Hum Mol Genet 2009.[51] Heijmans BT, Tobi EW, Ste<strong>in</strong> AD, Putter H, Blauw GJ, Susser ES, et al. Persistent epigenetic differences associatedwith prenatal exposure to fam<strong>in</strong>e <strong>in</strong> humans. Proc Natl Acad Sci USA 2008;105:17046e9.[52] Park JH, Stoffers DA, Nicholls RD, Simmons RA. Development of type 2 diabetes follow<strong>in</strong>g <strong>in</strong>trauter<strong>in</strong>e growthretardation <strong>in</strong> rats is associated with progressive epigenetic silenc<strong>in</strong>g of Pdx1. J Cl<strong>in</strong> Invest 2008;118:2316e24.[53] Sandovici IS, NH. Dekker-Nitert M, Ackers-Johnson M, Jones RH, O’Neill LP, Marquez VE, et al. Dynamicepigenetic regulation by early-diet and ag<strong>in</strong>g of the type 2 diabetes susceptibility gene Hnf4a <strong>in</strong> pancreaticislets. Proc Natl Acad Sci USA 2011.[54] Thompson RF, Fazzari MJ, Niu H, Barzilai N, Simmons RA, Greally JM. Experimental <strong>in</strong>trauter<strong>in</strong>e growthrestriction <strong>in</strong>duces alterations <strong>in</strong> DNA methylation and gene expression <strong>in</strong> pancreatic islets of rats. J Biol Chem2010;285:15111e8.[55] P<strong>in</strong>ney SE, Jaeckle Santos LJ, Han Y, Stoffers DA, Simmons RA. Exend<strong>in</strong>-4 <strong>in</strong>creases histone acetylase activityand reverses epigenetic modifications that silence Pdx1 <strong>in</strong> the <strong>in</strong>trauter<strong>in</strong>e growth retarded rat. Diabetologia2011;54:2606e14.[56] Stoffers DA, Ferrer J, Clarke WL, Habener JF. Early-onset type-II diabetes mellitus (MODY4) l<strong>in</strong>ked to IPF1. NatGenet 1997;17:138e9.[57] Ahlgren U, Jonsson J, Jonsson L, Simu K, Edlund H. beta-cell-specific <strong>in</strong>activation of the mouse Ipf1/Pdx1 generesults <strong>in</strong> loss of the beta-cell phenotype and maturity onset diabetes. Gene Dev 1998;12:1763e8.[58] Kelley DE, He J, Menshikova EV, Ritov VB. Dysfunction of mitochondria <strong>in</strong> human skeletal muscle <strong>in</strong> type 2diabetes. Diabetes 2002;51:2944e50.[59] Mootha VK, L<strong>in</strong>dgren CM, Eriksson KF, Subramanian A, Sihag S, Lehar J, et al. PGC-1alpha-responsive genes<strong>in</strong>volved <strong>in</strong> oxidative phosphorylation are coord<strong>in</strong>ately downregulated <strong>in</strong> human diabetes. Nat Genet2003;34:267e73.[60] Huang X, Eriksson KF, Vaag A, Lehtovirta M, Hansson M, Laurila E, et al. Insul<strong>in</strong>-regulated mitochondrial geneexpression is associated with glucose flux <strong>in</strong> human skeletal muscle. Diabetes 1999;48:1508e14.[61] Patti ME, Butte AJ, Crunkhorn S, Cusi K, Berria R, Kashyap S, et al. Coord<strong>in</strong>ated reduction of genes of oxidativemetabolism <strong>in</strong> humans with <strong>in</strong>sul<strong>in</strong> resistance and diabetes: Potential role of PGC1 and NRF1. Proc Natl AcadSci USA 2003;100:8466e71.[62] Sreekumar R, Halvatsiotis P, Schimke JC, Nair KS. Gene expression profile <strong>in</strong> skeletal muscle of type 2 diabetesand the effect of <strong>in</strong>sul<strong>in</strong> treatment. Diabetes 2002;51:1913e20.[63] Ronn T, Poulsen P, Hansson O, Holmkvist J, Almgren P, Nilsson P, et al. Age <strong>in</strong>fluences DNA methylation andgene expression of COX7A1 <strong>in</strong> human skeletal muscle. Diabetologia 2008;51:1159e68.


CHAPTER 16<strong>Epigenetics</strong> of Diabetes <strong>in</strong> <strong>Human</strong>s[64] L<strong>in</strong>g C, Poulsen P, Simonsson S, Ronn T, Holmkvist J, Almgren P, et al. Genetic and epigenetic factors areassociated with expression of respiratory cha<strong>in</strong> component NDUFB6 <strong>in</strong> human skeletal muscle. J Cl<strong>in</strong> Invest2007;117:3427e35.[65] Barres R, Osler ME, Yan J, Rune A, Fritz T, Caidahl K, et al. Non-CpG methylation of the PGC-1alpha promoterthrough DNMT3B controls mitochondrial density. Cell Metab 2009;10:189e98.[66] Brons C, Jacobsen S, Nilsson E, Ronn T, Jensen CB, Storgaard H, et al. Deoxyribonucleic acid methylation andgene expression of PPARGC1A <strong>in</strong> human muscle is <strong>in</strong>fluenced by high-fat overfeed<strong>in</strong>g <strong>in</strong> a birth-weightdependentmanner. J Cl<strong>in</strong> Endocr<strong>in</strong>ol Metab 2010;95:3048e56.[67] Ozanne SE, Constancia M. Mechanisms of disease: the developmental orig<strong>in</strong>s of disease and the role of theepigenotype. Nat Cl<strong>in</strong> Pract 2007;3:539e46.[68] Lillycrop KA, Phillips ES, Jackson AA, Hanson MA, Burdge GC. Dietary prote<strong>in</strong> restriction of pregnant rats<strong>in</strong>duces and folic acid supplementation prevents epigenetic modification of hepatic gene expression <strong>in</strong> theoffspr<strong>in</strong>g. J Nutr 2005;135:1382e6.[69] Lillycrop KA, Slater-Jefferies JL, Hanson MA, Godfrey KM, Jackson AA, Burdge GC. Induction of alteredepigenetic regulation of the hepatic glucocorticoid receptor <strong>in</strong> the offspr<strong>in</strong>g of rats fed a prote<strong>in</strong>-restricted dietdur<strong>in</strong>g pregnancy suggests that reduced DNA methyltransferase-1 expression is <strong>in</strong>volved <strong>in</strong> impaired DNAmethylation and changes <strong>in</strong> histone modifications. Br J Nutr 2007;97:1064e73.[70] Lillycrop KA, Phillips ES, Torrens C, Hanson MA, Jackson AA, Burdge GC. Feed<strong>in</strong>g pregnant rats a prote<strong>in</strong>restricteddiet persistently alters the methylation of specific cytos<strong>in</strong>es <strong>in</strong> the hepatic PPAR alpha promoter ofthe offspr<strong>in</strong>g. Br J Nutr 2008;100:278e82.329


This page <strong>in</strong>tentionally left blank


CHAPTER 17The Potential ofEpigenetic Compounds<strong>in</strong> Treat<strong>in</strong>g DiabetesSteven G. GraySt James’s Hospital, Dubl<strong>in</strong>, IrelandCHAPTER OUTLINE17.1 The Problem of Diabetes 33217.2 <strong>Epigenetics</strong> 33217.2.1 DNA CpG Methylation 33217.2.2 Histone PTMs 33317.2.3 microRNAs/EpimicroRNAs33417.3 Aberrant Epigenetic Regulation ofGene Expression or Prote<strong>in</strong>Function as a Cause ofDiabetes 33417.4 Aberrant <strong>Epigenetics</strong> with<strong>in</strong> theDiabetic Sett<strong>in</strong>g 33417.4.1 Aberrant DNA CpG Methylationand Diabetes 33417.4.2 Aberrant Chromat<strong>in</strong> RegulatoryMach<strong>in</strong>ery and Diabetes 334Effects of HDACI on diabetogeneexpression 336Class I e Hormones 336Class II e Transcriptionfactors 337Class III e Transporters 33817.4.3 Aberrant microRNAs/EpimicroRNAsand Diabetes 339Effects of <strong>in</strong>sul<strong>in</strong> onmiRNAs 341miRNAs and Diabetes 341Effects of obesity onmiRNAs 341Effects of epigenetic <strong>in</strong>hibitorson miRNAs 34117.4.4 Effects of Insul<strong>in</strong> Resistance/Metabolic Syndrome onEpigenetic Modify<strong>in</strong>gEnzymes 34217.5 Non-Epigenetic Effects of HistoneModifier Prote<strong>in</strong>s with DiabetesPathogenesis 34217.5.1 Inflammation <strong>in</strong> DiabetesPathogenesis 34217.5.2 ER Stress/Chaperones andDiabetes Pathogenesis 343L<strong>in</strong>k<strong>in</strong>g HATS/HDAC activitiesto chaperone activities 344L<strong>in</strong>k<strong>in</strong>g KATS/HDAC activitiesto ER stress pathways 34517.6 Potential for the Use of HDACi toAmeliorate or Treat Symptoms ofDiabetes Pathogenesis 34617.6.1 Currently DevelopedDrugs 34617.6.2 Therapeutic Implications 34717.6.3 Pancreatic Islet Development,and Protection Us<strong>in</strong>g HistoneDeacetylase Inhibitors, or NFkBBlockade 34717.6.4 Modulation of Th17-MediatedAutoimmunity 34817.6.5 Stem Cells 34817.6.6 The ability of phenylbutyrate toact as a chaperoneand alleviate ER stress 349331T. Tollefsbol (Ed): <strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>. DOI: 10.1016/B978-0-12-388415-2.00017-2Copyright Ó 2012 Elsevier Inc. All rights reserved.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>17.6.7 Is HDACi Specificity anIssue? 35017.6.8 Compounds Which TargetLys<strong>in</strong>e Methylases/Demethylases 35017.6.9 Nutrition-Based NaturalCompounds as TherapeuticAgents 350Sulforaphane 351Curcum<strong>in</strong> 351Resveratrol 35117.6.10 miRNA-/siRNA-BasedTherapeutics 35217.7 Conclusions 353References 35317.1 THE PROBLEM OF DIABETESDiabetes is a lifelong (chronic) disease <strong>in</strong> which high levels of the sugar glucose are present <strong>in</strong>the blood. The body produces a hormone, <strong>in</strong>sul<strong>in</strong>, which regulates blood sugar levels bymov<strong>in</strong>g glucose from the bloodstream <strong>in</strong>to muscle, fat, and liver cells, to be used as fuel.Diabetes is essentially a consequence of the body’s failure to regulate blood sugar causedprimarily by hav<strong>in</strong>g (a) too little <strong>in</strong>sul<strong>in</strong>, (b) develop<strong>in</strong>g resistance to <strong>in</strong>sul<strong>in</strong>, or (c) both.Complications associated with diabetes <strong>in</strong>clude kidney failure, non-traumatic lower-limbamputations, bl<strong>in</strong>dness and diabetes is a major cause of heart disease and stroke [1,2].Globally, diabetes (and <strong>in</strong> particular type 2 diabetes) represents a major challenge to worldhealth. Accord<strong>in</strong>g to the WHO more than 220 million people worldwide have diabetes, and itprojects that diabetes deaths will double between 2005 and 2030 [3].332In 2010 for the USA alone, diabetes was calculated to affect 25.8 million people, represent<strong>in</strong>gapproximately 8.3% of the population [4]. Estimated global healthcare expenditures to treatand prevent diabetes and its complications are expected to total at least US Dollar (USD) 376billion <strong>in</strong> 2010. By 2030, this number is projected to exceed some USD490 billion [5].Economically, the cost of diabetes to nations has significant consequences. For example it isestimated that <strong>in</strong> the period 2006e2015, Ch<strong>in</strong>a will lose $558 billion <strong>in</strong> foregone national<strong>in</strong>come due to heart disease, stroke, and diabetes alone [3].Diabetes is a complex syndrome of dysregulation of carbohydrate and lipid metabolism dueprimarily to beta cell dysfunction associated with a variable degree of <strong>in</strong>sul<strong>in</strong> resistance. It isclear that a complex <strong>in</strong>terplay between environmental, nutritional, and genetic factors playa role <strong>in</strong> diabetes pathogenesis. Nevertheless, it is my contention that a common thread, thatof histone and transcription factor/prote<strong>in</strong> acetylation l<strong>in</strong>ks many of the currently identifiedpathways known to be <strong>in</strong>volved with diabetes pathogenesis. Over the follow<strong>in</strong>g sections, I willattempt to l<strong>in</strong>k these diverse observations <strong>in</strong>to a s<strong>in</strong>gle unified concept, on the therapeuticpotential of target<strong>in</strong>g HDACs for the treatment of diabetes.17.2 EPIGENETICSEpigenetic regulation of gene expression <strong>in</strong>volves stable and heritable changes <strong>in</strong> geneexpression which are not due to changes <strong>in</strong> the primary DNA sequence. Current knownepigenetic mechanisms <strong>in</strong>volve the follow<strong>in</strong>g: DNA CpG methylation, histone posttranslationalmodifications (PTMs), gene impr<strong>in</strong>t<strong>in</strong>g, and non-cod<strong>in</strong>g RNA (ncRNA)(Figure 17.1).17.2.1 DNA CpG MethylationDNA CpG methylation was the first epigenetic mechanism identified. In mammalian cells,DNA methylation occurs ma<strong>in</strong>ly at the C5 position of CpG d<strong>in</strong>ucleotides and is carried out by


CHAPTER 17The Potential of Epigenetic Compounds <strong>in</strong> Treat<strong>in</strong>g Diabetes(A)(B)(D)(C)FIGURE 17.1Overview of epigenetic mechanisms. Four ma<strong>in</strong>mechanisms for epigenetic regulation of geneexpression have been characterized. (A) DNAmethylation, (B) covalent histone modifications, (C) noncovalentmechanisms (e.g.; <strong>in</strong>corporation of histonevariants) and (D) non-cod<strong>in</strong>g RNAs (ncRNAs) <strong>in</strong>clud<strong>in</strong>gmicroRNAs (miRNAs). DNMT ¼ DNA methyltransferase;KAT ¼ lys<strong>in</strong>e acetyltransferase; KDM ¼ lys<strong>in</strong>edemethylase; KMT ¼ lys<strong>in</strong>e methyltransferase; MeCP¼ methyl CpG b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>; PTMS ¼ posttranslationalmodifications; S ¼ sumoylation; U ¼ubiquit<strong>in</strong>ation; Ac ¼ acetylation; Me ¼ methylation.This figure is reproduced <strong>in</strong> the color plate section.two general classes of enzymatic activities e ma<strong>in</strong>tenance methylation and de novo methylation.The enzymes which transfer these methyl groups are the DNA methyltransferases(DNMTs) [6]. The enzymes and mechanisms for demethylation rema<strong>in</strong> to be elucidated, withbase excision repair emerg<strong>in</strong>g as the lead<strong>in</strong>g candidate [7].17.2.2 Histone PTMsThe “histone code” is a well-established hypothesis describ<strong>in</strong>g the idea that specific patterns ofpost-translational modifications to histones act like a molecular “code” recognized and usedby non-histone prote<strong>in</strong>s to regulate specific chromat<strong>in</strong> functions [8e10]. This <strong>in</strong>itial conceptmay however be too simplistic, and it has now been suggested that this “code” should beconsidered to be more like a “language”, whereby PTMs provide more “nuanced” or “contextdependent”effects [11].333These modifications <strong>in</strong>clude acetylation, methylation, phosphorylation, sumoylation, andubiquit<strong>in</strong>ation, and various families of prote<strong>in</strong>s have been identified which function to placeor remove these PTMs. The best studied of these families are the lys<strong>in</strong>e acetyltransferases orK-acetyltransferases (KATs), histone deacetylases (HDACs), K-methyltransferases (KMTs), andK-demethylases (KDMs) [12].K-acetyltransferases (KATs), also known as histone acetyltransferases [12], function to covalentlyadd acetyl groups to lys<strong>in</strong>e residues on prote<strong>in</strong>s, and histone deacetylases (HDACs)function to remove acetyl groups [13]. K-methyltransferases (KMTs) add mono-, di-, ortrimethyl groups to lys<strong>in</strong>e residues [14], while HDACs and K-demethylases (KDMs) removethese respective modifications [12]. These PTMs play important roles on many prote<strong>in</strong>s <strong>in</strong>addition to histones and may <strong>in</strong> fact <strong>in</strong>volve a “prote<strong>in</strong> code” exemplified by the prote<strong>in</strong>s p53[15] and NFkB [16,17]. The importance of these “non-epigenetic” modifications <strong>in</strong> theregulation of cellular processes can be exemplified by a recent study that found 3600 acetylationsites on 1750 prote<strong>in</strong>s. To put these <strong>in</strong> context only 61 acetylation events were found onhistones [18]. Nevertheless, if one considers all of the possible comb<strong>in</strong>atorial possibilities forhistone modifications, the known modifications on histone H3 alone could produce over onemillion dist<strong>in</strong>ct post-translational “signatures” [19].


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>17.2.3 microRNAs/Epi-microRNAsmicroRNAs (miRNAs) are specialized forms of non-cod<strong>in</strong>g RNA (ncRNA). They consist ofsmall, approximately 22 nucleotide s<strong>in</strong>gle-stranded RNA molecules that regulate geneexpression <strong>in</strong> cells by directly b<strong>in</strong>d<strong>in</strong>g to and either degrad<strong>in</strong>g or translationally repress<strong>in</strong>gtargets and are emerg<strong>in</strong>g as key regulators of most, if not all, physiological processes. AlteredmiRNA expression is implicated <strong>in</strong> both diabetes [20] and obesity [21]. Some of these miRNAshave been shown to target epigenetic regulators which has led to this subset of miRNAs be<strong>in</strong>gdescribed as “epi-miRNAs” [22].17.3 ABERRANT EPIGENETIC REGULATION OF GENE EXPRESSIONOR PROTEIN FUNCTION AS A CAUSE OF DIABETESPreviously the author has hypothesized that aberrant gene expression may be a fundamentalissue <strong>in</strong> the development of diabetes [23]. Three central mechanisms exist (depicted <strong>in</strong>Figure 17.1) whereby aberrant gene expression could affect the expression of critical “diabetogenes”.(a) At a basic level, the activities of these enzmes can directly affect chromat<strong>in</strong> attarget genes, and any alterations or disruptions of their activities could consequently lead toaberrant transcription of diabetogenes. (b) Alternatively, several prote<strong>in</strong>s, which have beenidentified as the causative factors <strong>in</strong> monogenic autosomal dom<strong>in</strong>ant forms of type 2 diabetes(MODY), have also been shown to associate with KATs/HDACs [23]. Indeed some of themutations identified <strong>in</strong> these prote<strong>in</strong>s result <strong>in</strong> loss of association with KATs/HDACs, or lead toa loss <strong>in</strong> KAT enzymatic activity [23] (Table 17.1). (c) Alterations to the activities of KATs/HDACs may functionally result <strong>in</strong> the aberrant regulation of transcription of sets of targetgenes essential for normal cellular homeostasis/development <strong>in</strong> diabetes pathogenesis.33417.4 ABERRANT EPIGENETICS WITHIN THE DIABETIC SETTINGThe follow<strong>in</strong>g sections will discuss some of the evidence l<strong>in</strong>k<strong>in</strong>g aberrant epigenetics withdiabetes pathogenesis.17.4.1 Aberrant DNA CpG Methylation and DiabetesDNA CpG methylation was essentially the first epigenetic regulatory mechanism identified,and aberrant DNA CpG methylation was <strong>in</strong>itially associated with transient neonatal diabetes[24,25]. Excit<strong>in</strong>g new genome-wide methylation analyses have now revealed the presence ofDNA methylation variations that predispose to both type 1 and type 2 diabetes [26e29],rais<strong>in</strong>g the possibility of epigenetic target<strong>in</strong>g of DNA methyltransferases. The transcriptionalcoactivator peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1a)isa critical regulator of cellular metabolic control [30]. Research has shown that levels of PGC-1aare decreased <strong>in</strong> the muscle of patients with type 2 diabetes [31]. Additional studies have nowshown that PGC-1a is also down-regulated <strong>in</strong> pancreatic islets of diabetics and that furthermoreDNA methylation is <strong>in</strong>volved with this down-regulation [32]. Intrigu<strong>in</strong>gly while L<strong>in</strong>gand colleagues found that this was due to DNA CpG methylation [32], non-CpG methylationhas also been shown to be essential for the down-regulation of PGC-1a mRNA <strong>in</strong> myotubesisolated from diabetic patients [33]. As such target<strong>in</strong>g DNMTs may be a potential therapeuticmodality <strong>in</strong> the treatment of patients with diabetes.17.4.2 Aberrant Chromat<strong>in</strong> Regulatory Mach<strong>in</strong>ery and DiabetesStrong evidence now l<strong>in</strong>ks aberrant expression/regulation of gene expression by chromat<strong>in</strong>modify<strong>in</strong>gprote<strong>in</strong>s <strong>in</strong>volved with histone/prote<strong>in</strong> PTMs. For <strong>in</strong>stance, genome-wide associationstudies (GWAS) identified chromosomal region 6q21 as be<strong>in</strong>g l<strong>in</strong>ked to both type 1 andtype 2 diabetes [34e37], and it is of <strong>in</strong>terest that HDAC2 maps to this region. In support of thisobservation, among six HDACs tested (HDAC-1 through -5 and HDAC-8) <strong>in</strong> two diabetes


CHAPTER 17The Potential of Epigenetic Compounds <strong>in</strong> Treat<strong>in</strong>g DiabetesTABLE 17.1 L<strong>in</strong>k<strong>in</strong>g HATs/HDACs to MODY Forms of DiabetesCondition Evidence to l<strong>in</strong>k HATs/HDACs to this form ReferencesMODY1(HNF-4a)MODY2(GK)MODY3(HNF-1a)MODY4(PDX-1)MODY5(HNF-1b)MODY6(NEUROD1)MODY7(KLF11)HNF-4a is directly acetylated by KAT3A (CBP)[23]HNF-4a target gene promoters are regulated by histone acetylationThe HNF-4a promoter is regulated by histone acetylationThe GK2 promoter is regulated by histone acetylation [23]Physically <strong>in</strong>teracts with lys<strong>in</strong>e acetyltransferasesHNF-1a dom<strong>in</strong>ant negative mutants have stronger association with KATs, but thiscauses decreased KAT activityPo<strong>in</strong>t mutations <strong>in</strong> HNF-1a have decreased aff<strong>in</strong>ity for KAT3B (p300)HNF-1a directs hyperacetylation at promoters <strong>in</strong> pancreatic isletsPDX-1 activity depends upon the particular chromat<strong>in</strong> environment it <strong>in</strong>teracts withInsul<strong>in</strong> gene activiation is mediated through PDX-1/KAT3B (p300) <strong>in</strong>teractionsThe PDX-1 promoter is regulated by histone acetylationActivation of PDX-1 function is directly enhanced by KAT3B (p300)Under conditions of low glucose PDX-1 associates with HDAC1 and HDAC2Under conditions of high glucose PDX-1 associates with KAT3B (p300)In a term<strong>in</strong>ally differentiated glucagonoma cell l<strong>in</strong>e, PDX-1 modulates histone H4acetylation to activate <strong>in</strong>sul<strong>in</strong> gene expressionMutations of HNF-1b result <strong>in</strong> loss of association with KAT3A (CBP) and KAT2B(P/CAF)HNF-1b/KAT2B or KAT3A complexes regulate target promotersIn diabetics mutations of NEUROD1 affect its ability to associate with KAT3A andKAT3BPromoter regions associated with NEUROD1 b<strong>in</strong>d<strong>in</strong>g colocalise with KAT3A/KAT3BNEUROD1 associates with KAT3A to drive <strong>in</strong>sul<strong>in</strong> transcriptionDirect acetylation of NEUROD1 is required for <strong>in</strong>sul<strong>in</strong> transcriptionNEUROD1 expression by INSM1 is regulated via recruitment of HDACsA novel KLF-KAT3B-regulated pathway for <strong>in</strong>sul<strong>in</strong> biosynthesis is disrupted by thec.-331 INS mutation found <strong>in</strong> neonatal diabetes mellitusKLF11, is a novel KAT3B-dependent regulator of Pdx-1 (MODY4) transcription <strong>in</strong>pancreatic islet beta cells[23][23][23][23][65][64]335models (STZ-<strong>in</strong>duced diabetic rats and db/db mice), only HDAC-2 activity was significantly<strong>in</strong>creased and was associated with renal <strong>in</strong>jury <strong>in</strong> these models, reflect<strong>in</strong>g the diabeticnephropathy observed <strong>in</strong> human diabetes patients [38].Indeed, one of the first <strong>in</strong>dicators that histone-modify<strong>in</strong>g enzymes may be a central element <strong>in</strong>diabetes came from a loss-of-function mouse model of KAT3A (CBP). In this model micewhich were heterozygous for the mutant displayed <strong>in</strong>creased <strong>in</strong>sul<strong>in</strong> sensitivity and glucosetolerance, even though they present with a marked lipidystrophy of white adipose tissue [39].Furthermore, knockout of another k-acetyltransferase KAT13B (SRC-3), prevents the developmentof obesity <strong>in</strong> mice, and improves <strong>in</strong>sul<strong>in</strong> sensitivity by reduc<strong>in</strong>g levels of acetylation ofPGC-1a [40]. KAT13B would appear to be an important regulator <strong>in</strong> diabetes pathogenesis asa knock-<strong>in</strong> model of KAT13B with various mutations display<strong>in</strong>g a phenotype with <strong>in</strong>creasedbodyweight and adiposity, coupled with reduced peripheral <strong>in</strong>sul<strong>in</strong> sensitivity [41].As previously stated above, elevated levels of HDAC-2 are associated with diabetic renal<strong>in</strong>jury. The HDAC family comprises 18 members separated <strong>in</strong>to four classes (classes IeIV)based on sequence identity and doma<strong>in</strong> organization [13,42]. The ”classical” HDACscomprise classes I, II, and IV and utilize Zn for their functional activity. The class III histonedeacetylase are also known as Sirtu<strong>in</strong>s or SIRTs. These HDACs, of which there are sevenmembers, are nicot<strong>in</strong>amide aden<strong>in</strong>e d<strong>in</strong>ucleotide (NAD)-dependent [43], and strong


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>evidence for their roles <strong>in</strong> diabetes pathogenesis is emerg<strong>in</strong>g. For example, mice which havebeen bred to overexpress SIRT1 <strong>in</strong> their forebra<strong>in</strong> (especially females), exhibit <strong>in</strong>creased fataccumulation accompanied by a significant up-regulation of adipogenic genes <strong>in</strong> whiteadipose tissue. Furthermore, glucose tolerance <strong>in</strong> these mice is impaired and is coupled witha decrease <strong>in</strong> Glut4 mRNA <strong>in</strong> their muscle [44]. In addition, specific overexpression of SIRT1<strong>in</strong> the liver ameliorates systemic <strong>in</strong>sul<strong>in</strong> resistance <strong>in</strong> mice via enhanced liver <strong>in</strong>sul<strong>in</strong> receptorsignal<strong>in</strong>g, lead<strong>in</strong>g to decreased hepatic gluconeogenesis and improved glucose tolerance[45]. Furthermore, <strong>in</strong> livers of mice fed on an HFD (high-fat diet) compared with controls,hyperacetylation of prote<strong>in</strong>s <strong>in</strong>volved <strong>in</strong> gluconeogenesis, mitochondrial oxidative metabolism,liver <strong>in</strong>jury, and the ER (endoplasmic reticulum) stress response, are observed whichwas found to be due to reduced SIRT3 activity [46]. SIRT6-null mice die from hypoglycemia,whereas <strong>in</strong> contrast, neural-specific deletion of Sirt6 <strong>in</strong> mice leads to obesity [47,48].Lys<strong>in</strong>e demethylases (KDMs) and lys<strong>in</strong>e methyltransferases (KMTs) may also play functionalroles <strong>in</strong> diabetes pathogenesis. For <strong>in</strong>stance, prior hyperglycemia ("hyperglycemic memory")causes <strong>in</strong>creased recruitment of KDM1 (also known as LSD1) to the NFkB p65 subunitpromoter [49]. Furthermore levels of KDM1 have been shown to be significantly decreased <strong>in</strong>the vascular smooth muscle cells of diabetic mice [50].KMT7 (also known as SET7/9) has been shown to be a coactivator of NFkB, and <strong>in</strong> diabeticmice macrophages show <strong>in</strong>creased recruitment of KMT7 at <strong>in</strong>flammatory genes [51], and theexpression of KMT7 is enriched <strong>in</strong> pancreatic beta cells [52,53].336Taken together, it is clear that many of the enzymes/prote<strong>in</strong>s <strong>in</strong>volved with regulat<strong>in</strong>g histone/prote<strong>in</strong> PTMs may play important roles <strong>in</strong> the pathogenesis of diabetes, and <strong>in</strong> particular thoseprote<strong>in</strong>s which regulate histone/prote<strong>in</strong> acetylation. In this regard, target<strong>in</strong>g HDACs maytherefore be important for therapeutically target<strong>in</strong>g diabetes. In the next section we will discusssome of the results obta<strong>in</strong>ed for target<strong>in</strong>g these prote<strong>in</strong>s with<strong>in</strong> the diabetic sett<strong>in</strong>g.EFFECTS OF HDACI ON DIABETOGENE EXPRESSIONThe potential epigenetic basis for diabetes pathogenesis has been discussed <strong>in</strong> the previouschapter. The importance of histone acetylation <strong>in</strong> the regulation of genes central to diabetespathogenesis can be highlighted by a recent genome-wide profil<strong>in</strong>g analysis of humanmesenchymal (bone marrow) stem cell-derived adipocytes. Us<strong>in</strong>g this technique Fraenkel andcolleagues exam<strong>in</strong>ed H3K56 acetylation (mediated by KAT3A (CBP) and KAT3B (p300), anddeacetylated by SIRT1, SIRT2 and SIRT3) <strong>in</strong> adipocytes [54,55]. Critically, while this modificationcould be found across half the genome, the highest levels of H3K56 acetylation wereassociated with transcription factors and prote<strong>in</strong>s <strong>in</strong> the adipok<strong>in</strong>e signal<strong>in</strong>g and type 2 diabetespathways [54]. A full discussion on the effects of histone deacetylase <strong>in</strong>hibitors on thesegenes/pathways central to diabetes pathogenesis is beyond the remit of this chapter, and forthe purposes of the follow<strong>in</strong>g sections I will limit discussion of the effects of HDACi to a smallsample of genes from various classes associated with diabetes and <strong>in</strong>sul<strong>in</strong> sensitivity.CLASS I e HORMONESInsul<strong>in</strong>Early studies identified chromat<strong>in</strong> remodel<strong>in</strong>g <strong>in</strong>volv<strong>in</strong>g histone acetylation as a criticalregulatory mechanism of <strong>in</strong>sul<strong>in</strong> biosynthesis <strong>in</strong> resoponse to glucose levels [56e60]. Furtherfunctional analyses have shown that other important regulators of <strong>in</strong>sul<strong>in</strong> expression<strong>in</strong>clud<strong>in</strong>g pancreatic and duodenal homeobox-1 (PDX-1) and neurogenic differentiationfactor 1/beta-cell E-box transactivator 2 (NeuroD/BETA2) either associate with or require theactivities of chromat<strong>in</strong>-modify<strong>in</strong>g enzymes to regulate <strong>in</strong>sul<strong>in</strong> expression [61,62]. Hypothalamicexpression of KAT3A (CBP) and CBP-b<strong>in</strong>d<strong>in</strong>g partner Special AT-rich sequence b<strong>in</strong>d<strong>in</strong>gprote<strong>in</strong> 1 (SATB-1) has been found to be highly correlated with lifespan across five stra<strong>in</strong>s ofmice, and the expression of these genes decreases with both age and diabetes <strong>in</strong> mice [63].


CHAPTER 17The Potential of Epigenetic Compounds <strong>in</strong> Treat<strong>in</strong>g DiabetesThe transcription factor KLF11 has also been shown to functionally associate with KAT3A(p300) <strong>in</strong> pancreatic beta cells to mediate the activation of Pdx-1, a master regulator of islet<strong>in</strong>sul<strong>in</strong>-produc<strong>in</strong>g activity [64]. In addition, a novel KLF-KAT3A (p300) pathway for theregulation of <strong>in</strong>sul<strong>in</strong> biosynthesis has recently been elucidated from studies of the homozygousc.-331 mutation with<strong>in</strong> the <strong>in</strong>sul<strong>in</strong> promoter associated with neonatal diabetesmellitus [65].A study exam<strong>in</strong><strong>in</strong>g the histone modifications found at the <strong>in</strong>sul<strong>in</strong> gene <strong>in</strong> freshly isolated isletsfrom multiple human donors found that <strong>in</strong> contrast to most genes where activat<strong>in</strong>g modificationstend to be concentrated with<strong>in</strong> 1 kb around the transcription start site; these markswere <strong>in</strong> fact distributed over the entire cod<strong>in</strong>g region of the <strong>in</strong>sul<strong>in</strong> gene. In addition almostuniformly elevated levels of histone acetylation and H3K4 dimethylation was found at the<strong>in</strong>sul<strong>in</strong> gene and was accompanied by islet-specific coord<strong>in</strong>ate expression of <strong>in</strong>sul<strong>in</strong> with twoneighbor<strong>in</strong>g genes, tyros<strong>in</strong>e hydroxylase (TH) and <strong>in</strong>sul<strong>in</strong>-like growth factor 2 (IGF2),confirm<strong>in</strong>g the essential role of histone acetylation <strong>in</strong> the regulation of <strong>in</strong>sul<strong>in</strong> expression <strong>in</strong>pancreatic beta cells [66].A l<strong>in</strong>k between the pro<strong>in</strong>flammatory cytok<strong>in</strong>e IL-1b, histone acetylation and <strong>in</strong>sul<strong>in</strong> expressionhas emerged. IL-1b has been shown to be an important mediator of pancreatic beta cellloss [67]. In a study exam<strong>in</strong><strong>in</strong>g the impact of hyperglycemia on histone acetylation at the<strong>in</strong>sul<strong>in</strong> gene promoter, histone acetylation, KAT3A (p300), and RNA polymerase II (pol II)b<strong>in</strong>d<strong>in</strong>g were still observed after 4 h <strong>in</strong> 16mM glucose, but could no longer be detected if IL-1bwas also present [68], <strong>in</strong>dicat<strong>in</strong>g that IL-1b was prevent<strong>in</strong>g appropriate regulation of <strong>in</strong>sul<strong>in</strong>gene expression and that target<strong>in</strong>g IL-1b may be a potential means to both prevent pancreaticbeta cell loss, and improve <strong>in</strong>sul<strong>in</strong> expression from pancreatic beta cells. Indeed, the epigenetictarget<strong>in</strong>g drug vor<strong>in</strong>ostat can ablate IL-1b expression [69] and this has been shown to bewith<strong>in</strong> the diabetic sett<strong>in</strong>g to prevent aga<strong>in</strong>st beta cell loss [70,71]. As such, epigenetic therapywith HDACi may have a threefold benefit by (a) prevent<strong>in</strong>g pancreatic beta cell loss, (b)improv<strong>in</strong>g <strong>in</strong>sul<strong>in</strong> expression, and (c) reduc<strong>in</strong>g pro<strong>in</strong>flammatory cues by decreas<strong>in</strong>g IL-1b.337Adiponect<strong>in</strong>Adiponect<strong>in</strong> (Adpn), the most abundant prote<strong>in</strong> secreted by white adipose tissue,regulates energy homeostasis and glucose and lipid metabolism, and functions as anantidiabetic adipok<strong>in</strong>e with <strong>in</strong>sul<strong>in</strong>-sensitiz<strong>in</strong>g, anti-<strong>in</strong>flammatory, antiatherosclerotic, andcardioprotective properties <strong>in</strong> obesity-related disorders such as <strong>in</strong>sul<strong>in</strong> resistance and type 2diabetes [72].Chromat<strong>in</strong> modifications <strong>in</strong>clud<strong>in</strong>g histone lys<strong>in</strong>e acetylation (H3Ac) and methylation(H3K4me2) are <strong>in</strong>volved with the coord<strong>in</strong>ated regulation of Adpn <strong>in</strong> adipoctyedifferentiation [73].Dur<strong>in</strong>g the development of <strong>in</strong>sul<strong>in</strong> resistance <strong>in</strong> NIH-3T3 adipocytes decreased acetylation atlys<strong>in</strong>e 9 of histone H3 (H3K9) is seen at the promoter of this gene [74], <strong>in</strong>dicat<strong>in</strong>g thatpotentially HDACi may be able to alleviate this. As a caveat however, treatment of mice andcells with VPA was actually found to suppress adiponect<strong>in</strong> expression <strong>in</strong> mature adipocytes,but adiponect<strong>in</strong> promoter-driven luciferase expression <strong>in</strong> fibroblasts [75], but more studieswill be required to determ<strong>in</strong>e if adiponect<strong>in</strong> can be successfully <strong>in</strong>duced <strong>in</strong> the diabetic sett<strong>in</strong>gby HDACi.CLASS II e TRANSCRIPTION FACTORSPPARg/PGC-1aThe nuclear hormone receptor peroxisome proliferator-activated receptor gamma (PPARg)plays central roles <strong>in</strong> both metabolism and adipogenesis. Ligands such as thiazolid<strong>in</strong>ediones(TZDs) that b<strong>in</strong>d to this receptor (primarily on adipocytes) exert <strong>in</strong>sul<strong>in</strong>-sensitiz<strong>in</strong>g and


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>anti-<strong>in</strong>flammatory effects, and are widely used to treat metabolic syndrome, especially type 2diabetes. A number of PPARg <strong>in</strong>teract<strong>in</strong>g partners have been identified, many of which areknown epigenetic regulators, <strong>in</strong>clud<strong>in</strong>g enzymes for histone acetylation/deacetylation andhistone methylation/demethylation and has recently been extensively reviewed by Sugii andEvans [76]. PPARg regulates the expression of many important diabetogenes. One recentexample <strong>in</strong>volves PPARg and histone acetylation <strong>in</strong> the regulation of glucose-dependent<strong>in</strong>sul<strong>in</strong>otropic polypeptide receptor (GIPR), which has important roles with <strong>in</strong>sul<strong>in</strong>otropicgrowth and survival of pancreatic b-cells and adipocyte metabolism [77].Peroxisome proliferator-activated receptor gamma coactivator alpha (PGC-1a) is a criticalprote<strong>in</strong> that regulates many pathways l<strong>in</strong>ked to energy homeostasis and metabolism [78],through its associations with KATs/HDACs [23]. Given that levels of PGC-1a have been shownto be reduced <strong>in</strong> the muscles of patients with type 2 diabetes [79], and that the HDACi TSA canup-regulate expression of PGC-1a [80], it will be important to see whether other HDACi canaffect PGC-1a <strong>in</strong> the diabetic sett<strong>in</strong>g.PGC-1a has been shown to associate with the transcription factor Forkhead box prote<strong>in</strong> O(FOXO) and hepatocyte nuclear factor 4 alpha (HNF4a) to activate gluconeogenic geneexpression <strong>in</strong> the liver and regulate glucose homeostasis [81]. In addition acetylation of FOXOitself modulates its promoter specificity and transcriptional activity, and is regulated via theclass III deacetylase SIRT1 [82,83]. In a recent development, it has now been demonstratedthat the class II HDACs (HDAC4, 5, and 7) also play roles (along with the class I HDAC3) <strong>in</strong>regulat<strong>in</strong>g FOXO transcriptional activity by acetylation, and that deplet<strong>in</strong>g class II HDACs <strong>in</strong>mouse models of type 2 diabetes ameliorates this hyperglycemia [84,85]. From this latest seriesof developments it would appear that target<strong>in</strong>g HDACs may be a unique mechanism capableof regulat<strong>in</strong>g hepatic glucose homeostasis.338CLASS III e TRANSPORTERSGlut4Glut4 is an <strong>in</strong>sul<strong>in</strong>-dependent glucose transporter first described <strong>in</strong> rat skeletal muscle byBirnbaum [86]. It is generally only expressed <strong>in</strong> muscle and adipose tissue, and is typicallystored <strong>in</strong> <strong>in</strong>tracellular lipid rafts <strong>in</strong> these cells, and rapidly translocates to the plasmamembrane <strong>in</strong> response to <strong>in</strong>sul<strong>in</strong> signal<strong>in</strong>g [87]. The first evidence that Glut4 may be epigeneticallyregulated came from studies show<strong>in</strong>g that constitutive localization of HDAC5 <strong>in</strong>tothe nucleus <strong>in</strong> cardiac tissue resulted <strong>in</strong> a significant (~threefold) decrease <strong>in</strong> Glut4 expression[88]. Down-regulation of Glut4 expression <strong>in</strong> the muscle tissue of <strong>in</strong>trauter<strong>in</strong>e growthrestricted(IUGR) mice was found to <strong>in</strong>volve HDAC1 and HDAC4 [89]. Further studies haveshown that the repression of Glut-4 transcription also <strong>in</strong>volves complexes <strong>in</strong>volv<strong>in</strong>g the Glut4enhancer factor (GEF), myocyte enhancer factor 2 (MEF2), and HDAC5 [90,91]. GLUT4 geneexpression <strong>in</strong> preadipocytes is differentiation-dependent, with full expression delayed untillate <strong>in</strong> the differentiation program, and three class II HDACs (HDAC4, HDAC5, and HDAC9)have been shown to regulate this repression [92], and we have confirmed that Glut4 expressioncan be <strong>in</strong>duced <strong>in</strong> human SGBS preadipocytes us<strong>in</strong>g HDACi, and also show that the CTCFtranscription factor may be <strong>in</strong>volved with regulat<strong>in</strong>g Glut4 [93]. As CTCF regulates genetranscription through the formation of higher-order chromat<strong>in</strong> structures [94], this wouldclearly <strong>in</strong>dicate an important role for chromat<strong>in</strong> modifications and higher-order structures <strong>in</strong>the regulation of Glut4.The potential utility for the use of HDACi <strong>in</strong> target<strong>in</strong>g Glut4 came from an <strong>in</strong>itial study whichshowed that <strong>in</strong>creased Glut4 prote<strong>in</strong> translocation and <strong>in</strong>sul<strong>in</strong>-<strong>in</strong>duced uptake occurred <strong>in</strong>muscle cells treated with a HDACi [95]. As shown <strong>in</strong> Figure 17.2, we have demonstrated thatGlut4 expression can be <strong>in</strong>duced <strong>in</strong> many tissue types <strong>in</strong>clud<strong>in</strong>g, lung, liver, kidney, muscle,and preadipocytes [93]. Furthermore, levels of Glut4 mRNA transcript could be susta<strong>in</strong>ed


CHAPTER 17The Potential of Epigenetic Compounds <strong>in</strong> Treat<strong>in</strong>g Diabetes(A) (B) (C) (D)FIGURE 11.2Effect of HDACi on Glut4. Effects of histone deacetylase <strong>in</strong>hibitors (HDACi) on Glut-4 <strong>in</strong> cell l<strong>in</strong>es derived from <strong>in</strong>sul<strong>in</strong> target tissues. Various cell l<strong>in</strong>es(A) HEK-293 (Kidney), (B) H9C2 (muscle), (C) SGBS (preadipocyte), and (D) HepG2 (liver) were treated with or without the histone deacetylase <strong>in</strong>hibitorTrichostat<strong>in</strong> A (TSA), and mRNA expression of Glut-4 was exam<strong>in</strong>ed by RT-PCR. Densitometric analysis of Glut-4 expression with Beta-act<strong>in</strong> levels usedfor normalization purposes was carried out for each. Data are expressed as mean SEM. Statistical analysis was performed us<strong>in</strong>g a Student’s T-test.(UT, untreated; TSA, Trichostat<strong>in</strong> A). Previously published <strong>in</strong> [93].follow<strong>in</strong>g removal of the HDACi [93], <strong>in</strong>dicat<strong>in</strong>g that the use of HDACi may provide longtermelevation of Glut4 <strong>in</strong> <strong>in</strong>sul<strong>in</strong> target tissues.33917.4.3 Aberrant microRNAs/Epi-microRNAs and DiabetesSpecific miRNAs have now been shown to control the expression of some of the knownepigenetic regulators. mIR-34a targets the NAD-dependent histone deacetylase SIRT-1 [96],and is also critical for mediat<strong>in</strong>g lipotoxicity <strong>in</strong>duced beta-cell dysfunction [97]. Critically,miR-34a levels are elevated, and SIRT1 prote<strong>in</strong> levels are reduced <strong>in</strong> diet-<strong>in</strong>duced obese mice[98], <strong>in</strong>dicat<strong>in</strong>g that miRNAs that modulate epigenetic regulators may be important <strong>in</strong> obesity.Another miRNA, mIR-217 has also been shown to target SIRT1 [96,99]. This miRNA isessentially specific to pancreatic cells [100], and as such may be a critical l<strong>in</strong>k <strong>in</strong> diabetespathogenesis. Another miRNA, miR-132 has been shown to modulate SIRT-1 levels <strong>in</strong>response to nutrient deprivation [101]. Furthermore, mIR-132 is regulated by hyperglycemia[102], shows dysregulated expression <strong>in</strong> obese patients [103], and its other known targets<strong>in</strong>clude the chromat<strong>in</strong> modifiers Mecp2, KAT3B (p300), pRB, and KDM5A (Jarid1a) [104,105].In pancreatic beta-cell islets, the expression of SIRT-1 and mIR-9 are also <strong>in</strong>timately l<strong>in</strong>ked.When mir-9 expression is high dur<strong>in</strong>g glucose-dependent <strong>in</strong>sul<strong>in</strong> secretion, levels of SIRT-1prote<strong>in</strong> are reduced [106].Other miRNAs directly target the epigenetic regulatory mach<strong>in</strong>ery. A family of miRNAs, themiR-29 family, has been shown to target the DNA methyltransferases DNMT3A and DNMT3B[107,108]. In addition to be<strong>in</strong>g overexpressed <strong>in</strong> diabetic rats, overexpression of this miRNAleads to <strong>in</strong>sul<strong>in</strong> resistance <strong>in</strong> 3T3-L1 adipocytes, largely through repression of <strong>in</strong>sul<strong>in</strong>stimulatedglucose uptake [109]. mIR-29c has also recently been shown to be a key microRNA<strong>in</strong> diabetic nephropathy under high glucose conditions where it targets Sprouty homolog 1,and <strong>in</strong> vivo knockdown of this miRNA prevents progression of diabetic nephropathy [110].Furthermore, miR-143 has also been shown to target DNMT3A [111], and has been implicated<strong>in</strong> adipocyte differentiation [112]. This result was confirmed by Lodish and colleagues who


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>further noted that miR-143 was a miRNA whose expression was significantly decreased <strong>in</strong> theobese state [21]. Two other microRNAs, miR-148a and miR-152, have now been shown toregulate DNMT1 [113]. Intrigu<strong>in</strong>gly, <strong>in</strong> a comparison of miRNA profiles of human mesenchymalstem cells derived from breast, face, and abdom<strong>in</strong>al adipose tissues, mIR-152 was oneof a number of differentially expressed microRNAs which separated the breast and abdom<strong>in</strong>alfrom the facial stem cells [114]. mIR-194 is a further miRNA which has been hypothesized totarget DNMT3A [115]. More recently, this miRNA and another miRNA, miR-132, have <strong>in</strong> factbeen shown to regulate the expression of MeCP2 [116], a methyl-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> which hasbeen shown to associate with HDACs to regulate transcription.miR-449a has been shown to target both histone deacetylase 1 (HDAC1) [117], E2F1 [118],and SIRT-1 [119], but has yet to be associated with diabetes pathogenesis.In another <strong>in</strong>sul<strong>in</strong> target tissue, miR-1 targets HDAC4 specifically to regulate skeletal muscleproliferation and differentiation [120]. This miRNA may also play a critical role <strong>in</strong> diabeticcardiomyopathy as under high-glucose conditions, this miRNA is significantly elevated <strong>in</strong>cardiomyocytes, and accelerates apoptosis by deplet<strong>in</strong>g the cells of Hsp60, an importantcomponent of the cell’s defense mechanisms aga<strong>in</strong>st diabetic myocardial <strong>in</strong>jury [121].mIR-27b plays important roles <strong>in</strong> adipocyte differentiation and has been shown to specificallytarget PPARg, a prote<strong>in</strong> known to associate with HDACs [122]. miR-27a has also been shownto target PPARg, and ectopic expression of miR-27a <strong>in</strong> 3T3-L1 pre-adipocytes repressedadipocyte differentiation through reduction of PPARg expression [123]. Another miRNA thatregulates PPARg, miR-519d, was found to be overexpressed <strong>in</strong> subcutaneous adipose tissue(SAT) from nondiabetic severely obese (n ¼ 20) compared to non-obese adults (n ¼ 8) [124].340The microRNA mIR-101 has been shown to target the lys<strong>in</strong>e methyltransferase KMT6 (akaEZH2) [125]. KMT6 has now been shown to regulate both pancreatic beta cell expression andregeneration <strong>in</strong> diabetes [126], and also to facilitate adipogenesis [127].Other miRNAs have been identified which regulate other important pathways on severallevels <strong>in</strong> diabetes and obesity. For <strong>in</strong>stance miRNA-223 has been shown to directly regulateGlut-4 expression [128], while miRNA-133 has been shown to <strong>in</strong>directly alter Glut4expression by regulat<strong>in</strong>g KLF15 (MODY7), an essential regulator of Glut-4 [129], and <strong>in</strong>sul<strong>in</strong>biosynthesis. mIR-696 has now been shown to regulate the transcriptional coactivator PGC-1a <strong>in</strong> skeletal muscle <strong>in</strong> response to physical activity [130]. This would <strong>in</strong>dicate that miRNAsmay be altered <strong>in</strong> response to exercise. Intrigu<strong>in</strong>gly, levels of critical miRNAs such as mIR-1(target HDAC4) have been shown to alter <strong>in</strong> skeletal muscle as an adaptation to acuteendurance exercise [131].Transcription factors strongly associated with diabetes, PDX-1 and NeuroD1, have been shownto regulate the miRNA miR-375 [132]. This microRNA has now been shown to regulate3 0 -phospho<strong>in</strong>ositide-dependent prote<strong>in</strong> k<strong>in</strong>ase-1 and as such glucose-<strong>in</strong>duced biologicalresponses <strong>in</strong> pancreatic b-cells [133]. mIR-696 has now been shown to regulate the transcriptionalcoactivator PGC-1a <strong>in</strong> skeletal muscle <strong>in</strong> response to physical activity [130].mIR-103 and mIR-107 have also been shown to be critical regulators of <strong>in</strong>sul<strong>in</strong> sensitivity viatheir regulation of caveol<strong>in</strong>-1. Loss of these miRNAs results <strong>in</strong> improved glucose homeostasisand <strong>in</strong>sul<strong>in</strong> sensitivity, while their overexpression <strong>in</strong> either liver or fat is sufficient to <strong>in</strong>duceimpaired glucose homeostasis [134]. In a study of glucose-regulated miRNAs from pancreaticbcells, Özcan and colleagues demonstrated that <strong>in</strong> MIN6 pancreatic-b cells, expression of 61 of108 detectable miRNAs (56%) was altered <strong>in</strong> response to glucose [135].F<strong>in</strong>ally, critical roles for miRNAs have now been shown for pancreatic development. miRNA-375 was orig<strong>in</strong>ally <strong>in</strong>dicated as be<strong>in</strong>g a critical regulator of pancreatic islet development [136].Further studies have now revealed that four different islet-specific microRNAs (miR-7, miR-9,


CHAPTER 17The Potential of Epigenetic Compounds <strong>in</strong> Treat<strong>in</strong>g DiabetesmiR-375, and miR-376) are expressed at high levels dur<strong>in</strong>g human pancreatic islet development[137].EFFECTS OF INSULIN ON miRNAsInsul<strong>in</strong> can also modulate miRNA expression. A comparative microRNA (miRNA) expressionprofile of human skeletal muscle biopsies before and after a 3-h euglycemic-hyper<strong>in</strong>sul<strong>in</strong>emicclamp found that <strong>in</strong>sul<strong>in</strong> down-regulated the expressions of 39 dist<strong>in</strong>ct miRNAs <strong>in</strong> humanskeletal muscle [138].miRNAS AND DIABETESOne study exam<strong>in</strong><strong>in</strong>g gene and miRNA expression changes <strong>in</strong> skeletal muscle <strong>in</strong>sul<strong>in</strong> resistance<strong>in</strong> patients with type 2 diabetes (average age ~54 years) found that while the musclemRNA transcriptome is <strong>in</strong>variant with respect to <strong>in</strong>sul<strong>in</strong> or glucose homeostasis, a third ofmiRNAs detected <strong>in</strong> muscle were altered <strong>in</strong> disease (n ¼ 62), with many chang<strong>in</strong>g prior to theonset of cl<strong>in</strong>ical diabetes [139].EFFECTS OF OBESITY ON miRNAsIn a study profil<strong>in</strong>g approximately 220 miRNAs <strong>in</strong> pancreatic islets, adipose tissue, and liverfrom diabetes-resistant (B6) and diabetes-susceptible (BTBR) mice, more than half of themiRNAs profiled were expressed <strong>in</strong> all three tissues. In addition, many miRNAs <strong>in</strong> each tissueshowed significant changes <strong>in</strong> response to genetic obesity. In liver there were approximately 40miRNAs that were down-regulated <strong>in</strong> response to obesity <strong>in</strong> B6 but not BTBR mice, <strong>in</strong>dicat<strong>in</strong>gthat genetic differences between the mouse stra<strong>in</strong>s play a critical role <strong>in</strong> miRNA regulation, andthat genetics may play critical roles <strong>in</strong> miRNA expression [140].In a separate study of global miRNA expression <strong>in</strong> human adipocytes dur<strong>in</strong>g differentiation and<strong>in</strong> subcutaneous fat samples from non-obese (n ¼ 6) and obese with (n ¼ 9) and without (n ¼13) type 2 diabetes mellitus (DM-2) women, approximately 50 miRNAs (6.2%) significantlydiffered between fat cells from lean and obese subjects. Seventy miRNAs (8.8%) were highly andsignificantly up- or down-regulated <strong>in</strong> mature adipocytes as compared to preadipocytes.Seventeen miRNAs (2.1%) were correlated with anthropometrical (BMI) and/or metabolic(fast<strong>in</strong>g glucose and/or triglycerides) parameters, while 11 miRNAs (1.4%) were significantlyderegulated <strong>in</strong> subcutaneous fat from obese subjects with and without DM-2 [141].341In various mouse models of obesity, miR-143 has been shown to impair <strong>in</strong>sul<strong>in</strong>-stimulated AKTactivation and glucose homeostasis, and if mice are made deficient for this miRNA, they areprotected from the development of obesity-associated <strong>in</strong>sul<strong>in</strong> resistance [142]. Furthermore, <strong>in</strong>preadipocytes overexpression of this miRNA has been shown to <strong>in</strong>crease adipogenesis [21].EFFECTS OF EPIGENETIC INHIBITORS ON miRNAsmiRNAs themselves have been shown to be epigenetically regulated [143]. Indeed thepotential to epigenetically target or modulate miRNAs, which may play a role <strong>in</strong> diabetespathogenesis, came from a study on the effects of the histone deacetylase <strong>in</strong>hibitor (HDACi)trichostat<strong>in</strong> A (TSA) on miRNA expression <strong>in</strong> <strong>in</strong> vivo hepatocyte cultures of rat hepatocytes. Inthis study mIR-143 was shown to be the most down-regulated miRNA after 7 days of treatmentwith TSA [144], demonstrat<strong>in</strong>g the potential to functionally modulate critical miRNAs <strong>in</strong>diabetes pathogenesis through HDACi. Another miRNA (mIR-449a) is epigenetically regulatedvia histone H3 Lys27 trimethylation (H3K27me3), and can be dramatically <strong>in</strong>ducedfollow<strong>in</strong>g treatments with a comb<strong>in</strong>ation of a histone methyltransferase <strong>in</strong>hibitor (DZNep)and a HDACi (TSA) [118], and while this miRNA has yet to be implicated <strong>in</strong> diabetes pathogenesis,this result demonstrates the possibility that target<strong>in</strong>g miRNAs associated with diabetespathogenesis could be achieved via epigenetic therapies.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>17.4.4 Effects of Insul<strong>in</strong> Resistance/Metabolic Syndrome on EpigeneticModify<strong>in</strong>g EnzymesUs<strong>in</strong>g peripheral blood mononuclear cells (PBMCs) as an ex vivo surrogate model systemAvogaro and colleagues exam<strong>in</strong>ed SIRT1eSIRT7 gene and prote<strong>in</strong> expression <strong>in</strong> PBMCs of 54subjects (41 with normal glucose tolerance and 13 with metabolic syndrome). Their analysisshowed that SIRT1 levels (both mRNA and prote<strong>in</strong>) were down-regulated <strong>in</strong> the patients withmetabolic syndrome/<strong>in</strong>sul<strong>in</strong> resistance [145].17.5 NON-EPIGENETIC EFFECTS OF HISTONE MODIFIERPROTEINS WITH DIABETES PATHOGENESISWhilst epigenetics <strong>in</strong> its purist form relates to the regulation of gene expression through directmodifications of DNA or chromat<strong>in</strong>, the enzymes <strong>in</strong>volved with this epigenetic regulation alsoplay important roles <strong>in</strong> regulat<strong>in</strong>g various other cellular responses through post-translationalmodification of non-histone prote<strong>in</strong>s [146]. As such, a fourth mechanism can be envisaged<strong>in</strong>volv<strong>in</strong>g a "prote<strong>in</strong> code or language" where the activities of KATs/HDACs and <strong>in</strong>deed otherepigenetic modify<strong>in</strong>g enzymes such as lys<strong>in</strong>e methyltransferases, on such prote<strong>in</strong>s may affecttheir functions such that the aberrant regulation of target genes <strong>in</strong> diabetes pathogenesis mayalso occur [51,147]. This has important implications for us<strong>in</strong>g drugs such as HDACi, as <strong>in</strong>addition to their effects on chromat<strong>in</strong>, they also elicit important additional effects on otherpathways. In the next sections I will detail the potential utility of HDACi to target "nonepigenetic"events on the follow<strong>in</strong>g important elements of diabetes pathogenesis namely<strong>in</strong>flammation, and ER stress/chaperone activity.34217.5.1 Inflammation <strong>in</strong> Diabetes PathogenesisInflammation is a critical component traditionally associated with the destruction of thepancreatic beta cell <strong>in</strong> type 1 diabetes, but <strong>in</strong>creas<strong>in</strong>gly l<strong>in</strong>ked to type 2 diabetes pathogenesis[148]. The major <strong>in</strong>flammation mechanism associated with diabetes pathogenesis <strong>in</strong>volves theNFkB pathway [23]. The NFkB-Rel family consists of five subunits, but NFkB typically consistsof a heterodimeric prote<strong>in</strong> comprised of a p50 and a p65(RelA) subunit. Prote<strong>in</strong> acetylationvia lys<strong>in</strong>e acetylases would appear to be a central regulator of NFkB-mediated responses [17].The lys<strong>in</strong>e acetyltransferases KAT3A (p300) and KAT3B (CBP) act as key coactivators <strong>in</strong>regulat<strong>in</strong>g NFkB-driven gene expression via <strong>in</strong>teractions with the p65(RelA) subunit[149e151]. Likewise, KAT13A (SRC-1) potentiates NFkB transactivation through <strong>in</strong>teractionswith the other subunit p50 [152]. Sirt1 also regulates NFkB transactivation by physically<strong>in</strong>teract<strong>in</strong>g with p65(RelA) and <strong>in</strong>hibit<strong>in</strong>g transcription by deacetylat<strong>in</strong>g a critical lys<strong>in</strong>e atposition 310 [153].It has also been shown that NFkB transcription requires IKKa to phosphorylate SMRT whichstimulates the exchange of corepressor for coactivator complexes. In this regard, loss ofHDAC3 is observed follow<strong>in</strong>g this phosphorylation event [154].HDACi have been shown to <strong>in</strong>hibit NFkB-mediated responses [155]. Importantly, <strong>in</strong> relationto diabetes pathogenesis HDACi have also been found to dampen NFkB transactivation <strong>in</strong>studies <strong>in</strong>volv<strong>in</strong>g pancreatic beta cells (INS-1) [156].Two pro<strong>in</strong>flammatory genes associated with diabetes are IL-6 and Il-8 [157,158]. NFkB hasalso been shown to utilize the lys<strong>in</strong>e acetyltransferase activity of KAT3A/KAT3B (CBP/p300) tostimulate transcription of these genes [159] (Figure 17.3).Initial observations had l<strong>in</strong>ked the <strong>in</strong>hibitor kappa B k<strong>in</strong>ase complex (IKK) to the developmentof <strong>in</strong>sul<strong>in</strong> resistance through phosphorylation of IRS-1 by IKKb lead<strong>in</strong>g to impaireddownstream signal<strong>in</strong>g of PI3k<strong>in</strong>ase pathways [160]. Subsequently, two IKKb-mediated<strong>in</strong>flammation models have now directly l<strong>in</strong>ked IKKb-mediated <strong>in</strong>flammatory responses to the


CHAPTER 17The Potential of Epigenetic Compounds <strong>in</strong> Treat<strong>in</strong>g DiabetesFIGURE 17.3Simplified overview of how HDACi could target diabetes.Diagram summariz<strong>in</strong>g the available evidence l<strong>in</strong>k<strong>in</strong>gaberrant epigenetics such as (i) aberrant diabetogeneregulation, (ii) aberrant expression of miRNAs, (iii)aberrant expression of epigenetic regulators, (iv)aberrant non-epigenetic post-translational modificationof prote<strong>in</strong>s, result<strong>in</strong>g <strong>in</strong> <strong>in</strong>appropriate gene expression,cellular and tissue dysfunction, and the development ofdiabetes. This figure is reproduced <strong>in</strong> the color platesection.development of obesity-<strong>in</strong>duced <strong>in</strong>sul<strong>in</strong> resistance [161,162]. Other evidence l<strong>in</strong>k<strong>in</strong>g theNFkB pathway to HATs/HDACs have come from studies which demonstrate that the sequestrationof the p65 subunit of NFkB by the I-kappa-B alpha (IKBA) prote<strong>in</strong> also resulted <strong>in</strong> thecytoplasmic translocation of the nuclear corepressors N-CoR and SMRT [163], and from datademonstrat<strong>in</strong>g that diabetes-<strong>in</strong>duced extracellular matrix prote<strong>in</strong> expression was associatedboth with <strong>in</strong>creased histone acetyltransferase activity and NFkB activation [164].The pro<strong>in</strong>flammatory cytok<strong>in</strong>e IL-1b has been described as the gatekeeper of <strong>in</strong>flammation[165]. Indeed this cytok<strong>in</strong>e mediates important events dur<strong>in</strong>g type 1 diabetes pathogenesisboth <strong>in</strong> vivo and <strong>in</strong> vitro <strong>in</strong>clud<strong>in</strong>g the <strong>in</strong>duction of beta-cell islet apoptosis where itreduced the number of docked <strong>in</strong>sul<strong>in</strong> granules <strong>in</strong> live pancreas beta cells by 60% [67]. Ithas also been demonstrated that IL-1b utilizes NFkB to <strong>in</strong>duce a susta<strong>in</strong>ed decrease ofspecific beta-cell prote<strong>in</strong>s like <strong>in</strong>sul<strong>in</strong>, GLUT-2 and PDX-1, and regulates the entrance ofislet beta-cells <strong>in</strong>to the cell death program [166]. It has now been shown that <strong>in</strong> pancreaticbeta cells and isolated rat islets pro<strong>in</strong>flammatory cytok<strong>in</strong>es (<strong>in</strong>clud<strong>in</strong>g IL-1b) alteredtheexpression of HDAC- 1, -2, -6, and -11, and that furthermore this regulation was reducedupon the use of HDACi [167].343It is clear from the above that NFkB-mediated <strong>in</strong>flammation is central to pancreatic beta cellloss, and that epigenetic modifications may be essential to this process.17.5.2 ER Stress/Chaperones and Diabetes PathogenesisChaperones are important prote<strong>in</strong>s which function to assist the non-covalent fold<strong>in</strong>g/unfold<strong>in</strong>g and the assembly/disassembly of other macromolecular structures, but do not occur<strong>in</strong> these structures when the latter are perform<strong>in</strong>g their normal biological functions. The firstidentified chaperones assist <strong>in</strong> the correct assembly of nucleosomes, but we now know thatsuch chaperones also contribute to (i) the complex balance between nucleosome assemblyand reassembly dur<strong>in</strong>g transcription, and (ii) may equally be <strong>in</strong>volved as much <strong>in</strong> histoneeviction as <strong>in</strong> chromat<strong>in</strong> assembly [168]. Chaperones also act to prevent newly synthesizedpolypeptide cha<strong>in</strong>s and assembled subunits from aggregat<strong>in</strong>g <strong>in</strong>to non-functional structures.Many chaperones are heat shock prote<strong>in</strong>s (HSP), which are prote<strong>in</strong>s expressed <strong>in</strong> response toelevated temperatures or other cellular stresses. Two important families of these chaperonesare HSP70 [169], and HSP90 [170,171]. Many of these chaperones can be found <strong>in</strong> the


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>endoplasmic reticulum (ER), a large organelle consist<strong>in</strong>g of a network of <strong>in</strong>terconnected,closed membrane-bound vesicles. The ER is the site for the synthesis, fold<strong>in</strong>g, and modificationof secretory and cell-surface prote<strong>in</strong>s and provides many essential cellular functions,<strong>in</strong>clud<strong>in</strong>g the production of cell membrane components, prote<strong>in</strong>s, lipids, and sterols [172].Only correctly folded prote<strong>in</strong>s are transported out of the ER, and <strong>in</strong>completely folded prote<strong>in</strong>sare reta<strong>in</strong>ed with<strong>in</strong> the ER to either complete the fold<strong>in</strong>g process or to be targeted fordestruction [173]. Due to its important roles this organelle is vital to cellular homeostasis.Intrigu<strong>in</strong>gly some HSP prote<strong>in</strong>s have also been described as glucose <strong>in</strong>ducible. These areGlucose-Regulated Prote<strong>in</strong>, 78-KD, GRP78 (also known as HSPA5) [174]; Glucose-RegulatedProte<strong>in</strong>, 94-KD; GRP94 (also known as HSP90 Beta 1) [175]. This may have importantimplications <strong>in</strong> the pathogenesis of diabetes, where ER stress and glucose homeostasis playimportant roles [176e178].The ma<strong>in</strong>tenance of cellular homeostasis requires that a cell be able to sense, and respond tocircumvent, any stress elicited. There are many ways by which endogenous or exogenous stresscan occur <strong>in</strong> a cell; these <strong>in</strong>clude pathogenic <strong>in</strong>fection, chemical <strong>in</strong>sult, genetic mutation,nutrient deprivation, excess nutrients, and even normal differentiation. The process of mutantprote<strong>in</strong> fold<strong>in</strong>g is particularly sensitive to such <strong>in</strong>sults. Adaptive programs exist <strong>in</strong> the cellularcompartments responsible for prote<strong>in</strong>s procession and fold<strong>in</strong>g, which enable the detectionand correction of fold<strong>in</strong>g errors [179].344However, many conditions can <strong>in</strong>terfere with normal ER function lead<strong>in</strong>g to a situation calledER stress. Thus, ER stress can arise from a disturbance <strong>in</strong> prote<strong>in</strong> fold<strong>in</strong>g which results <strong>in</strong> anaccumulation of unfolded or misfolded prote<strong>in</strong>s with<strong>in</strong> the organelle [180]. If this occurs, theER has evolved specialized mechanisms that promote proper fold<strong>in</strong>g of aberrant prote<strong>in</strong>, thusprevent<strong>in</strong>g its aggregation. Simply put, when ER stress occurs, the ER responds by <strong>in</strong>duc<strong>in</strong>g theexpression of specific genes <strong>in</strong> an attempt to restore normal function and to ma<strong>in</strong>ta<strong>in</strong> cellularhomeostasis [181]. The pr<strong>in</strong>cipal mechanisms of endoplasmic reticulum (ER) stress responses<strong>in</strong>volve: (1) ER-Associated prote<strong>in</strong> Degradation (ERAD), (2) Endoplasmic overload response(EOR), (3) Unfolded prote<strong>in</strong> response (UPR), and (4) Cellular Death pathway [182e185].This four-stage model def<strong>in</strong>es the role of ER stress <strong>in</strong> the onset of cl<strong>in</strong>ical manifestations. TwoER stress-<strong>in</strong>duced signal transduction pathways have been described: the UPR [183] and theEOR [186] which attempt to re-establish normal ER function [187]. However, excessive orprolonged ER stress may overwhelm the cell and subsequently <strong>in</strong>itiate cell death via apoptosisor autophagy [188]. Critically, ER stress has been implicated as a major element <strong>in</strong> diabetespathogenesis [177,189e192].LINKING HATS/HDAC ACTIVITIES TO CHAPERONE ACTIVITIESProte<strong>in</strong>s with chaperone activity have now been shown to functionally associate withcomplexes conta<strong>in</strong><strong>in</strong>g HATs/HDACs. Early coimmunoprecipitation studies l<strong>in</strong>ked the class IHDACs (HDACs1-3) with the chaperone HSP70, while HSP60 was found to only be associatedwith HDAC1, and HSP90 was not found associated with any of the class I HDACs [193].Recently specific chaperones have been identified <strong>in</strong> a multifunctional KATechaperonecomplex that both acetylates histone H3 and deposits histones H3 and H4 onto DNA, thusfunctionally l<strong>in</strong>k<strong>in</strong>g chaperone activity, histone modification, and nucleosome assemblydur<strong>in</strong>g replication, chromosome position<strong>in</strong>g, and double-strand break (DSB) DNA repair[55,168,194e213], and that lys<strong>in</strong>e acetyltransferase activity is important <strong>in</strong> stabiliz<strong>in</strong>g thechaperone/histone H3/H4 complex [214]. The Jun dimerization prote<strong>in</strong>-2 (JDP2) has beenshown to have both <strong>in</strong>tr<strong>in</strong>isic chaperone activity and the ability to <strong>in</strong>hibit KAT3B (p300)-mediated acetylation of core histones <strong>in</strong> vitro and <strong>in</strong> vivo. This factor may therefore controltranscription via direct regulation of the modification of histones and the assembly of chromat<strong>in</strong>[215]. Another prote<strong>in</strong> with histone chaperone activity nucleophosph<strong>in</strong>, has beenshown to be acetylated by KATs both <strong>in</strong> vitro and <strong>in</strong> vivo, and this acetylation enhances its


CHAPTER 17The Potential of Epigenetic Compounds <strong>in</strong> Treat<strong>in</strong>g Diabetesability to regulate chromat<strong>in</strong> transcription [216], and is <strong>in</strong> agreement with the previousobservations by Turner and colleagues that the abilities of HDACs to remodel chromat<strong>in</strong> areenhanced by chaperones [193].Furthermore, regulation of various receptors and signal<strong>in</strong>g pathways often affected <strong>in</strong> diabetesrequires coord<strong>in</strong>ated acetylation of histone chaperones <strong>in</strong>clud<strong>in</strong>g androgen receptor signal<strong>in</strong>g[217], aryl hydrocarbon receptor (AhR) signal<strong>in</strong>g [218], estrogen receptor (ER) signal<strong>in</strong>g[219,220], glucocorticoid receptor (GR) signal<strong>in</strong>g [221], and the AKT/PI3K<strong>in</strong>ase signal<strong>in</strong>gpathway [222].LINKING KATS/HDAC ACTIVITIES TO ER STRESS PATHWAYSAggresomes are a cellular response to misfolded prote<strong>in</strong> accumulation as a consequence of ERstress. Emerg<strong>in</strong>g evidence l<strong>in</strong>ks both the activities of KATs/HDACs to ER stress, and furtherevidence <strong>in</strong>dicates that agents which target these prote<strong>in</strong>s may have utility <strong>in</strong> alleviat<strong>in</strong>g ER stress(Box 17.1). For <strong>in</strong>stance, dur<strong>in</strong>g cytokerat<strong>in</strong> aggresome (Mallory bodies) formation <strong>in</strong> hepatocytes,decreased lys<strong>in</strong>e acetyltransferase and <strong>in</strong>creased histone deacetylase activity was observed[223], while <strong>in</strong> a model of oxidative stress-<strong>in</strong>duced <strong>in</strong>clusion formation, treatment with theHDACi 4-phenylbutyrate was found to alleviate the formation of these <strong>in</strong>clusions [224].Further direct physical evidence for the association of KATs and HDACs with critical regulatoryelements with<strong>in</strong> the ER stress pathway is emerg<strong>in</strong>g. CHOP (C/EBP Homologous Prote<strong>in</strong>) is anER stress-<strong>in</strong>ducible prote<strong>in</strong> which plays a critical role <strong>in</strong> regulat<strong>in</strong>g programmed cell death.Regulation of the CHOP promoter <strong>in</strong>volves a complex conta<strong>in</strong><strong>in</strong>g JDP2 and HDAC3 [225].Inaddition, CHOP itself directly associates with the histone acetyltransferase KAT3B (p300) toregulate gene expression responses <strong>in</strong> response to ER stress [226], and importantly, <strong>in</strong>hibitionof HDACs prevents the degradation of the CHOP prote<strong>in</strong> [226].345BOX 17.1 PATHWAYS LINKING KATS/HDACS TO DIABETESDirect AssociationsGWAS associations (6q21 harbors HDAC2) l<strong>in</strong>ked to type 1 and type 2 diabetesMODY forms of diabetes l<strong>in</strong>ked directly to chromat<strong>in</strong> remodel<strong>in</strong>g activities (see Table 17.1)Mouse mutant models of chromat<strong>in</strong> remodel<strong>in</strong>g enzymes have <strong>in</strong>creased levels of adiposityand markers related to diabetes pathogenesisPancreas DevelopmentA large body of data l<strong>in</strong>ks pancreatic development with chromat<strong>in</strong> modifiers such as HDACsStem Cell DifferentiationA significant body of evidence suggests HDACi are useful <strong>in</strong> differentiat<strong>in</strong>g stem cellsInflammationMany roles for KATs/HDACs have been identified <strong>in</strong> the regulation of <strong>in</strong>flammatory processes<strong>in</strong>clud<strong>in</strong>g the regulation of NFkBDiabetogenesMany genes associated with diabetes pathogenesis are regulated by HDACsInsul<strong>in</strong> Resistance/SensitivityGenes such as Glut4 can be <strong>in</strong>duced via HDACi. HDACi can enhance <strong>in</strong>sul<strong>in</strong>-<strong>in</strong>duced glucoseuptakeER Stress/ChaperoneHDACi such as phenylbutyrate can act as chemical chaperones to alleviate ER stress <strong>in</strong> thepancreas


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>In the context of diabetes one of the central regulators of ER stress responses is a glucoseresponsivegene, Glucose Regulated Prote<strong>in</strong> 78 (GRP78 or BiP). This prosurvival ER chaperoneis <strong>in</strong>duced under conditions of ER stress, and has been shown to be regulated via histone PTMsand chromat<strong>in</strong> remodel<strong>in</strong>g through the activities of prote<strong>in</strong> arg<strong>in</strong><strong>in</strong>e methyltransferases(PRMT1) and K-acetyltransferases (KAT3B) [227]. Similar results have been observed for otherER-stress responsive promoters, whereby <strong>in</strong>creased histone H4 hyperacetylation was observedfollow<strong>in</strong>g <strong>in</strong>duction of ER stress [228]. The B lymphocyte-<strong>in</strong>duced maturation prote<strong>in</strong>-1(BLIMP-1) is another prote<strong>in</strong> associated with cellular stress. In some cellular models BLIMP-1is rapidly up-regulated dur<strong>in</strong>g the UPR [229]. This repressor prote<strong>in</strong> has been shown to directlyassociate with histone deacetylases to repress transcription [230], and this may <strong>in</strong>dicate thatBLIMP-1 may utilize histone deacetylases to repress important genes dur<strong>in</strong>g ER stress.Hepatic regulation of lipid and glucose homeostasis has been shown to <strong>in</strong>volve the activitiesof an ER stress <strong>in</strong>ducer, activat<strong>in</strong>g transcription factor 6 (ATF6). In response to ER stresscaused by glucose starvation, ATF6 translocates from the ER to the golgi, where it is cleaved byS1P and S2P proteases. It then translocates to the nucleus whereupon it abrogates SREBP2-mediated sterogenesis and lipogenesis by form<strong>in</strong>g a repressive complex with SREBP andHDAC1 [231].346As previously stated the GR signal<strong>in</strong>g pathway has been shown to be affected by the activities ofHDACs and chaperones. GR signal<strong>in</strong>g is critical for ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g glucose homeostasis <strong>in</strong> stress,<strong>in</strong>flammation, and dur<strong>in</strong>g fast<strong>in</strong>g [232], and a multiprote<strong>in</strong> HSP90/HSP70-based chaperonecomplex has been shown to be critical for regulat<strong>in</strong>g the steroid b<strong>in</strong>d<strong>in</strong>g, traffick<strong>in</strong>g, andturnover of the GR [233]. Recent studies have shown that the acetylation status of HSP90 iscritical to GR function. In cells deficient for HDAC6, HSP90-dependent maturation of theglucocorticoid receptor (GR) is compromised, result<strong>in</strong>g <strong>in</strong> GR defective <strong>in</strong> ligand b<strong>in</strong>d<strong>in</strong>g,nuclear translocation, and transcriptional activation [221]. Additional studies us<strong>in</strong>g an siRNA“knock-down” approach found that depletion of HDAC6 resulted <strong>in</strong> a markedly decreasedability to assemble stable GR/HSP90 heterocomplexes [234].In addition, the GR has been shown to functionally associate with HDACs to mediaterepression of gene expression [235e237]. This has important implications <strong>in</strong> preadipocytedifferentiation where glucocorticoid treatment strongly potentiates differentiation, by stimulat<strong>in</strong>gthe titration of the corepressor histone deacetylase 1 (HDAC1) from the C/EBPbpromoter [238,239].Clearly, the activities of histone/prote<strong>in</strong>-modify<strong>in</strong>g enzymes such as lys<strong>in</strong>e acetyltransferasesand histone deacetylases play important roles <strong>in</strong> ER stress-mediated responses, and mayfunctionally contribute to diabetes pathogenesis. It is also clear that target<strong>in</strong>g these enzymesmay also have some potential <strong>in</strong> alleviat<strong>in</strong>g the ER stress associated with diabetes pathogenesis.The follow<strong>in</strong>g section details how target<strong>in</strong>g HDACi <strong>in</strong> particular may be of benefit <strong>in</strong> thetreatment/management of diabetes.17.6 POTENTIAL FOR THE USE OF HDACi TO AMELIORATEOR TREAT SYMPTOMS OF DIABETES PATHOGENESISHDACi has the potential to be used to ameliorate or treat symptoms of diabetes pathogenesis.We will now discuss this further.17.6.1 Currently Developed DrugsDue to their highly conserved active doma<strong>in</strong>, histone deacetylases have been extensivelystudied for the development of <strong>in</strong>hibitors. Most of the currently designed <strong>in</strong>hibitors fall <strong>in</strong>tofour broad classes, short-cha<strong>in</strong> fatty acids (SCFAs), hydroxamates, benzamates, and cyclictetrapeptides, which for the most part target the class I and class II HDACs [240]. Several<strong>in</strong>hibitors of the class III HDACs (Sirtu<strong>in</strong>s) have now been synthesized and also show


CHAPTER 17The Potential of Epigenetic Compounds <strong>in</strong> Treat<strong>in</strong>g Diabetestherapeutic potential [241e246]. Additionally natural prodrugs which target histone deacetylaseshave also been isolated and <strong>in</strong>clude sulforaphane (SFN), diallyl disulfide (DADS),and resveratrol [247e249].17.6.2 Therapeutic ImplicationsMany histone deacetylase <strong>in</strong>hibitors have entered cl<strong>in</strong>ical trials primarily for the treatment ofcancer. Generally, most of the current <strong>in</strong>hibitors are well tolerated with<strong>in</strong> the cl<strong>in</strong>ical sett<strong>in</strong>g[23,250,251]. Several old drugs which have recently been discovered to target histone deacetylasesalready have FDA approval (e.g. valproic acid (VPA) [252]), and recent cl<strong>in</strong>ical trials ofthis drug target<strong>in</strong>g solid tumors both alone and <strong>in</strong> comb<strong>in</strong>ation with other agents demonstratedreasonable toxicities [253e255]. One of the caveats for the use of VPA <strong>in</strong> the treatmentof diabetes is that known side effects of this drug are <strong>in</strong>creased obesity and <strong>in</strong>sul<strong>in</strong> resistance[75,256e262]. This may be due to VPAs effects on lept<strong>in</strong>. In adipocytes it was found that VPAreduced lept<strong>in</strong> mRNA levels while TSA did not, suggest<strong>in</strong>g that VPA therapy may be associatedwith altered lept<strong>in</strong> homeostasis contribut<strong>in</strong>g to weight ga<strong>in</strong> <strong>in</strong> vivo, and that other HDAC<strong>in</strong>hibitors may not cause similar effects <strong>in</strong> relation to obesity and <strong>in</strong>sul<strong>in</strong> resistance [263].VPAwas also found to decrease the expression of adiponect<strong>in</strong> <strong>in</strong> differentiated 3T3-L1 adipocytesand this may therefore expla<strong>in</strong> the <strong>in</strong>creased <strong>in</strong>sul<strong>in</strong> resistance <strong>in</strong> patients treated with VPA[75]. It has also been suggested that the hyper<strong>in</strong>sul<strong>in</strong>emia associated with VPA can beattributed to <strong>in</strong>hibition of <strong>in</strong>sul<strong>in</strong> metabolism <strong>in</strong> the liver [262].Another therapeutic approach to diabetes <strong>in</strong>volves us<strong>in</strong>g salicylates to reduce both <strong>in</strong>sul<strong>in</strong>resistance [264,265], and protect pancreatic beta cells from apoptosis and impaired function[266]. Salicylates are traditionally thought of as <strong>in</strong>hibitors of cyclooxygenases, however anearly report also <strong>in</strong>dicates that these drugs may also affect histone acetylation [267], morerecently confirmed <strong>in</strong> studies exam<strong>in</strong><strong>in</strong>g embryo development where treatments with sodiumsalicylate were found to <strong>in</strong>hibit HDACs activity [268]. In a recent study on the efficacy ofaspir<strong>in</strong> <strong>in</strong> treat<strong>in</strong>g a rat model of diabetes (STZ), the authors found that aspir<strong>in</strong> treatmentsresulted <strong>in</strong> a significant reduction of hepatic NFkB activation and serum TNF-a levels withimproved <strong>in</strong>sul<strong>in</strong> resistance compared to the diabetic group [269].347HDACi have been shown to have effects on adipocyte differentiation. Treatment of 3T3-L1cells with HDAC <strong>in</strong>hibitors (apicid<strong>in</strong>, TSA, or SAHA), led to a dramatic reduction <strong>in</strong> preadipocytedifferentiation <strong>in</strong>to adipocytes, however, <strong>in</strong> contrast, sodium butyrate (NaB) treatment<strong>in</strong>creased adipocyte differentiation [270,271].Two recent studies have demonstrated that HDACi also have the capacity to ameliorate diabeticnephropathy. In one study, long-term adm<strong>in</strong>istration of vor<strong>in</strong>ostat decreased album<strong>in</strong>uria,mesangial collagen IV deposition, and oxidative-nitrosative stress <strong>in</strong> STZ diabetic mice,and attenuated renal <strong>in</strong>jury [272], while the other study exam<strong>in</strong><strong>in</strong>g one of the earliest featuresof diabetic nephropathy (renal enlargement), found that vor<strong>in</strong>ostat was able to blunt renalgrowth and glomerular hypertrophy <strong>in</strong> STZ-<strong>in</strong>duced diabetes [273].17.6.3 Pancreatic Islet Development, and Protection Us<strong>in</strong>g HistoneDeacetylase Inhibitors, or NFkB BlockadeSeveral recent publications have demonstrated the ability of histone deacetylase <strong>in</strong>hibitors toprotect pancreatic beta cell apoptosis. In various diabetes models, nicot<strong>in</strong>amide has frequentlybeen observed to both ameliorate and/or accelerate the reversal of diabetes and prevent irreversibleB-cell damage [274e277]. However, the ENDIT cl<strong>in</strong>ical trial assess<strong>in</strong>g whether thepretreatment with nicot<strong>in</strong>amide of non-diabetic <strong>in</strong>dividuals predisposed to the developmentof diabetes could prevent or delay cl<strong>in</strong>ical onset of diabetes was <strong>in</strong>effective at the dose used[278], but did however reduce high secretion of IFN-g <strong>in</strong> high-risk <strong>in</strong>dividuals [279]. Insul<strong>in</strong>secret<strong>in</strong>gcells exposed long term to either nicot<strong>in</strong>amide or sodium butyrate were found to


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>have reduced viability and <strong>in</strong>sul<strong>in</strong> sensitivity, yet enhanced <strong>in</strong>sul<strong>in</strong> secretory responsiveness toa wide range of beta cell stimulators [280]. Both trichostat<strong>in</strong> A (TSA) and suberoylanilidehydroxamic acid (SAHA) were shown to prevent cytok<strong>in</strong>e-<strong>in</strong>duced toxicity <strong>in</strong> pancreatic betacells [156], while another HDACi (THS-78-5) has been shown to prevent IL-1b-<strong>in</strong>ducedmetabolic dysfunction <strong>in</strong> pancreatic beta-cells [71]. Most recently, the HDACi ITF2357 wasshown to normalize streptozotoc<strong>in</strong> (STZ)-<strong>in</strong>duced hyperglycemia at the cl<strong>in</strong>ically relevantdoses of 1.25e2.5 mg/kg, and protects pancreatic b cells <strong>in</strong> vivo and <strong>in</strong> vitro [70].Islet transplantation is a widely pursued potential therapy for the treatment of patients withdiabetes. However, currently this is limited by difficulties <strong>in</strong> their isolation from donors andma<strong>in</strong>tenance <strong>in</strong> culture. A novel mechanism to enhance this has recently been elicited wherebyHDACi (TSA and VPA) were found to rejuvenate isolated islets, enhanc<strong>in</strong>g the expression ofkey genes (<strong>in</strong>clud<strong>in</strong>g <strong>in</strong>sul<strong>in</strong> 1, <strong>in</strong>sul<strong>in</strong> 2), with <strong>in</strong>creased glucose-stimulated <strong>in</strong>sul<strong>in</strong> secretionand engraftment efficacy <strong>in</strong> xenogeneic transplantation model [281].17.6.4 Modulation of Th17-Mediated AutoimmunityEffector T-cells show great plasticity. Th17 cells are acknowledged to be <strong>in</strong>strumental <strong>in</strong> theresponse aga<strong>in</strong>st microbial <strong>in</strong>fection, but are also associated with autoimmune <strong>in</strong>flammatoryprocesses particularly with type 1 diabetes development, and now also with type 2 diabetes[282e287]. The HDACi TSA dramatically reduced the emergence of IL-17-produc<strong>in</strong>g cells fromTregs [288], imply<strong>in</strong>g (a) that Treg differentiation <strong>in</strong>to IL-17-produc<strong>in</strong>g cells depends onhistone/prote<strong>in</strong> deacetylase activity and (b) that HDACi may be useful <strong>in</strong> dampen<strong>in</strong>g downTh17-mediated autoimmunity <strong>in</strong> diabetes pathogenesis. In agreement with this, treatment ofa NOD mouse model of autoimmune diabetes dur<strong>in</strong>g the transition from prediabetic to diabeticstage with a HDACi (TSA) was found to effectively reduce the <strong>in</strong>cidence of diabetes [289].34817.6.5 Stem CellsA currently hotly pursued therapeutic avenue for diabetes centers on embryonic stem (ES) celltechnology [290]. Evidence is emerg<strong>in</strong>g <strong>in</strong>dicat<strong>in</strong>g that histone deacetylases may be animportant consideration <strong>in</strong> the development of this technology. Indeed histone deacetylaseactivity has been shown to be required for ES cell differentiation [291]. The importance ofHDAC <strong>in</strong>hibitors <strong>in</strong> differentiat<strong>in</strong>g embryonic stem cells <strong>in</strong> general has been reviewed recentlyelsewhere and the reader is directed to the follow<strong>in</strong>g reviews [292,293].Nicot<strong>in</strong>amide, a SIRT-specific <strong>in</strong>hibitor, was also used to differentiate embryonic stem cells <strong>in</strong>tostructures resembl<strong>in</strong>g pancreatic islets and which secreted <strong>in</strong>sul<strong>in</strong> [294]. Indeed a role for SIRT1is the differentiation of stem cells. Levels of SIRT1 are precisely down-regulated dur<strong>in</strong>g humanembryonic stem cell differentiation at both mRNA and prote<strong>in</strong> levels by a pathway at thearg<strong>in</strong><strong>in</strong>e methyltransferase coactivator-associated arg<strong>in</strong><strong>in</strong>e methyltransferase 1 (CARM1), and itis this down-regulation that leads to the reactivation of key developmental genes [295].Under appropriate culture conditions, bone marrow stem cells (BMSC) cultured <strong>in</strong> the presenceof the histone deacetylase <strong>in</strong>hibitor trichostat<strong>in</strong> A can be differentiated <strong>in</strong>to islet-like clusterssimilar to the cells of the islets of the pancreas and capable of secret<strong>in</strong>g <strong>in</strong>sul<strong>in</strong> [296]. Us<strong>in</strong>gBMSCs derived from diabetic patients, similar results were obta<strong>in</strong>ed us<strong>in</strong>g nicot<strong>in</strong>amide asone of the f<strong>in</strong>al steps <strong>in</strong> the differentiation process [297].Two recent articles have utilized the histone deacetylase <strong>in</strong>hibitor sodium butyrate to (a)stimulate early pancreatic development <strong>in</strong> embryonic stem cells [298], and (b) generate isletlikeclusters from human embryonic stem cells grown under feeder-free conditions [299].<strong>Human</strong> islet-derived precursor cells (hIPCs), are mesenchymal stem cells derived <strong>in</strong> vitro fromadult pancreas,and because they can be expanded prior to differentiation they offer thepotential to be a major source of transplantable islet cells if they can be differentiatedsuccessfully <strong>in</strong>to <strong>in</strong>sul<strong>in</strong>-produc<strong>in</strong>g cells. However, an issue with most hIPCs is that dur<strong>in</strong>g


CHAPTER 17The Potential of Epigenetic Compounds <strong>in</strong> Treat<strong>in</strong>g Diabetesproliferative expansion, <strong>in</strong>sul<strong>in</strong> mRNA transcript becomes undetectable and cannot be<strong>in</strong>duced, a phenomenon consistent with epigenetic silenc<strong>in</strong>g of the <strong>in</strong>sul<strong>in</strong> gene [300,301].In an analysis of the chromat<strong>in</strong> from hIPCs, patterns of histone modifications over the <strong>in</strong>sul<strong>in</strong>gene <strong>in</strong> human islets and hIPCs were compared aga<strong>in</strong>st HeLa and human bone marrowderivedmesenchymal stem cells (hBM-MSCs), neither of which expresses <strong>in</strong>sul<strong>in</strong>. The <strong>in</strong>sul<strong>in</strong>gene <strong>in</strong> islets were found to display high levels of histone modifications (H4 hyperacetylationand dimethylation of H3 lys<strong>in</strong>e 4) typical of active genes which were not present <strong>in</strong> HeLa andhBM-MSCs, which <strong>in</strong>stead had elevated levels of H3 lys<strong>in</strong>e 9 dimethylation (H3K9me2),a mark of <strong>in</strong>active genes. hIPCs also showed significant levels of active chromat<strong>in</strong> modifications,as much as half those seen <strong>in</strong> islets, and show no measurable H3 K9me2. If cells wereexpanded from a m<strong>in</strong>or population of mesenchymal stromal cells found <strong>in</strong> islets, these cellsexhibited the same histone modifications as established hIPCs. The authors therefore concludethat hIPCs, which do not express the <strong>in</strong>sul<strong>in</strong> gene, nonetheless uniquely exhibit epigeneticmarks that could poise them for activation of <strong>in</strong>sul<strong>in</strong> expression [300]. Even with the absenceof <strong>in</strong>sul<strong>in</strong> expression transplanted hIPCs <strong>in</strong> mice matured <strong>in</strong>to functional cells that secretedhuman C-peptide <strong>in</strong> response to glucose, and transcripts for <strong>in</strong>sul<strong>in</strong>, glucagon, and somatostat<strong>in</strong><strong>in</strong> recovered grafts <strong>in</strong>creased with time <strong>in</strong> vivo, reach<strong>in</strong>g levels approximately 1% ofthose <strong>in</strong> adult islets [302].When taken together, it is clear that studies with epigenetic <strong>in</strong>hibitors such as HDACi arewarranted <strong>in</strong> hIPCs and other stem cells as they may be of great utility <strong>in</strong> enhanc<strong>in</strong>g differentiationof patient stem cells <strong>in</strong>to islet cells, which could be then used for transplantation.17.6.6 The ability of phenylbutyrate to act as a chaperoneand alleviate ER stressA significant body of work has shown that phenylbutyrate and valproate can both function as“chemical chaperones” to alleviate ER stress. For example, 4-PBA relieves ER stress <strong>in</strong> cell l<strong>in</strong>emodels of cystic fibrosis [303,304], alpha-1 antitryps<strong>in</strong> Z prote<strong>in</strong> (alpha1-ATZ) liver disease[305], and may act as a chemical chaperone to relieve ER stress <strong>in</strong>duced <strong>in</strong> models of ischemia[306,307], cataract formation [308], and ret<strong>in</strong>itis pigmentosa [309]. PB also prevents ER stressmediatedaggregate formation <strong>in</strong> a model of hereditary haemochromatosis (HH) [310], andalleviates ER stress <strong>in</strong> Park<strong>in</strong>son’s disease [311]. VPA also alleviates ER stress, as cells treatedwith VPA have up-regulated expression of Grp78/BiP, Grp94, and calreticul<strong>in</strong> [312,313]result<strong>in</strong>g <strong>in</strong> neuroprotection [314].349In a cell l<strong>in</strong>e model of misfolded low-density lipoprote<strong>in</strong> receptors, PB was shown to restorereceptor functionality, and shuttle them to the cell surface. The authors concluded from theseresults that phenylbutyrate did not just solely mediate this response by its ability to <strong>in</strong>ducegene expression of prote<strong>in</strong>s <strong>in</strong>volved <strong>in</strong> <strong>in</strong>tracellular transport, but could also mediate thiseffect via a direct chemical chaperone activity [315].Phenylbutyrate has also been shown to alleviate ER stress with<strong>in</strong> the diabetic sett<strong>in</strong>g. For<strong>in</strong>stance, obesity is a major factor <strong>in</strong> the development of diabetes [316], and the adiposederivedhormone, lept<strong>in</strong>, is a critical regulator which functions to <strong>in</strong>hibit food <strong>in</strong>take andprevent body weight ga<strong>in</strong> [317,318]. However, the use of lept<strong>in</strong> as a therapy for the treatmentof obesity has been hampered by the fact that the majority of obese patients demonstrate“lept<strong>in</strong> resistance”, lead<strong>in</strong>g to the notion that lept<strong>in</strong> resistance may be one of the ma<strong>in</strong> causesof obesity [318]. Several reports have now shown that ER stress is a major factor underp<strong>in</strong>n<strong>in</strong>glept<strong>in</strong> resistance, and treatment with 4-phenylbutyrate has been shown to alleviatethe ER stress and restore lept<strong>in</strong> sensitivity [319e321]. Increased glycemia and reducedmelaton<strong>in</strong> (Mel) levels have been recently shown to coexist <strong>in</strong> diabetic patients at the end ofthe night period. In a rat model mimick<strong>in</strong>g this situation, the absence of melaton<strong>in</strong> <strong>in</strong>ducednight-time hepatic <strong>in</strong>sul<strong>in</strong> resistance and <strong>in</strong>creased gluconeogenesis due to stimulation of


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>nocturnal unfolded prote<strong>in</strong> response, which could be alleviated us<strong>in</strong>g phenylbutyrate [322].Furthermore, 4-phenybutyrate has also been shown to both relieve ER stress and restoreglucose homeostasis by the restoration of systemic <strong>in</strong>sul<strong>in</strong> sensitivity, resolution of fatty liverdisease, and enhancement of <strong>in</strong>sul<strong>in</strong> action <strong>in</strong> liver, muscle, and adipose tissues <strong>in</strong> a mousemodel of type 2 diabetes [323]. This has led to the <strong>in</strong>stigation of a phase IV cl<strong>in</strong>ical trial(NCT00533559) by the University Health Network, Toronto, to exam<strong>in</strong>e the effect ofBuphenyl Ò (phenylbutyrate) on fatty acid-<strong>in</strong>duced impairment of glucose-stimulated <strong>in</strong>sul<strong>in</strong>secretion, which has recently been completed and this small study <strong>in</strong>volv<strong>in</strong>g eight patientsfound that Buphenyl was able to partially ameliorate both <strong>in</strong>sul<strong>in</strong> resistance and b-celldysfunction <strong>in</strong> these patients, demonstrat<strong>in</strong>g the potential utility of this HDACi/chemicalchaperone <strong>in</strong> the treatment of diabetes [324]. F<strong>in</strong>ally treatment of experimentally <strong>in</strong>duceddiabetic mice that had undergone islet transplants with phenylbutyrate postoperatively wasfound to enhance islet engraftment with a higher cumulative cure rate of diabetes (p


CHAPTER 17The Potential of Epigenetic Compounds <strong>in</strong> Treat<strong>in</strong>g Diabetesdevelop<strong>in</strong>g diabetes, or treat patients for whom diabetes-related comorbidities or complicationsprevent or reduce their ability to exercise. A large body of evidence exists show<strong>in</strong>g thatmany natural compounds can <strong>in</strong>hibit the epigenetic mach<strong>in</strong>ery, and <strong>in</strong>clude well-knowncompounds which can act as HDACi [333e336].SULFORAPHANESulforaphane is an organic isothiocyanate (ITC) found <strong>in</strong> cruciferous plants such as broccoli,and with<strong>in</strong> the diabetic sett<strong>in</strong>g, sulforaphane has been shown to (a) protect b-cell damage bysuppress<strong>in</strong>g NFkB pathways [337], and (b) <strong>in</strong> studies of islet transplantation, pre-treatmentwith sulforaphane 24 hours prior to transplantation was found to lead to improved long-termislet function <strong>in</strong> vivo [338]. Some <strong>in</strong>dividuals caution aga<strong>in</strong>st the use of such dietarycompounds as these may not be specific enough and may tend to target prote<strong>in</strong>s moreessential to an organism than specific disease genes [339]. Nevertheless a nutrition-basedapproach for delivery of specific target<strong>in</strong>g therapies may have potential benefit <strong>in</strong> patientssuffer<strong>in</strong>g from diabetes. Indeed a recent study found that sulforaphane when used at nutritionallevels protected mesenchymal stem cells from apoptosis and senescence and promotedtheir proliferation [340].CURCUMINOne of the most <strong>in</strong>tensively studied natural compounds for its potential role <strong>in</strong> treat<strong>in</strong>g diabetesis the compound curcum<strong>in</strong>. Derived from turmeric, this compound shows manypleiotropic effects, one of which is to <strong>in</strong>hibit histone deacetylases [341,342]. Conversely it hasalso been shown to <strong>in</strong>hibit lys<strong>in</strong>e acetyltransferases [342e344].Curcum<strong>in</strong> has antidiabetic effects <strong>in</strong> adipocytes, pancreatic cells, hepatic stellate cells,macrophages, and muscle cells, revers<strong>in</strong>g <strong>in</strong>sul<strong>in</strong> resistance, hyperglycemia, and hyperlipidemia,by suppress<strong>in</strong>g the pro<strong>in</strong>flammatory transcription factors such as NFkB and activatesPPARg lead<strong>in</strong>g to the down-regulation of adipok<strong>in</strong>es, and the up-regulation of adiponect<strong>in</strong>and other gene products [345].351Indeed one of the first case studies of the use of this compound <strong>in</strong> patients was <strong>in</strong> a diabeticpatient almost four decades ago [346]. In a more recent small patient study, <strong>in</strong>gestion of 6 g ofa curcum<strong>in</strong> preparation <strong>in</strong>creased postprandial serum <strong>in</strong>sul<strong>in</strong> levels, but had no significanteffect on overall glucose response as measured by an oral glucose tolerance test [347]. One ofthe ongo<strong>in</strong>g issues with the use of curcum<strong>in</strong> <strong>in</strong> humans concerns its poor absorption and thelarge quantity needed to be effective. However, it has been shown that curcum<strong>in</strong> has <strong>in</strong>creasedbioavailability when comb<strong>in</strong>ed with phospholipids. When complexed with soy lecitih<strong>in</strong>curcum<strong>in</strong> (Meriva Ò ) has 29-fold <strong>in</strong>creased absorption <strong>in</strong> human patients [348]. Other technologiessuch as nanoparticles have also been shown to improve the delivery of curcum<strong>in</strong>[349]. A phase I cl<strong>in</strong>ical trial <strong>in</strong>volv<strong>in</strong>g nano-curcum<strong>in</strong> has been <strong>in</strong>itiated <strong>in</strong> patients withadvanced malignancies to identify the maximum dose limits (Cl<strong>in</strong>icaltrial.govNCT01201694).Taken together further studies with curcum<strong>in</strong> are warranted to determ<strong>in</strong>e whether anutritional-based compound such as curcum<strong>in</strong> may have either chemopreventative benefit<strong>in</strong> patients at risk of develop<strong>in</strong>g diabetes, or may have a role <strong>in</strong> the management of patientswith prediabetes or type 2 diabetes.RESVERATROLResveratrol is another natural compound which has been extensively studied for its potentialutility <strong>in</strong> the management of diabetes. This compound is thought to be an activator of Sirt1[248], but this has s<strong>in</strong>ce been called <strong>in</strong>to question [350]. Potential beneficial effects ofresveratrol <strong>in</strong>clude protection of b cells <strong>in</strong> diabetes, reduction of <strong>in</strong>sul<strong>in</strong> secretion, <strong>in</strong>hibitionof cytok<strong>in</strong>e action, and attenuation of the oxidative damage of the pancreatic tissue by


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>resveratrol, and <strong>in</strong> studies of animals with <strong>in</strong>sul<strong>in</strong> resistance resveratrol may also improve<strong>in</strong>sul<strong>in</strong> action <strong>in</strong>volv<strong>in</strong>g reduced adiposity, changes <strong>in</strong> gene expression, and changes <strong>in</strong> theactivities of some enzymes [351].Recent studies have also evaluated safety and potential mechanisms of activity follow<strong>in</strong>gmultiple dos<strong>in</strong>g, and have found resveratrol to be safe and reasonably well-tolerated at dosesof up to 5 g/day, although it is anticipated that the doses used <strong>in</strong> future trials will be significantlyless than this amount. In 2008, an unpublished phase 1b cl<strong>in</strong>ical trial of a reformulatedversion of resveratrol (SRT501) <strong>in</strong> patients with type 2 diabetes, test<strong>in</strong>g either 1.25 or 2.5 gramsof SRT501 given twice daily, found that the patient group receiv<strong>in</strong>g 2.5 grams twice a day hadsignificantly lower blood glucose levels as determ<strong>in</strong>ed through an oral glucose tolerance test(OGTT) at the test’s 2-hour timepo<strong>in</strong>t, as compared with the placebo group. Several cl<strong>in</strong>icaltrials are currently ongo<strong>in</strong>g or have been completed us<strong>in</strong>g resveratrol or SRT501 <strong>in</strong> the diabeticsett<strong>in</strong>g (Cl<strong>in</strong>icaltrials.gov identifiers NCT01158417, NCT01375959, NCT01354977,NCT01150955, NCT00823381, NCT00998504, NCT01038089), but to my knowledge theresults of these trials have as yet to be published. It must also be noted that the cl<strong>in</strong>icaldevelopment of SRT501 by GlaxoSmithKl<strong>in</strong>e PLC has been discont<strong>in</strong>ued due to the developmentof kidney complications <strong>in</strong> malignant myeloma patients undertak<strong>in</strong>g a small cl<strong>in</strong>icaltrial of this drug. As such, care must be taken <strong>in</strong> evaluat<strong>in</strong>g the potential benefits of resveratrolsupplements <strong>in</strong> patients with diabetes, particularly given that resveratrol is also available assupplement pills and liquids, <strong>in</strong> which it is sometimes comb<strong>in</strong>ed with vitam<strong>in</strong>s and/or other<strong>in</strong>gredients. The supplements are generally labeled as conta<strong>in</strong><strong>in</strong>g from 20 to 500 mg per tabletor capsule. However, the purity of these products is unknown, and because dietary supplementsare loosely regulated, it should not be assumed that the labeled dosage is accurate.352Clearly, further work will be required to establish whether resveratrol may be an effectivetreatment for diabetes.GlaxoSmithKl<strong>in</strong>e PLC have <strong>in</strong>stead focused on the development of new small-moleculechemical entities that activate SIRT1 that are structurally dist<strong>in</strong>ct from resveratrol. Threecompounds have been identified which have entered various cl<strong>in</strong>ical trials (SRT2104,SRT2379, and SRT3025). In precl<strong>in</strong>ical studies, SRT2104 demonstrated a moderate lower<strong>in</strong>g ofblood glucose, reduced weight ga<strong>in</strong>, <strong>in</strong>creased energy expenditure, improved exercise tolerance,and improved <strong>in</strong>sul<strong>in</strong> sensitivity <strong>in</strong> the diet-<strong>in</strong>duced obesity model of type 2 diabetes.Two phase II cl<strong>in</strong>ical trials of this compound have been completed <strong>in</strong> patients with diabetes(Cl<strong>in</strong>icalTrials.gov identifiers NCT01018017, NCT01031108), but to my knowledge the resultsof these have yet to be published.17.6.10 miRNA-/siRNA-Based TherapeuticsGiven the emerg<strong>in</strong>g body of evidence l<strong>in</strong>k<strong>in</strong>g aberrant miRNA expression with diabetespathogenesis, it is becom<strong>in</strong>g <strong>in</strong>creas<strong>in</strong>gly clear that these ncRNAs may have utility <strong>in</strong> either themanagement or treatment of this disease. The possibility of us<strong>in</strong>g miRNA and/or siRNA totarget diabetes has recently been extensively reviewed [352,353]. For example, dur<strong>in</strong>g thedifferentiation of human embryonic stem cells to hepatocytes, the cells are <strong>in</strong>duced <strong>in</strong>todef<strong>in</strong>itive endoderm (DE) cells before be<strong>in</strong>g further differentiated to hepatocytes. At the DEstage it has been shown that levels of mIR-9 are significantly elevated [354]. As the pancreas isan endodermal tissue and human embryonic stem cells can be <strong>in</strong>duced to form pancreaticendoderm [355], it may be possible to <strong>in</strong>duce hESCs to form pancreatic endoderm throughoverexpression of this miRNA.Clearly, this is an excit<strong>in</strong>g area of epigenetic regulation of gene expression which may havegreat utility <strong>in</strong> the treatment of diabetes, and will require further studies. One area which willwarrant further studies will be to exam<strong>in</strong>e whether epi-miRNAs (those miRNAs which targetthe epigenetic mach<strong>in</strong>ery) can be used to epigenetically target diabetes. As stable miRNAs are


CHAPTER 17The Potential of Epigenetic Compounds <strong>in</strong> Treat<strong>in</strong>g Diabetesfound circulat<strong>in</strong>g <strong>in</strong> patient serum either freely or conta<strong>in</strong>ed <strong>in</strong> exosomes [356], the nature androle of these <strong>in</strong> diabetes pathogenesis and the potential to utilize these to treat diabetespathogenesis is <strong>in</strong>creas<strong>in</strong>gly becom<strong>in</strong>g important. Furthermore, those miRNAs which areaberrantly overexpressed <strong>in</strong> diabetes could also be targeted us<strong>in</strong>g antagomirs [357].17.7 CONCLUSIONSIt is clear that epigenetic regulation is a central tenet of critical pathways <strong>in</strong> diabetespathogenesis.From the significant body of work presented <strong>in</strong> this review, it is clear that further studies will berequired to exam<strong>in</strong>e the therapeutic potential of compounds which target the epigeneticmach<strong>in</strong>ery <strong>in</strong> the treatment/management of diabetes pathogenesis. In the short term, HDAC<strong>in</strong>hibitors would appear to be the current best candidates. However, as these <strong>in</strong>hibitors affectmany genes it is hoped that the balance of genes altered by such treatments would be tippedfrom unfavorable to favorable diabetogenes. Indeed the next generation of these <strong>in</strong>hibitorsmay have better specificity and efficacy. Currently miRNA/siRNA-based therapeutics are <strong>in</strong>their <strong>in</strong>fancy, but show great promise for the future. A greater understand<strong>in</strong>g of miRNA targetidentification/validation, their roles <strong>in</strong> diabetes pathogenesis, and mechanisms of specificdelivery will be required before they can be evaluated as a therapeutic modality. F<strong>in</strong>ally, asnutritional compounds have been shown to act as HDACi, future studies will be required totest the possibility of <strong>in</strong>corporat<strong>in</strong>g nutritional-based <strong>in</strong>terventions as an adjuvant orchemopreventative option <strong>in</strong> the management and treatment of diabetes.References[1] American Diabetes Association. Standards of medical care <strong>in</strong> diabetes e 2011. Diabetes Care 2011;34(Suppl. 1):S11e61.[2] Encyclopedia ADAMM. Diabetes. Atlanta (GA): A.D.A.M., Inc; 2011.[3] WHO. Diabetes Fact sheet N 312. World Health Organization; 2011.[4] CDC. National diabetes fact sheet: national estimates and general <strong>in</strong>formation on diabetes and prediabetes <strong>in</strong>the United States, 2011. In: Prevention CfDCa, editor. Department of Health and <strong>Human</strong> Services, Centers for<strong>Disease</strong> Control and Prevention. Atlanta, GA: U.S; 2011.[5] IDF. Diabetes Atlas. International Diabetes Federation; 2009.[6] Denis H, Ndlovu MN, Fuks F. Regulation of mammalian DNA methyltransferases: a route to new mechanisms.EMBO Rep 2011;12:647e56.[7] Chen ZX, Riggs AD. DNA methylation and demethylation <strong>in</strong> mammals. J Biol Chem 2011;286:18347e53.[8] Jenuwe<strong>in</strong> T, Allis CD. Translat<strong>in</strong>g the histone code. <strong>Science</strong> 2001;293:1074e80.[9] Strahl BD, Allis CD. The language of covalent histone modifications. Nature 2000;403:41e5.[10] Turner BM. Def<strong>in</strong><strong>in</strong>g an epigenetic code. Nat Cell Biol 2007;9:2e6.[11] Lee JS, Smith E, Shilatifard A. The language of histone crosstalk. Cell 2010;142:682e5.[12] Allis CD, Berger SL, Cote J, Dent S, Jenuwien T, Kouzarides T, et al. New nomenclature for chromat<strong>in</strong>modify<strong>in</strong>genzymes. Cell 2007;131:633e6.[13] Gray SG, Ekström TJ. The <strong>Human</strong> Histone Deacetylase Family. Exp Cell Res 2001;262:75e83.[14] Albert M, Hel<strong>in</strong> K. Histone methyltransferases <strong>in</strong> cancer. Sem<strong>in</strong> Cell Dev Biol 2010;21:209e20.[15] Sims 3rd RJ, Re<strong>in</strong>berg D. Is there a code embedded <strong>in</strong> prote<strong>in</strong>s that is based on post-translational modifications?Nat Rev Mol Cell Biol 2008;9:815e20.[16] Calao M, Burny A, Quivy V, Dekon<strong>in</strong>ck A, Van L<strong>in</strong>t C. A pervasive role of histone acetyltransferases anddeacetylases <strong>in</strong> an NF-kappaB-signal<strong>in</strong>g code. Trends Biochem Sci 2008;33:339e49.[17] Mankan AK, Lawless MW, Gray SG, Kelleher D, McManus R. NF-kappaB regulation: the nuclear response.J Cell Mol Med 2009;13:631e43.[18] Choudhary C, Kumar C, Gnad F, Nielsen ML, Rehman M, Walther TC, et al. Lys<strong>in</strong>e acetylation targets prote<strong>in</strong>complexes and co-regulates major cellular functions. <strong>Science</strong> 2009;325:834e40.[19] Cyr AR, Domann FE. The redox basis of epigenetic modifications: from mechanisms to functional consequences.Antioxid Redox Signal 2011;15:551e89.353


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>354[20] Kolfschoten IG, Roggli E, Nesca V, Regazzi R. Role and therapeutic potential of microRNAs <strong>in</strong> diabetes.Diabetes Obes Metab 2009;11(Suppl. 4):118e29.[21] Xie H, Lim B, Lodish HF. MicroRNAs <strong>in</strong>duced dur<strong>in</strong>g adipogenesis that accelerate fat cell development aredownregulated <strong>in</strong> obesity. Diabetes 2009;58:1050e7.[22] Fabbri M, Cal<strong>in</strong> GA. <strong>Epigenetics</strong> and miRNAs <strong>in</strong> human cancer. Adv Genet 2010;70:87e99.[23] Gray SG, De Meyts P. Role of histone and transcription factor acetylation <strong>in</strong> diabetes pathogenesis. DiabetesMetab Res Rev 2005;21:416e33.[24] Temple IK, Shield JP. Transient neonatal diabetes, a disorder of impr<strong>in</strong>t<strong>in</strong>g. J Med Genet 2002;39:872e5.[25] Mackay DJ, Temple IK. Transient neonatal diabetes mellitus type 1. Am J Med Genet C Sem<strong>in</strong> Med Genet2010;154C:335e42.[26] Li Y, Zhao M, Hou C, Liang G, Yang L, Tan Y, et al. Abnormal DNA methylation <strong>in</strong> CD4(þ) T cells frompatients with latent autoimmune diabetes <strong>in</strong> adults. Diabetes Res Cl<strong>in</strong> Pract 2011.[27] Pirola L, Balcerczyk A, Tothill RW, Haviv I, Kaspi A, Lunke S, et al. Genome-wide analysis dist<strong>in</strong>guisheshyperglycemia regulated epigenetic signatures of primary vascular cells. Genome Res 2011;21:1601e15.[28] Rakyan VK, Beyan H, Down TA, Hawa MI, Maslau S, Aden D, et al. Identification of type 1 diabetes-associatedDNA methylation variable positions that precede disease diagnosis. PLoS Genet 2011;7:e1002300.[29] Toperoff G, Aran D, Kark JD, Rosenberg M, Dubnikov T, Nissan B, et al. Genome-wide survey reveals predispos<strong>in</strong>gdiabetes type 2-related DNA methylation variations <strong>in</strong> human peripheral blood. Hum Mol Genet 2011.[30] Jen<strong>in</strong>ga EH, Schoonjans K, Auwerx J. Reversible acetylation of PGC-1: connect<strong>in</strong>g energy sensors and effectorsto guarantee metabolic flexibility. Oncogene 2010;29:4617e24.[31] Mootha VK, L<strong>in</strong>dgren CM, Eriksson KF, Subramanian A, Sihag S, Lehar J, et al. PGC-1alpha-responsive genes<strong>in</strong>volved <strong>in</strong> oxidative phosphorylation are coord<strong>in</strong>ately downregulated <strong>in</strong> human diabetes. Nat Genet2003;34:267e73.[32] L<strong>in</strong>g C, Del Guerra S, Lupi R, Ronn T, Granhall C, Luthman H, et al. Epigenetic regulation of PPARGC1A <strong>in</strong>human type 2 diabetic islets and effect on <strong>in</strong>sul<strong>in</strong> secretion. Diabetologia 2008;51:615e22.[33] Barres R, Osler ME, Yan J, Rune A, Fritz T, Caidahl K, et al. Non-CpG methylation of the PGC-1alpha promoterthrough DNMT3B controls mitochondrial density. Cell Metab 2009;10:189e98.[34] Cox NJ, Wapelhorst B, Morrison VA, Johnson L, P<strong>in</strong>chuk L, Spielman RS, et al. Seven regions of the genomeshow evidence of l<strong>in</strong>kage to type 1 diabetes <strong>in</strong> a consensus analysis of 767 multiplex families. Am J Hum Genet2001;69:820e30.[35] Nerup J, Pociot F. A genomewide scan for type 1-diabetes susceptibility <strong>in</strong> Scand<strong>in</strong>avian families: identificationof new loci with evidence of <strong>in</strong>teractions. Am J Hum Genet 2001;69:1301e13.[36] Xiang K, Wang Y, Zheng T, Jia W, Li J, Chen L, et al. Genome-wide search for type 2 diabetes/impaired glucosehomeostasis susceptibility genes <strong>in</strong> the Ch<strong>in</strong>ese: significant l<strong>in</strong>kage to chromosome 6q21-q23 and chromosome1q21-q24. Diabetes 2004;53:228e34.[37] Concannon P, Erlich HA, Julier C, Morahan G, Nerup J, Pociot F, et al. Type 1 diabetes: evidence forsusceptibility loci from four genome-wide l<strong>in</strong>kage scans <strong>in</strong> 1,435 multiplex families. Diabetes2005;54:2995e3001.[38] Noh H, Oh EY, Seo JY, Yu MR, Kim YO, Ha H, et al. Histone deacetylase-2 is a key regulator of diabetes- andtransform<strong>in</strong>g growth factor-beta1-<strong>in</strong>duced renal <strong>in</strong>jury. Am J Physiol Renal Physiol 2009;297:F729e739.[39] Yamauchi T, Oike Y, Kamon J, Waki H, Komeda K, Tsuchida A, et al. Increased <strong>in</strong>sul<strong>in</strong> sensitivity despitelipodystrophy <strong>in</strong> Crebbp heterozygous mice. Nat Genet 2002;30:221e6.[40] Coste A, Louet JF, Lagouge M, Ler<strong>in</strong> C, Antal MC, Meziane H, et al. The genetic ablation of SRC-3 protectsaga<strong>in</strong>st obesity and improves <strong>in</strong>sul<strong>in</strong> sensitivity by reduc<strong>in</strong>g the acetylation of PGC-1{alpha}. Proc Natl AcadSci USA 2008;105:17187e92.[41] York B, Yu C, Sagen JV, Liu Z, Nikolai BC, Wu RC, et al. Reprogramm<strong>in</strong>g the posttranslational code of SRC-3confers a switch <strong>in</strong> mammalian systems biology. Proc Natl Acad Sci USA 2010;107:11122e7.[42] Witt O, Deubzer HE, Milde T, Oehme I. HDAC family: What are the cancer relevant targets? Cancer Lett2009;277:8e21.[43] Guarente L. Frankl<strong>in</strong> H. Epste<strong>in</strong> Lecture: Sirtu<strong>in</strong>s, ag<strong>in</strong>g, and medic<strong>in</strong>e. N Engl J Med 2011;364:2235e44.[44] Wu D, Qiu Y, Gao X, Yuan XB, Zhai Q. Overexpression of SIRT1 <strong>in</strong> Mouse Forebra<strong>in</strong> Impairs Lipid/GlucoseMetabolism and Motor Function. PLoS One 2011;6:e21759.[45] Li Y, Xu S, Giles A, Nakamura K, Lee JW, Hou X, et al. Hepatic overexpression of SIRT1 <strong>in</strong> mice attenuatesendoplasmic reticulum stress and <strong>in</strong>sul<strong>in</strong> resistance <strong>in</strong> the liver. Faseb J 2011;25:1664e79.[46] Kendrick AA, Choudhury M, Rahman SM, McCurdy CE, Friederich M, Hove Van, et al. Fatty liver is associatedwith reduced SIRT3 activity and mitochondrial prote<strong>in</strong> hyperacetylation. Biochem J 2011;433:505e14.[47] Schwer B, Schumacher B, Lombard DB, Xiao C, Kurtev MV, Gao J, et al. Neural sirtu<strong>in</strong> 6 (Sirt6) ablationattenuates somatic growth and causes obesity. Proc Natl Acad Sci USA 2010;107:21790e4.


CHAPTER 17The Potential of Epigenetic Compounds <strong>in</strong> Treat<strong>in</strong>g Diabetes[48] Xiao C, Kim HS, Lahusen T, Wang RH, Xu X, Gavrilova O, et al. SIRT6 deficiency results <strong>in</strong> severe hypoglycemiaby enhanc<strong>in</strong>g both basal and <strong>in</strong>sul<strong>in</strong>-stimulated glucose uptake <strong>in</strong> mice. J Biol Chem 2010;285:36776e84.[49] Brasacchio D, Okabe J, Tikellis C, Balcerczyk A, George P, Baker EK, et al. Hyperglycemia <strong>in</strong>duces a dynamiccooperativity of histone methylase and demethylase enzymes associated with gene-activat<strong>in</strong>g epigenetic marksthat coexist on the lys<strong>in</strong>e tail. Diabetes 2009;58:1229e36.[50] Reddy MA, Villeneuve LM, Wang M, Lant<strong>in</strong>g L, Natarajan R. Role of the lys<strong>in</strong>e-specific demethylase 1 <strong>in</strong> thepro<strong>in</strong>flammatory phenotype of vascular smooth muscle cells of diabetic mice. Circ Res 2008;103:615e23.[51] Li Y, Reddy MA, Miao F, Shanmugam N, Yee JK, Hawk<strong>in</strong>s D, et al. Role of the histone H3 lys<strong>in</strong>e 4 methyltransferase,SET7/9, <strong>in</strong> the regulation of NF-kappaB-dependent <strong>in</strong>flammatory genes. Relevance to diabetes and<strong>in</strong>flammation. J Biol Chem 2008;283:26771e81.[52] Deer<strong>in</strong>g TG, Ogihara T, Trace AP, Maier B, Mirmira RG. Methyltransferase Set7/9 ma<strong>in</strong>ta<strong>in</strong>s transcription andeuchromat<strong>in</strong> structure at islet-enriched genes. Diabetes 2009;58:185e93.[53] Ogihara T, Vanderford NL, Maier B, Ste<strong>in</strong> RW, Mirmira RG. Expression and function of Set7/9 <strong>in</strong> pancreaticislets. Islets 2009;1:269e72.[54] Lo KA, Bauchmann MK, Baumann AP, Donahue CJ, Thiede MA, Hayes LS, et al. Genome-Wide Profil<strong>in</strong>g ofH3K56 Acetylation and Transcription Factor B<strong>in</strong>d<strong>in</strong>g Sites <strong>in</strong> <strong>Human</strong> Adipocytes. PLoS One 2011;6:e19778.[55] Vempati RK, Jayani RS, Notani D, Sengupta A, Galande S, Haldar D. p300-mediated acetylation of histone H3lys<strong>in</strong>e 56 functions <strong>in</strong> DNA damage response <strong>in</strong> mammals. J Biol Chem 2010;285:28553e64.[56] Evans-Mol<strong>in</strong>a C, Garmey JC, Ketchum R, Brayman KL, Deng S, Mirmira RG. Glucose regulation of <strong>in</strong>sul<strong>in</strong> genetranscription and pre-mRNA process<strong>in</strong>g <strong>in</strong> human islets. Diabetes 2007;56:827e35.[57] Mosley AL, Corbett JA, Ozcan S. Glucose regulation of <strong>in</strong>sul<strong>in</strong> gene expression requires the recruitment ofp300 by the beta-cell-specific transcription factor Pdx-1. Mol Endocr<strong>in</strong>ol 2004;18:2279e90.[58] Mosley AL, Ozcan S. Glucose regulates <strong>in</strong>sul<strong>in</strong> gene transcription by hyperacetylation of histone h4. J BiolChem 2003;278:19660e6.[59] Qiu Y, Guo M, Huang S, Ste<strong>in</strong> R. Insul<strong>in</strong> gene transcription is mediated by <strong>in</strong>teractions between the p300coactivator and PDX-1, BETA2, and E47. Mol Cell Biol 2002;22:412e20.[60] Qiu Y, Sharma A, Ste<strong>in</strong> R. p300 mediates transcriptional stimulation by the basic helix-loop-helix activators ofthe <strong>in</strong>sul<strong>in</strong> gene. Mol Cell Biol 1998;18:2957e64.[61] Mosley AL, Ozcan S. The pancreatic duodenal homeobox-1 prote<strong>in</strong> (Pdx-1) <strong>in</strong>teracts with histone deacetylasesHdac-1 and Hdac-2 on low levels of glucose. J Biol Chem 2004;279:54241e7.[62] Qiu Y, Guo M, Huang S, Ste<strong>in</strong> R. Acetylation of the BETA2 Transcription Factor by p300-associated Factor IsImportant <strong>in</strong> Insul<strong>in</strong> Gene Expression. J Biol Chem 2004;279:9796e802.[63] Zhang M, Poplawski M, Yen K, Cheng H, Bloss E, Zhu X, et al. Role of CBP and SATB-1 <strong>in</strong> ag<strong>in</strong>g, dietaryrestriction, and <strong>in</strong>sul<strong>in</strong>-like signal<strong>in</strong>g. PLoS Biol 2009;7:e1000245.[64] Fernandez-Zapico ME, van Velk<strong>in</strong>burgh JC, Gutierrez-Aguilar R, Neve B, Froguel P, Urrutia R, et al. MODY7gene, KLF11, is a novel p300-dependent regulator of Pdx-1 (MODY4) transcription <strong>in</strong> pancreatic islet betacells. J Biol Chem 2009;284:36482e90.[65] Bonnefond A, Lomberk G, Buttar N, Busiah K, Vaillant E, Lobbens S, et al. Disruption of a novel KLF-p300-regulated pathway for <strong>in</strong>sul<strong>in</strong> biosynthesis revealed by studies of the c.-331 INS mutation found <strong>in</strong> neonataldiabetes mellitus. J Biol Chem 2011.[66] Mutskov V, Felsenfeld G. The human <strong>in</strong>sul<strong>in</strong> gene is part of a large open chromat<strong>in</strong> doma<strong>in</strong> specific for humanislets. Proc Natl Acad Sci USA 2009;106:17419e24.[67] Ohara-Imaizumi M, Cardozo AK, Kikuta T, Eizirik DL, Nagamatsu S. The cytok<strong>in</strong>e <strong>in</strong>terleuk<strong>in</strong>-1beta reducesthe dock<strong>in</strong>g and fusion of <strong>in</strong>sul<strong>in</strong> granules <strong>in</strong> pancreatic beta-cells, preferentially decreas<strong>in</strong>g the first phase ofexocytosis. J Biol Chem 2004;279:41271e4.[68] Lawrence MC, Shao C, McGlynn K, Nazirudd<strong>in</strong> B, Levy MF, Cobb MH. Multiple chromat<strong>in</strong>-bound prote<strong>in</strong>k<strong>in</strong>ases assemble factors that regulate <strong>in</strong>sul<strong>in</strong> gene transcription. Proc Natl Acad Sci USA2009;106:22181e6.[69] Leoni F, Zaliani A, Bertol<strong>in</strong>i G, Porro G, Pagani P, Pozzi P, et al. The antitumor histone deacetylase <strong>in</strong>hibitorsuberoylanilide hydroxamic acid exhibits anti<strong>in</strong>flammatory properties via suppression of cytok<strong>in</strong>es. Proc NatlAcad Sci USA 2002;99:2995e3000.[70] Lewis EC, Blaabjerg L, Storl<strong>in</strong>g J, Ronn SG, Mascagni P, D<strong>in</strong>arello CA, et al. The Oral Histone DeacetylaseInhibitor ITF2357 Reduces Cytok<strong>in</strong>es and Protects Islet beta Cells In Vivo and In Vitro. Mol Med2011;17:369e77.[71] Susick L, Senanayake T, Veluthakal R, Woster PM, Kowluru A. A novel histone deacetylase <strong>in</strong>hibitor prevents IL-1beta <strong>in</strong>duced metabolic dysfunction <strong>in</strong> pancreatic beta-cells. J Cell Mol Med 2009;13:1877e85.[72] Brochu-Gaudreau K, Rehfeldt C, Blou<strong>in</strong> R, Bordignon V, Murphy BD, Pal<strong>in</strong> MF. Adiponect<strong>in</strong> action from headto toe. Endocr<strong>in</strong>e 2010;37:11e32.355


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>356[73] Musri MM, Corom<strong>in</strong>ola H, Casamitjana R, Gomis R, Parrizas M. Histone H3 lys<strong>in</strong>e 4 dimethylation signals thetranscriptional competence of the adiponect<strong>in</strong> promoter <strong>in</strong> preadipocytes. J Biol Chem 2006;281:17180e8.[74] Sakurai N, Mochizuki K, Goda T. Modifications of histone H3 at lys<strong>in</strong>e 9 on the adiponect<strong>in</strong> gene <strong>in</strong> 3T3-L1adipocytes. J Nutr Sci Vitam<strong>in</strong>ol (Tokyo) 2009;55:131e8.[75] Qiao L, Schaack J, Shao J. Suppression of adiponect<strong>in</strong> gene expression by histone deacetylase <strong>in</strong>hibitorvalproic acid. Endocr<strong>in</strong>ology 2006;147:865e74.[76] Sugii S, Evans RM. Epigenetic codes of PPARgamma <strong>in</strong> metabolic disease. FEBS Lett 2011;585:2121e8.[77] Kim SJ, Nian C, McIntosh CH. Adipocyte expression of the glucose-dependent <strong>in</strong>sul<strong>in</strong>otropic polypeptidereceptor <strong>in</strong>volves gene regulation by PPARgamma and histone acetylation. J Lipid Res 2011;52:759e70.[78] Handsch<strong>in</strong> C, Spiegelman BM. Peroxisome proliferator-activated receptor gamma coactivator 1 coactivators,energy homeostasis, and metabolism. Endocr Rev 2006;27:728e35.[79] Patti ME, Butte AJ, Crunkhorn S, Cusi K, Berria R, Kashyap S, et al. Coord<strong>in</strong>ated reduction of genes of oxidativemetabolism <strong>in</strong> humans with <strong>in</strong>sul<strong>in</strong> resistance and diabetes: Potential role of PGC1 and NRF1. Proc Natl AcadSci USA 2003;100:8466e71.[80] Crunkhorn S, Dearie F, Mantzoros C, Gami H, da Silva WS, Esp<strong>in</strong>oza D, et al. Peroxisome proliferator activatorreceptor gamma coactivator-1 expression is reduced <strong>in</strong> obesity: potential pathogenic role of saturated fattyacids and p38 mitogen-activated prote<strong>in</strong> k<strong>in</strong>ase activation. J Biol Chem 2007;282:15439e50.[81] Karpac J, Jasper H. Metabolic homeostasis: HDACs take center stage. Cell 2011;145:497e9.[82] Calnan DR, Brunet A. The FoxO code. Oncogene 2008;27:2276e88.[83] Haigis MC, S<strong>in</strong>clair DA. Mammalian sirtu<strong>in</strong>s: biological <strong>in</strong>sights and disease relevance. Annu Rev Pathol2010;5:253e95.[84] Mihaylova MM, Vasquez DS, Ravnskjaer K, Denechaud PD, Yu RT, Alvarez JG, et al. Class IIa histone deacetylasesare hormone-activated regulators of FOXO and mammalian glucose homeostasis. Cell2011;145:607e21.[85] Wang B, Moya N, Niessen S, Hoover H, Mihaylova MM, Shaw RJ, et al. A hormone-dependent moduleregulat<strong>in</strong>g energy balance. Cell 2011;145:596e606.[86] Birnbaum MJ. Identification of a novel gene encod<strong>in</strong>g an <strong>in</strong>sul<strong>in</strong>-responsive glucose transporter prote<strong>in</strong>. Cell1989;57:305e15.[87] Garvey WT, Maianu L, Zhu JH, Brechtel-Hook G, Wallace P, Baron AD. Evidence for defects <strong>in</strong> the traffick<strong>in</strong>gand translocation of GLUT4 glucose transporters <strong>in</strong> skeletal muscle as a cause of human <strong>in</strong>sul<strong>in</strong> resistance.J Cl<strong>in</strong> Invest 1998;101:2377e86.[88] Czubryt MP, McAnally J, Fishman GI, Olson EN. Regulation of peroxisome proliferator-activated receptorgamma coactivator 1 alpha (PGC-1 alpha ) and mitochondrial function by MEF2 and HDAC5. Proc Natl AcadSci USA 2003;100:1711e6.[89] Raychaudhuri N, Raychaudhuri S, Thamotharan M, Devaskar SU. Histone code modifications repressglucose transporter 4 expression <strong>in</strong> the <strong>in</strong>trauter<strong>in</strong>e growth-restricted offspr<strong>in</strong>g. J Biol Chem2008;283:13611e26.[90] Sparl<strong>in</strong>g DP, Griesel BA, Weems J, Olson AL. GLUT4 enhancer factor (GEF) <strong>in</strong>teracts with MEF2A and HDAC5to regulate the GLUT4 promoter <strong>in</strong> adipocytes. J Biol Chem 2008;283:7429e37.[91] McGee SL, van Denderen BJ, Howlett KF, Mollica J, Schertzer JD, Kemp BE, et al. AMP-activated prote<strong>in</strong> k<strong>in</strong>aseregulates GLUT4 transcription by phosphorylat<strong>in</strong>g histone deacetylase 5. Diabetes 2008;57:860e7.[92] Weems J, Olson AL. Class II histone deacetylases limit GLUT4 gene expression dur<strong>in</strong>g adipocyte differentiation.J Biol Chem 2011;286:460e8.[93] O’Byrne KJ, Baird A-M, Kilmart<strong>in</strong> L, Leonard J, Sacevich C, Gray SG. Epigenetic Regulation of GlucoseTransporters <strong>in</strong> Non-Small Cell Lung Cancer. Cancers 2011;3:1550e65.[94] Phillips JE, Corces VG. CTCF: master weaver of the genome. Cell 2009;137:1194e211.[95] Takigawa-Imamura H, Sek<strong>in</strong>e T, Murata M, Takayama K, Nakazawa K, Nakagawa J. Stimulation of glucoseuptake <strong>in</strong> muscle cells by prolonged treatment with scriptide, a histone deacetylase <strong>in</strong>hibitor. Biosci BiotechnolBiochem 2003;67:1499e506.[96] Yamakuchi M, Ferlito M, Lowenste<strong>in</strong> CJ. miR-34a repression of SIRT1 regulates apoptosis. Proc Natl Acad SciUSA 2008;105:13421e6.[97] Lovis P, Roggli E, Laybutt DR, Gattesco S, Yang JY, Widmann C, et al. Alterations <strong>in</strong> microRNA expressioncontribute to fatty acid-<strong>in</strong>duced pancreatic beta-cell dysfunction. Diabetes 2008;57:2728e36.[98] Lee J, Padhye A, Sharma A, Song G, Miao J, Mo YY, et al. A pathway <strong>in</strong>volv<strong>in</strong>g farnesoid X receptor and smallheterodimer partner positively regulates hepatic sirtu<strong>in</strong> 1 levels via microRNA-34a <strong>in</strong>hibition. J Biol Chem2010;285:12604e11.[99] Mengh<strong>in</strong>i R, Casagrande V, Cardell<strong>in</strong>i M, Martelli E, Terr<strong>in</strong>oni A, Amati F, et al. MicroRNA 217 modulatesendothelial cell senescence via silent <strong>in</strong>formation regulator 1. Circulation 2009;120:1524e32.


CHAPTER 17The Potential of Epigenetic Compounds <strong>in</strong> Treat<strong>in</strong>g Diabetes[100] Szafranska AE, Davison TS, John J, Cannon T, Sipos B, Maghnouj A, et al. MicroRNA expression alterationsare l<strong>in</strong>ked to tumorigenesis and non-neoplastic processes <strong>in</strong> pancreatic ductal adenocarc<strong>in</strong>oma. Oncogene2007;26:4442e52.[101] Strum JC, Johnson JH, Ward J, Xie H, Feild J, Hester A, et al. MicroRNA 132 regulates nutritional stress<strong>in</strong>ducedchemok<strong>in</strong>e production through repression of SirT1. Mol Endocr<strong>in</strong>ol 2009;23:1876e84.[102] Esguerra JL, Bolmeson C, Cilio CM, Eliasson L. Differential glucose-regulation of microRNAs <strong>in</strong> pancreaticislets of non-obese type 2 diabetes model Goto-Kakizaki rat. PLoS One 2011;6:e18613.[103] Heneghan HM, Miller N, McAnena OJ, O’Brien T, Ker<strong>in</strong> MJ. Differential miRNA expression <strong>in</strong> omentaladipose tissue and <strong>in</strong> the circulation of obese patients identifies novel metabolic biomarkers. J Cl<strong>in</strong> Endocr<strong>in</strong>olMetab 2011;96:E846e850.[104] Park JK, Henry JC, Jiang J, Esau C, Gusev Y, Lerner MR, et al. miR-132 and miR-212 are <strong>in</strong>creased <strong>in</strong>pancreatic cancer and target the ret<strong>in</strong>oblastoma tumor suppressor. Biochem Biophys Res Commun2011;406:518e23.[105] Alvarez-Saavedra M, Antoun G, Yanagiya A, Oliva-Hernandez R, Cornejo-Palma D, Perez-Iratxeta C, et al.miRNA-132 orchestrates chromat<strong>in</strong> remodel<strong>in</strong>g and translational control of the circadian clock. Hum MolGenet 2011;20:731e51.[106] Ramachandran D, Roy U, Garg S, Ghosh S, Pathak S, Kolthur-Seetharam U. Sirt1 and mir-9 expression isregulated dur<strong>in</strong>g glucose-stimulated <strong>in</strong>sul<strong>in</strong> secretion <strong>in</strong> pancreatic beta-islets. Febs J 2011;278:1167e74.[107] Fabbri M, Garzon R, Cimm<strong>in</strong>o A, Liu Z, Zanesi N, Callegari E, et al. MicroRNA-29 family reverts aberrantmethylation <strong>in</strong> lung cancer by target<strong>in</strong>g DNA methyltransferases 3A and 3B. Proc Natl Acad Sci USA2007;104:15805e10.[108] Pass HI, Goparaju C, Ivanov S, Don<strong>in</strong>gton J, Carbone M, Hoshen M, et al. hsa-miR-29c* is l<strong>in</strong>ked to theprognosis of malignant pleural mesothelioma. Cancer Res 2010;70:1916e24.[109] He A, Zhu L, Gupta N, Chang Y, Fang F. Overexpression of micro ribonucleic acid 29, highly up-regulated <strong>in</strong>diabetic rats, leads to <strong>in</strong>sul<strong>in</strong> resistance <strong>in</strong> 3T3-L1 adipocytes. Mol Endocr<strong>in</strong>ol 2007;21:2785e94.[110] Long J, Wang Y, Wang W, Chang BH, Danesh FR. MicroRNA-29c is a signature microRNA under high glucoseconditions that targets Sprouty homolog 1, and its <strong>in</strong> vivo knockdown prevents progression of diabeticnephropathy. J Biol Chem 2011;286:11837e48.[111] Ng EK, Tsang WP, Ng SS, J<strong>in</strong> HC, Yu J, Li JJ, et al. MicroRNA-143 targets DNA methyltransferases 3A <strong>in</strong>colorectal cancer. Br J Cancer 2009;101:699e706.[112] Esau C, Kang X, Peralta E, Hanson E, Marcusson EG, Ravichandran LV, et al. MicroRNA-143 regulatesadipocyte differentiation. J Biol Chem 2004;279:52361e5.[113] Braconi C, Huang N, Patel T. MicroRNA-dependent regulation of DNA methyltransferase-1 and tumorsuppressor gene expression by <strong>in</strong>terleuk<strong>in</strong>-6 <strong>in</strong> human malignant cholangiocytes. Hepatology2010;51:881e90.[114] Wang KH, Kao AP, S<strong>in</strong>gh S, Yu SL, Kao LP, Tsai ZY, et al. Comparative expression profiles of mRNAs andmicroRNAs among human mesenchymal stem cells derived from breast, face, and abdom<strong>in</strong>al adipose tissues.Kaohsiung J Med Sci 2010;26:113e22.[115] Koturbash I, Boyko A, Rodriguez-Juarez R, McDonald RJ, Tryndyak VP, Kovalchuk I, et al. Role of epigeneticeffectors <strong>in</strong> ma<strong>in</strong>tenance of the long-term persistent bystander effect <strong>in</strong> spleen <strong>in</strong> vivo. Carc<strong>in</strong>ogenesis2007;28:1831e8.[116] Kle<strong>in</strong> ME, Lioy DT, Ma L, Impey S, Mandel G, Goodman RH. Homeostatic regulation of MeCP2 expression bya CREB-<strong>in</strong>duced microRNA. Nat Neurosci 2007;10:1513e4.[117] Noonan EJ, Place RF, Pookot D, Basak S, Whitson JM, Hirata H, et al. miR-449a targets HDAC-1 and <strong>in</strong>ducesgrowth arrest <strong>in</strong> prostate cancer. Oncogene 2009;28:1714e24.[118] Yang X, Feng M, Jiang X, Wu Z, Li Z, Aau M, et al. miR-449a and miR-449b are direct transcriptional targets ofE2F1 and negatively regulate pRb-E2F1 activity through a feedback loop by target<strong>in</strong>g CDK6 and CDC25A.Genes Dev 2009;23:2388e93.[119] Bou Kheir T, Futoma-Kazmierczak E, Jacobsen A, Krogh A, Bardram L, Hother C, et al. miR-449 <strong>in</strong>hibits cellproliferation and is down-regulated <strong>in</strong> gastric cancer. Mol Cancer 2011;10:29.[120] Chen JF, Mandel EM, Thomson JM, Wu Q, Callis TE, Hammond SM, et al. The role of microRNA-1 andmicroRNA-133 <strong>in</strong> skeletal muscle proliferation and differentiation. Nat Genet 2006;38:228e33.[121] Shan ZX, L<strong>in</strong> QX, Deng CY, Zhu JN, Mai LP, Liu JL, et al. miR-1/miR-206 regulate Hsp60 expressioncontribut<strong>in</strong>g to glucose-mediated apoptosis <strong>in</strong> cardiomyocytes. FEBS Lett 2010;584:3592e600.[122] Karbiener M, Fischer C, Nowitsch S, Opriessnig P, Papak C, Ailhaud G, et al. microRNA miR-27b impairshuman adipocyte differentiation and targets PPARgamma. Biochem Biophys Res Commun2009;390:247e51.[123] Kim SY, Kim AY, Lee HW, Son YH, Lee GY, Lee JW, et al. miR-27a is a negative regulator of adipocytedifferentiation via suppress<strong>in</strong>g PPARgamma expression. Biochem Biophys Res Commun 2010;392:323e8.357


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>358[124] Mart<strong>in</strong>elli R, Nardelli C, Pilone V, Buonomo T, Liguori R, Castano I, et al. miR-519d Overexpression IsAssociated With <strong>Human</strong> Obesity. Silver Spr<strong>in</strong>g: Obesity; 2010.[125] Varambally S, Cao Q, Mani RS, Shankar S, Wang X, Ateeq B, et al. Genomic loss of microRNA-101 leads tooverexpression of histone methyltransferase EZH2 <strong>in</strong> cancer. <strong>Science</strong> 2008;322:1695e9.[126] Chen H, Gu X, Su IH, Bott<strong>in</strong>o R, Contreras JL, Tarakhovsky A, et al. Polycomb prote<strong>in</strong> Ezh2 regulatespancreatic beta-cell Ink4a/Arf expression and regeneration <strong>in</strong> diabetes mellitus. Genes Dev 2009;23:975e85.[127] Wang L, J<strong>in</strong> Q, Lee JE, Su IH, Ge K. Histone H3K27 methyltransferase Ezh2 represses Wnt genes to facilitateadipogenesis. Proc Natl Acad Sci USA 2010;107:7317e22.[128] Lu H, Buchan RJ, Cook SA. MicroRNA-223 regulates Glut4 expression and cardiomyocyte glucose metabolism.Cardiovasc Res 2010;86:410e20.[129] Horie T, Ono K, Nishi H, Iwanaga Y, Nagao K, K<strong>in</strong>oshita M, et al. MicroRNA-133 regulates the expression ofGLUT4 by target<strong>in</strong>g KLF15 and is <strong>in</strong>volved <strong>in</strong> metabolic control <strong>in</strong> cardiac myocytes. Biochem Biophys ResCommun 2009;389:315e20.[130] Aoi W, Naito Y, Mizushima K, Takanami Y, Kawai Y, Ichikawa H, et al. The microRNA miR-696 regulates PGC-1{alpha} <strong>in</strong> mouse skeletal muscle <strong>in</strong> response to physical activity. Am J Physiol Endocr<strong>in</strong>ol Metab2010;298:E799e806.[131] Safdar A, Abadi A, Akhtar M, Hett<strong>in</strong>ga BP, Tarnopolsky MA. miRNA <strong>in</strong> the regulation of skeletal muscleadaptation to acute endurance exercise <strong>in</strong> C57Bl/6J male mice. PLoS One 2009;4:e5610.[132] Keller DM, McWeeney S, Arsenlis A, Drou<strong>in</strong> J, Wright CV, Wang H, et al. Characterization of pancreatictranscription factor Pdx-1 b<strong>in</strong>d<strong>in</strong>g sites us<strong>in</strong>g promoter microarray and serial analysis of chromat<strong>in</strong> occupancy.J Biol Chem 2007;282:32084e92.[133] El Ouaamari A, Baroukh N, Martens GA, Lebrun P, Pipeleers D, van Obberghen E. miR-375 targets 3’-phospho<strong>in</strong>ositide-dependent prote<strong>in</strong> k<strong>in</strong>ase-1 and regulates glucose-<strong>in</strong>duced biological responses <strong>in</strong>pancreatic beta-cells. Diabetes 2008;57:2708e17.[134] Trajkovski M, Hausser J, Soutschek J, Bhat B, Ak<strong>in</strong> A, Zavolan M, et al. MicroRNAs 103 and 107 regulate<strong>in</strong>sul<strong>in</strong> sensitivity. Nature 2011;474:649e53.[135] Tang X, Muniappan L, Tang G, Ozcan S. Identification of glucose-regulated miRNAs from pancreatic {beta}cells reveals a role for miR-30d <strong>in</strong> <strong>in</strong>sul<strong>in</strong> transcription. Rna 2009;15:287e93.[136] Kloosterman WP, Lagendijk AK, Kett<strong>in</strong>g RF, Moulton JD, Plasterk RH. Targeted <strong>in</strong>hibition of miRNA maturationwith morphol<strong>in</strong>os reveals a role for miR-375 <strong>in</strong> pancreatic islet development. PLoS Biol 2007;5:e203.[137] Joglekar MV, Joglekar VM, Hardikar AA. Expression of islet-specific microRNAs dur<strong>in</strong>g human pancreaticdevelopment. Gene Expr Patterns 2009;9:109e13.[138] Granjon A, Gust<strong>in</strong> MP, Rieusset J, Lefai E, Meugnier E, Guller I, et al. The microRNA signature <strong>in</strong> response to<strong>in</strong>sul<strong>in</strong> reveals its implication <strong>in</strong> the transcriptional action of <strong>in</strong>sul<strong>in</strong> <strong>in</strong> human skeletal muscle and the roleof a sterol regulatory element-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>-1c/myocyte enhancer factor 2C pathway. Diabetes2009;58:2555e64.[139] Gallagher IJ, Scheele C, Keller P, Nielsen AR, Remenyi J, Fischer CP, et al. Integration of microRNA changes <strong>in</strong> vivoidentifies novel molecular features of muscle <strong>in</strong>sul<strong>in</strong> resistance <strong>in</strong> type 2 diabetes. Genome Med 2010;2:9.[140] Zhao E, Keller MP, Rabaglia ME, Oler AT, Stapleton DS, Schueler KL, et al. Obesity and genetics regulatemicroRNAs <strong>in</strong> islets, liver, and adipose of diabetic mice. Mamm Genome 2009;20:476e85.[141] Ortega FJ, Moreno-Navarrete JM, Pardo G, Sabater M, Hummel M, Ferrer A, et al. MiRNA expression profile ofhuman subcutaneous adipose and dur<strong>in</strong>g adipocyte differentiation. PLoS One 2010;5:e9022.[142] Jordan SD, Kruger M, Willmes DM, Redemann N, Wunderlich FT, Bronneke HS, et al. Obesity-<strong>in</strong>ducedoverexpression of miRNA-143 <strong>in</strong>hibits <strong>in</strong>sul<strong>in</strong>-stimulated AKT activation and impairs glucose metabolism.Nat Cell Biol 2011;13:434e46.[143] Iorio MV, Piovan C, Croce CM. Interplay between microRNAs and the epigenetic mach<strong>in</strong>ery: An <strong>in</strong>tricatenetwork. Biochim Biophys Acta 2010.[144] Bolleyn J, Fraczek J, V<strong>in</strong>ken M, Lizarraga D, Gaj S, van Delft JH, et al. Effect of Trichostat<strong>in</strong> A on miRNAexpression <strong>in</strong> cultures of primary rat hepatocytes. Toxicol In Vitro 2011.[145] de Kreutzenberg SV, Ceolotto G, Papparella I, Bortoluzzi A, Semplic<strong>in</strong>i A, Dalla Man C, et al. Downregulationof the longevity-associated prote<strong>in</strong> sirtu<strong>in</strong> 1 <strong>in</strong> <strong>in</strong>sul<strong>in</strong> resistance and metabolic syndrome: potentialbiochemical mechanisms. Diabetes 2010;59:1006e15.[146] Arif M, Senapati P, Shandilya J, Kundu TK. Prote<strong>in</strong> lys<strong>in</strong>e acetylation <strong>in</strong> cellular function and its role <strong>in</strong> cancermanifestation. Biochim Biophys Acta 2010;1799:702e16.[147] Lee J, Saha PK, Yang QH, Lee S, Park JY, Suh Y, et al. Targeted <strong>in</strong>activation of MLL3 histone H3-Lys-4methyltransferase activity <strong>in</strong> the mouse reveals vital roles for MLL3 <strong>in</strong> adipogenesis. Proc Natl Acad Sci USA2008;105:19229e34.[148] Donath MY, Shoelson SE. Type 2 diabetes as an <strong>in</strong>flammatory disease. Nat Rev Immunol 2011;11:98e107.


CHAPTER 17The Potential of Epigenetic Compounds <strong>in</strong> Treat<strong>in</strong>g Diabetes[149] Gerritsen ME, Williams AJ, Neish AS, Moore S, Shi Y, Coll<strong>in</strong>s T. CREB-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>/p300 are transcriptionalcoactivators of p65. Proc Natl Acad Sci USA 1997;94:2927e32.[150] Perk<strong>in</strong>s ND, Felzien LK, Betts JC, Leung K, Beach DH, Nabel GJ. Regulation of NF-kappaB by cycl<strong>in</strong>dependentk<strong>in</strong>ases associated with the p300 coactivator. <strong>Science</strong> 1997;275:523e7.[151] Wadgaonkar R, Phelps KM, Haque Z, Williams AJ, Silverman ES, Coll<strong>in</strong>s T. CREB-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> is a nuclear<strong>in</strong>tegrator of nuclear factor-kappaB and p53 signal<strong>in</strong>g. J Biol Chem 1999;274:1879e82.[152] Na SY, Lee SK, Han SJ, Choi HS, Im SY, Lee JW. Steroid receptor coactivator-1 <strong>in</strong>teracts with the p50 subunitand coactivates nuclear factor kappaB-mediated transactivations. J Biol Chem 1998;273:10831e4.[153] Yeung F, Hoberg JE, Ramsey CS, Keller MD, Jones DR, Frye RA, et al. Modulation of NF-kappaB-dependenttranscription and cell survival by the SIRT1 deacetylase. Embo J 2004;23:2369e80.[154] Hoberg JE, Yeung F, Mayo MW. SMRT derepression by the IkappaB k<strong>in</strong>ase alpha: a prerequisite to NF-kappaBtranscription and survival. Mol Cell 2004;16:245e55.[155] Fabre C, Grosjean J, Tailler M, Boehrer S, Ades L, Perfett<strong>in</strong>i JL, et al. A novel effect of DNA methyltransferaseand histone deacetylase <strong>in</strong>hibitors: NFkappaB <strong>in</strong>hibition <strong>in</strong> malignant myeloblasts. Cell Cycle2008;7:2139e45.[156] Larsen L, Tonnesen M, Ronn SG, Storl<strong>in</strong>g J, Jorgensen S, Mascagni P, et al. Inhibition of histone deacetylasesprevents cytok<strong>in</strong>e-<strong>in</strong>duced toxicity <strong>in</strong> beta cells. Diabetologia 2007;50:779e89.[157] Dandona P, Aljada A, Bandyopadhyay A. Inflammation: the l<strong>in</strong>k between <strong>in</strong>sul<strong>in</strong> resistance, obesity anddiabetes. Trends Immunol 2004;25:4e7.[158] Esposito K, Nappo F, Giugliano F, Di Palo C, Ciotola M, Barbieri M, et al. Cytok<strong>in</strong>e milieu tends toward<strong>in</strong>flammation <strong>in</strong> type 2 diabetes. Diabetes Care 2003;26:1647.[159] Vanden Berghe W, De Bosscher K, Boone E, Plaisance S, Haegeman G. The nuclear factor-kappaB engagesCBP/p300 and histone acetyltransferase activity for transcriptional activation of the <strong>in</strong>terleuk<strong>in</strong>-6 genepromoter. J Biol Chem 1999;274:32091e8.[160] Gao Z, Hwang D, Bataille F, Lefevre M, York D, Quon MJ, et al. Ser<strong>in</strong>e phosphorylation of <strong>in</strong>sul<strong>in</strong> receptorsubstrate 1 by <strong>in</strong>hibitor kappa B k<strong>in</strong>ase complex. J Biol Chem 2002;277:48115e21.[161] Arkan MC, Hevener AL, Greten FR, Maeda S, Li ZW, Long JM, et al. IKK-beta l<strong>in</strong>ks <strong>in</strong>flammation to obesity<strong>in</strong>duced<strong>in</strong>sul<strong>in</strong> resistance. Nat Med 2005;11:191e8.[162] Cai D, Yuan M, Frantz DF, Melendez PA, Hansen L, Lee J, et al. Local and systemic <strong>in</strong>sul<strong>in</strong> resistance result<strong>in</strong>gfrom hepatic activation of IKK-beta and NF-kappaB. Nat Med 2005;11:183e90.[163] Esp<strong>in</strong>osa L, Ingles-Esteve J, Robert-Moreno A, Bigas A. IkappaBalpha and p65 regulate the cytoplasmicshuttl<strong>in</strong>g of nuclear corepressors: cross-talk between Notch and NFkappaB pathways. Mol Biol Cell2003;14:491e502.[164] Kaur H, Chen S, X<strong>in</strong> X, Chiu J, Khan ZA, Chakrabarti S. Diabetes-<strong>in</strong>duced extracellular matrix prote<strong>in</strong>expression is mediated by transcription coactivator p300. Diabetes 2006;55:3104e11.[165] D<strong>in</strong>arello CA. A cl<strong>in</strong>ical perspective of IL-1beta as the gatekeeper of <strong>in</strong>flammation. Eur J Immunol2011;41:1203e17.[166] Papaccio G, Graziano A, d’Aqu<strong>in</strong>o R, Valiante S, Naro F. A biphasic role of nuclear transcription factor (NF)-kappaB <strong>in</strong> the islet beta-cell apoptosis <strong>in</strong>duced by <strong>in</strong>terleuk<strong>in</strong> (IL)-1beta. J Cell Physiol 2004.[167] Lundh M, Christensen DP, Rasmussen DN, Mascagni P, D<strong>in</strong>arello CA, Billestrup N, et al. Lys<strong>in</strong>e deacetylasesare produced <strong>in</strong> pancreatic beta cells and are differentially regulated by pro<strong>in</strong>flammatory cytok<strong>in</strong>es. Diabetologia2010;53:2569e78.[168] Park YJ, Luger K. Histone chaperones <strong>in</strong> nucleosome eviction and histone exchange. Curr Op<strong>in</strong> Struct Biol2008;18:282e9.[169] Meimaridou E, Gooljar SB, Chapple JP. From hatch<strong>in</strong>g to dispatch<strong>in</strong>g: the multiple cellular roles of theHsp70 molecular chaperone mach<strong>in</strong>ery. J Mol Endocr<strong>in</strong>ol 2009;42:1e9.[170] Barg<strong>in</strong>ear MF, Van Poznak C, Rosen N, Modi S, Hudis CA, Budman DR. The heat shock prote<strong>in</strong> 90 chaperonecomplex: an evolv<strong>in</strong>g therapeutic target. Curr Cancer Drug Targets 2008;8:522e32.[171] Wand<strong>in</strong>ger SK, Richter K, Buchner J. The Hsp90 chaperone mach<strong>in</strong>ery. J Biol Chem 2008;283:18473e7.[172] Hebert DN, Mol<strong>in</strong>ari M. In and out of the ER: prote<strong>in</strong> fold<strong>in</strong>g, quality control, degradation, and relatedhuman diseases. Physiol Rev 2007;87:1377e408.[173] Rapoport TA. Prote<strong>in</strong> translocation across the eukaryotic endoplasmic reticulum and bacterial plasmamembranes. Nature 2007;450:663e9.[174] Qu<strong>in</strong>ones QJ, de Ridder GG, Pizzo SV. GRP78: a chaperone with diverse roles beyond the endoplasmicreticulum. Histol Histopathol 2008;23:1409e16.[175] Paris S, Denis H, Delaive E, Dieu M, Dumont V, N<strong>in</strong>ane N, et al. Up-regulation of 94-kDa glucose-regulatedprote<strong>in</strong> by hypoxia-<strong>in</strong>ducible factor-1 <strong>in</strong> human endothelial cells <strong>in</strong> response to hypoxia. FEBS Lett2005;579:105e14.359


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>360[176] Cryer PE. The barrier of hypoglycemia <strong>in</strong> diabetes. Diabetes 2008;57:3169e76.[177] Eizirik DL, Cardozo AK, Cnop M. The role for endoplasmic reticulum stress <strong>in</strong> diabetes mellitus. Endocr Rev2008;29:42e61.[178] Scheuner D, Kaufman RJ. The unfolded prote<strong>in</strong> response: a pathway that l<strong>in</strong>ks <strong>in</strong>sul<strong>in</strong> demand with beta-cellfailure and diabetes. Endocr Rev 2008;29:317e33.[179] Rutkowski DT, Kaufman RJ. A trip to the ER: cop<strong>in</strong>g with stress. Trends Cell Biol 2004;14:20e8.[180] Lai E, Teodoro T, Volchuk A. Endoplasmic reticulum stress: signal<strong>in</strong>g the unfolded prote<strong>in</strong> response. Physiology(Bethesda) 2007;22:193e201.[181] Caruso ME, Chevet E. Systems biology of the endoplasmic reticulum stress response. Subcell Biochem2007;43:277e98.[182] Kim I, Xu W, Reed JC. Cell death and endoplasmic reticulum stress: disease relevance and therapeuticopportunities. Nat Rev Drug Discov 2008;7:1013e30.[183] Ron D, Walter P. Signal <strong>in</strong>tegration <strong>in</strong> the endoplasmic reticulum unfolded prote<strong>in</strong> response. Nat Rev MolCell Biol 2007;8:519e29.[184] Tabas I, Ron D. Integrat<strong>in</strong>g the mechanisms of apoptosis <strong>in</strong>duced by endoplasmic reticulum stress. Nat CellBiol 2011;13:184e90.[185] Vembar SS, Brodsky JL. One step at a time: endoplasmic reticulum-associated degradation. Nat Rev Mol CellBiol 2008;9:944e57.[186] Pahl HL, Baeuerle PA. The ER-overload response: activation of NF-kappa B. Trends Biochem Sci1997;22:63e7.[187] Xu C, Bailly-Maitre B, Reed JC. Endoplasmic reticulum stress: cell life and death decisions. J Cl<strong>in</strong> Invest2005;115:2656e64.[188] Heath-Engel HM, Chang NC, Shore GC. The endoplasmic reticulum <strong>in</strong> apoptosis and autophagy: role of theBCL-2 prote<strong>in</strong> family. Oncogene 2008;27:6419e33.[189] Kaufman RJ, Back SH, Song B, Han J, Hassler J. The unfolded prote<strong>in</strong> response is required to ma<strong>in</strong>ta<strong>in</strong> the<strong>in</strong>tegrity of the endoplasmic reticulum, prevent oxidative stress and preserve differentiation <strong>in</strong> beta-cells.Diabetes Obes Metab 12(Suppl. 2):99e107.[190] Lei X, Zhang S, Emani B, Barbour SE, Ramanadham S. A l<strong>in</strong>k between endoplasmic reticulum stress-<strong>in</strong>ducedbeta-cell apoptosis and the group VIA Ca2þ-<strong>in</strong>dependent phospholipase A2 (iPLA2beta). Diabetes ObesMetab 12(Suppl. 2):93e98[191] Leibowitz G, Bachar E, Shaked M, S<strong>in</strong>ai A, Ketz<strong>in</strong>el-Gilad M, Cerasi E, et-al. Glucose regulation of beta-cellstress <strong>in</strong> type 2 diabetes. Diabetes Obes Metab 12(Suppl. 2):66e75[192] Oslowski CM, Urano FA. switch from life to death <strong>in</strong> endoplasmic reticulum stressed beta-cells. DiabetesObes Metab 12(Suppl. 2):58e65[193] Johnson CA, White DA, Lavender JS, O’Neill LP, Turner BM. <strong>Human</strong> Class I Histone Deacetylase ComplexesShow Enhanced Catalytic Activity <strong>in</strong> the Presence of ATP and Co-immunoprecipitate with the ATP-dependentChaperone Prote<strong>in</strong> Hsp70. J Biol Chem 2002;277:9590e7.[194] Battu A, Ray A, Wani AA. ASF1A and ATM regulate H3K56-mediated cell-cycle checkpo<strong>in</strong>t recovery <strong>in</strong>response to UV irradiation. Nucleic Acids Res 2011.[195] M<strong>in</strong>ard LV, L<strong>in</strong> LJ, Schultz MC. SWI/SNF and Asf1 Independently Promote Derepression of the DNA DamageResponse Genes under Conditions of Replication Stress. PLoS One 2011;6:e21633.[196] Das C, Lucia MS, Hansen KC, Tyler JK. CBP/p300-mediated acetylation of histone H3 on lys<strong>in</strong>e 56. Nature2009;459:113e7.[197] L<strong>in</strong> LJ, M<strong>in</strong>ard LV, Johnston GC, S<strong>in</strong>ger RA, Schultz MC. Asf1 can promote trimethylation of H3 K36 by Set2.Mol Cell Biol 2010;30:1116e29.[198] Berndsen CE, Tsubota T, L<strong>in</strong>dner SE, Lee S, Holton JM, Kaufman PD, et al. Molecular functions of the histoneacetyltransferase chaperone complex Rtt109-Vps75. Nat Struct Mol Biol 2008;15:948e56.[199] Chen CC, Carson JJ, Feser J, Tambur<strong>in</strong>i B, Zabaronick S, L<strong>in</strong>ger J, et al. Acetylated lys<strong>in</strong>e 56 on histoneH3 drives chromat<strong>in</strong> assembly after repair and signals for the completion of repair. Cell2008;134:231e43.[200] Hiraga S, Botsios S, Donaldson AD. Histone H3 lys<strong>in</strong>e 56 acetylation by Rtt109 is crucial for chromosomeposition<strong>in</strong>g. J Cell Biol 2008;183:641e51.[201] Kaplan T, Liu CL, Erkmann JA, Holik J, Grunste<strong>in</strong> M, Kaufman PD, et al. Cell cycle- and chaperone-mediatedregulation of H3K56ac <strong>in</strong>corporation <strong>in</strong> yeast. PLoS Genet 2008;4:e1000270.[202] Li Q, Zhou H, Wurtele H, Davies B, Horazdovsky B, Verreault A, et al. Acetylation of histone H3 lys<strong>in</strong>e 56regulates replication-coupled nucleosome assembly. Cell 2008;134:244e55.[203] Miller A, Yang B, Foster T, Kirchmaier AL. Proliferat<strong>in</strong>g cell nuclear antigen and ASF1 modulate silentchromat<strong>in</strong> <strong>in</strong> Saccharomyces cerevisiae via lys<strong>in</strong>e 56 on histone H3. Genetics 2008;179:793e809.


CHAPTER 17The Potential of Epigenetic Compounds <strong>in</strong> Treat<strong>in</strong>g Diabetes[204] Tang Y, Meeth K, Jiang E, Luo C, Marmorste<strong>in</strong> R. Structure of Vps75 and implications for histone chaperonefunction. Proc Natl Acad Sci USA 2008;105:12206e11.[205] Williams SK, Truong D, Tyler JK. Acetylation <strong>in</strong> the globular core of histone H3 on lys<strong>in</strong>e-56 promoteschromat<strong>in</strong> disassembly dur<strong>in</strong>g transcriptional activation. Proc Natl Acad Sci USA 2008;105:9000e5.[206] Adk<strong>in</strong>s MW, Carson JJ, English CM, Ramey CJ, Tyler JK. The histone chaperone anti-silenc<strong>in</strong>g function1 stimulates the acetylation of newly synthesized histone H3 <strong>in</strong> S-phase. J Biol Chem2007;282:1334e40.[207] Fill<strong>in</strong>gham J, Recht J, Silva AC, Suter B, Emili A, Stagljar I, et al. Chaperone control of the activity andspecificity of the histone H3 acetyltransferase Rtt109. Mol Cell Biol 2008;28:4342e53.[208] Han J, Zhou H, Li Z, Xu RM, Zhang Z. Acetylation of lys<strong>in</strong>e 56 of histone H3 catalyzed by RTT109 andregulated by ASF1 is required for replisome <strong>in</strong>tegrity. J Biol Chem 2007;282:28587e96.[209] LeRoy G, Rickards B, Fl<strong>in</strong>t SJ. The double bromodoma<strong>in</strong> prote<strong>in</strong>s Brd2 and Brd3 couple histone acetylationto transcription. Mol Cell 2008;30:51e60.[210] Munemasa Y, Suzuki T, Aizawa K, Miyamoto S, Imai Y, Matsumura T, et al. Promoter region-specific histone<strong>in</strong>corporation by the novel histone chaperone ANP32B and DNA-b<strong>in</strong>d<strong>in</strong>g factor KLF5. Mol Cell Biol2008;28:1171e81.[211] Recht J, Tsubota T, Tanny JC, Diaz RL, Berger JM, Zhang X, et al. Histone chaperone Asf1 is required forhistone H3 lys<strong>in</strong>e 56 acetylation, a modification associated with S phase <strong>in</strong> mitosis and meiosis. Proc NatlAcad Sci USA 2006;103:6988e93.[212] Rufiange A, Jacques PE, Bhat W, Robert F, Nourani A. Genome-wide replication-<strong>in</strong>dependent histone H3exchange occurs predom<strong>in</strong>antly at promoters and implicates H3 K56 acetylation and Asf1. Mol Cell2007;27:393e405.[213] Schneider J, Bajwa P, Johnson FC, Bhaumik SR, Shilatifard A. Rtt109 is required for proper H3K56 acetylation:a chromat<strong>in</strong> mark associated with the elongat<strong>in</strong>g RNA polymerase II. J Biol Chem2006;281:37270e4.[214] Barman HK, Takami Y, Nishijima H, Shibahara K, Sanematsu F, Nakayama T. Histone acetyltransferase-1regulates <strong>in</strong>tegrity of cytosolic histone H3-H4 conta<strong>in</strong><strong>in</strong>g complex. Biochem Biophys Res Commun2008;373:624e30.[215] J<strong>in</strong> C, Kato K, Chimura T, Yamasaki T, Nakade K, Murata T, et al. Regulation of histone acetylation andnucleosome assembly by transcription factor JDP2. Nat Struct Mol Biol 2006;13:331e8.[216] Swam<strong>in</strong>athan V, Kishore AH, Febitha KK, Kundu TK. <strong>Human</strong> histone chaperone nucleophosm<strong>in</strong> enhancesacetylation-dependent chromat<strong>in</strong> transcription. Mol Cell Biol 2005;25:7534e45.[217] Thomas M, Dadgar N, Aphale A, Harrell JM, Kunkel R, Pratt WB, et al. Androgen receptor acetylation sitemutations cause traffick<strong>in</strong>g defects, misfold<strong>in</strong>g, and aggregation similar to expanded glutam<strong>in</strong>e tracts. J BiolChem 2004;279:8389e95.[218] Kekatpure VD, Dannenberg AJ, Subbaramaiah K. HDAC6 modulates HSP90 chaperone activity and regulatesactivation of aryl hydrocarbon receptor signal<strong>in</strong>g. J Biol Chem 2009;284:7436e45.[219] Fiskus W, Ren Y, Mohapatra A, Bali P, Mandawat A, Rao R, et al. Hydroxamic acid analogue histone deacetylase<strong>in</strong>hibitors attenuate estrogen receptor-alpha levels and transcriptional activity: a result of hyperacetylationand <strong>in</strong>hibition of chaperone function of heat shock prote<strong>in</strong> 90. Cl<strong>in</strong> Cancer Res2007;13:4882e90.[220] Suuronen T, Ojala J, Hytt<strong>in</strong>en JM, Kaarniranta K, Thornell A, Kyrylenko S, et al. Regulation of ER alphasignal<strong>in</strong>g pathway <strong>in</strong> neuronal HN10 cells: role of prote<strong>in</strong> acetylation and Hsp90. Neurochem Res2008;33:1768e75.[221] Kovacs JJ, Murphy PJ, Gaillard S, Zhao X, Wu JT, Nicchitta CV, et al. HDAC6 regulates Hsp90 acetylation andchaperone-dependent activation of glucocorticoid receptor. Mol Cell 2005;18:601e7.[222] Giust<strong>in</strong>iani J, Daire V, Cantaloube I, Durand G, Pous C, Perdiz D, et al. Tubul<strong>in</strong> acetylation favors Hsp90recruitment to microtubules and stimulates the signal<strong>in</strong>g function of the Hsp90 clients Akt/PKB and p53.Cell Signal 2008;21:529e39.[223] Bardag-Gorce F, Dedes J, French BA, Oliva JV, Li J, French SW. Mallory body formation is associated withepigenetic phenotypic change <strong>in</strong> hepatocytes <strong>in</strong> vivo. Exp Mol Pathol 2007;83:160e8.[224] Hanada S, Harada M, Kumemura H, Bishr Omary M, Koga H, Kawaguchi T, et al. Oxidative stress<strong>in</strong>duces the endoplasmic reticulum stress and facilitates <strong>in</strong>clusion formation <strong>in</strong> cultured cells. J Hepatol2007;47:93e102.[225] Cherasse Y, Chaveroux C, Jousse C, Maur<strong>in</strong> AC, Carraro V, Parry L, et al. Role of the repressor JDP2 <strong>in</strong> theam<strong>in</strong>o acid-regulated transcription of CHOP. FEBS Lett 2008;582:1537e41.[226] Ohoka N, Hattori T, Kitagawa M, Onozaki K, Hayashi H. Critical and Functional Regulation ofCHOP (C/EBP Homologous Prote<strong>in</strong>) through the N-term<strong>in</strong>al Portion. J Biol Chem2007;282:35687e94.361


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>362[227] Baumeister P, Luo S, Skarnes WC, Sui G, Seto E, Shi Y, et al. Endoplasmic reticulum stress <strong>in</strong>duction of theGrp78/BiP promoter: activat<strong>in</strong>g mechanisms mediated by YY1 and its <strong>in</strong>teractive chromat<strong>in</strong> modifiers. MolCell Biol 2005;25:4529e40.[228] Donati G, Imbriano C, Mantovani R. Dynamic recruitment of transcription factors and epigenetic changes onthe ER stress response gene promoters. Nucleic Acids Res 2006;34:3116e27.[229] Doody GM, Stephenson S, Tooze RM. BLIMP-1 is a target of cellular stress and downstream of the unfoldedprote<strong>in</strong> response. Eur J Immunol 2006;36:1572e82.[230] Yu J, Angel<strong>in</strong>-Duclos C, Greenwood J, Liao J, Calame K. Transcriptional repression by blimp-1 (PRDI-BF1)<strong>in</strong>volves recruitment of histone deacetylase. Mol Cell Biol 2000;20:2592e603.[231] Zeng L, Lu M, Mori K, Luo S, Lee AS, Zhu Y, et al. ATF6 modulates SREBP2-mediated lipogenesis. Embo J2004;23:950e8.[232] Sugden MC, Holness MJ. Role of nuclear receptors <strong>in</strong> the modulation of <strong>in</strong>sul<strong>in</strong> secretion <strong>in</strong> lipid-<strong>in</strong>duced<strong>in</strong>sul<strong>in</strong> resistance. Biochem Soc Trans 2008;36:891e900.[233] Pratt WB, Morishima Y, Murphy M, Harrell M. Chaperon<strong>in</strong>g of glucocorticoid receptors. Handb Exp Pharmacol2006:111e38.[234] Murphy PJ, Morishima Y, Kovacs JJ, Yao TP, Pratt WB. Regulation of the dynamics of hsp90 action onthe glucocorticoid receptor by acetylation/deacetylation of the chaperone. J Biol Chem 2005;280:33792e9.[235] Amat R, Solanes G, Giralt M, Villarroya F. SIRT1 is <strong>in</strong>volved <strong>in</strong> glucocorticoid-mediated control of uncoupl<strong>in</strong>gprote<strong>in</strong>-3 gene transcription. J Biol Chem 2007;282:34066e76.[236] Ito K, Barnes PJ, Adcock IM. Glucocorticoid receptor recruitment of histone deacetylase 2 <strong>in</strong>hibits<strong>in</strong>terleuk<strong>in</strong>-1beta-<strong>in</strong>duced histone H4 acetylation on lys<strong>in</strong>es 8 and 12. Mol Cell Biol 2000;20:6891e903.[237] Kelly A, Bowen H, Jee YK, Mahfiche N, Soh C, Lee T, et al. The glucocorticoid receptor beta isoform canmediate transcriptional repression by recruit<strong>in</strong>g histone deacetylases. J Allergy Cl<strong>in</strong> Immunol2008;121:203e8. e201.[238] Wiper-Bergeron N, Salem HA, Toml<strong>in</strong>son JJ, Wu D, Hache RJ. Glucocorticoid-stimulated preadipocytedifferentiation is mediated through acetylation of C/EBPbeta by GCN5. Proc Natl Acad Sci USA2007;104:2703e8.[239] Wiper-Bergeron N, Wu D, Pope L, Schild-Poulter C, Hache RJ. Stimulation of preadipocyte differentiation bysteroid through target<strong>in</strong>g of an HDAC1 complex. EMBO J 2003;22:2135e45.[240] Bolden JE, Peart MJ, Johnstone RW. Anticancer activities of histone deacetylase <strong>in</strong>hibitors. Nat Rev DrugDiscov 2006;5:769e84.[241] Outeiro TF, Kontopoulos E, Altman S, Kufareva I, Strathearn KE, Amore AM, et al. Sirtu<strong>in</strong> 2 Inhibitors Rescuea-Synucle<strong>in</strong>-Mediated Toxicity <strong>in</strong> Models of Park<strong>in</strong>son’s <strong>Disease</strong>. <strong>Science</strong> 2007;317:516e9.[242] Schuetz A, M<strong>in</strong> J, Antoshenko T, Wang CL, Allali-Hassani A, Dong A, et al. Structural basis of <strong>in</strong>hibition of thehuman NADþ-dependent deacetylase SIRT5 by suram<strong>in</strong>. Structure 2007;15:377e89.[243] Kiviranta PH, Leppanen J, Kyrylenko S, Salo HS, Lahtela-Kakkonen M, Tervo AJ, et al. N, N’-Bisbenzylidenebenzene-1,4-diam<strong>in</strong>esand N, N’-Bisbenzylidenenaphthalene-1,4-diam<strong>in</strong>es as Sirtu<strong>in</strong> Type 2 (SIRT2).Inhibitors. J Med Chem 2006;49:7907e11.[244] Tervo AJ, Suuronen T, Kyrylenko S, Kuusisto E, Kiviranta PH, Salm<strong>in</strong>en A, et al. Discover<strong>in</strong>g <strong>in</strong>hibitors ofhuman sirtu<strong>in</strong> type 2: novel structural scaffolds. J Med Chem 2006;49:7239e41.[245] Olaharski AJ, R<strong>in</strong>e J, Marshall BL, Babiarz J, Zhang L, Verd<strong>in</strong> E, et al. The flavor<strong>in</strong>g agent dihydrocoumar<strong>in</strong>reverses epigenetic silenc<strong>in</strong>g and <strong>in</strong>hibits sirtu<strong>in</strong> deacetylases. PLoS Genet 2005;1:e77.[246] Mai A, Massa S, Lavu S, Pezzi R, Simeoni S, Ragno R, et al. Design, synthesis, and biological evaluation ofsirt<strong>in</strong>ol analogues as class III histone/prote<strong>in</strong> deacetylase (Sirtu<strong>in</strong>) <strong>in</strong>hibitors. J Med Chem2005;48:7789e95.[247] Dashwood RH, Ho E. Dietary histone deacetylase <strong>in</strong>hibitors: From cells to mice to man. Sem<strong>in</strong> Cancer Biol2007;17:363e9.[248] Howitz KT, Bitterman KJ, Cohen HY, Lamm<strong>in</strong>g DW, Lavu S, Wood JG, et al. Small molecule activators ofsirtu<strong>in</strong>s extend Saccharomyces cerevisiae lifespan. Nature 2003;425:191e6.[249] Kaeberle<strong>in</strong> M, McDonagh T, Heltweg B, Hixon J, Westman EA, Caldwell SD, et al. Substrate-specific activationof sirtu<strong>in</strong>s by resveratrol. J Biol Chem 2005;280:17038e45.[250] Gray SG. Histone acetyltransferases and histone deacetylases <strong>in</strong> gene regulation and as drug targets. In:DevitaJr VT, Hellman S, Rosenberg SA, editors. Progress <strong>in</strong> Oncology. Sudbury: Jones and Bartlett; 2004.p. 99e128.[251] Tambaro F, Dell’Aversana C, Carafa V, Nebbioso A, Radic B, Ferrara F, et al. Histone deacetylase <strong>in</strong>hibitors:cl<strong>in</strong>ical implications for hematological malignancies. Cl<strong>in</strong> Epigenet 2010:25e44.


CHAPTER 17The Potential of Epigenetic Compounds <strong>in</strong> Treat<strong>in</strong>g Diabetes[252] Blaheta RA, C<strong>in</strong>atl Jr J. Anti-tumor mechanisms of valproate: a novel role for an old drug. Med Res Rev2002;22:492e511.[253] Soriano AO, Yang H, Faderl S, Estrov Z, Giles F, Ravandi F, et al. Safety and cl<strong>in</strong>ical activity of the comb<strong>in</strong>ationof 5-azacytid<strong>in</strong>e, valproic acid and all-trans ret<strong>in</strong>oic acid <strong>in</strong> acute myeloid leukemia and myelodysplasticsyndrome. Blood 2007;110:2302e8.[254] Atmaca A, Al-Batran SE, Maurer A, Neumann A, He<strong>in</strong>zel T, Hentsch B, et al. Valproic acid (VPA) <strong>in</strong> patientswith refractory advanced cancer: a dose escalat<strong>in</strong>g phase I cl<strong>in</strong>ical trial. Br J Cancer 2007;97:177e82.[255] Münster P, Marchion D, Bicaku E, Schmitt M, Lee JH, DeConti R, et al. Phase I trial of histone deacetylase<strong>in</strong>hibition by valproic acid followed by the topoisomerase II <strong>in</strong>hibitor epirubic<strong>in</strong> <strong>in</strong> advanced solid tumors:a cl<strong>in</strong>ical and translational study. J Cl<strong>in</strong> Oncol 2007;25:1979e85.[256] Greco R, Lat<strong>in</strong>i G, Chiarelli F, Iannetti P, Verrotti A. Lept<strong>in</strong>, ghrel<strong>in</strong>, and adiponect<strong>in</strong> <strong>in</strong> epileptic patientstreated with valproic acid. Neurology 2005;65:1808e9.[257] Pylvanen V, Knip M, Pakar<strong>in</strong>en AJ, Turkka J, Kotila M, Rattya J, et al. Fast<strong>in</strong>g serum <strong>in</strong>sul<strong>in</strong> and lipid levels <strong>in</strong>men with epilepsy. Neurology 2003;60:571e4.[258] Pylvanen V, Pakar<strong>in</strong>en A, Knip M, Isojarvi J. Characterization of <strong>in</strong>sul<strong>in</strong> secretion <strong>in</strong> Valproate-treated patientswith epilepsy. Epilepsia 2006;47:1460e4.[259] Pylvanen V, Pakar<strong>in</strong>en A, Knip M, Isojarvi J. Insul<strong>in</strong>-related metabolic changes dur<strong>in</strong>g treatment withvalproate <strong>in</strong> patients with epilepsy. Epilepsy Behav 2006;8:643e8.[260] Tan H, Orbak Z, Kantarci M, Kocak N, Karaca L. Valproate-<strong>in</strong>duced <strong>in</strong>sul<strong>in</strong> resistance <strong>in</strong> prepubertal girls withepilepsy. J Pediatr Endocr<strong>in</strong>ol Metab 2005;18:985e9.[261] Verrotti A, di Corcia G, Sallad<strong>in</strong>i C, Trotta D, Morgese G, Chiarelli F. Serum <strong>in</strong>sul<strong>in</strong> and lept<strong>in</strong> levels andvalproate-associated obesity. Epilepsia 2003;44:1606.[262] Verrotti A, D’Egidio C, Mohn A, Coppola G, Chiarelli F. Weight ga<strong>in</strong> follow<strong>in</strong>g treatment with valproic acid:pathogenetic mechanisms and cl<strong>in</strong>ical implications. Obes Rev 2011;12:e32e43.[263] Lagace DC, McLeod RS, Nachtigal MW. Valproic acid <strong>in</strong>hibits lept<strong>in</strong> secretion and reduces lept<strong>in</strong> messengerribonucleic acid levels <strong>in</strong> adipocytes. Endocr<strong>in</strong>ology 2004;145:5493e503.[264] Persegh<strong>in</strong> G, Petersen K, Shulman GI. Cellular mechanism of <strong>in</strong>sul<strong>in</strong> resistance: potential l<strong>in</strong>ks with<strong>in</strong>flammation. Int J Obes Relat Metab Disord 27 (Suppl.3)/ 2003(S6-11):S6e11.[265] Shoelson SE, Lee J, Yuan M. Inflammation and the IKK beta/I kappa B/NF-kappa B axis <strong>in</strong> obesity- anddiet-<strong>in</strong>duced <strong>in</strong>sul<strong>in</strong> resistance. Int J Obes Relat Metab Disord 2003;27:49e52.[266] Zeender E, Maedler K, Bosco D, Berney T, Donath MY, Halban PA. Pioglitazone and sodium salicylate protecthuman beta-cells aga<strong>in</strong>st apoptosis and impaired function <strong>in</strong>duced by glucose and <strong>in</strong>terleuk<strong>in</strong>-1beta. J Cl<strong>in</strong>Endocr<strong>in</strong>ol Metab 2004;89:5059e66.[267] P<strong>in</strong>ckard RN, Hawk<strong>in</strong>s D, Farr RS. In vitro acetylation of plasma prote<strong>in</strong>s, enzymes and DNA by aspir<strong>in</strong>.Nature 1968;219:68e9.[268] Di Renzo F, Cappelletti G, Broccia ML, Giav<strong>in</strong>i E, Menegola E. The <strong>in</strong>hibition of embryonic histone deacetylasesas the possible mechanism account<strong>in</strong>g for axial skeletal malformations <strong>in</strong>duced by sodium salicylate.Toxicol Sci 2008;104:397e404.[269] Sun X, Han F, Yi J, Han L, Wang B. Effect of Aspir<strong>in</strong> on the Expression of Hepatocyte NF-kappaB and SerumTNF-alpha <strong>in</strong> Streptozotoc<strong>in</strong>-Induced Type 2 Diabetic Rats. J Korean Med Sci 2011;26:765e70.[270] Kim SN, Choi HY, Kim YK. Regulation of adipocyte differentiation by histone deacetylase <strong>in</strong>hibitors. ArchPharm Res 2009;32:535e41.[271] Yoo EJ, Chung JJ, Choe SS, Kim KH, Kim JB. Down-regulation of histone deacetylases stimulates adipocytedifferentiation. J Biol Chem 2006;281:6608e15.[272] Advani A, Huang Q, Thai K, Advani SL, White KE, Kelly DJ, et al. Long-term adm<strong>in</strong>istration of the histonedeacetylase <strong>in</strong>hibitor vor<strong>in</strong>ostat attenuates renal <strong>in</strong>jury <strong>in</strong> experimental diabetes through an endothelial nitricoxide synthase-dependent mechanism. Am J Pathol 2011;178:2205e14.[273] Gilbert RE, Huang Q, Thai K, Advani SL, Lee K, Yuen DA, et al. Histone deacetylase <strong>in</strong>hibition attenuatesdiabetes-associated kidney growth: potential role for epigenetic modification of the epidermal growth factorreceptor. Kidney Int 2011;79:1312e21.[274] Sche<strong>in</strong> PS, Cooney DA, Vernon ML. The use of nicot<strong>in</strong>amide to modify the toxicity of streptozotoc<strong>in</strong> diabeteswithout loss of antitumor activity. Cancer Res 1967;27:2324e32.[275] Stauffacher W, Burr I, Gutzeit A, Beaven D, Velem<strong>in</strong>sky J, Renold AE. Streptozotoc<strong>in</strong> diabetes: time course ofirreversible B-cell damage; further observations on prevention by nicot<strong>in</strong>amide. Proc Soc Exp Biol Med1970;133:194e200.[276] Sandler S, Andersson A, Korsgren O, Tollemar J, Petersson B, Groth CG, et al. Tissue culture of human fetalpancreas. Effects of nicot<strong>in</strong>amide on <strong>in</strong>sul<strong>in</strong> production and formation of isletlike cell clusters. Diabetes1989; 38(suppl.1):168e71 .363


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>364[277] Korsgren O, Andersson A, Sandler S. Pretreatment of fetal porc<strong>in</strong>e pancreas <strong>in</strong> culture with nicot<strong>in</strong>amideaccelerates reversal of diabetes after transplantation to nude mice. Surgery 1993;113:205e14.[278] Gale EA, B<strong>in</strong>gley PJ, Emmett CL, Collier T. European Nicot<strong>in</strong>amide Diabetes Intervention Trial (ENDIT):a randomised controlled trial of <strong>in</strong>tervention before the onset of type 1 diabetes. Lancet 2004;363:925e31.[279] Hedman M, Ludvigsson J, Faresjo MK. Nicot<strong>in</strong>amide reduces high secretion of IFN-gamma <strong>in</strong>high-risk relatives even though it does not prevent type 1 diabetes. J Interferon Cytok<strong>in</strong>e Res 2006;26:207e13.[280] Liu HK, Green BD, Flatt PR, McClenaghan NH, McCluskey JT. Effects of long-term exposure to nicot<strong>in</strong>amideand sodium butyrate on growth, viability, and the function of clonal <strong>in</strong>sul<strong>in</strong> secret<strong>in</strong>g cells. Endocr Res2004;30:61e8.[281] Sh<strong>in</strong> JS, M<strong>in</strong> BH, Lim JY, Kim BK, Han HJ, Yoon KH, et al. Novel culture technique <strong>in</strong>volv<strong>in</strong>g an histonedeacetylase <strong>in</strong>hibitor reduces the marg<strong>in</strong>al islet mass to correct streptozotoc<strong>in</strong>-<strong>in</strong>duced diabetes. CellTransplant 2011;20:1321e32.[282] Honkanen J, Niem<strong>in</strong>en JK, Gao R, Luopajarvi K, Salo HM, Ilonen J, et al. IL-17 immunity <strong>in</strong> human type 1diabetes. J Immunol 2010;185:1959e67.[283] Jagannathan-Bogdan M, McDonnell ME, Sh<strong>in</strong> H, Rehman Q, Hasturk H, Apovian CM, et al. Elevatedpro<strong>in</strong>flammatory cytok<strong>in</strong>e production by a skewed T cell compartment requires monocytes and promotes<strong>in</strong>flammation <strong>in</strong> type 2 diabetes. J Immunol 2011;186:1162e72.[284] Sutherland AP, Van Belle T, Wurster AL, Suto A, Michaud M, Zhang D, et al. Interleuk<strong>in</strong>-21 is required for thedevelopment of type 1 diabetes <strong>in</strong> NOD mice. Diabetes 2009;58:1144e55.[285] Spolski R, Kashyap M, Rob<strong>in</strong>son C, Yu Z, Leonard WJ. IL-21 signal<strong>in</strong>g is critical for the development of type Idiabetes <strong>in</strong> the NOD mouse. Proc Natl Acad Sci USA 2008;105:14028e33.[286] McGuire HM, Walters S, Vogelzang A, Lee CM, Webster KE, Sprent J, et al. Interleuk<strong>in</strong>-21 is criticallyrequired <strong>in</strong> autoimmune and allogeneic responses to islet tissue <strong>in</strong> mur<strong>in</strong>e models. Diabetes 2011;60:867e75.[287] Arif S, Moore F, Marks K, Bouckenooghe T, Dayan CM, Planas R, et al. Peripheral and Islet Interleuk<strong>in</strong>-17Pathway Activation Characterizes <strong>Human</strong> Autoimmune Diabetes and Promotes Cytok<strong>in</strong>e-Mediated {b }-CellDeath. Diabetes 2011;60:2112e9.[288] Koenen HJ, Smeets RL, V<strong>in</strong>k PM, van Rijssen E, Boots AM, Joosten I. <strong>Human</strong> CD25highFoxp3pos regulatoryT cells differentiate <strong>in</strong>to IL-17-produc<strong>in</strong>g cells. Blood 2008;112:2340e52.[289] Patel T, Patel V, S<strong>in</strong>gh R, Jayaraman S. Chromat<strong>in</strong> remodel<strong>in</strong>g resets the immune system to protect aga<strong>in</strong>stautoimmune diabetes <strong>in</strong> mice. Immunol Cell Biol 2011;89:640e9.[290] Gangaram-Panday ST, Faas MM, de Vos P. Towards stem-cell therapy <strong>in</strong> the endocr<strong>in</strong>e pancreas. Trends MolMed 2007;13:164e73.[291] Lee JH, Hart SR, Skalnik DG. Histone deacetylase activity is required for embryonic stem cell differentiation.Genesis 2004;38:32e8.[292] Romagnani P, Lasagni L, Mazz<strong>in</strong>ghi B, Lazzeri E, Romagnani S. Pharmacological modulation of stem cellfunction. Curr Med Chem 2007;14:1129e39.[293] Okazaki K, Maltepe E. Oxygen, epigenetics and stem cell fate. Regen Med 2006;1:71e83.[294] Lumelsky N, Blondel O, Laeng P, Velasco I, Rav<strong>in</strong> R, McKay R. Differentiation of embryonic stem cells to<strong>in</strong>sul<strong>in</strong>-secret<strong>in</strong>g structures similar to pancreatic islets. <strong>Science</strong> 2001;292:1389e94.[295] Calvanese V, Lara E, Suarez-Alvarez B, Abu Dawud R, Vazquez-Chantada M, Mart<strong>in</strong>ez-Chantar ML, et al.Sirtu<strong>in</strong> 1 regulation of developmental genes dur<strong>in</strong>g differentiation of stem cells. Proc Natl Acad Sci USA2011;107:13736e41.[296] Tayaramma T, Ma B, Rohde M, Mayer H. Chromat<strong>in</strong>-remodel<strong>in</strong>g factors allow differentiation of bone marrowcells <strong>in</strong>to <strong>in</strong>sul<strong>in</strong>-produc<strong>in</strong>g cells. Stem Cells 2006;24:2858e67.[297] Sun Y, Chen L, Hou XG, Hou WK, Dong JJ, Sun L, et al. Differentiation of bone marrow-derived mesenchymalstem cells from diabetic patients <strong>in</strong>to <strong>in</strong>sul<strong>in</strong>-produc<strong>in</strong>g cells <strong>in</strong> vitro. Ch<strong>in</strong> Med J (Engl) 2007;120:771e6.[298] Goicoa S, Alvarez S, Ricordi C, Inverardi L, Dom<strong>in</strong>guez-Bendala J. Sodium butyrate activates genes of earlypancreatic development <strong>in</strong> embryonic stem cells. Clon<strong>in</strong>g Stem Cells 2006;8:140e9.[299] Jiang J, Au M, Lu K, Eshpeter A, Korbutt G, Fisk G, et al. Generation of Insul<strong>in</strong>-produc<strong>in</strong>g Islet-like Clustersfrom <strong>Human</strong> Embryonic Stem Cells. Stem Cells 2007;25:1940e53.[300] Mutskov V, Raaka BM, Felsenfeld G, Gershengorn MC. The human <strong>in</strong>sul<strong>in</strong> gene displays transcriptionallyactive epigenetic marks <strong>in</strong> islet-derived mesenchymal precursor cells <strong>in</strong> the absence of <strong>in</strong>sul<strong>in</strong> expression.Stem Cells 2007;25:3223e33.[301] Wilson LM, Wong SH, Yu N, Geras-Raaka E, Raaka BM, Gershengorn MC. Insul<strong>in</strong> but not glucagon gene issilenced <strong>in</strong> human pancreas-derived mesenchymal stem cells. Stem Cells 2009;27:2703e11.


CHAPTER 17The Potential of Epigenetic Compounds <strong>in</strong> Treat<strong>in</strong>g Diabetes[302] Davani B, Ikonomou L, Raaka BM, Geras-Raaka E, Morton RA, Marcus-Samuels B, et al. <strong>Human</strong> islet-derivedprecursor cells are mesenchymal stromal cells that differentiate and mature to hormone-express<strong>in</strong>g cells<strong>in</strong> vivo. Stem Cells 2007;25:3215e22.[303] Rubenste<strong>in</strong> RC, Zeitl<strong>in</strong> PL. Sodium 4-phenylbutyrate downregulates Hsc70: implications for <strong>in</strong>tracellulartraffick<strong>in</strong>g of DeltaF508-CFTR. Am J Physiol Cell Physiol 2000;278:C259e67.[304] Vij N, Fang S, Zeitl<strong>in</strong> PL. Selective <strong>in</strong>hibition of endoplasmic reticulum-associated degradation rescuesDeltaF508-cystic fibrosis transmembrane regulator and suppresses <strong>in</strong>terleuk<strong>in</strong>-8 levels: therapeutic implications.J Biol Chem 2006;281:17369e78.[305] Burrows JA, Willis LK, Perlmutter DH. Chemical chaperones mediate <strong>in</strong>creased secretion of mutant alpha1-antitryps<strong>in</strong> (alpha 1-AT) Z: A potential pharmacological strategy for prevention of liver <strong>in</strong>jury andemphysema <strong>in</strong> alpha 1-AT deficiency. Proc Natl Acad Sci USA 2000;97:1796e801.[306] Qi X, Hosoi T, Okuma Y, Kaneko M, Nomura Y. Sodium 4-phenylbutyrate protects aga<strong>in</strong>st cerebral ischemic<strong>in</strong>jury. Mol Pharmacol 2004;66:899e908.[307] Vilatoba M, Eckste<strong>in</strong> C, Bilbao G, Smyth CA, Jenk<strong>in</strong>s S, Thompson JA, et al. Sodium 4-phenylbutyrateprotects aga<strong>in</strong>st liver ischemia reperfusion <strong>in</strong>jury by <strong>in</strong>hibition of endoplasmic reticulum-stress mediatedapoptosis. Surgery 2005;138:342e51.[308] Mulhern ML, Madson CJ, Kador PF, Randazzo J, Sh<strong>in</strong>ohara T. Cellular osmolytes reduce lens epithelial celldeath and alleviate cataract formation <strong>in</strong> galactosemic rats. Mol Vis 2007;13:1397e405.[309] Bonapace G, Waheed A, Shah GN, Sly WS. Chemical chaperones protect from effects of apoptosis-<strong>in</strong>duc<strong>in</strong>gmutation <strong>in</strong> carbonic anhydrase IV identified <strong>in</strong> ret<strong>in</strong>itis pigmentosa 17. Proc Natl Acad Sci USA2004;101:12300e5.[310] de Almeida SF, Picarote G, Flem<strong>in</strong>g JV, Carmo-Fonseca M, Azevedo JE,. de Sousa M.Chemical chaperonesreduce endoplasmic reticulum stress and prevent mutant HFE aggregate formation. J Biol Chem2007;282:27905e12.[311] Kubota K, Ni<strong>in</strong>uma Y, Kaneko M, Okuma Y, Sugai M, Omura T, et al. Suppressive effects of4-phenylbutyrate on the aggregation of Pael receptors and endoplasmic reticulum stress. J Neurochem2006;97:1259e68.[312] Shao L, Sun X, Xu L, Young LT, Wang JF. Mood stabiliz<strong>in</strong>g drug lithium <strong>in</strong>creases expression of endoplasmicreticulum stress prote<strong>in</strong>s <strong>in</strong> primary cultured rat cerebral cortical cells. Life Sci 2006;78:1317e23.[313] Wang JF, Bown C, Young LT. Differential display PCR reveals novel targets for the mood-stabiliz<strong>in</strong>g drugvalproate <strong>in</strong>clud<strong>in</strong>g the molecular chaperone GRP78. Mol Pharmacol 1999;55:521e7.[314] Cui J, Shao L, Young LT, Wang JF. Role of glutathione <strong>in</strong> neuroprotective effects of mood stabiliz<strong>in</strong>g drugslithium and valproate. Neuroscience 2007;144:1447e53.[315] Tveten K, Holla OL, Ranheim T, Berge KE, Leren TP, Kulseth MA. 4-Phenylbutyrate restores the functionalityof a misfolded mutant low-density lipoprote<strong>in</strong> receptor. Febs J 2007;274:1881e93.[316] Chiang DJ, Pritchard MT, Nagy LE. Obesity, diabetes mellitus, and liver fibrosis. Am J Physiol Gastro<strong>in</strong>testLiver Physiol 2011;300:G697e702.[317] Morton GJ, Schwartz MW. Lept<strong>in</strong> and the central nervous system control of glucose metabolism. Physiol Rev2011;91:389e411.[318] Myers Jr MG, Leibel RL, Seeley RJ, Schwartz MW. Obesity and lept<strong>in</strong> resistance: dist<strong>in</strong>guish<strong>in</strong>g cause fromeffect. Trends Endocr<strong>in</strong>ol Metab 2010;21:643e51.[319] Hosoi T, Sasaki M, Miyahara T, Hashimoto C, Matsuo S, Yoshii M, et al. Endoplasmic reticulum stress <strong>in</strong>duceslept<strong>in</strong> resistance. Mol Pharmacol 2008;74:1610e9.[320] Ozcan L, Erg<strong>in</strong> AS, Lu A, Chung J, Sarkar S, Nie D, et al. Endoplasmic reticulum stress plays a central role <strong>in</strong>development of lept<strong>in</strong> resistance. Cell Metab 2009;9:35e51.[321] Won JC, Jang PG, Namkoong C, Koh EH, Kim SK, Park JY, et al. Central adm<strong>in</strong>istration of an endoplasmicreticulum stress <strong>in</strong>ducer <strong>in</strong>hibits the anorexigenic effects of lept<strong>in</strong> and <strong>in</strong>sul<strong>in</strong>. Obesity (Silver Spr<strong>in</strong>g)2009;17:1861e5.[322] Nogueira TC, Lellis-Santos C, Jesus DS, Taneda M, Rodrigues SC, Amaral FG, et al. Absence of melaton<strong>in</strong><strong>in</strong>duces night-time hepatic <strong>in</strong>sul<strong>in</strong> resistance and <strong>in</strong>creased gluconeogenesis due to stimulation of nocturnalunfolded prote<strong>in</strong> response. Endocr<strong>in</strong>ology 2011;152:1253e63.[323] Ozcan U, Yilmaz E, Ozcan L, Furuhashi M, Vaillancourt E, Smith RO, et al. Chemical chaperones reduceER stress and restore glucose homeostasis <strong>in</strong> a mouse model of type 2 diabetes. <strong>Science</strong>2006;313:1137e40.[324] Xiao C, Giacca A, Lewis GF. Sodium phenylbutyrate, a drug with known capacity to reduce endoplasmicreticulum stress, partially alleviates lipid-<strong>in</strong>duced <strong>in</strong>sul<strong>in</strong> resistance and beta-cell dysfunction <strong>in</strong> humans.Diabetes 2011;60:918e24.[325] Hsu BR, Chen ST, Fu SH. Enhanc<strong>in</strong>g engraftment of islets us<strong>in</strong>g perioperative sodium 4-phenylbutyrate. IntImmunopharmacol 2006;6:1952e9.365


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>366[326] Zhou N, Moradei O, Raeppel S, Leit S, Frechette S, Gaudette F, et al. Discovery of N-(2-am<strong>in</strong>ophenyl)-4-[(4-pyrid<strong>in</strong>-3-ylpyrimid<strong>in</strong>-2-ylam<strong>in</strong>o)methyl]benzamide (MGCD0103), an orally active histone deacetylase<strong>in</strong>hibitor. J Med Chem 2008;51:4072e5.[327] Balasubramanian S, Ramos J, Luo W, Sirisawad M, Verner E, Buggy JJ. A novel histone deacetylase 8(HDAC8)-specific <strong>in</strong>hibitor PCI-34051 <strong>in</strong>duces apoptosis <strong>in</strong> T-cell lymphomas. Leukemia2008;22:1026e34.[328] Napper AD, Hixon J, McDonagh T, Keavey K, Pons JF, Barker J, et al. Discovery of <strong>in</strong>doles as potent andselective <strong>in</strong>hibitors of the deacetylase SIRT1. J Med Chem 2005;48:8045e54.[329] Cam<strong>in</strong>s A, Sureda FX, Junyent F, Verdaguer E, Folch J, Pelegri C, et al. Sirtu<strong>in</strong> activators: design<strong>in</strong>g moleculesto extend life span. Biochim Biophys Acta 2010;1799:740e9.[330] Miranda TB, Cortez CC, Yoo CB, Liang G, Abe M, Kelly TK, et al. DZNep is a global histone methylation <strong>in</strong>hibitorthat reactivates developmental genes not silenced by DNA methylation. Mol Cancer Ther 2009;8:1579e88.[331] Suzuki T, Miyata N. Lys<strong>in</strong>e Demethylases Inhibitors. J Med Chem 2011;54:8236e50.[332] Lizcano F, Romero C, Vargas D. Regulation of adipogenesis by nuclear receptor PPARgamma is modulated bythe histone demethylase JMJD2C. Genet Mol Biol 2011;34:19e24.[333] Huang J, Plass C, Gerhauser C. Cancer Chemoprevention by Target<strong>in</strong>g the Epigenome. Curr Drug Targets2010;12:1925e56.[334] L<strong>in</strong>k A, Balaguer F, Goel A. Cancer chemoprevention by dietary polyphenols: promis<strong>in</strong>g role for epigenetics.Biochem Pharmacol 2010;80:1771e92.[335] Meeran SM, Ahmed A, Tollefsbol TO. Epigenetic targets of bioactive dietary components for cancerprevention and therapy. Cl<strong>in</strong> <strong>Epigenetics</strong> 2010;1:101e16.[336] Spannhoff A, Kim YK, Raynal NJ, Gharibyan V, Su MB, Zhou YY, et al. Histone deacetylase <strong>in</strong>hibitor activity<strong>in</strong> royal jelly might facilitate caste switch<strong>in</strong>g <strong>in</strong> bees. EMBO Rep 2011;12:238e43.[337] Song MY, Kim EK, Moon WS, Park JW, Kim HJ, So HS, et al. Sulforaphane protects aga<strong>in</strong>st cytok<strong>in</strong>e- andstreptozotoc<strong>in</strong>-<strong>in</strong>duced beta-cell damage by suppress<strong>in</strong>g the NF-kappaB pathway. Toxicol Appl Pharmacol2009;235:57e67.[338] Solowiej E, Solowiej J, Godlewski M, Motyl T, Perkowska-Ptas<strong>in</strong>ska A, Jaskiewicz K, et al. Application ofsulforaphane: histopathological study of <strong>in</strong>traportal transplanted pancreatic islets <strong>in</strong>to livers of diabetic rats.Transplant Proc 2006;38:282e3.[339] Dancik V, Seiler KP, Young DW, Schreiber SL, Clemons PA. Dist<strong>in</strong>ct biological network properties betweenthe targets of natural products and disease genes. J Am Chem Soc 2010;132:9259e61.[340] Zanichelli F, Capasso S, Cipollaro M, Pagnotta E, Carteni M, Casale F, et al. Dose-dependent effects ofR-sulforaphane isothiocyanate on the biology of human mesenchymal stem cells, at dietary amounts, itpromotes cell proliferation and reduces senescence and apoptosis, while at anti-cancer drug doses, it hasa cytotoxic effect. Age (Dordr) 2012;34:281e93.[341] Lee SJ, Krauthauser C, Maduskuie V, Fawcett PT, Olson JM, Rajasekaran SA. Curcum<strong>in</strong>-<strong>in</strong>duced HDAC<strong>in</strong>hibition and attenuation of medulloblastoma growth <strong>in</strong> vitro and <strong>in</strong> vivo. BMC Cancer 2011;11:144.[342] Chen Y, Shu W, Chen W, Wu Q, Liu H, Cui G. Curcum<strong>in</strong>, both histone deacetylase and p300/CBP-specific<strong>in</strong>hibitor, represses the activity of nuclear factor kappa B and Notch 1 <strong>in</strong> Raji cells. Basic Cl<strong>in</strong> PharmacolToxicol 2007;101:427e33.[343] Balasubramanyam K, Varier RA, Altaf M, Swam<strong>in</strong>athan V, Siddappa NB, Ranga U, et al. Curcum<strong>in</strong>, a novelp300/CREB-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>-specific <strong>in</strong>hibitor of acetyltransferase, represses the acetylation of histone/nonhistone prote<strong>in</strong>s and histone acetyltransferase-dependent chromat<strong>in</strong> transcription. J Biol Chem2004;279:51163e71.[344] Marcu MG, Jung YJ, Lee S, Chung EJ, Lee MJ, Trepel J, et al. Curcum<strong>in</strong> is an <strong>in</strong>hibitor of p300 histoneacetylatransferase. Med Chem 2006;2:169e74.[345] Aggarwal BB. Target<strong>in</strong>g <strong>in</strong>flammation-<strong>in</strong>duced obesity and metabolic diseases by curcum<strong>in</strong> and othernutraceuticals. Annu Rev Nutr 2010;30:173e99.[346] Sr<strong>in</strong>ivasan M. Effect of curcum<strong>in</strong> on blood sugar as seen <strong>in</strong> a diabetic subject. Indian J Med Sci 1972;26:269e70.[347] Wickenberg J, Ingemansson SL, Hlebowicz J. Effects of Curcuma longa (turmeric) on postprandial plasmaglucose and <strong>in</strong>sul<strong>in</strong> <strong>in</strong> healthy subjects. Nutr J 9:43[348] Cuomo J, Append<strong>in</strong>o G, Dern AS, Schneider E, McK<strong>in</strong>non TP, Brown MJ, et al. Comparative absorption ofa standardized curcum<strong>in</strong>oid mixture and its lecith<strong>in</strong> formulation. J Nat Prod 2011;74:664e9.[349] Nair HB, Sung B, Yadav VR, Kannappan R, Chaturvedi MM, Aggarwal BB. Delivery of anti<strong>in</strong>flammatorynutraceuticals by nanoparticles for the prevention and treatment of cancer. Biochem Pharmacol2010;80:1833e43.[350] Beher D, Wu J, Cum<strong>in</strong>e S, Kim KW, Lu SC, Atangan L, et al. Resveratrol is not a direct activator of SIRT1enzyme activity. Chem Biol Drug Des 2009;74:619e24.


CHAPTER 17The Potential of Epigenetic Compounds <strong>in</strong> Treat<strong>in</strong>g Diabetes[351] Szkudelski T, Szkudelska K. Anti-diabetic effects of resveratrol. Ann N Y Acad Sci 2011;1215:34e9.[352] Czech MP, Aouadi M, Tesz GJ. RNAi-based therapeutic strategies for metabolic disease. Nat Rev Endocr<strong>in</strong>ol 2011.[353] Guay C, Roggli E, Nesca V, Jacovetti C, Regazzi R. Diabetes mellitus, a microRNA-related disease? Transl Res2011;157:253e64.[354] Kim N, Kim H, Jung I, Kim Y, Kim D, Han YM. Expression profiles of miRNAs <strong>in</strong> human embryonic stem cellsdur<strong>in</strong>g hepatocyte differentiation. Hepatol Res 2011;41:170e83.[355] Kroon E, Mart<strong>in</strong>son LA, Kadoya K, Bang AG, Kelly OG, Eliazer S, et al. Pancreatic endoderm derived fromhuman embryonic stem cells generates glucose-responsive <strong>in</strong>sul<strong>in</strong>-secret<strong>in</strong>g cells <strong>in</strong> vivo. Nat Biotechnol2008;26:443e52.[356] Wittmann J, Jack HM. Serum microRNAs as powerful cancer biomarkers. Biochim Biophys Acta2010;1806:200e7.[357] Mattes J, Yang M, Foster PS. Regulation of microRNA by antagomirs: a new class of pharmacologicalantagonists for the specific regulation of gene function? Am J Respir Cell Mol Biol 2007;36:8e12.367


This page <strong>in</strong>tentionally left blank


CHAPTER 18Epigenetic Aberrations <strong>in</strong><strong>Human</strong> Allergic <strong>Disease</strong>sManori Amarasekera 1 , David Mart<strong>in</strong>o 2 , Meri K. Tulic 1 , Richard Saffery 2 , Susan Prescott 11 University of Western Australia, Subiaco, WA, Australia2 Murdoch Children’s Research Institute, Parkville, VIC, AustraliaCHAPTER OUTLINE18.1 Introduction and Context: TheRis<strong>in</strong>g Prevalence of Allergic<strong>Disease</strong>s 36918.2 Mechanisms of AllergicResponse 37018.3 Fetal life: The Critical Period ofImmune Development 37118.4 Developmental Differences <strong>in</strong>Gene Expression <strong>in</strong> Allergic<strong>Disease</strong> 37118.5 Epigenetic Regulation ofImmune Development 37218.6 Factors that Modulate Allergic<strong>Disease</strong> Risk ThroughEpigenetic Mechanisms 37418.6.1 Epigenetic Effects ofBacterial Exposure onImmuneDevelopment 37418.6.2 Epigenetic Effects ofMaternal Diet on ImmuneFunction 37618.6.3 Epigenetic Effects ofTobacco Smoke 37718.6.4 Effects of Air Pollutantsand Other OutdoorPollutants on<strong>Epigenetics</strong> 37818.6.5 Other Maternal Factorsthat May ModulateAllergic Propensity <strong>in</strong> theNewborn ThroughEpigeneticMechanisms 37918.7 Conclusions 379References 38036918.1 INTRODUCTION AND CONTEXT: THE RISING PREVALENCEOF ALLERGIC DISEASESThe prevalence of allergic diseases such as asthma, allergic rh<strong>in</strong>itis, and eczema has risen at analarm<strong>in</strong>g rate over the past 4e5 decades [1,2]. This has been clearly associated with the markedenvironmental changes associated with transition to more modern lifestyles. Moreover, theparallel rise <strong>in</strong> a wide range of other immune diseases dur<strong>in</strong>g this short period providesadditional strong evidence that the immune system is highly susceptible to these environmentalchanges [3]. Furthermore, there is mount<strong>in</strong>g evidence that the effects of environmentalchange are potentially greatest dur<strong>in</strong>g critical periods of life, when epigenetic modifications <strong>in</strong>immune gene expression can alter subsequent disease susceptibility.T. Tollefsbol (Ed): <strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>. DOI: 10.1016/B978-0-12-388415-2.00018-4Copyright Ó 2012 Elsevier Inc. All rights reserved.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>The “allergy epidemic” was first evident <strong>in</strong> <strong>in</strong>dustrially developed countries <strong>in</strong>itially as a surgeof respiratory diseases such as asthma, ris<strong>in</strong>g at approximately 5% per year towards the newmillennium [1]. The burden of these disorders is enormous, with more than 40% of thepopulation <strong>in</strong> developed countries experienc<strong>in</strong>g allergic symptoms [4,5]. While the prevalenceof asthma and rh<strong>in</strong>itis may have reached a plateau, or may even have begun to decl<strong>in</strong>e <strong>in</strong> someregions [6e8], the global burden of these diseases cont<strong>in</strong>ues to rise as the prevalence ofrespiratory allergies <strong>in</strong>creases <strong>in</strong> develop<strong>in</strong>g countries as they undergo economic and lifestyletransition [6]. Of further concern, is an apparent “second wave” of allergic disease, manifest bya much more recent rise <strong>in</strong> food allergy, now loom<strong>in</strong>g as an epidemic <strong>in</strong> developed countries[9]. Food allergy was still uncommon at the time of the “first wave” of respiratory allergicdisease, only emerg<strong>in</strong>g as a significant problem <strong>in</strong> the last 10e15 years. The reason for thisearlier and more dramatic presentation of the allergic phenotype is not clear, but as thisappears l<strong>in</strong>ked with ongo<strong>in</strong>g environmental change, the same trends can also be anticipated <strong>in</strong>develop<strong>in</strong>g regions. This rise <strong>in</strong> disease burden is most evident <strong>in</strong> <strong>in</strong>fants and children under3 years of age, further highlight<strong>in</strong>g the likely role of early environmental exposures. Of yetfurther concern, there is also evidence of <strong>in</strong>creas<strong>in</strong>g disease persistence. Food allergies (such asegg and milk allergy) which were previously transient <strong>in</strong> nature, are now more likely to persist<strong>in</strong>to late childhood and adolescence [10]. Less common presentations of food allergy, forexample eos<strong>in</strong>ophilic eosophagitis, have also <strong>in</strong>creased <strong>in</strong> recently [11]. Collectively, thesechanges <strong>in</strong> disease patterns are plac<strong>in</strong>g grow<strong>in</strong>g demands on healthcare systems globally.370While environmental change may be driv<strong>in</strong>g the recent rise <strong>in</strong> disease, differences <strong>in</strong> susceptibilityand familial aggregation of allergic diseases also implies a genetic contribution to therisk of these diseases. Variations <strong>in</strong> genetic susceptibility cannot expla<strong>in</strong> the rise <strong>in</strong> disease, butthere was hope that identification of specific atopy/asthma genes could provide valuable<strong>in</strong>sight <strong>in</strong>to the causal pathways and disease pathogenesis. Although a large number ofcandidate genes have been associated with the asthma/allergy phenotype, the f<strong>in</strong>d<strong>in</strong>gs havebeen highly variable with poor reproducibility between populations [12,13]. A study carriedout <strong>in</strong> early 1990s reported that monozygotic tw<strong>in</strong>s showed a greater concordance thandizygotic tw<strong>in</strong>s, whether reared apart or together, for asthma and rh<strong>in</strong>itis, <strong>in</strong>dicat<strong>in</strong>g heritabilityas the major factor on expression of these disorders [14]. However, subsequent tw<strong>in</strong>studies, with higher levels of with<strong>in</strong>-pair discordance, revealed that environmental factors areequally or even more important <strong>in</strong> the development of disease [15,16]. This reflects thesignificant heterogeneity of these conditions that arise as a result of multiple and variablegenetic and environmental <strong>in</strong>fluences. It is important to elucidate how environmentalmodifiers confer changes to gene expression to fully understand the geneeenvironment<strong>in</strong>teractions.18.2 MECHANISMS OF ALLERGIC RESPONSEDifferentiation of naive CD4 þ T cells <strong>in</strong>to type 2 helper (Th2) cells to an “<strong>in</strong>nocuous”environmental antigen (allergen) is a hallmark of the allergic response, and produces thepropensity for IgE production (atopy) to that specific allergen. Once this pattern of response isestablished, subsequent exposure to the allergen leads to crossl<strong>in</strong>k<strong>in</strong>g of IgE on mast cells andan <strong>in</strong>flammatory cascade that culm<strong>in</strong>ates <strong>in</strong> the release of histam<strong>in</strong>e and other mediatorswhich produce the many signs and symptoms of allergic disease. The cl<strong>in</strong>ical pattern andseverity vary accord<strong>in</strong>g to the route of the allergen exposure, dose of the allergen, the pattern oftissues affected, and other <strong>in</strong>dividual factors. Although the culm<strong>in</strong>at<strong>in</strong>g events <strong>in</strong> the IgEcascade and the underly<strong>in</strong>g characteristics of the Th2 cellular response are well characterised[17] 17 , the factors <strong>in</strong>itiat<strong>in</strong>g and driv<strong>in</strong>g this process are less clear. Naive CD4 þ T cells have thecapacity to differentiate <strong>in</strong>to a range of effector cells or regulatory cells depend<strong>in</strong>g on the localmilieu at the time of allergen/antigen encounter (as discussed <strong>in</strong> more detail below). Forexample, the presence of <strong>in</strong>terleuk<strong>in</strong> 12 (IL-12) secreted from antigen-present<strong>in</strong>g cells (APC)


CHAPTER 18Epigenetic Aberrations <strong>in</strong> <strong>Human</strong> Allergic <strong>Disease</strong>spromotes differentiation <strong>in</strong>to Th1 subset. On the other hand, relative absence of IL-12 and thepresence of IL-4 <strong>in</strong> the local microenvironment promote Th2 differentiation over the Th1pathway. While the environmental changes favor<strong>in</strong>g Th2 differentiation are not fully understood,decl<strong>in</strong><strong>in</strong>g microbial exposure (a potent stimulant for APC <strong>in</strong>duced IL-12 production)has been a lead<strong>in</strong>g candidate. The range of other environmental factors which can also modifyIL-12 production or T-cell differentiation, <strong>in</strong>cludes smok<strong>in</strong>g [19], vitam<strong>in</strong> D [19], and antioxidants[20], which have all been implicated <strong>in</strong> the rise <strong>in</strong> allergic disease. Therefore, theprocesses regulat<strong>in</strong>g T-cell gene expression dur<strong>in</strong>g differentiation and maturation are of central<strong>in</strong>terest, particularly with emerg<strong>in</strong>g evidence that some of the environmental factors regulat<strong>in</strong>gthe differentiation process have epigenetic effects which could <strong>in</strong>fluence CD4 þ T-cell l<strong>in</strong>eagecommitment and subsequent allergic propensity, as further discussed below.18.3 FETAL LIFE: THE CRITICAL PERIOD OF IMMUNEDEVELOPMENTAs allergic disease is often first manifest <strong>in</strong> early childhood, it is clear that preced<strong>in</strong>g events <strong>in</strong>development are important. Early life therefore represents a critical period when geneeenvironmental <strong>in</strong>teractions play a determ<strong>in</strong><strong>in</strong>g role <strong>in</strong> specify<strong>in</strong>g immune tolerance. Cleardifferences <strong>in</strong> the neonatal immune function of children who subsequently develop allergystrongly suggests that these disorders have their orig<strong>in</strong>s <strong>in</strong> fetal life [21e26]. The DevelopmentalOrig<strong>in</strong>s of Health and <strong>Disease</strong> (DOHaD) hypothesis proposes that prenatal exposures have thepotential to modify a range of developmental processes <strong>in</strong> the fetus, and these exposures mayprogram susceptibility to many chronic diseases <strong>in</strong> later life [27,28]. There is some evidence thatthese changes <strong>in</strong> disease predisposition are the result of altered fetal gene expression <strong>in</strong>ducedthrough epigenetic changes. Although this has been best studied <strong>in</strong> the context of cardiovascularand metabolic diseases, epigenetic effects of environmental changes are also now be<strong>in</strong>g <strong>in</strong>vestigatedas a mechanism of the dramatic rise <strong>in</strong> allergic diseases [29e31].Complex immunological mechanisms have evolved to allow fetal and maternal immunesystems to coexist dur<strong>in</strong>g pregnancy. The maternal immune system adapts <strong>in</strong> a subtle way toa “Th2-state” <strong>in</strong> order to down-regulate Th1-mediated IFN-g responses to fetal antigens whichcan adversely affect the develop<strong>in</strong>g fetus [32e34]. Reflect<strong>in</strong>g this, neonatal cytok<strong>in</strong>e productionis dom<strong>in</strong>ated by Th2 cytok<strong>in</strong>es, with relative suppression of IFN-g production [35]. Thisdown-regulation of Th1 response <strong>in</strong> the fetal environment is generally attributed to theproduction of Th1-antagonistic mediators produced by the placenta; however there is alsoevidence of direct epigenetic control of gene transcription (further discussed below). RegulatoryT cells (Tregs) expand dur<strong>in</strong>g pregnancy and are recruited to the fetoematernal <strong>in</strong>terfacewhere they orchestrate immune tolerance towards the fetus [36] which may also be underepigenetic control. Together, these observations suggest a role for epigenetic regulation <strong>in</strong> theestablishment and ma<strong>in</strong>tenance of the fetal environment.371While the transition <strong>in</strong> early gene expression patterns from fetal to postnatal patterns isdevelopmentally regulated, environmental forces, such as microbial exposure (which isknown to promote Th1 and Treg differentiation), also appear to play a critical role <strong>in</strong> thesuccess of this process [37]. A better understand<strong>in</strong>g of these effects is important for develop<strong>in</strong>gstrategies to prevent or suppress the allergic phenotype.18.4 DEVELOPMENTAL DIFFERENCES IN GENE EXPRESSION INALLERGIC DISEASEMany presymptomatic differences of immune maturation pathways have been observedbetween allergic and non-allergic children. Of these, relative immaturity of neonatal Th1immune function has been one of the prom<strong>in</strong>ent antecedents of allergic disease [21,38].Although Th1 responses are generally suppressed at birth, this appears to be more marked <strong>in</strong>


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong><strong>in</strong>dividuals who develop subsequent allergic disease [21,38]. This is also coupled witha delayed postnatal maturation of Th1 immunity <strong>in</strong> high-risk children [22,26]. Differences <strong>in</strong><strong>in</strong>nate immunity [26] and Treg function [39,40] are also observed at birth between allergicand non-allergic children. These differences could reflect both genetic predisposition andenvironmental exposures <strong>in</strong> pregnancy at a time when the fetal immune system is potentiallymore vulnerable to epigenetic changes <strong>in</strong> gene expression, as discussed further below.18.5 EPIGENETIC REGULATION OF IMMUNE DEVELOPMENTDevelopment of the immune system, like all other systems/organs, is under epigenetic control.Changes <strong>in</strong> epigenetic profile have been observed <strong>in</strong> developmental maturation of T cells withage [41]. In the fetus, there is low-level methylation of CpG sites with<strong>in</strong> the promoter regionsof the IFNG and Th2 cytok<strong>in</strong>e loci (gene silenc<strong>in</strong>g) <strong>in</strong> naive CD4 þ T cells [42]. With age there isprogressive demethylation of IFNG which is accompanied by an <strong>in</strong>creased capacity of IFNgproduction by adult naive CD4 þ T cells.372The best evidence of epigenetic regulation of immune pathways has been observed for T-celldifferentiation [43e45]. Polarization of naive CD4 þ T cells by the cytok<strong>in</strong>e milieu ismediated through activation of signal<strong>in</strong>g pathways and transcription factors that are dist<strong>in</strong>ctfor Th1 and Th2 subsets. IL-4 activation of STAT-6 signal<strong>in</strong>g and the expression of thetranscription factor GATA-3 promotes Th2 differentiation. On the other hand, IL-12-<strong>in</strong>duced differentiation <strong>in</strong>to IFN-g produc<strong>in</strong>g Th1 cells is mediated through the activation ofSTAT-4 signal<strong>in</strong>g and the transcription factor T-bet. Regulation of these Th cell l<strong>in</strong>eages isgoverned by reciprocal <strong>in</strong>hibition; i.e. IL-4 activated GATA-3 <strong>in</strong>hibits Th1 cytok<strong>in</strong>e expression,while <strong>in</strong>hibition of Th2 cytok<strong>in</strong>e expression by T-bet ensures that once committednaive T helper cells generally differentiate <strong>in</strong> one dom<strong>in</strong>ant direction [46,47]. More recentlyanother transcription factor, Runt-related transcriptional factor 3 (RUNX3), was found to be<strong>in</strong>duced by T-bet, which together enhances expression of IFNG while repress<strong>in</strong>g IL4expression [48].In naive CD4 þ T cells, both IFNG and IL4 are methylated, result<strong>in</strong>g <strong>in</strong> chromat<strong>in</strong> remodel<strong>in</strong>gthat is transcriptionally non-permissive [43,45]. Dur<strong>in</strong>g Th1 l<strong>in</strong>eage commitment thepromoter region of the IFNG gene undergoes progressive demethylation (activation), whilethe IL4 is hypermethylated (silenced), thus <strong>in</strong>duc<strong>in</strong>g chromat<strong>in</strong> remodel<strong>in</strong>g compatible withIFNG but not IL4 expression. On the other hand, differentiation of naive CD4 þ cells to Th2cells <strong>in</strong>volves hypomethylation of the IL4 and concomitant silenc<strong>in</strong>g (through methylation) ofIFNG [42e44,46,49]. GATA-3-mediated chromat<strong>in</strong> remodel<strong>in</strong>g at the Th2 cytok<strong>in</strong>e loci (IL4/IL5/IL13/RAD50) is also essential for the Th2 l<strong>in</strong>eage commitment and ma<strong>in</strong>tenance of Th2phenotype through cell division [50]. Changes <strong>in</strong> histone modifications follow<strong>in</strong>g T-cellreceptor (TCR) signal<strong>in</strong>g appear to be <strong>in</strong>volved <strong>in</strong> this process. These modifications areorchestrated by site-specific enzymes <strong>in</strong>clud<strong>in</strong>g histone acetyltranferases (HATs) and histonedeacetylases (HDACs) [46]. Acetylation of histone by HAT is generally associated with geneexpression, whereas removal of acetyl group by HDAC is associated with more closed chromat<strong>in</strong>structure that makes the gene transcriptionally non-permissive. The importance ofendogenous HDAC activity on Th cell differentiation was demonstrated by a shift <strong>in</strong> recall(memory) responses toward a Th2 phenotype when HDAC activity was <strong>in</strong>hibited bytrichostat<strong>in</strong>-A (TSA). Bronchial biopsies from untreated asthmatics revealed a higher level ofHAT and lower level of HDAC activity [51]. Of note, treatment with <strong>in</strong>haled steroids wasassociated with a reverse <strong>in</strong> HAT/HDAC levels to that of controls, suggest<strong>in</strong>g therapeuticpathways also <strong>in</strong>volve epigenetic modulation.IL-2, another important cytok<strong>in</strong>e <strong>in</strong> ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g survival and proliferation of activated T cells,provides another example of the epigenetic regulation of immune pathways. In naive T cells,


CHAPTER 18Epigenetic Aberrations <strong>in</strong> <strong>Human</strong> Allergic <strong>Disease</strong>sthe IL2 promoter region is also methylated, undergo<strong>in</strong>g demethylation follow<strong>in</strong>g activation ofthe T cell with enhanc<strong>in</strong>g IL2 expression [52,53].Regulatory T-cell differentiation is also under epigenetic control [54,55]. Expression of theforkhead transcription factor 3 (FOXP3) is known to be critical for the development andregulatory functions of Tregs. The regulation of FOXP3 expression is not completely understood,however, recent f<strong>in</strong>d<strong>in</strong>gs <strong>in</strong>dicate a pivotal role for DNA methylation at both promoterand enhancer sequences [55]. In undifferentiated naive T cells, CpG sites with<strong>in</strong> a CpG islandassociated with the FOXP3 promoter are highly methylated and upon stimulation by TGF-b,these methylation marks are removed, ensur<strong>in</strong>g stable expression of FOXP3 and differentiation<strong>in</strong>to Treg cell l<strong>in</strong>eage.A more recently recognized dist<strong>in</strong>ct helper-T cell subset, the Th17 l<strong>in</strong>eage associated witha number of autoimmune diseases as well as with severe forms of allergic diseases [56e58]also appears to be regulated through changes <strong>in</strong> histone acetylation [59]. Although thedevelopmental role of this IL-17-produc<strong>in</strong>g l<strong>in</strong>eage is not clear, a recent study revealed thatTh17 cells can differentiate from Treg cells, and that HDAC <strong>in</strong>hibitor TSA had a profoundnegative effect on the emergence of these IL-17-produc<strong>in</strong>g cells [60].These observations have led to speculation that factors that <strong>in</strong>crease methylation (i.e. of IFNGand FOXP3) may <strong>in</strong>crease the risk of disease by silenc<strong>in</strong>g the Th1 and Treg pathways. Whilethis notion may appear simplistic, there are now a number of studies pursu<strong>in</strong>g this generalconcept that environmental changes can alter patterns of gene methylation.While there is solid evidence that epigenetic mach<strong>in</strong>ery regulates genes/pathways directlyl<strong>in</strong>ked to allergic response, epigenetic regulation of “master” transcription factors (such asNFkB) <strong>in</strong>directly controls a wider range of downstream immune and <strong>in</strong>flammatory responses[61,62]. For example, glucocorticoid receptor (GR) function is regulated by HDAC2 which issensitive to oxidative stress [63]. Reduced GR function up-regulates the NFkB activity [64]which <strong>in</strong> turn enhances the expression of <strong>in</strong>flammatory genes such as IL8 [63]. This highlightshow disease risk may be modulated at multiple levels.373In addition to histone modifications and DNA methylation, other important gene regulatorynetworks contribute to the control of gene expression, <strong>in</strong>clud<strong>in</strong>g microRNAs (miRNAs), small<strong>in</strong>terfer<strong>in</strong>g RNAs (siRNAs), and long non-cod<strong>in</strong>g RNAs (ncRNA). There is accumulat<strong>in</strong>gevidence that miRNAs are important for T-cell differentiation [65,66] and may be l<strong>in</strong>ked todevelopment of <strong>in</strong>flammatory disease [67,68]. S<strong>in</strong>ce miRNAs often target hundreds of genes[69], they may be more important epigenetic regulators that mediate environmental assaults<strong>in</strong> specific <strong>in</strong>flammatory pathways.Although early models of “l<strong>in</strong>eage commitment” proposed dist<strong>in</strong>ct term<strong>in</strong>ally differentiatedTh subsets, more recent evidence argues aga<strong>in</strong>st this more static view [70]. While CD4 þ cellsdifferentiate accord<strong>in</strong>g to their local cytok<strong>in</strong>e milieu dur<strong>in</strong>g stimulation, they reta<strong>in</strong> a degree ofcellular plasticity [70e72]. Epigenetic modifications are undoubtedly <strong>in</strong>volved <strong>in</strong> regulat<strong>in</strong>gthe switch that controls signature-cytok<strong>in</strong>e genes determ<strong>in</strong><strong>in</strong>g naive CD4 þ cell fates, but maybe equally important <strong>in</strong> contribut<strong>in</strong>g to the potential cellular flexibility.Now there is a grow<strong>in</strong>g body of data <strong>in</strong> support of epigenetic regulation of cellular plasticity ofTh cells [70]. While Foxp3 is essential for the ma<strong>in</strong>tenance of suppressive function <strong>in</strong> Treg cells,it has been shown that depletion of Foxp3 expression results <strong>in</strong> acquisition of “effector”functions by these cells with concomitant loss of regulatory properties [73,74]. These observationshave led to the speculation that “Th-effector” functions <strong>in</strong> Treg cells are cont<strong>in</strong>uouslyma<strong>in</strong>ta<strong>in</strong>ed <strong>in</strong> a dormant state by an active Foxp3-mediated mechanism. Recently, Beyer et al.reported that repression of “Th-effector” functions of Treg cells are mostly mediated by SATB1,a chromat<strong>in</strong> organizer and a transcription factor, and appear to be under direct transcriptionalcontrol of Foxp3 (at SATB1 locus) as well as <strong>in</strong>directly by Foxp3-dependent miRNA [75].


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>Furthermore, release of SATB1 from Foxp3 control was able to reprogram the Treg to ga<strong>in</strong>“effector” function while los<strong>in</strong>g its suppressive function. This shows that Treg populationrepresents a differentiated cell l<strong>in</strong>eage committed to a specific function but reta<strong>in</strong>s developmentalplasticity that may be mediated through epigenetic mechanisms.Challeng<strong>in</strong>g traditional models of epigenetic control of T-cell l<strong>in</strong>eage commitment, Wei et al.mapped active and repressive histone marks genome-wide across a spectrum of Th1, Th2,Th17, natural and <strong>in</strong>duced Treg phenotypes [76]. Modifications at signature cytok<strong>in</strong>e genesconformed to previous models of T-cell commitment, however, master transcription factors,such as the gene encod<strong>in</strong>g T-bet (TBX21) and GATA3, were found to exhibit a mixture of activeand repressive (bivalent) chromat<strong>in</strong> states across these phenotypes. It is speculated thatbivalent epigenetic marks <strong>in</strong> master regulators of the Th differentiation ma<strong>in</strong>ta<strong>in</strong> these transcriptionfactors at a “poised” state for expression <strong>in</strong> non-express<strong>in</strong>g cell l<strong>in</strong>eages and underappropriate conditions they can be <strong>in</strong>duced lead<strong>in</strong>g to an alternate cell fate [76,77]. This showsthat epigenetic mechanisms play a dual role <strong>in</strong> Th differentiation: ensure a “committed” stateof Th-cell response upon activation while conferr<strong>in</strong>g cellular plasticity.18.6 FACTORS THAT MODULATE ALLERGIC DISEASE RISKTHROUGH EPIGENETIC MECHANISMSThere is <strong>in</strong>tense <strong>in</strong>terest <strong>in</strong> the prenatal factors that may modify optimal patterns of immune geneactivation or silenc<strong>in</strong>g. A range of exposures already implicated <strong>in</strong> the rise <strong>in</strong> allergic disease havebeen shown to have potential epigenetic effects on fetal immune function, <strong>in</strong>clud<strong>in</strong>g microbialexposure, maternal diet, and pollutants [37,38] (Figure 18.1). Table 18.1 summarizes the environmentalfactors that modulate disease risk possibly through epigenetic modifications.37418.6.1 Epigenetic Effects of Bacterial Exposure on ImmuneDevelopmentDecl<strong>in</strong><strong>in</strong>g microbial exposure has long been implicated <strong>in</strong> the rise <strong>in</strong> allergic diseases [3],although the orig<strong>in</strong>al “hygiene hypothesis” was focused more on the allergy-protective effectsof <strong>in</strong>fectious exposures rather than more general microbial burden [79]. The <strong>in</strong>itial explanationsfor this protective effect centered around the ability of microbial components to upregulateIFN-g production and <strong>in</strong>hibit pro-allergic Th2 response [3]. This notion is supportedFIGURE 18.1Environmental <strong>in</strong>fluences on develop<strong>in</strong>g immune system:This illustrates the environmental factors that have beenshown to modify epigenetic profile and gene expressiondur<strong>in</strong>g early development. This figure is reproduced <strong>in</strong>the color plate section.


CHAPTER 18Epigenetic Aberrations <strong>in</strong> <strong>Human</strong> Allergic <strong>Disease</strong>sTABLE 18.1 Environmental Factors Known to be Associated with Epigenetic ModificationsEnvironmentalFactorEpigenetic ModificationEffect of Epigenetic Changes onAllergyReferencePrenatal exposure tomicrobial productsMaternal high-folatedietExposure to tobaccosmokePrenatal exposure totobacco smokeExposure to blackcarbon particlesPrenatal exposure toPAHExposure to dieselexhaust particlesPrenatal exposure toleadH4 acetylation of IFNG promoter <strong>in</strong>miceDemethylation of Treg-specificdemethylated region (TSDR) <strong>in</strong>neonatesMethylation changes <strong>in</strong> 82 gene loci<strong>in</strong>clud<strong>in</strong>g RUNX3 <strong>in</strong> miceSuppress HDAC activity and overallHDAC activity <strong>in</strong> alveolarmacrophages and bronchial biopsies<strong>in</strong> healthy smokersHypomethyalation of MAOBpromorter <strong>in</strong> circulat<strong>in</strong>g platelets andPBMCs <strong>in</strong> smokersGlobal DNA hypomethylation,hypermethylation of AXL andPTPRO, varied pattern of LINE1methylation pattern by child’s GSPT1haplotypeHypomethylation of LINE1 ofleukocyte DNA of elderly peopleHypermethylation of ACSL3 <strong>in</strong> cordblood mononuclear cellsHypermethylation of IFNG promoterand hypomethylation of IL4promorter <strong>in</strong> splenic CD4 þ cells <strong>in</strong>miceGlobal DNA hypomethylation <strong>in</strong>neonatesIncrease <strong>in</strong> IFNG expression [80]Induce FOXP3 expression [87]Increase <strong>in</strong> airwayhyperresponsiveness, airwayeos<strong>in</strong>ophilia, and production of<strong>in</strong>flammatory cytok<strong>in</strong>esIncrease <strong>in</strong> expression of<strong>in</strong>flammatory mediators GM-CSF,IL-8, and TNF-aReduced response to corticosteroidsSignificance to allergy is not yetknownRelevance to pathogenesis of allergicdisease is not yet knownRelevance to allergy has not yetidentifiedDirect relevance to allergic diseasepathogenesis has not been def<strong>in</strong>edIncrease <strong>in</strong> the production of IgEupon <strong>in</strong>tranasal adm<strong>in</strong>istration ofAspergillus fumigatesOutcome on allergic disease is notyet known[94][63][111][116][121][126][133][146]375by the impaired postnatal maturation of IFN-g pathways <strong>in</strong> allergic disease [22,26]. Furthermore,an array of microbial exposures (such as enteric flora, farm animals, and house dustendotox<strong>in</strong>) has been shown to both <strong>in</strong>crease early IFN-g production (Th1 response) anddecrease the risk of allergic disease. Given the fact that ultimate regulation of IFN-g expressionis l<strong>in</strong>ked to methylation/demethylation events <strong>in</strong> the IFNG promoter <strong>in</strong> CD4 þ T cells, it istempt<strong>in</strong>g to speculate that microbial exposures <strong>in</strong>duce demethylation/acetylation of IFNG <strong>in</strong>CD4 þ T cells [31]. Prelim<strong>in</strong>ary results from an animal model provide some evidence that this ispossible. Non-pathogenic microbial stra<strong>in</strong>s (Ac<strong>in</strong>etobacter lwoffi) isolated from farm<strong>in</strong>genvironments can <strong>in</strong>duce epigenetic effects when adm<strong>in</strong>istered to pregnant animals andprotect the offspr<strong>in</strong>g from experimental postnatal asthma [80]. This effect depends on<strong>in</strong>creased expression of IFN-g mediated by an <strong>in</strong>crease <strong>in</strong> H4 acetylation of the IFNGpromoter. Notably, these effects were abolished by <strong>in</strong>hibition of histone acetylation follow<strong>in</strong>ggarc<strong>in</strong>ol treatment. Even though there is no direct evidence for such association <strong>in</strong> humans,bacterial, viral, and parasitic agents have been shown to hold the capacity of <strong>in</strong>duc<strong>in</strong>gmethylation events <strong>in</strong> host DNA [81e84]. This is an area for future research with clearimplications on therapeutic as well as preventive strategies for allergic diseases.While the postnatal microbial exposure has the most obvious implications for the develop<strong>in</strong>gimmune system, there is emerg<strong>in</strong>g evidence that effects of microbial exposure may beg<strong>in</strong> muchearlier with maternal microbial exposure show<strong>in</strong>g potential to modulate fetal immune


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>function. Both human and animal studies clearly demonstrate that <strong>in</strong> utero exposure to bothpathogenic and non-pathogenic microbial products can prevent allergic outcomes <strong>in</strong> theoffspr<strong>in</strong>g, <strong>in</strong>dependent of postnatal exposure [85e88]. This protective effect has been associatedwith enhanced neonatal Treg numbers and function [87] along with <strong>in</strong>creased IFNGexpression [80]. Interest<strong>in</strong>gly, exposure to microbial products <strong>in</strong> a farm<strong>in</strong>g environmentappeared to stimulate FOXP3 expression <strong>in</strong> neonates by demethylat<strong>in</strong>g an evolutionaryconserved element with<strong>in</strong> the FOXP3 locus, Treg-specific demethylated region (TSDR) [87].These f<strong>in</strong>d<strong>in</strong>gs together, are highly suggestive of microbial exposure dur<strong>in</strong>g pregnancy canmodify fetal immune responses through epigenetic mechanisms [81e83].37618.6.2 Epigenetic Effects of Maternal Diet on Immune FunctionModern diets differ <strong>in</strong> many aspects from more traditional diets with more processed andsynthetic foods and less fresh fish, fruits, and vegetables. Of immediate relevance here, thisdietary pattern <strong>in</strong> pregnancy appears to provide less tolerogenic conditions dur<strong>in</strong>g earlyimmune development promot<strong>in</strong>g allergic outcomes <strong>in</strong> the offspr<strong>in</strong>g [89]. The specific nutritionalchanges implicated <strong>in</strong> the ris<strong>in</strong>g prevalence of asthma and other allergic diseases <strong>in</strong>cludesa decl<strong>in</strong>e <strong>in</strong> consumption of polyunsaturated fatty acids (PUFA) [90], soluble fiber [91], antioxidants,and other vitam<strong>in</strong>s [92,93]. Indeed, diet and nutrition <strong>in</strong> pregnancy have beena dom<strong>in</strong>ant basis for notions of the “developmental orig<strong>in</strong>s” of many diseases [28]. The firstevidence that maternal dietary changes <strong>in</strong> pregnancy can alter immune function and allergicoutcomes through epigenetic modifications came from animal studies. A diet rich <strong>in</strong> methyldonors (folate) fed to pregnant mice <strong>in</strong>duced allergic airway disease and a Th2 phenotype to <strong>in</strong>the offspr<strong>in</strong>g (F1 generation) [94]. This folate-rich maternal diet <strong>in</strong>duced methylation changes<strong>in</strong> 82-gene loci <strong>in</strong> the offspr<strong>in</strong>g, result<strong>in</strong>g <strong>in</strong> <strong>in</strong>creased airway hyperresponsiveness, airwayeos<strong>in</strong>ophilia, and production of <strong>in</strong>flammatory cytok<strong>in</strong>es. This trait was then <strong>in</strong>herited to thesubsequent F2 generation, demonstrat<strong>in</strong>g the transgenerational effects of environmentalmodification. Among these genes, RUNX3, a gene known to regulate T-lymphocyte developmentand to suppress airways <strong>in</strong>flammation [95] was hypomethylated with concordant transcriptionalsilenc<strong>in</strong>g of this gene <strong>in</strong> the progeny [94]. While some human studies reported thatfolic acid supplementation dur<strong>in</strong>g pregnancy is associated with an <strong>in</strong>creased risk of asthma andrespiratory <strong>in</strong>fections <strong>in</strong> <strong>in</strong>fants [96], a recent Dutch study revealed no association betweenmaternal folic acid supplementation and allergic outcomes <strong>in</strong> neonates [97]. However, until thisis fully explored <strong>in</strong> human studies and the mechanistic pathways are clearly del<strong>in</strong>eated, it is notappropriate to change the current practice of folate fortification to prevent neural tube defects.The role of vitam<strong>in</strong> D as an immune-modulatory substance is currently under much debate.Epidemiological associations between vitam<strong>in</strong> D levels and allergic diseases rema<strong>in</strong> <strong>in</strong>conclusive.Vitam<strong>in</strong> D <strong>in</strong>take dur<strong>in</strong>g pregnancy has been associated with either <strong>in</strong>creased risk[98] or decreased risk [99,100] of allergic disease <strong>in</strong> <strong>in</strong>fants. At a cellular level, 1a,25-dihydroxyvitam<strong>in</strong> D 3 (active metabolite of vitam<strong>in</strong> D) also appears to have diverse actions onnuclear factor kappa B (NF-kB)-driven transcription of <strong>in</strong>flammatory genes [101,102]. However,it appears that HDAC activity is required for vitam<strong>in</strong> D-mediated NF-kB modulation [101].Supplementation of fish oil (n-3 PUFA) is associated with effects on immune function of theoffspr<strong>in</strong>g [90]. However, at this stage it is not clear whether it is related to epigenetic modulation.Similarly, antioxidants have the capacity to <strong>in</strong>duce T-cell tolerance [103] and enhancethe production of IL-12 by dendritic cells [104]. It can be postulated that through these effectsantioxidants can favor the development of Th1 cells while suppress<strong>in</strong>g the Th2 development.The effect of dietary antioxidants dur<strong>in</strong>g pregnancy on fetal immune development is limited[105]. Evidence that oxidative stressors can modify the disease risk through epigeneticmechanisms suggests a role for these pathways [106].Purified compounds isolated from garlic and broccoli have been reported to have epigeneticeffects [107]. These bioactive compounds are found to be associated with HDAC <strong>in</strong>hibitory


CHAPTER 18Epigenetic Aberrations <strong>in</strong> <strong>Human</strong> Allergic <strong>Disease</strong>sactivity <strong>in</strong> animal models and may be related to <strong>in</strong>creased cancer risk. Based on the immunemodulatory property of these extracts, these common dietary components may be an additionalsource of epigenome modifiers <strong>in</strong> allergy risk and warrant further study.18.6.3 Epigenetic Effects of Tobacco SmokeExposure to tobacco smoke represents a major risk factor for an array of diseases <strong>in</strong>clud<strong>in</strong>gasthma [108], chronic obstructive pulmonary disease (COPD) [109], and lung cancers [110].There is mount<strong>in</strong>g evidence that epigenetic modifications <strong>in</strong>duced by tobacco smoke areassociated with the development of these chronic diseases [78]. One possible epigenetic effectof tobacco smoke <strong>in</strong> the pathogenesis of respiratory <strong>in</strong>flammatory diseases is throughperturb<strong>in</strong>g the balance between the HAT/HDAC homeostasis of the airway immune cells.Bronchial biopsies and alveolar macrophages taken from healthy smokers and age-matchedhealthy non-smokers reveal that tobacco smoke suppresses the HDAC2 expression and overallHDAC activity and enhanced the expression of <strong>in</strong>flammatory mediators GM-CSF, IL-8, andTNF-a [63]. Of note, cigarette smok<strong>in</strong>g reduces the response to corticosteroids by decreas<strong>in</strong>gHDAC activity <strong>in</strong> key <strong>in</strong>flammatory cells such as alveolar macrophages, expla<strong>in</strong><strong>in</strong>g theattenuated response to steroid therapy <strong>in</strong> patients with COPD (which also has a strong l<strong>in</strong>kwith chronic exposure to tobacco smoke). The HDAC <strong>in</strong>hibitor TSA has been shown to reversethe pro<strong>in</strong>flammatory changes and glucocorticoid responsiveness <strong>in</strong> the macrophages [63],imply<strong>in</strong>g their usefulness as an adjuvant drug for the treatment of asthma.In addition to alter<strong>in</strong>g the HAT/HDAC balance, tobacco smoke can modulate the DNAmethylation status of regulatory regions <strong>in</strong> a number of genes. Direct evidence formodified DNA methylation of epigenetic tags comes from a report that smok<strong>in</strong>g leads tohypomethylation of monoam<strong>in</strong>e oxidase (MAO) type B promoter <strong>in</strong> circulat<strong>in</strong>g platelets[111]. Furthermore, the authors describe similar results for PBMC of smokers and implysmok<strong>in</strong>g-<strong>in</strong>duced MAO gene deregulation could have a more general impact than vasculareffects.377Exposure to cigarette smoke <strong>in</strong> pregnancy has many adverse effects on the fetus, <strong>in</strong>clud<strong>in</strong>geffects on lung function and asthma risk [112,113]. Smok<strong>in</strong>g <strong>in</strong> the last trimester has beenassociated with early onset of airway hyperreactivity (likely asthma) by the age of 1 year [114].Moreover, both maternal and grandmaternal smok<strong>in</strong>g dur<strong>in</strong>g pregnancy are associated with<strong>in</strong>creased risk of childhood asthma, suggest<strong>in</strong>g a persistent heritable effect [115]. Comparisonof DNA methylation pattern of buccal cells from children born accord<strong>in</strong>g to maternal smok<strong>in</strong>ghabits dur<strong>in</strong>g pregnancy, revealed that exposure to tobacco smoke was associated with globalDNA hypomethylation. Exposed children had significantly lower methylation of short <strong>in</strong>terspersednucleotide element AluYb8, a surrogate marker of global DNA methylation [116]. Inaddition, the study revealed that smok<strong>in</strong>g affects epigenetic marks <strong>in</strong> gene specific manner.Us<strong>in</strong>g a CpG loci screen, eight genes were found differentially methylated <strong>in</strong> exposed childrenas opposed to unexposed children. Two genes, AXL and PTPRO, were validated by pyrosequenc<strong>in</strong>gand showed significant hypermethylation <strong>in</strong> exposed children but their significance<strong>in</strong> relation to asthma pathogenesis rema<strong>in</strong>s unclear. Moreover, methylation status of the DNArepetitive element LINE1 was observed only <strong>in</strong> children with common GSTM1-null genotype,while methylation pattern <strong>in</strong> exposed children varied with a common GSPT1 haplotypeimply<strong>in</strong>g the genetic <strong>in</strong>fluence over the epigeneticeenvironmental <strong>in</strong>teractions. It is <strong>in</strong>terest<strong>in</strong>gthat maternal smok<strong>in</strong>g had dual effects on fetal methylation profile; global DNA hypomethylationand hypermethylation of some specific genes [116]. Global hypomethylationcould result from DNA damage caused by smoke-<strong>in</strong>duced oxidative stress to DNA that<strong>in</strong>terferes with the b<strong>in</strong>d<strong>in</strong>g of DNA methyltransferase (DNMT) prevent<strong>in</strong>g DNA methylation[117]. The effect of tobacco smoke exposure on methylation of specific genes could possibly bedue to de novo methylation <strong>in</strong> specific gene promoters, perhaps by <strong>in</strong>complete erasure dur<strong>in</strong>gmethylation reprogramm<strong>in</strong>g that occurs <strong>in</strong> the embryo after fertilization [118].


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>37818.6.4 Effects of Air Pollutants and Other Outdoor Pollutants on<strong>Epigenetics</strong>Airborne pollutants such as particulate matter (PM) and noxious gases <strong>in</strong>clud<strong>in</strong>g benzene havebeen shown to be associated with asthma and other respiratory diseases [119,120]. Theseagents cause exacerbation of asthma symptoms <strong>in</strong> affected <strong>in</strong>dividuals but a causative l<strong>in</strong>k toasthma has not been well def<strong>in</strong>ed. Ambient level of black carbon particles, a marker of trafficpollution, has been consistently associated with a variety of adverse health outcomes andexposure even for a short duration was associated with hypomethylation of LINE1 but not Alu<strong>in</strong> blood DNA samples taken from a cohort of elderly people [121]. Polycyclic aromatichydrocarbons (PAH) are one of the most widespread organic pollutants and also a majorcomponent of PM of urban aerosol. In addition to their presence <strong>in</strong> oil, coal, and tar depositsthey are also formed by the <strong>in</strong>complete combustion of carbon-conta<strong>in</strong><strong>in</strong>g fuels such as wood,coal, diesel, fat, tobacco, and <strong>in</strong>cense. Grilled, smoked, or barbecued meats also appear toconta<strong>in</strong> high levels of PAH [122,123]. In addition to its carc<strong>in</strong>ogenic properties [124], it hasbeen found not only to impair functions of airway cells and smooth muscle cells but alsodim<strong>in</strong>ish responsiveness to standard therapy given to asthmatics [125]. Recently, a novelepigenetic marker for PAH-associated asthma has been identified and cord blood mononuclearcells (CBMC) of children born to mothers who had been exposed to considerable levelof PAH showed hypermethylation of the acyl-CoA synthetase long-chian family member 3(ACSL3) promoter [126]. Furthermore, the exposure level was highly correlated with <strong>in</strong>creasedrisk of asthma symptoms <strong>in</strong> the offspr<strong>in</strong>g before age 5 years. ACSL3 genes are expressed <strong>in</strong>lungs and thymus tissue and encode key enzymes <strong>in</strong> fatty acid metabolism [127,128]. Giventhe fact that ACSL3 is located <strong>in</strong> 2q36.1 which is associated with regions of the asthmasusceptibility loci <strong>in</strong> specific populations [129,130], it is tempt<strong>in</strong>g to speculate that hypermethylationof this gene <strong>in</strong> lung tissues can potentially <strong>in</strong>fluence the fatty acid metabolism andphospholipid composition of the membranes and lung function. The direct relevance of thisf<strong>in</strong>d<strong>in</strong>g to asthma pathogenesis has not been def<strong>in</strong>ed but epidemiological studies haverevealed the alterations <strong>in</strong> fatty acid composition <strong>in</strong> the diet [131] and cord blood [132] areassociated with the <strong>in</strong>creased risk of asthma.Diesel exhaust particles (DEP), <strong>in</strong> addition to a source of PAH, give rise to an array of chemicalsdispersed <strong>in</strong> the air as ultraf<strong>in</strong>e particles. Exposure of mice to <strong>in</strong>haled DEP for 3 weeks canaugment the production of IgE upon <strong>in</strong>tranasal adm<strong>in</strong>istration of Aspergillus fumigates [133].Hypersensitization occurred through hypermethylation of IFNG promoter with concomitanthypomethylation of IL4 promoter <strong>in</strong> DNA from splenic CD4 þ cells. The effects of PM could bemediated <strong>in</strong> the airways through <strong>in</strong>duction of oxidative stress. Treatment of A549 cells(adenocarc<strong>in</strong>omic human alveolar basal epithelial cells) with either PM-10 or H 2 O 2 <strong>in</strong>creasedthe expression and release of IL-8, which <strong>in</strong>creased the HAT activity, hence remodeled the IL8promoter region [134] suggest<strong>in</strong>g PM exert their effects through chromat<strong>in</strong> remodel<strong>in</strong>g of thesusceptible genes.Benzene, toluene, xylene, and other volatile organic compounds like PM are associated withadverse health effects <strong>in</strong>clud<strong>in</strong>g asthma. Children exposed to benzene have an <strong>in</strong>creased risk ofasthma symptoms [135] and this could possibly be mediated through chang<strong>in</strong>g the DNAmethylation profile s<strong>in</strong>ce exposure to benzene <strong>in</strong>duced changes <strong>in</strong> DNA methylation <strong>in</strong>a global and gene-specific manner [136].Although less is known about the immune effects of other pollutants released to the environmentby agricultural and modern <strong>in</strong>dustrial processes, it has been shown that highly lipidsoluble substances such as polychlor<strong>in</strong>ated biphenyl compounds, organochlor<strong>in</strong>e pesticide,diox<strong>in</strong>s, and phthalates accumulate <strong>in</strong> human tissue with age [137] possibly throughcontam<strong>in</strong>ated water, food, and cloth<strong>in</strong>g. Of most concern, some of these products have beenmeasured <strong>in</strong> breast milk, cord blood, and placental tissue [138e140] emphasiz<strong>in</strong>g the possibleadverse outcome <strong>in</strong> early development and subsequent disease pathogenesis <strong>in</strong> offspr<strong>in</strong>g.


CHAPTER 18Epigenetic Aberrations <strong>in</strong> <strong>Human</strong> Allergic <strong>Disease</strong>sHigher levels of organic pollutants were associated with higher levels of cord blood IgEantibodies [141]. At higher levels these products can have immunosuppressive effects <strong>in</strong>humans [142], whereas at low levels some appear to selectively <strong>in</strong>hibit type 1 immuneresponses [143], lead<strong>in</strong>g to speculation that this could possibly favor allergic (type 2) immuneresponses. Many of these organic pollutants now have been associated with modified epigenetictags <strong>in</strong> humans, as evidenced by variations <strong>in</strong> global DNA hypomethylation patterns withpersistent low-dose exposure [144]. More evidence for epigenetic alterations <strong>in</strong>duced byexposure to organic pollutants comes from rodent studies [145]. Prenatal exposure to lead wasassociated with global DNA hypomethylation <strong>in</strong> a human study suggest<strong>in</strong>g epigenome of thedevelop<strong>in</strong>g fetus can be <strong>in</strong>fluenced by maternal cumulative lead burden [146]. This may<strong>in</strong>fluence long-term epigenetic programm<strong>in</strong>g and disease susceptibility throughout the lifecourse. The levels of many pollutants are decl<strong>in</strong><strong>in</strong>g <strong>in</strong> some regions as a result of restrictionsimposed on the use of pesticides and other toxic chemicals, and this is reflected <strong>in</strong> decl<strong>in</strong><strong>in</strong>glevels measured <strong>in</strong> adipose tissue [147]. Nevertheless, the effects of these factors should not beignored, as epigenetic effects may potentially reflect exposure of subsequent several generationsand this relationship may be obscured <strong>in</strong> cross-sectional epidemiological studies.18.6.5 Other Maternal Factors that May Modulate Allergic Propensity <strong>in</strong>the Newborn Through Epigenetic MechanismsIn the context of asthma and allergic diseases, the maternal phenotype appears to be a majorfactor determ<strong>in</strong><strong>in</strong>g the subsequent outcomes <strong>in</strong> the offspr<strong>in</strong>g. Maternal asthma and allergicstatus has a stronger effect than paternal allergy on both allergic diseases and Th1-IFNgproduction <strong>in</strong> the neonate [148]. Lower IFNg responses to HLA-DR-mismatched fetal antigenshave also been observed <strong>in</strong> allergic mothers compared to non-allergic mothers [149]. This mayaffect the cytok<strong>in</strong>e milieu at the fetoematernal <strong>in</strong>terface and could be a mechanistic l<strong>in</strong>k ofattenuated Th1 responses commonly observed <strong>in</strong> <strong>in</strong>fants born to atopic mothers [38]. Ris<strong>in</strong>grates of maternal allergy mean that the endogenous effects of the maternal allergic phenotypehave the potential to amplify the effects of a proallergic exogenous environment. The underly<strong>in</strong>gmechanisms, yet unclear, may <strong>in</strong>volve epigenetic modifications of the specific immune genes.379The hypothalamicepituitaryeadrenal (HPA) axis is a major component of the neuroendocr<strong>in</strong>esystem. This axis controls many body processes and plays a major role <strong>in</strong> controll<strong>in</strong>g stressresponses. The immune system and the HPA axis are closely l<strong>in</strong>ked, particularly through theeffects of glucocorticoids which connect these two critical systems. In pregnancy, the placentalimmune system is, at least <strong>in</strong> part, regulated by glucocorticoids. Under adverse (stressful)conditions HPA axis activation can <strong>in</strong>duce up-regulation of placental Th1 cytok<strong>in</strong>e production,result<strong>in</strong>g <strong>in</strong> poor fetal outcomes. In animal models, it has been shown that maternal stress canalter placental gene expression, <strong>in</strong> particular, genes <strong>in</strong>volved <strong>in</strong> DNA methylation and histonemodification and cell cycle regulation [150] and strongly suggest that maternal stress can <strong>in</strong>duceepigenetic effects on fetal immune function with implications for the subsequent risk of disease<strong>in</strong> childhood. This is still poorly understood and an important area for ongo<strong>in</strong>g research.18.7 CONCLUSIONSExposure to a plethora of environmental factors (microbial exposure, maternal diet andsmok<strong>in</strong>g) dur<strong>in</strong>g critical periods of early immune development, has the potential to modifythe fetal immune development and the risk of subsequent disease. Notions of plasticity <strong>in</strong> geneexpression that may be epigenetically modified by the early environment provide a new modelto understand the geneeenvironmental <strong>in</strong>teractions that contribute to the ris<strong>in</strong>g prevalence ofasthma, allergy, and other immune diseases. Of greatest significance, this epigenetic plasticitymay pave the way to develop novel early <strong>in</strong>terventions to curb the epidemic of immunedisease, ideally through primary prevention <strong>in</strong> early life.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>The discovery of epigenetics as a key mechanism modulat<strong>in</strong>g immune mach<strong>in</strong>ery hasprofoundly changed perspectives and research approaches to allergy disease. However, manyunanswered questions need to be addressed before these f<strong>in</strong>d<strong>in</strong>gs will be of any therapeuticvalue, <strong>in</strong>clud<strong>in</strong>g: can epigenetic profiles be used to accurately predict disease risk andsusceptibility to treatment at the <strong>in</strong>dividual and population levels? How long will epigeneticmemory last and can we reverse any events that occurred <strong>in</strong> early life at a later stage? Can weerase the epigenetic marks passed through generations by modulat<strong>in</strong>g the environment of thenext generations or with therapeutic <strong>in</strong>terventions?380References[1] Devereux G. The <strong>in</strong>crease <strong>in</strong> allergic disease: environment and susceptibility. Proceed<strong>in</strong>gs of a symposiumheld at the Royal Society of Ed<strong>in</strong>burgh, 4th June 2002. Cl<strong>in</strong> Exp Allergy 2003;33:394e406.[2] Downs SH, Marks GB, Sporik R, Belosouva EG, Car NG, Peat JK. Cont<strong>in</strong>ued <strong>in</strong>crease <strong>in</strong> the prevalence ofasthma and atopy. Arch Dis Child 2001;84:20e3.[3] Bach JF. The effect of <strong>in</strong>fections on susceptibility to autoimmune and allergic diseases. N Engl J Med2002;347:911e20.[4] Peat JK, van den Berg RH, Green WF, Mellis CM, Leeder SR, Woolcock AJ. Chang<strong>in</strong>g prevalence of asthma <strong>in</strong>Australian children. BMJ 1994;308:1591e6.[5] Hopper JL, Jenk<strong>in</strong>s MA, Carl<strong>in</strong> JB, Giles GG. Increase <strong>in</strong> the self-reported prevalence of asthma and hay fever<strong>in</strong> adults over the last generation: a matched parent-offspr<strong>in</strong>g study. Aust J Public Health 1995;19:120e4.[6] Pearce N, Ait-Khaled N, Beasley R, Mallol J, Keil U, Mitchell E, et al. Worldwide trends <strong>in</strong> the prevalence ofasthma symptoms: phase III of the International Study of Asthma and Allergies <strong>in</strong> Childhood (ISAAC).Thorax 2007;62:758e66.[7] Robertson CF, Roberts MF, Kappers JH. Asthma prevalence <strong>in</strong> Melbourne schoolchildren: have we reached thepeak? Med J Aust 2004;180:273e6.[8] Bjorksten B, Clayton T, Ellwood P, Stewart A, Strachan D. Worldwide time trends for symptoms of rh<strong>in</strong>itis andconjunctivitis: Phase III of the International Study of Asthma and Allergies <strong>in</strong> Childhood. Pediatr AllergyImmunol 2008;19:110e24.[9] Prescott S, Allen KJ. Food allergy: rid<strong>in</strong>g the second wave of the allergy epidemic. Pediatr Allergy Immunol2011;22:155e60.[10] Skripak JM, Matsui EC, Mudd K, Wood RA. The natural history of IgE-mediated cow’s milk allergy. J AllergyCl<strong>in</strong> Immunol 2007;120:1172e7.[11] Straumann A, Simon HU. Eos<strong>in</strong>ophilic esophagitis: escalat<strong>in</strong>g epidemiology? J Allergy Cl<strong>in</strong> Immunol2005;115:418e9.[12] Holloway JW, Yang IA, Holgate ST. Genetics of allergic disease. J Allergy Cl<strong>in</strong> Immunol 2010;125:S81e94.[13] Weiss ST, Raby BA, Rogers A. Asthma genetics and genomics 2009. Curr Op<strong>in</strong> Genet Dev 2009;19:279e82.[14] Hanson B, McGue M, Roitman-Johnson B, Segal NL, Bouchard Jr TJ, Blumenthal MN. Atopic disease andimmunoglobul<strong>in</strong> E <strong>in</strong> tw<strong>in</strong>s reared apart and together. Am J Hum Genet 1991;48:873e9.[15] Skadhauge LR, Christensen K, Kyvik KO, Sigsgaard T. Genetic and environmental <strong>in</strong>fluence on asthma:a population-based study of 11,688 Danish tw<strong>in</strong> pairs. Eur Respir J 1999;13:8e14.[16] Liu X, Zhang S, Tsai HJ, Hong X, Wang B, Fang Y, et al. Genetic and environmental contributions to allergensensitization <strong>in</strong> a Ch<strong>in</strong>ese tw<strong>in</strong> study. Cl<strong>in</strong> Exp Allergy 2009;39:991e8.[17] Holgate ST. The epidemic of allergy and asthma. Nature 1999;402:B2e4.[18] Noakes PS, Holt PG, Prescott SL. Maternal smok<strong>in</strong>g <strong>in</strong> pregnancy alters neonatal cytok<strong>in</strong>e responses. Allergy2003;58:1053e8.[19] D’Ambrosio D, Cippitelli M, Cocciolo MG, Mazzeo D, Di Lucia P, Lang R, et al. Inhibition of IL-12production by 1,25-dihydroxyvitam<strong>in</strong> D3. Involvement of NF-kappaB downregulation <strong>in</strong> transcriptionalrepression of the p40 gene. J Cl<strong>in</strong> Invest 1998;101:252e62.[20] Murata Y, Shimamura T, Hamuro J. The polarization of T(h)1/T(h)2 balance is dependent on the <strong>in</strong>tracellularthiol redox status of macrophages due to the dist<strong>in</strong>ctive cytok<strong>in</strong>e production. Int Immunol 2002;14:201e12.[21] Tang ML, Kemp AS, Thorburn J, Hill DJ. Reduced <strong>in</strong>terferon-gamma secretion <strong>in</strong> neonates and subsequentatopy. Lancet 1994;344:983e5.[22] Prescott SL, Macaubas C, Smallacombe T, Holt BJ, Sly PD, Holt PG. Development of allergen-specific T-cellmemory <strong>in</strong> atopic and normal children. Lancet 1999;353:196e200.


CHAPTER 18Epigenetic Aberrations <strong>in</strong> <strong>Human</strong> Allergic <strong>Disease</strong>s[23] Amoudruz P, Holmlund U, Malmstrom V, Trollmo C, Bremme K, Scheynius A, et al. Neonatal immune responsesto microbial stimuli: is there an <strong>in</strong>fluence of maternal allergy? J Allergy Cl<strong>in</strong> Immunol 2005;115:1304e10.[24] Schaub B, Campo M, He H, Perk<strong>in</strong>s D, Gillman MW, Gold DR, et al. Neonatal immune responses to TLR2stimulation: <strong>in</strong>fluence of maternal atopy on Foxp3 and IL-10 expression. Respir Res 2006;7:40.[25] Prescott SL, Noakes P, Chow BW, Breckler L, Thornton CA, Hollams EM, et al. Presymptomatic differences <strong>in</strong>Toll-like receptor function <strong>in</strong> <strong>in</strong>fants who have allergy. J Allergy Cl<strong>in</strong> Immunol 2008;122:391e9:399e1e5.[26] Tulic MK, Hodder M, Forsberg A, McCarthy S, Richman T, D’Vaz N, et al. Differences <strong>in</strong> <strong>in</strong>nate immunefunction between allergic and nonallergic children: new <strong>in</strong>sights <strong>in</strong>to immune ontogeny. J Allergy Cl<strong>in</strong>Immunol 2011;127:470e8:e1.[27] Barker DJ, Fall CH. Fetal and <strong>in</strong>fant orig<strong>in</strong>s of cardiovascular disease. Arch Dis Child 1993;68:797e9.[28] Waterland RA, Michels KB. Epigenetic epidemiology of the developmental orig<strong>in</strong>s hypothesis. Annu Rev Nutr2007;27:363e88.[29] Miller RL, Ho SM. Environmental epigenetics and asthma: current concepts and call for studies. Am J RespirCrit Care Med 2008;177:567e73.[30] Bousquet J, Jacot W, Yssel H, Vignola AM, Humbert M. Epigenetic <strong>in</strong>heritance of fetal genes <strong>in</strong> allergicasthma. Allergy 2004;59:138e47.[31] Vuillerm<strong>in</strong> PJ, Ponsonby AL, Saffery R, Tang ML, Ellis JA, Sly P, et al. Microbial exposure, <strong>in</strong>terferon gammagene demethylation <strong>in</strong> naive T-cells, and the risk of allergic disease. Allergy 2009;64:348e53.[32] Breckler LA, Hale J, Jung W, Westcott L, Dunstan JA, Thornton CA, et al. Modulation of <strong>in</strong> vivo and <strong>in</strong> vitrocytok<strong>in</strong>e production over the course of pregnancy <strong>in</strong> allergic and non-allergic mothers. Pediatr AllergyImmunol 2010;21:14e21.[33] Wegmann TG, L<strong>in</strong> H, Guilbert L, Mosmann TR. Bidirectional cytok<strong>in</strong>e <strong>in</strong>teractions <strong>in</strong> the maternal-fetalrelationship: is successful pregnancy a TH2 phenomenon? Immunol Today 1993;14:353e6.[34] Breckler LA, Hale J, Taylor A, Dunstan JA, Thornton CA, Prescott SL. Pregnancy IFN-gamma responses tofoetal alloantigens are altered by maternal allergy and gravidity status. Allergy 2008;63:1473e80.[35] Prescott SL, Macaubas C, Holt BJ, Smallacombe TB, Loh R, Sly PD, et al. Transplacental prim<strong>in</strong>g of the humanimmune system to environmental allergens: universal skew<strong>in</strong>g of <strong>in</strong>itial T cell responses toward the Th2cytok<strong>in</strong>e profile. J Immunol 1998;160:4730e7.[36] Schumacher A, Brachwitz N, Sohr S, Engeland K, Langwisch S, Dolaptchieva M, et al. <strong>Human</strong> chorionicgonadotrop<strong>in</strong> attracts regulatory T cells <strong>in</strong>to the fetal-maternal <strong>in</strong>terface dur<strong>in</strong>g early human pregnancy.J Immunol 2009;182:5488e97.[37] Mart<strong>in</strong>o DJ, Prescott SL. Silent mysteries: epigenetic paradigms could hold the key to conquer<strong>in</strong>g theepidemic of allergy and immune disease. Allergy 2010;65:7e15.[38] R<strong>in</strong>as U, Horneff G, Wahn V. Interferon-gamma production by cord-blood mononuclear cells is reduced <strong>in</strong>newborns with a family history of atopic disease and is <strong>in</strong>dependent from cord blood IgE-levels. PediatrAllergy Immunol 1993;4:60e4.[39] Schaub B, Liu J, Hoppler S, Haug S, Sattler C, Lluis A, et al. Impairment of T-regulatory cells <strong>in</strong> cord blood ofatopic mothers. J Allergy Cl<strong>in</strong> Immunol 2008;121:1491e9:1499e1e13.[40] Smith M, Tourigny MR, Noakes P, Thornton CA, Tulic MK, Prescott SL. Children with egg allergy haveevidence of reduced neonatal CD4(þ)CD25(þ)CD127(lo/-) regulatory T cell function. J Allergy Cl<strong>in</strong>Immunol 2008;121:1460e6:1466e1e7.[41] Mart<strong>in</strong>o DJ, Tulic MK, Gordon L, Hodder M, Richman T, Metcalfe J, et al. Evidence for age-related and<strong>in</strong>dividual-specific changes <strong>in</strong> DNA methylation profile of mononuclear cells dur<strong>in</strong>g early immune development<strong>in</strong> humans. <strong>Epigenetics</strong> 2011;6.[42] White GP, Watt PM, Holt BJ, Holt PG. Differential patterns of methylation of the IFN-gamma promoter atCpG and non-CpG sites underlie differences <strong>in</strong> IFN-gamma gene expression between human neonatal andadult CD45RO- T cells. J Immunol 2002;168:2820e7.[43] Fields PE, Kim ST, Flavell RA. Cutt<strong>in</strong>g edge: changes <strong>in</strong> histone acetylation at the IL-4 and IFN-gamma lociaccompany Th1/Th2 differentiation. J Immunol 2002;169:647e50.[44] Lee DU, Agarwal S, Rao A. Th2 l<strong>in</strong>eage commitment and efficient IL-4 production <strong>in</strong>volves extended demethylationof the IL-4 gene. Immunity 2002;16:649e60.[45] White GP, Hollams EM, Yerkovich ST, Bosco A, Holt BJ, Bassami MR, et al. CpG methylation patterns <strong>in</strong> theIFNgamma promoter <strong>in</strong> naive T cells: variations dur<strong>in</strong>g Th1 and Th2 differentiation and between atopics andnon-atopics. Pediatr Allergy Immunol 2006;17:557e64.[46] Sawalha AH. <strong>Epigenetics</strong> and T-cell immunity. Autoimmunity 2008;41:245e52.[47] Zhu J. Transcriptional regulation of Th2 cell differentiation. Immunol Cell Biol 2010;88:244e9.[48] Djuretic IM, Levanon D, Negreanu V, Groner Y, Rao A, Ansel KM. Transcription factors T-bet and Runx3cooperate to activate Ifng and silence Il4 <strong>in</strong> T helper type 1 cells. Nat Immunol 2007;8:145e53.381


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>382[49] Sh<strong>in</strong> HJ, Park HY, Jeong SJ, Park HW, Kim YK, Cho SH, et al. STAT4 expression <strong>in</strong> human T cells is regulatedby DNA methylation but not by promoter polymorphism. J Immunol 2005;175:7143e50.[50] Yamashita M, Ukai-Tadenuma M, Miyamoto T, Sugaya K, Hosokawa H, Hasegawa A, et al. Essential role ofGATA3 for the ma<strong>in</strong>tenance of type 2 helper T (Th2) cytok<strong>in</strong>e production and chromat<strong>in</strong> remodel<strong>in</strong>g at theTh2 cytok<strong>in</strong>e gene loci. J Biol Chem 2004;279:26983e90.[51] Ito K, Caramori G, Lim S, Oates T, Chung KF, Barnes PJ, et al. Expression and activity of histone deacetylases<strong>in</strong> human asthmatic airways. Am J Respir Crit Care Med 2002;166:392e6.[52] Bruniquel D, Schwartz RH. Selective, stable demethylation of the <strong>in</strong>terleuk<strong>in</strong>-2 gene enhances transcriptionby an active process. Nat Immunol 2003;4:235e40.[53] Bird A. Il2 transcription unleashed by active DNA demethylation. Nat Immunol 2003;4:208e9.[54] Polansky JK, Kretschmer K, Freyer J, Floess S, Garbe A, Baron U, et al. DNA methylation controls Foxp3 geneexpression. Eur J Immunol 2008;38:1654e63.[55] Janson PC, W<strong>in</strong>erdal ME, Marits P, Thorn M, Ohlsson R, W<strong>in</strong>qvist O. FOXP3 promoter demethylation revealsthe committed Treg population <strong>in</strong> humans. PLoS One 2008;3:e1612.[56] Hu Y, Shen F, Crell<strong>in</strong> NK, Ouyang W. The IL-17 pathway as a major therapeutic target <strong>in</strong> autoimmunediseases. Ann N Y Acad Sci 2011;1217:60e76.[57] Toh ML, Miossec P. The role of T cells <strong>in</strong> rheumatoid arthritis: new subsets and new targets. Curr Op<strong>in</strong>Rheumatol 2007;19:284e8.[58] Stock<strong>in</strong>ger B, Veldhoen M. Differentiation and function of Th17 T cells. Curr Op<strong>in</strong> Immunol 2007;19:281e6.[59] Akimzhanov AM, Yang XO, Dong C. Chromat<strong>in</strong> remodel<strong>in</strong>g of <strong>in</strong>terleuk<strong>in</strong>-17 (IL-17)-IL-17F cytok<strong>in</strong>e genelocus dur<strong>in</strong>g <strong>in</strong>flammatory helper T cell differentiation. J Biol Chem 2007;282:5969e72.[60] Koenen HJ, Smeets RL, V<strong>in</strong>k PM, van Rijssen E, Boots AM, Joosten I. <strong>Human</strong> CD25highFoxp3pos regulatoryT cells differentiate <strong>in</strong>to IL-17-produc<strong>in</strong>g cells. Blood 2008;112:2340e52.[61] Bonizzi G, Kar<strong>in</strong> M. The two NF-kappaB activation pathways and their role <strong>in</strong> <strong>in</strong>nate and adaptive immunity.Trends Immunol 2004;25:280e8.[62] Okamoto T. The epigenetic alteration of synovial cell gene expression <strong>in</strong> rheumatoid arthritis and the roles ofnuclear factor kappaB and Notch signal<strong>in</strong>g pathways. Mod Rheumatol 2005;15:79e86.[63] Ito K, Lim S, Caramori G, Chung KF, Barnes PJ, Adcock IM. Cigarette smok<strong>in</strong>g reduces histone deacetylase 2expression, enhances cytok<strong>in</strong>e expression, and <strong>in</strong>hibits glucocorticoid actions <strong>in</strong> alveolar macrophages.FASEB J 2001;15:1110e2.[64] De Bosscher K, Vanden Berghe W, Haegeman G. The <strong>in</strong>terplay between the glucocorticoid receptor and nuclearfactor-kappaB or activator prote<strong>in</strong>-1: molecular mechanisms for gene repression. Endocr Rev2003;24:488e522.[65] Lu TX, Hartner J, Lim EJ, Fabry V, M<strong>in</strong>gler MK, Cole ET, et al. MicroRNA-21 limits <strong>in</strong> vivo immune responsemediatedactivation of the IL-12/IFN-gamma pathway, Th1 polarization, and the severity of delayed-typehypersensitivity. J Immunol 2011;187:3362e73.[66] Ste<strong>in</strong>er DF, Thomas MF, Hu JK, Yang Z, Babiarz JE, Allen CD, et al. MicroRNA-29 regulates T-box transcriptionfactors and <strong>in</strong>terferon-gamma production <strong>in</strong> helper T cells. Immunity 2011;35:169e81.[67] Lu TX, Munitz A, Rothenberg ME. MicroRNA-21 is up-regulated <strong>in</strong> allergic airway <strong>in</strong>flammation and regulatesIL-12p35 expression. J Immunol 2009;182:4994e5002.[68] Tsitsiou E, Williams AE, Moschos SA, Patel K, Rossios C, Jiang X, et al. Transcriptome analysis shows activationof circulat<strong>in</strong>g CD8(þ) T cells <strong>in</strong> patients with severe asthma. J Allergy Cl<strong>in</strong> Immunol 2011.[69] Cannell IG, Kong YW, Bushell M. How do microRNAs regulate gene expression? Biochem Soc Trans2008;36:1224e31.[70] O’Shea JJ, Paul WE. Mechanisms underly<strong>in</strong>g l<strong>in</strong>eage commitment and plasticity of helper CD4þ T cells.<strong>Science</strong> 2010;327:1098e102.[71] Yang XO, Nurieva R, Mart<strong>in</strong>ez GJ, Kang HS, Chung Y, Pappu BP, et al. Molecular antagonism and plasticity ofregulatory and <strong>in</strong>flammatory T cell programs. Immunity 2008;29:44e56.[72] Xu L, Kitani A, Fuss I, Strober W. Cutt<strong>in</strong>g edge: regulatory T cells <strong>in</strong>duce CD4þCD25-Foxp3- T cells or are self<strong>in</strong>ducedto become Th17 cells <strong>in</strong> the absence of exogenous TGF-beta. J Immunol 2007;178:6725e9.[73] Williams LM, Rudensky AY. Ma<strong>in</strong>tenance of the Foxp3-dependent developmental program <strong>in</strong> mature regulatoryT cells requires cont<strong>in</strong>ued expression of Foxp3. Nat Immunol 2007;8:277e84.[74] Lahl K, Mayer CT, Bopp T, Huehn J, Loddenkemper C, Eberl G, et al. Nonfunctional regulatory T cellsand defective control of Th2 cytok<strong>in</strong>e production <strong>in</strong> natural scurfy mutant mice. J Immunol 2009;183:5662e72.[75] Beyer M, Thabet Y, Muller RU, Sadlon T, Classen S, Lahl K, et al. Repression of the genome organizer SATB1 <strong>in</strong>regulatory T cells is required for suppressive function and <strong>in</strong>hibition of effector differentiation. Nat Immunol2011;12:898e907.


CHAPTER 18Epigenetic Aberrations <strong>in</strong> <strong>Human</strong> Allergic <strong>Disease</strong>s[76] Wei G, Wei L, Zhu J, Zang C, Hu-Li J, Yao Z, et al. Global mapp<strong>in</strong>g of H3K4me3 and H3K27me3 reveals specificityand plasticity <strong>in</strong> l<strong>in</strong>eage fate determ<strong>in</strong>ation of differentiat<strong>in</strong>g CD4þ T cells. Immunity 2009;30:155e67.[77] Northrup DL, Zhao K. Application of ChIP-Seq and related techniques to the study of immune function.Immunity 2011;34:830e42.[78] Ho SM. Environmental epigenetics of asthma: an update. J Allergy Cl<strong>in</strong> Immunol 2010;126:453e65.[79] Strachan DP. Hay fever, hygiene, and household size. BMJ 1989;299:1259e60.[80] Brand S, Teich R, Dicke T, Harb H, Yildirim AO, Tost J, et al. Epigenetic regulation <strong>in</strong> mur<strong>in</strong>e offspr<strong>in</strong>g asa novel mechanism for transmaternal asthma protection <strong>in</strong>duced by microbes. J Allergy Cl<strong>in</strong> Immunol2011;128:618e25:e1e7.[81] Bobetsis YA, Barros SP, L<strong>in</strong> DM, Weidman JR, Dol<strong>in</strong>oy DC, Jirtle RL, et al. Bacterial <strong>in</strong>fection promotes DNAhypermethylation. J Dent Res 2007;86:169e74.[82] Gutierrez MI, Siraj AK, Khaled H, Koon N, El-Rifai W, Bhatia K. CpG island methylation <strong>in</strong> Schistosoma- andnon-Schistosoma-associated bladder cancer. Mod Pathol 2004;17:1268e74.[83] Kitajima Y, Ohtaka K, Mitsuno M, Tanaka M, Sato S, Nakafusa Y, et al. Helicobacter pylori <strong>in</strong>fection is an<strong>in</strong>dependent risk factor for Runx3 methylation <strong>in</strong> gastric cancer. Oncol Rep 2008;19:197e202.[84] Foster SL, Hargreaves DC, Medzhitov R. Gene-specific control of <strong>in</strong>flammation by TLR-<strong>in</strong>duced chromat<strong>in</strong>modifications. Nature 2007;447:972e8.[85] Blumer N, Herz U, Wegmann M, Renz H. Prenatal lipopolysaccharide-exposure prevents allergic sensitizationand airway <strong>in</strong>flammation, but not airway responsiveness <strong>in</strong> a mur<strong>in</strong>e model of experimental asthma. Cl<strong>in</strong>Exp Allergy 2005;35:397e402.[86] Blumer N, Sel S, Virna S, Patrascan CC, Zimmermann S, Herz U, et al. Per<strong>in</strong>atal maternal application ofLactobacillus rhamnosus GG suppresses allergic airway <strong>in</strong>flammation <strong>in</strong> mouse offspr<strong>in</strong>g. Cl<strong>in</strong> Exp Allergy2007;37:348e57.[87] Schaub B, Liu J, Hoppler S, Schleich I, Huehn J, Olek S, et al. Maternal farm exposure modulates neonatalimmune mechanisms through regulatory T cells. J Allergy Cl<strong>in</strong> Immunol 2009;123:774e82:e5.[88] Douwes J, Cheng S, Travier N, Cohet C, Nies<strong>in</strong>k A, McKenzie J, et al. Farm exposure <strong>in</strong> utero may protectaga<strong>in</strong>st asthma, hay fever and eczema. Eur Respir J 2008;32:603e11.[89] West CE, Videky DJ, Prescott SL. Role of diet <strong>in</strong> the development of immune tolerance <strong>in</strong> the context ofallergic disease. Curr Op<strong>in</strong> Pediatr 2010;22:635e41.[90] Dunstan JA, Mori TA, Barden A, Beil<strong>in</strong> LJ, Taylor AL, Holt PG, et al. Fish oil supplementation <strong>in</strong> pregnancymodifies neonatal allergen-specific immune responses and cl<strong>in</strong>ical outcomes <strong>in</strong> <strong>in</strong>fants at high risk of atopy:a randomized, controlled trial. J Allergy Cl<strong>in</strong> Immunol 2003;112:1178e84.[91] Maslowski KM, Vieira AT, Ng A, Kranich J, Sierro F, Yu D, et al. Regulation of <strong>in</strong>flammatory responses by gutmicrobiota and chemoattractant receptor GPR43. Nature 2009;461:1282e6.[92] Miller RL. Prenatal maternal diet affects asthma risk <strong>in</strong> offspr<strong>in</strong>g. J Cl<strong>in</strong> Invest 2008;118:3265e8.[93] Devereux G, Litonjua AA, Turner SW, Craig LC, McNeill G, Mart<strong>in</strong>dale S, et al. Maternal vitam<strong>in</strong> D <strong>in</strong>takedur<strong>in</strong>g pregnancy and early childhood wheez<strong>in</strong>g. Am J Cl<strong>in</strong> Nutr 2007;85:853e9.[94] Holl<strong>in</strong>gsworth JW, Maruoka S, Boon K, Garantziotis S, Li Z, Tomfohr J, et al. In utero supplementation withmethyl donors enhances allergic airway disease <strong>in</strong> mice. J Cl<strong>in</strong> Invest 2008;118:3462e9.[95] Fa<strong>in</strong>aru O, Shseyov D, Hantisteanu S, Groner Y. Accelerated chemok<strong>in</strong>e receptor 7-mediated dendritic cellmigration <strong>in</strong> Runx3 knockout mice and the spontaneous development of asthma-like disease. Proc Natl AcadSci USA 2005;102:10598e603.[96] Haberg SE, London SJ, Stigum H, Nafstad P, Nystad W. Folic acid supplements <strong>in</strong> pregnancy and earlychildhood respiratory health. Arch Dis Child 2009;94:180e4.[97] Magdelijns FJ, Mommers M, Penders J, Smits L, Thijs C. Folic Acid use <strong>in</strong> pregnancy and the development ofatopy, asthma, and lung function <strong>in</strong> childhood. Pediatrics 2011;128:e135e44.[98] Gale CR, Rob<strong>in</strong>son SM, Harvey NC, Javaid MK, Jiang B, Martyn CN, et al. Maternal vitam<strong>in</strong> D status dur<strong>in</strong>gpregnancy and child outcomes. Eur J Cl<strong>in</strong> Nutr 2008;62:68e77.[99] Miyake Y, Sasaki S, Tanaka K, Hirota Y. Dairy food, calcium and vitam<strong>in</strong> D <strong>in</strong>take <strong>in</strong> pregnancy, and wheezeand eczema <strong>in</strong> <strong>in</strong>fants. Eur Respir J 2010;35:1228e34.[100] Erkkola M, Kaila M, Nwaru BI, Kronberg-Kippila C, Ahonen S, Nevala<strong>in</strong>en J, et al. Maternal vitam<strong>in</strong> D <strong>in</strong>takedur<strong>in</strong>g pregnancy is <strong>in</strong>versely associated with asthma and allergic rh<strong>in</strong>itis <strong>in</strong> 5-year-old children. Cl<strong>in</strong> ExpAllergy 2009;39:875e82.[101] Tse AK, Zhu GY, Wan CK, Shen XL, Yu ZL, Fong WF. 1alpha,25-Dihydroxyvitam<strong>in</strong> D3 <strong>in</strong>hibits transcriptionalpotential of nuclear factor kappa B <strong>in</strong> breast cancer cells. Mol Immunol 2010;47:1728e38.[102] Riis JL, Johansen C, Gesser B, Moller K, Larsen CG, Kragballe K, et al. 1alpha,25(OH)(2)D(3) regulatesNF-kappaB DNA b<strong>in</strong>d<strong>in</strong>g activity <strong>in</strong> cultured normal human kerat<strong>in</strong>ocytes through an <strong>in</strong>crease <strong>in</strong> Ikappa-Balpha expression. Arch Dermatol Res 2004;296:195e202.383


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>384[103] Tan PH, Sagoo P, Chan C, Yates JB, Campbell J, Beutelspacher SC, et al. Inhibition of NF-kappa B andoxidative pathways <strong>in</strong> human dendritic cells by antioxidative vitam<strong>in</strong>s generates regulatory T cells. J Immunol2005;174:7633e44.[104] Utsugi M, Dobashi K, Ishizuka T, Endou K, Hamuro J, Murata Y, et al. c-Jun N-term<strong>in</strong>al k<strong>in</strong>ase negativelyregulates lipopolysaccharide-<strong>in</strong>duced IL-12 production <strong>in</strong> human macrophages: role of mitogen-activatedprote<strong>in</strong> k<strong>in</strong>ase <strong>in</strong> glutathione redox regulation of IL-12 production. J Immunol 2003;171:628e35.[105] Devereux G, Barker RN, Seaton A. Antenatal determ<strong>in</strong>ants of neonatal immune responses to allergens. Cl<strong>in</strong>Exp Allergy 2002;32:43e50.[106] Rahman I, Marwick J, Kirkham P. Redox modulation of chromat<strong>in</strong> remodel<strong>in</strong>g: impact on histone acetylationand deacetylation, NF-kappaB and pro-<strong>in</strong>flammatory gene expression. Biochem Pharmacol 2004;68:1255e67.[107] Nian H, Delage B, Ho E, Dashwood RH. Modulation of histone deacetylase activity by dietary isothiocyanatesand allyl sulfides: studies with sulforaphane and garlic organosulfur compounds. Environ Mol Mutagen2009;50:213e21.[108] McLeish AC, Zvolensky MJ. Asthma and cigarette smok<strong>in</strong>g: a review of the empirical literature. J Asthma2010;47:345e61.[109] L<strong>in</strong> JL, Thomas PS. Current perspectives of oxidative stress and its measurement <strong>in</strong> chronic obstructivepulmonary disease. COPD 2010;7:291e306.[110] Papadopoulos A, Guida F, Cenee S, Cyr D, Schmaus A, Radoi L, et al. Cigarette smok<strong>in</strong>g and lung cancer <strong>in</strong>women: Results of the French ICARE case-control study. Lung Cancer 2011.[111] Launay JM, Del P<strong>in</strong>o M, Chironi G, Callebert J, Peoc’h K, Megnien JL, et al. Smok<strong>in</strong>g <strong>in</strong>duces long-last<strong>in</strong>geffects through a monoam<strong>in</strong>e-oxidase epigenetic regulation. PLoS One 2009;4:e7959.[112] Magnusson LL, Olesen AB, Wennborg H, Olsen J. Wheez<strong>in</strong>g, asthma, hayfever, and atopic eczema <strong>in</strong>childhood follow<strong>in</strong>g exposure to tobacco smoke <strong>in</strong> fetal life. Cl<strong>in</strong> Exp Allergy 2005;35:1550e6.[113] Bisgaard H, Loland L, Holst KK, Pipper CB. Prenatal determ<strong>in</strong>ants of neonatal lung function <strong>in</strong> high-risknewborns. J Allergy Cl<strong>in</strong> Immunol 2009;123:651e7:657e1e4.[114] Sears MR, Holdaway MD, Flannery EM, Herbison GP, Silva PA. Parental and neonatal risk factors for atopy,airway hyper-responsiveness, and asthma. Arch Dis Child 1996;75:392e8.[115] Li YF, Langholz B, Salam MT, Gilliland FD. Maternal and grandmaternal smok<strong>in</strong>g patterns are associated withearly childhood asthma. Chest 2005;127:1232e41.[116] Breton CV, Byun HM, Wenten M, Pan F, Yang A, Gilliland FD. Prenatal tobacco smoke exposure affects globaland gene-specific DNA methylation. Am J Respir Crit Care Med 2009;180:462e7.[117] Franco R, Schoneveld O, Georgakilas AG, Panayiotidis MI. Oxidative stress, DNA methylation and carc<strong>in</strong>ogenesis.Cancer Lett 2008;266:6e11.[118] Chong S, Whitelaw E. Epigenetic germl<strong>in</strong>e <strong>in</strong>heritance. Curr Op<strong>in</strong> Genet Dev 2004;14:692e6.[119] Peden DB, Bush RK. Advances <strong>in</strong> environmental and occupational respiratory diseases <strong>in</strong> 2009. J Allergy Cl<strong>in</strong>Immunol 2010;125:559e62.[120] D’Amato G, Cecchi L, D’Amato M, Liccardi G. Urban air pollution and climate change as environmental riskfactors of respiratory allergy: an update. J Investig Allergol Cl<strong>in</strong> Immunol 2010;20:95e102. quiz follow<strong>in</strong>g102.[121] Baccarelli A, Wright RO, Bollati V, Tarant<strong>in</strong>i L, Litonjua AA, Suh HH, et al. Rapid DNA methylation changesafter exposure to traffic particles. Am J Respir Crit Care Med 2009;179:572e8.[122] Liu G, Niu Z, Van Niekerk D, Xue J, Zheng L. Polycyclic aromatic hydrocarbons (PAHs) from coalcombustion: emissions, analysis, and toxicology. Rev Environ Contam Toxicol 2008;192:1e28.[123] Simko P. Factors affect<strong>in</strong>g elim<strong>in</strong>ation of polycyclic aromatic hydrocarbons from smoked meat foods andliquid smoke flavor<strong>in</strong>gs. Mol Nutr Food Res 2005;49:637e47.[124] Brody JG, Moysich KB, Humblet O, Attfield KR, Beehler GP, Rudel RA. Environmental pollutants and breastcancer: epidemiologic studies. Cancer 2007;109:2667e711.[125] Factor P, Akhmedov AT, McDonald JD, Qu A, Wu J, Jiang H, et al. Polycyclic Aromatic Hydrocarbons Impairb 2AR Function <strong>in</strong> Airway Epithelial and Smooth Muscle Cells. Am J Respir Cell Mol Biol 2011;45:1045e9.[126] Perera F, Tang WY, Herbstman J, Tang D, Lev<strong>in</strong> L, Miller R, et al. Relation of DNA methylation of 5’-CpGisland of ACSL3 to transplacental exposure to airborne polycyclic aromatic hydrocarbons and childhoodasthma. PLoS One 2009;4:e4488.[127] Mashek DG, Bornfeldt KE, Coleman RA, Berger J, Bernlohr DA, Black P, et al. Revised nomenclature for themammalian long-cha<strong>in</strong> acyl-CoA synthetase gene family. J Lipid Res 2004;45:1958e61.[128] M<strong>in</strong>ekura H, Kang MJ, Inagaki Y, Suzuki H, Sato H, Fuj<strong>in</strong>o T, et al. Genomic organization and transcriptionunits of the human acyl-CoA synthetase 3 gene. Gene 2001;278:185e92.[129] Bouzigon E, Siroux V, Dizier MH, Lema<strong>in</strong>que A, Pison C, Lathrop M, et al. Scores of asthma and asthmaseverity reveal new regions of l<strong>in</strong>kage <strong>in</strong> EGEA study families. Eur Respir J 2007;30:253e9.


CHAPTER 18Epigenetic Aberrations <strong>in</strong> <strong>Human</strong> Allergic <strong>Disease</strong>s[130] Choudhry S, Taub M, Mei R, Rodriguez-Santana J, Rodriguez-C<strong>in</strong>tron W, Shriver MD, et al. Genome-widescreen for asthma <strong>in</strong> Puerto Ricans: evidence for association with 5q23 region. Hum Genet2008;123:455e68.[131] Woods RK, Raven JM, Walters EH, Abramson MJ, Thien FC. Fatty acid levels and risk of asthma <strong>in</strong> youngadults. Thorax 2004;59:105e10.[132] Beck M, Zelczak G, Lentze MJ. Abnormal fatty acid composition <strong>in</strong> umbilical cord blood of <strong>in</strong>fants at highrisk of atopic disease. Acta Paediatr 2000;89:279e84.[133] Liu J, Ballaney M, Al-alem U, Quan C, J<strong>in</strong> X, Perera F, et al. Comb<strong>in</strong>ed <strong>in</strong>haled diesel exhaust particles andallergen exposure alter methylation of T helper genes and IgE production <strong>in</strong> vivo. Toxicol Sci2008;102:76e81.[134] Gilmour PS, Rahman I, Donaldson K, MacNee W. Histone acetylation regulates epithelial IL-8 releasemediated by oxidative stress from environmental particles. Am J Physiol Lung Cell Mol Physiol2003;284:L533e40.[135] Nicolai T, Carr D, Weiland SK, Duhme H, von Ehrenste<strong>in</strong> O, Wagner C, et al. Urban traffic and pollutantexposure related to respiratory outcomes and atopy <strong>in</strong> a large sample of children. Eur Respir J2003;21:956e63.[136] Bollati V, Baccarelli A, Hou L, Bonz<strong>in</strong>i M, Fust<strong>in</strong>oni S, Cavallo D, et al. Changes <strong>in</strong> DNA methylation patterns<strong>in</strong> subjects exposed to low-dose benzene. Cancer Res 2007;67:876e80.[137] Smeds A, Saukko P. Identification and quantification of polychlor<strong>in</strong>ated biphenyls and some endocr<strong>in</strong>edisrupt<strong>in</strong>g pesticides <strong>in</strong> human adipose tissue from F<strong>in</strong>land. Chemosphere 2001;44:1463e71.[138] Stacey CI, Perriman WS, Whitney S. Organochlor<strong>in</strong>e pesticide residue levels <strong>in</strong> human milk: WesternAustralia, 1979-1980. Arch Environ Health 1985;40:102e8.[139] Rha<strong>in</strong>ds M, Levallois P, Dewailly E, Ayotte P. Lead, mercury, and organochlor<strong>in</strong>e compound levels <strong>in</strong> cordblood <strong>in</strong> Quebec, Canada. Arch Environ Health 1999;54:40e7.[140] Ataniyazova OA, Baumann RA, Liem AK, Mukhopadhyay UA, Vogelaar EF, Boersma ER. Levels of certa<strong>in</strong>metals, organochlor<strong>in</strong>e pesticides and diox<strong>in</strong>s <strong>in</strong> cord blood, maternal blood, human milk and somecommonly used nutrients <strong>in</strong> the surround<strong>in</strong>gs of the Aral Sea (Karakalpakstan, Republic of Uzbekistan). ActaPaediatr 2001;90:801e8.[141] Reichrtova E, Ciznar P, Prachar V, Palkovicova L, Ven<strong>in</strong>gerova M. Cord serum immunoglobul<strong>in</strong> E related tothe environmental contam<strong>in</strong>ation of human placentas with organochlor<strong>in</strong>e compounds. Environ HealthPerspect 1999;107:895e9.[142] Daniel V, Huber W, Bauer K, Opelz G. Impaired <strong>in</strong>-vitro lymphocyte responses <strong>in</strong> patients with elevatedpentachlorophenol (PCP) blood levels. Arch Environ Health 1995;50:287e92.[143] Daniel V, Huber W, Bauer K, Suesal C, Conradt C, Opelz G. Associations of blood levels of PCB, HCHS, andHCB with numbers of lymphocyte subpopulations, <strong>in</strong> vitro lymphocyte response, plasma cytok<strong>in</strong>e levels,and immunoglobul<strong>in</strong> autoantibodies. Environ Health Perspect 2001;109:173e8.[144] Kim KY, Kim DS, Lee SK, Lee IK, Kang JH, Chang YS, et al. Association of low-dose exposure to persistentorganic pollutants with global DNA hypomethylation <strong>in</strong> healthy Koreans. Environ Health Perspect2010;118:370e4.[145] Herceg Z. <strong>Epigenetics</strong> and cancer: towards an evaluation of the impact of environmental and dietary factors.Mutagenesis 2007;22:91e103.[146] Pilsner JR, Hu H, Ett<strong>in</strong>ger A, Sanchez BN, Wright RO, Cantonw<strong>in</strong>e D, et al. Influence of prenatallead exposure on genomic methylation of cord blood DNA. Environ Health Perspect 2009;117:1466e71.[147] Noakes PS, Taylor P, Wilk<strong>in</strong>son S, Prescott SL. The relationship between persistent organic pollutants <strong>in</strong>maternal and neonatal tissues and immune responses to allergens: A novel exploratory study. Chemosphere2006;63:1304e11.[148] Prescott SL, Holt PG, Jenmalm M, Bjorksten B. Effects of maternal allergen-specific IgG <strong>in</strong> cord blood on earlypostnatal development of allergen-specific T-cell immunity. Allergy 2000;55:470e5.[149] Prescott SL, Breckler LA, Witt CS, Smith L, Dunstan JA, Christiansen FT. Allergic women show reduced Thelper type 1 alloresponses to fetal human leucocyte antigen mismatch dur<strong>in</strong>g pregnancy. Cl<strong>in</strong> Exp Immunol2010;159:65e72.[150] Gheorghe CP, Goyal R, Mittal A, Longo LD. Gene expression <strong>in</strong> the placenta: maternal stress and epigeneticresponses. Int J Dev Biol 2010;54:507e23.385


This page <strong>in</strong>tentionally left blank


CHAPTER 19Therapy of Airway<strong>Disease</strong>: EpigeneticPotentialPeter J. BarnesNational Heart & Lung Institute, Imperial College, London, UKCHAPTER OUTLINE19.1 Introduction 38719.2 Histone Acetylation andInflammatory GeneRegulation 38819.3 Acetylation of Non-histoneProte<strong>in</strong>s 38819.4 Corticosteroids SuppressInflammation via EpigeneticMechanisms 38919.5 Molecular Mechanisms ofCorticosteroidResistance 39019.6 Theophyll<strong>in</strong>e as an EpigeneticModulator 39019.7 Other Drugs 39119.8 Future Directions 391References 39238719.1 INTRODUCTIONBoth asthma and COPD <strong>in</strong>volve the <strong>in</strong>creased expression of multiple <strong>in</strong>flammatory genes[1e3]. There is <strong>in</strong>creas<strong>in</strong>g recognition that epigenetics may play an important role <strong>in</strong> theregulation of <strong>in</strong>flammatory genes <strong>in</strong> diseases. S<strong>in</strong>ce epigenetic changes can be longstand<strong>in</strong>gand may be passed to the offspr<strong>in</strong>g this is likely to be important <strong>in</strong> understand<strong>in</strong>g the chronicityof <strong>in</strong>flammation and how environmental factors which affect the mother (such ascigarette smok<strong>in</strong>g and diet) may affect the progeny. It has also been recognized that currentlyused treatments, such as corticosteroids, also work, at least <strong>in</strong> part, through epigeneticmechanisms. Understand<strong>in</strong>g these epigenetic pathways may identify novel targets for thedevelopment of future therapy [4e6]. Histone acetylation has been studied <strong>in</strong> some detail <strong>in</strong>relation to the expression of <strong>in</strong>flammatory genes [7]. Histones may also be <strong>in</strong>fluenced by otherpost-translational modifications, <strong>in</strong>clud<strong>in</strong>g phosphorylation through the action of variousk<strong>in</strong>ases, methylation via histone methyltransferases, tyros<strong>in</strong>e nitration, ubiquit<strong>in</strong>ation andSUMOylation (SUMO ¼ small ubiquit<strong>in</strong> modifier prote<strong>in</strong>), result<strong>in</strong>g <strong>in</strong> changes <strong>in</strong> geneexpression (Figure 19.1). Indeed these modifications may take place sequentially so that onemodification then makes it possible for the next occurr<strong>in</strong>g and this “histone code” mayaccount for cell specificity <strong>in</strong> <strong>in</strong>flammatory gene regulation [8]. For example, regulation of thegene encod<strong>in</strong>g transform<strong>in</strong>g growth factor (TGF)-b1 <strong>in</strong>volves successive acetylation andmethylation at different lys<strong>in</strong>e and arg<strong>in</strong><strong>in</strong>e residues on histones-3 and -4 [9]. Various exist<strong>in</strong>gT. Tollefsbol (Ed): <strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>. DOI: 10.1016/B978-0-12-388415-2.00019-6Copyright Ó 2012 Elsevier Inc. All rights reserved.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>FIGURE 19.1Post-translational histone modification. Histone-3 and -4 may be modified <strong>in</strong> several ways <strong>in</strong>clud<strong>in</strong>g acetylation (Ac) of lys<strong>in</strong>e(Lys) residues by histone acetyltransferases (HAT) and deacetylation by histone deacetylases (HDAC); phosphorylation (P) ofser<strong>in</strong>e (Ser) residues by k<strong>in</strong>ases and dephosphorylation by phosphatases; nitration (NO) of tyros<strong>in</strong>e (Tyr) residues byperoxynitrite and denitration by denitrases; ubiquit<strong>in</strong>ation (Ub) by E3 ubiquit<strong>in</strong> ligases and reversal by deubiquit<strong>in</strong>ases;SUMOylation (Su) of Lys residues; methylation (Me) of Lys and arg<strong>in</strong><strong>in</strong>e residues by histone methyltransferases (HMT) anddemethylases. Drugs have the potential to <strong>in</strong>teract with any of these modifications, particularly by effects on the signal<strong>in</strong>gpathways that regulate the modify<strong>in</strong>g enzymes. This figure is reproduced <strong>in</strong> the color plate section.drugs may <strong>in</strong>terfere with these histone modification signal<strong>in</strong>g pathways and better understand<strong>in</strong>gof these molecular mechanisms may identify novel molecular targets for discover<strong>in</strong>gnew anti-<strong>in</strong>flammatory therapies <strong>in</strong> the future.38819.2 HISTONE ACETYLATION AND INFLAMMATORY GENEREGULATIONGene expression is regulated by various coactivator molecules, such as CREB-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>(CBP; CREB ¼ cyclic AMP response element b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>) and p300, all of which have<strong>in</strong>tr<strong>in</strong>sic histone acetyltransferase (HAT) activity. Expression of <strong>in</strong>flammatory genes is regulatedby <strong>in</strong>creased acetylation of histone 4 [10]. In this way epigenetic factors play a critical role<strong>in</strong> chronic <strong>in</strong>flammation [11]. In asthma patients there is evidence for <strong>in</strong>creased acetylation ofhistone-4, consistent with <strong>in</strong>creased expression of multiple <strong>in</strong>flammatory genes [12]. Many ofthese genes are regulated by the transcription factor nuclear factor-kB (NF-kB), which may alsobe acetylated. In COPD peripheral lung, airway biopsies and alveolar macrophages there is an<strong>in</strong>crease <strong>in</strong> the acetylation of histones associated with the promoter region of <strong>in</strong>flammatorygenes, such as CXCL8 (<strong>in</strong>terleuk<strong>in</strong>-8) that are regulated by NF-kB and the degree of acetylation<strong>in</strong>creases with disease severity [13]. This <strong>in</strong>creased acetylation of histones associated with<strong>in</strong>flammatory genes is not due to an <strong>in</strong>crease <strong>in</strong> HAT activity <strong>in</strong> lungs or macrophages as <strong>in</strong>asthma, but due to decreased histone deacetylase (HDAC) activity.Histone acetylation is reversed by HDACs. There are 11 HDAC isoenzymes that deacetylatehistones and other prote<strong>in</strong>s with<strong>in</strong> the nucleus and specific HDACs appear to be differentiallyregulated and to regulate different groups of genes [14]. HDACs play a critical role <strong>in</strong> thesuppression of gene expression by revers<strong>in</strong>g the hyperacetylation of core histones. For theregulation of <strong>in</strong>flammatory genes HDAC2 appears to be of critical importance [10,15]. Theexpression of <strong>in</strong>flammatory genes is determ<strong>in</strong>ed by a balance between histone acetylation(which activates transcription) and deacetylation which switches off transcription.19.3 ACETYLATION OF NON-HISTONE PROTEINSIt has been <strong>in</strong>creas<strong>in</strong>gly recognized that many regulatory prote<strong>in</strong>s, particularly transcriptionfactors and nuclear receptors, are also regulated by acetylation that is controlled by HATs andHDACs and may also play a key role <strong>in</strong> the regulation of <strong>in</strong>flammatory genes [16]. Thepro<strong>in</strong>flammatory transcription factor NF-kB may be acetylated at several Lys residues on p65


CHAPTER 19Therapy of Airway <strong>Disease</strong>: Epigenetic Potentialand this appears to enhance its ability to activate certa<strong>in</strong> <strong>in</strong>flammatory genes, whereas other<strong>in</strong>flammatory genes may be less activated. There is a complex <strong>in</strong>terplay between acetylationand ubiquit<strong>in</strong>ation which leads to loss of p65 prote<strong>in</strong> [17]. Acetylation also plays a key role <strong>in</strong>the regulation of androgen and estrogen receptors, and this is also the case for glucocorticoidreceptors (GR) [15]. GR is acetylated with<strong>in</strong> the nucleus at specific lys<strong>in</strong>e residues close to theh<strong>in</strong>ge region of the receptor and only b<strong>in</strong>ds to its DNA-b<strong>in</strong>d<strong>in</strong>g site <strong>in</strong> its acetylated form.However, <strong>in</strong> order to <strong>in</strong>hibit NF-kB-activated genes it is necessary to deacetylate the receptorand this is achieved by HDAC2 (Figure 19.2).19.4 CORTICOSTEROIDS SUPPRESS INFLAMMATION VIAEPIGENETIC MECHANISMSThere have been major advances <strong>in</strong> understand<strong>in</strong>g the molecular mechanisms wherebyglucocorticoids suppress <strong>in</strong>flammation, which also provides important <strong>in</strong>sights <strong>in</strong>to howcorticosteroid-resistance may arise [18, 19]. A major mechanism of action of corticosteroids<strong>in</strong>volves changes <strong>in</strong> histone acetylation to regulate <strong>in</strong>flammatory and anti-<strong>in</strong>flammatory genes.Corticosteroids diffuse across the cell membrane and b<strong>in</strong>d to glucocorticoid receptors (GR) <strong>in</strong>the cytoplasm. Upon ligand b<strong>in</strong>d<strong>in</strong>g, GR are activated and released from chaperone prote<strong>in</strong>s(heat shock prote<strong>in</strong> 90 and others) and rapidly translocate to the nucleus where they exert theirmolecular effects through the activation and suppression of multiple genes <strong>in</strong>volved <strong>in</strong> the<strong>in</strong>flammatory process. GR homodimerize and b<strong>in</strong>d to glucocorticoid response elements(GRE) <strong>in</strong> the promoter region of glucocorticoid-responsive genes and this <strong>in</strong>teraction switcheson (or occasionally switches off) gene transcription. Genes that are switched on by glucocorticoids<strong>in</strong>clude genes encod<strong>in</strong>g b 2 -adrenergic receptors and the anti-<strong>in</strong>flammatory prote<strong>in</strong>ssecretory leukoprotease <strong>in</strong>hibitor and mitogen-activated prote<strong>in</strong> k<strong>in</strong>ase phosphatase-1 (MKP-1), which <strong>in</strong>hibits MAP k<strong>in</strong>ase pathways. However, the major action of corticosteroids is toswitch off multiple activated <strong>in</strong>flammatory genes that encode for cytok<strong>in</strong>es, chemok<strong>in</strong>es,adhesion molecules, <strong>in</strong>flammatory enzymes, and receptors, which are regulated by pro<strong>in</strong>flammatorytranscription factors, such as NF-kB and activator prote<strong>in</strong>-1 (AP-1). These transcriptionfactors activate <strong>in</strong>flammatory genes through histone acetylation, whereas activatedGR reverse this process by <strong>in</strong>teract<strong>in</strong>g with corepressor molecules to attenuate NF-kB-associatedcoactivator activity, thus reduc<strong>in</strong>g histone acetylation [10, 18]. Reduction of histone389FIGURE 19.2Acetylation of the glucocorticoid receptor (GR). After corticosteroid b<strong>in</strong>d<strong>in</strong>g to GR the receptor translocates to the nucleus whereit is acetylated by a histone acetyltransferase (HAT), which is necessary for GR to b<strong>in</strong>d to its glucocorticoid receptor recognitionelement (GRE) <strong>in</strong> the promoter region of steroid-sensitive genes, which <strong>in</strong>clude the genes that mediate the side effects ofcorticosteroids, such as osteocalc<strong>in</strong>. It is necessary for the acetylated GR to be deacetylated by histone deacetylase 2 (HDAC2)<strong>in</strong> order to <strong>in</strong>hibit activated nuclear factor-kB (NF-kB) to suppress activated <strong>in</strong>flammatory genes. This figure is reproduced <strong>in</strong>the color plate section.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>acetylation ma<strong>in</strong>ly occurs through the specific recruitment of HDAC2 to the activated<strong>in</strong>flammatory gene complex by activated GR, thereby result<strong>in</strong>g <strong>in</strong> effective suppression ofactivated <strong>in</strong>flammatory genes with<strong>in</strong> the nucleus. GR becomes acetylated upon ligand b<strong>in</strong>d<strong>in</strong>g,allow<strong>in</strong>g it to b<strong>in</strong>d to GREs and HDAC2 can target acetylated GR thereby allow<strong>in</strong>g it toassociate with the NF-kB complex [15] (Figure 19.2).19.5 MOLECULAR MECHANISMS OF CORTICOSTEROIDRESISTANCESeveral dist<strong>in</strong>ct molecular mechanisms contribut<strong>in</strong>g to decreased anti-<strong>in</strong>flammatory effects ofglucocorticoids have now been identified, so that there is heterogeneity of mechanisms evenwith<strong>in</strong> a s<strong>in</strong>gle disease [20]. However, similar molecular mechanisms have also been identified<strong>in</strong> different <strong>in</strong>flammatory diseases, <strong>in</strong>dicat<strong>in</strong>g that there may be common therapeuticapproaches to these diseases <strong>in</strong> the future [19]. Histone acetylation plays a critical role <strong>in</strong> theregulation of <strong>in</strong>flammatory genes and corticosteroids switch on corticosteroid-responsivegenes, such as MKP-1, via acetylation of specific lys<strong>in</strong>e residues (K5 and K16) on histone-4[21]. In a small proportion of patients with corticosteroid-resistant asthma, GR translocatesnormally to the nucleus after dexamethasone exposure but fails to acetylate K5 so thattransactivation of genes does not occur [22].390As discussed above, recruitment of HDAC2 to activated <strong>in</strong>flammatory genes is a majormechanism of <strong>in</strong>flammatory gene repression by corticosteroids and reduced HDAC2 activityand expression is reduced <strong>in</strong> some diseases where patients respond poorly. For example,HDAC2 is markedly reduced <strong>in</strong> alveolar macrophages, airways, and peripheral lung <strong>in</strong> patientswith COPD [13], and similar changes are found <strong>in</strong> peripheral blood mononuclear cells andalveolar macrophages of patients with refractory asthma [23] and <strong>in</strong> the airways of smok<strong>in</strong>gasthmatics. The corticosteroid resistance of COPD bronchoalveolar macrophages iscompletely reversed by overexpress<strong>in</strong>g HDAC2 (us<strong>in</strong>g a plasmid vector) to the level seen <strong>in</strong>control subjects [15]. The mechanisms for HDAC2 reduction <strong>in</strong> COPD are now be<strong>in</strong>g elucidated[24]. Oxidative and nitrative stress result <strong>in</strong> the formation of peroxynitrite, whichnitrates tyros<strong>in</strong>e residues on HDAC2 result<strong>in</strong>g <strong>in</strong> its <strong>in</strong>activation, ubiquit<strong>in</strong>ation, and degradation[25]. Oxidative stress also activates phospho<strong>in</strong>ositide-3-k<strong>in</strong>ase (PI3K)-d, which leads tophosphorylation and <strong>in</strong>activation of HDAC2 (Figure 19.3) [26]. This suggests that oxidativestress may be an important mechanism of corticosteroid resistance and is <strong>in</strong>creased <strong>in</strong> mostsevere and corticosteroid-resistant <strong>in</strong>flammatory diseases.19.6 THEOPHYLLINE AS AN EPIGENETIC MODULATORAn attractive therapeutic option is to reverse the cause of corticosteroid resistance by <strong>in</strong>hibit<strong>in</strong>gthe specific molecular pathways <strong>in</strong>volved. Theophyll<strong>in</strong>e is a drug used to treat asthma andCOPD for over 80 years and was previously used as a bronchodilator through <strong>in</strong>hibition ofphosphodiesterases <strong>in</strong> airway smooth muscle. However, the use of theophyll<strong>in</strong>e has decl<strong>in</strong>edrecently as side effects are common <strong>in</strong> the high doses needed for bronchodilatation and theseare also mediated by phosphodiesterase <strong>in</strong>hibition, as well as through adenos<strong>in</strong>e receptorantagonism. Recently, low concentrations of theophyll<strong>in</strong>e have been found to havea completely different pharmacological effect. Theophyll<strong>in</strong>e <strong>in</strong> low concentrations causesselective activation of HDAC2, and restores the reduced HDAC2 activity <strong>in</strong> COPD macrophagesback to normal thereby revers<strong>in</strong>g corticosteroid resistance [27,28]. In cigarette-smokeexposedmice, which develop corticosteroid-resistant <strong>in</strong>flammation, oral theophyll<strong>in</strong>e is alsoeffective <strong>in</strong> revers<strong>in</strong>g resistance [26]. A cl<strong>in</strong>ical study has demonstrated that low doses of oraltheophyll<strong>in</strong>e <strong>in</strong>crease the anti-<strong>in</strong>flammatory effects of an <strong>in</strong>haled corticosteroid <strong>in</strong> patientswith COPD [29]. In smok<strong>in</strong>g asthmatics, who are also corticosteroid-resistant, a low dose oftheophyll<strong>in</strong>e appears to be effective <strong>in</strong> revers<strong>in</strong>g resistance [30] and accelerates recovery of an


CHAPTER 19Therapy of Airway <strong>Disease</strong>: Epigenetic PotentialFIGURE 19.3Mechanisms for decreased histone deacetylase(HDAC)2 <strong>in</strong> COPD and its reversal. Superoxide anions ( . O 2 - ) and nitric oxide (NO)generated by cigarette smoke and <strong>in</strong>flammatory cells comb<strong>in</strong>e to form peroxynitrite. NO production from <strong>in</strong>flammatory cells isderived from <strong>in</strong>ducible NO synthase (iNOS) <strong>in</strong> response to <strong>in</strong>flammatory stimuli. Peroxynitrite nitrates HDAC2 at a tyros<strong>in</strong>e (Tyr)residue with<strong>in</strong> the catalytic site, which <strong>in</strong>activates HDAC2 and also leads to its ubiquit<strong>in</strong>ation (Ub) and proteasomal degradation.Oxidative stress also activates a phospho<strong>in</strong>ositide-3-k<strong>in</strong>ase (PI3K)-d pathway that phosphorylates (P) and <strong>in</strong>activates HDAC2.Loss of HDAC function then results <strong>in</strong> enhanced <strong>in</strong>flammatory gene expression and blocks the anti-<strong>in</strong>flammatory action ofcorticosteroids. HDAC2 function may be restored by antioxidants (<strong>in</strong>clud<strong>in</strong>g Nrf2 activators), iNOS <strong>in</strong>hibitors or peroxynitritescavengers, which reduce tyros<strong>in</strong>e nitration or by theophyll<strong>in</strong>e, nortriptyl<strong>in</strong>e, curcum<strong>in</strong>, or selective PI3K-d <strong>in</strong>hibitors whichrestore HDAC function to normal through <strong>in</strong>hibition of PI3Kd. HDAC2 mRNA may also be <strong>in</strong>creased by macrolides. This figure isreproduced <strong>in</strong> the color plate section.acute COPD exacerbation, an effect which is accompanied by <strong>in</strong>creased HDAC activity ionsputum macrophages and reduced <strong>in</strong>flammatory mediators [31].The molecular mechanism of action of theophyll<strong>in</strong>e <strong>in</strong> restor<strong>in</strong>g HDAC2 appears to be viaselective <strong>in</strong>hibition of PI3Kd, which is activated by oxidative stress <strong>in</strong> COPD patients [26, 32,33]. Selective PI3Kd <strong>in</strong>hibitors are similarly effective and these drugs are currently <strong>in</strong> cl<strong>in</strong>icaldevelopment for other diseases.39119.7 OTHER DRUGSThe tricyclic antidepressant nortriptyl<strong>in</strong>e also <strong>in</strong>creased HDAC2 by selectively <strong>in</strong>hibit<strong>in</strong>g PI3Kdand is an alternative therapeutic approach [34]. S<strong>in</strong>ce oxidative stress appears to me animportant mechanism <strong>in</strong> reduc<strong>in</strong>g HDAC2 and leads to corticosteroid resistance, antioxidantsshould also be effective and the Nrf2 activator sulforaphane, which <strong>in</strong>creases the expression ofendogenous antioxidant genes, is also very effective <strong>in</strong> revers<strong>in</strong>g corticosteroid resistance<strong>in</strong>duced by oxidative stress. The naturally occurr<strong>in</strong>g compound curcum<strong>in</strong> is also effective <strong>in</strong><strong>in</strong>creas<strong>in</strong>g HDAC activity and revers<strong>in</strong>g corticosteroid resistance [35]. In the future, noveldrugs which <strong>in</strong>crease HDAC2 may be developed when the molecular signal<strong>in</strong>g pathways thatregulate HDAC2 are better understood [36].It has long been recognized that macrolides have anti-<strong>in</strong>flammatory effects that may be<strong>in</strong>dependent of their antibiotic effects. Macrolides appear to <strong>in</strong>hibit <strong>in</strong>flammation by <strong>in</strong>hibit<strong>in</strong>gNF-kB and other transcription factors. A non-antibiotic macrolide (EM-703) reversescorticosteroid resistance due to oxidative stress by <strong>in</strong>creas<strong>in</strong>g HDAC2 activity [37]. Severalnon-antibiotic macrolides are now <strong>in</strong> development as anti-<strong>in</strong>flammatory therapies.19.8 FUTURE DIRECTIONSThe recognition that histone acetylation activates <strong>in</strong>flammatory genes and can be modified byanti-<strong>in</strong>flammatory therapies, such as corticosteroids, suggests that it may be possible to


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>identify novel targets that may lead to the development of novel therapies. Epigenetic mechanismsmay also account for the corticosteroid resistance of COPD, smok<strong>in</strong>g asthma, andsevere asthma, lead<strong>in</strong>g to the development of new treatments with the ability to reverse thiscorticosteroid resistance. This is already exemplified by the ability of low concentrations oftheophyll<strong>in</strong>e to reverse corticosteroid resistance <strong>in</strong> COPD cells <strong>in</strong> vitro, smok<strong>in</strong>g animals<strong>in</strong> vivo, and <strong>in</strong> patients with asthma and smok<strong>in</strong>g asthmatics. The molecular mechanism ofaction of theophyll<strong>in</strong>e appears to be through <strong>in</strong>hibition of PI3Kd and selective PI3Kd <strong>in</strong>hibitorsmimic the effects of theophyll<strong>in</strong>e. Several PI3Kd <strong>in</strong>hibitors are now <strong>in</strong> cl<strong>in</strong>ical development.Other drugs may also <strong>in</strong>teract with this pathway, open<strong>in</strong>g up the possibility of a newtherapeutic strategy for treat<strong>in</strong>g COPD and severe asthma.392While the role of histone acetylation and deacetylation <strong>in</strong> the regulation of <strong>in</strong>flammatorygenes has been explored, the role of other histone modifications has hardly been <strong>in</strong>vestigated.DNA and histone methylation is associated with gene repression, so may be <strong>in</strong>volved <strong>in</strong>switch<strong>in</strong>g off <strong>in</strong>flammatory genes. The methylation <strong>in</strong>hibitor 5-azacytid<strong>in</strong>e <strong>in</strong>creasesexpression of <strong>in</strong>flammatory genes <strong>in</strong> vitro and this is partially reversed by a corticosteroid,<strong>in</strong>dicat<strong>in</strong>g that methylation may be <strong>in</strong>volved [38]. Methylation of lys<strong>in</strong>e (K) and arg<strong>in</strong><strong>in</strong>eresidues on histones may occur as a result of histone methyltransferases. Histone methylationis complex as methylation of K4 on histone-3 is associated with gene activation, whereasmethylation of K9 is associated with gene repression. H3-K4 trimethylation is associated with<strong>in</strong>creased expression of the anti-<strong>in</strong>flammatory gene-secretory leukocyte protease <strong>in</strong>hibitorand this is <strong>in</strong>hibited by 5-azacytid<strong>in</strong>e [39]. Demethylase <strong>in</strong>hibitors may have anti<strong>in</strong>flammatorypotential as <strong>in</strong>hibitors of <strong>in</strong>flammatory gene expression. Several specifichistone lys<strong>in</strong>e demethylases have now been characterized, mak<strong>in</strong>g it possible to nowidentify selective <strong>in</strong>hibitors [40]. Similarly, the therapeutic potential of affect<strong>in</strong>g otherhistone modifications, such as phosphorylation, tyros<strong>in</strong>e nitration, ubiquit<strong>in</strong>ation, andSUMOylation, has hardly been explored.References[1] Barnes PJ. Pathophysiology of allergic <strong>in</strong>flammation. Immunol Rev 2011;242:31e50.[2] Barnes PJ. Immunology of asthma and chronic obstructive pulmonary disease. Nat Immunol Rev2008;8:183e92.[3] Barnes PJ, Shapiro SD, Pauwels RA. Chronic obstructive pulmonary disease: molecular and cellular mechanisms.Eur Respir J 2003;22:672e88.[4] Egger G, Liang G, Aparicio A, Jones PA. <strong>Epigenetics</strong> <strong>in</strong> human disease and prospects for epigenetic therapy.Nature 2004;429:457e63.[5] Schwartz DA. <strong>Epigenetics</strong> and environmental lung disease. Proc Am Thorac Soc 2010;7:123e5.[6] Barnes PJ. Target<strong>in</strong>g the epigenome <strong>in</strong> the treatment of asthma and chronic obstructive pulmonary disease.Proc Am Thorac Soc 2009;6:693e6.[7] Barnes PJ, Adcock IM, Ito K. Histone acetylation and deacetylation: importance <strong>in</strong> <strong>in</strong>flammatory lung diseases.Eur Respir J 2005;25:552e63.[8] Berger SL. The complex language of chromat<strong>in</strong> regulation dur<strong>in</strong>g transcription. Nature 2007;447:407e12.[9] Lee KY, Ito K, Hayashi R, Jazrawi EP, Barnes PJ, Adcock IM. NF-kB and activator prote<strong>in</strong> 1 response elementsand the role of histone modifications <strong>in</strong> IL-1b-Induced TGF-b1 gene transcription. J Immunol2006;176:603e15.[10] Ito K, Barnes PJ, Adcock IM. Glucocorticoid receptor recruitment of histone deacetylase 2 <strong>in</strong>hibits IL-1b<strong>in</strong>ducedhistone H4 acetylation on lys<strong>in</strong>es 8 and 12. Mol Cell Biol 2000;20:6891e903.[11] Adcock IM, Ford P, Barnes PJ, Ito K. <strong>Epigenetics</strong> and airways disease. Respir Res 2006;7:21.[12] Ito K, Caramori G, Lim S, Oates T, Chung KF, Barnes PJ, et al. Expression and activity of histone deacetylases(HDACs) <strong>in</strong> human asthmatic airways. Am J Respir Crit Care Med 2002;166:392e6.[13] Ito K, Ito M, Elliott WM, Cosio B, Caramori G, Kon OM, et al. Decreased histone deacetylase activity <strong>in</strong> chronicobstructive pulmonary disease. New Engl J Med 2005;352:1967e76.[14] Thiagal<strong>in</strong>gam S, Cheng KH, Lee HJ, M<strong>in</strong>eva N, Thiagal<strong>in</strong>gam A, Ponte JF. Histone deacetylases: unique players<strong>in</strong> shap<strong>in</strong>g the epigenetic histone code. Ann N Y Acad Sci 2003;983:84e100.


CHAPTER 19Therapy of Airway <strong>Disease</strong>: Epigenetic Potential[15] Ito K, Yamamura S, Essilfie-Quaye S, Cosio B, Ito M, Barnes PJ, et al. Histone deacetylase 2-mediated deacetylationof the glucocorticoid receptor enables NF-kB suppression. J Exp Med 2006;203:7e13.[16] Popov VM, Wang C, Shirley LA, Rosenberg A, Li S, Nevala<strong>in</strong>en M, et al. The functional significance of nuclearreceptor acetylation. Steroids 2007;72:221e30.[17] Li H, Wittwer T, Weber A, Schneider H, Moreno R, Ma<strong>in</strong>e GN, et al. Regulation of NF-kappaB activity bycompetition between RelA acetylation and ubiquit<strong>in</strong>ation. Oncogene 2011;10.[18] Barnes PJ. Glucocorticosteroids: current and future directions. Br J Pharmacol 2011;163:29e43.[19] Barnes PJ, Adcock IM. Glucocorticoid resistance <strong>in</strong> <strong>in</strong>flammatory diseases. Lancet 2009;342:1905e17.[20] Adcock IM, Barnes PJ. Molecular mechanisms of corticosteroid resistance. Chest 2008;134:394e401.[21] Barnes PJ. How corticosteroids control <strong>in</strong>flammation. Br J Pharmacol 2006;148:245e54.[22] Matthews JG, Ito K, Barnes PJ, Adcock IM. Defective glucocorticoid receptor nuclear translocation and alteredhistone acetylation patterns <strong>in</strong> glucocorticoid-resistant patients. J Allergy Cl<strong>in</strong> Immunol 2004;113:1100e8.[23] Hew M, Bhavsar P, Torrego A, Meah S, Khorasani N, Barnes PJ, et al. Relative corticosteroid <strong>in</strong>sensitivity ofperipheral blood mononuclear cells <strong>in</strong> severe asthma. Am J Respir Crit Care Med 2006;174:134e41.[24] Barnes PJ. Role of HDAC2 <strong>in</strong> the pathophysiology of COPD. Ann Rev Physiol 2009;71:451e64.[25] Osoata G, Adcock IM, Barnes PJ, Ito K. Oxidative stress causes HDAC2 reduction by nitration, ubiquitnylationand proteasomall degradation. Proc Amer Thorac Soc 2005;2:A755.[26] To Y, Ito K, Kizawa Y, Failla M, Ito M, Kusama T, et al. Target<strong>in</strong>g phospho<strong>in</strong>ositide-3-k<strong>in</strong>ase-d with theophyll<strong>in</strong>ereverses corticosteroid <strong>in</strong>sensitivity <strong>in</strong> COPD. Am J Resp Crit Care Med 2010;182:897e904.[27] Ito K, Lim S, Caramori G, Cosio B, Chung KF, Adcock IM, et al. A molecular mechanism of action oftheophyll<strong>in</strong>e: Induction of histone deacetylase activity to decrease <strong>in</strong>flammatory gene expression. Proc NatlAcad Sci USA 2002;99:8921e6.[28] Cosio BG, Tsaprouni L, Ito K, Jazrawi E, Adcock IM, Barnes PJ. Theophyll<strong>in</strong>e restores histone deacetylaseactivity and steroid responses <strong>in</strong> COPD macrophages. J Exp Med 2004;200:689e95.[29] Ford PA, Durham AL, Russell REK, Gordon F, Adcock IM, Barnes PJ. Treatment effects of low dose theophyll<strong>in</strong>ecomb<strong>in</strong>ed with an <strong>in</strong>haled corticosteroid <strong>in</strong> COPD. Chest 2010;137:1338e44.[30] Spears M, Donnelly I, Jolly L, Brannigan M, Ito K, McSharry C, et al. Effect of theophyll<strong>in</strong>e plus beclometasoneon lung function <strong>in</strong> smokers with asthma-a pilot study. Eur Respir J 2009;33:1010e7.[31] Cosio BG, Iglesias A, Rios A, Noguera A, Sala E, Ito K, et al. Low-dose theophyll<strong>in</strong>e enhances the anti<strong>in</strong>flammatoryeffects of steroids dur<strong>in</strong>g exacerbations of chronic obstructive pulmonary disease. Thorax2009;64:424e9.[32] Marwick JA, Caramori G, Stevenson CC, Casolari P, Jazrawi E, Barnes PJ, et al. Inhibition of PI3Kd restoresglucocorticoid function <strong>in</strong> smok<strong>in</strong>g-<strong>in</strong>duced airway <strong>in</strong>flammation <strong>in</strong> mice. Am J Respir Crit Care Med2009;179:542e8.[33] Marwick JA, Wallis G, Meja K, Kuster B, Bouwmeester T, Chakravarty P, et al. Oxidative stress modulatestheophyll<strong>in</strong>e effects on steroid responsiveness. Biochem Biophys Res Commun 2008;377:797e802.[34] Mercado N, To Y, Ito K, Barnes PJ. Nortriptyl<strong>in</strong>e reverses corticosteroid <strong>in</strong>sensitivity by <strong>in</strong>hibition of PI3K-d.J Pharmacol Exp Ther 2011;337:465e70.[35] Meja KK, Rajendrasozhan S, Adenuga D, Biswas SK, Sundar IK, Spooner G, et al. Curcum<strong>in</strong> Restores CorticosteroidFunction <strong>in</strong> Monocytes Exposed to Oxidants by Ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g HDAC2. Am J Respir Cell Mol Biol2008;39:312e23.[36] Barnes PJ. Histone deacetylase-2 and airway disease. Ther Adv Respir Dis 2009;3:235e43.[37] Charron C, Sumakuza T, Oomura S, Ito K. EM-703, a non-antibacterial erythromyc<strong>in</strong> derivative, restoresHDAC2 activition dim<strong>in</strong>ished by hypoxia and oxidative stress. Proc Am Thorac Soc 2007;175:A640.[38] Kagoshima M, Wilcke T, Ito K, Tsaprouni L, Barnes PJ, Punchard N, et al. Glucocorticoid-mediated transrepressionis regulated by histone acetylation and DNA methylation. Eur J Pharmacol 2001;429:327e34.[39] Wada H, Kagoshima M, Ito K, Barnes PJ, Adcock IM. 5-Azacytid<strong>in</strong>e suppresses RNA polymerase II recruitmentto the SLPI gene. Biochem Biophys Res Commun 2005;331:93e9.[40] Anand R, Marmorste<strong>in</strong> R. Structure and mechanism of lys<strong>in</strong>e-specific demethylase enzymes. J Biol Chem2007;282:35425e9.393


This page <strong>in</strong>tentionally left blank


CHAPTER 20The Role of <strong>Epigenetics</strong> <strong>in</strong>Cardiovascular <strong>Disease</strong>Boda Zhou 1,2 , Andriana Margariti 1 , Q<strong>in</strong>gbo Xu 11 K<strong>in</strong>g’s College London BHF Centre, London, UK2 Pek<strong>in</strong>g University, Beij<strong>in</strong>g, Ch<strong>in</strong>aCHAPTER OUTLINE20.1 Introduction 39520.1.1 DNA Methylation 39620.1.2 Histone Acetylation 39620.2 <strong>Epigenetics</strong> and ECHomeostasis 39820.2.1 DNA Methylation and ECProliferation 39820.2.2 Histone Acetylation and ECProliferation 39820.2.3 Histone Acetylation and ECMigration 39920.2.4 DNA Methylation and ECApoptosis 40020.2.5 Histone Acetylation and ECSurvival 40020.3 <strong>Epigenetics</strong> and SMCHomeostasis 40120.3.1 DNA Methylation,Atherosclerosis, and SMCPhenotype Switch 40120.3.2 Histone Acetylation and SMCProliferation 40220.3.3 Histone Acetylation and SMCMigration 40320.3.4 Histone Acetylation and SMCApoptosis 40320.3.5 DNA Methylation and SMCDifferentiation 40420.3.6 Histone Acetylation andSmooth Muscle Cell-specificGene Expression 40420.4 <strong>Epigenetics</strong> andAtherosclerosis 40420.4.1 DNA Methylation <strong>in</strong>Atherosclerosis 40420.4.2 Histone Acetylation <strong>in</strong>Arteriosclerosis 40520.5 <strong>Epigenetics</strong> and HeartFailure 40520.5.1 HAT <strong>in</strong> VentricularRemodel<strong>in</strong>g 40620.5.2 HDAC <strong>in</strong> CardiacHypertrophy 40720.6 Biomarker and MicroRNA 40820.7 Summary and FuturePerspectives 409Acknowledgments 410References 41039520.1 INTRODUCTIONCardiovascular diseases, such as atherosclerosis and heart failure, are major public healthproblems and lead<strong>in</strong>g causes of mortality <strong>in</strong> Western countries [1]. Abundant research hasbeen carried out to understand the factors associated with this disease. Although, there hasbeen a substantial decl<strong>in</strong>e <strong>in</strong> the disease mortality <strong>in</strong> Europe and <strong>in</strong> the US over the past 30years [1], this is ma<strong>in</strong>ly due to improvements <strong>in</strong> quality of care and to the prevention of thedisease itself. As a result, understand<strong>in</strong>g the mechanisms which lead to the development ofcardiovascular disease keeps an important promise for the future.T. Tollefsbol (Ed): <strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>. DOI: 10.1016/B978-0-12-388415-2.00020-2Copyright Ó 2012 Elsevier Inc. All rights reserved.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>Functions of endothelial (ECs) and smooth muscle cells (SMCs), such as proliferation,migration, and apoptosis have an <strong>in</strong>dispensable role <strong>in</strong> atherosclerosis. Although the detailedmechanism of atherosclerosis is unclear, it is generally believed to be a multistep <strong>in</strong>flammatorydisease [2,3]. The <strong>in</strong>itial step is endothelial dysfunction <strong>in</strong>volv<strong>in</strong>g EC proliferation, migration,and apoptosis, which leads to <strong>in</strong>creased endothelial permeability to lipoprote<strong>in</strong>s, <strong>in</strong>creasedleukocyte migration, and adhesion result<strong>in</strong>g <strong>in</strong> fatty streak formation [2]. In this process, themigration as well as proliferation of SMCs play critical role <strong>in</strong> the formation of fatty streak.Then, as fatty streaks progress to advanced lesions fibrous caps are formed [2]. At last, <strong>in</strong> mostpatients with myocardial <strong>in</strong>farction the f<strong>in</strong>al step is th<strong>in</strong>n<strong>in</strong>g and rupture of fibrous caps, <strong>in</strong>which the apoptosis of SMCs is considered as a possible cause [4]. Also, other cell types such asmacrophages have a crucial role <strong>in</strong> this process [2].Cardiomyocyte hypertrophy plays a part <strong>in</strong> the development of heart failure and leftventricular hypertrophy, which can be <strong>in</strong>duced by hypertension, while myocardial hypertrophyis <strong>in</strong>itially an adaptive response. However, after susta<strong>in</strong>ed external load hearts canchange <strong>in</strong>to a state of decompensated hypertrophy result<strong>in</strong>g <strong>in</strong> dilation of the left ventricle(remodel<strong>in</strong>g) and loss of contractile function. The molecular mechanisms responsible formyocardial remodel<strong>in</strong>g and transition from compensated to decompensated hypertrophy arepoorly def<strong>in</strong>ed, recent research showed the <strong>in</strong>volvement of epigenetic modulations [5].39620.1.1 DNA Methylation<strong>Epigenetics</strong> is the study of heritable alterations <strong>in</strong> phenotypes and gene expression that occurwithout chang<strong>in</strong>g the DNA sequence, provid<strong>in</strong>g a rapid and reversible regulation of therepertoire of expressed genes [6]. Epigenetic process <strong>in</strong>cludes DNA methylation as well ashistone tails modification [7]. Methylation and acetylation are two major forms of histonemodification. Histone methylation is modulated by two enzymes: histone methyltransferases(HMTs) and histone demethylases. The acetylation status of histone is f<strong>in</strong>e tuned by histoneacetyltransferases (HATs) and histone deacetylases (HDACs). The role of epigenetics <strong>in</strong> cancerand neurological diseases has been extensively exam<strong>in</strong>ed [8]. However, the functions of DNAmethylation and histone acetylation <strong>in</strong> cardiovascular disease are not clear yet. This chapterwill discuss the impact of epigenetics on atherosclerosis and heart failure.DNA methylation is a covalent modification that <strong>in</strong> mammals occurs predom<strong>in</strong>antly atcytos<strong>in</strong>es followed by guan<strong>in</strong>es (i.e. CpG d<strong>in</strong>ucleotides) to form 5-methylcytos<strong>in</strong>es. CpGmethylation generally is associated with gene silenc<strong>in</strong>g. DNA methylation is a form ofepigenetic gene regulation that together with altered b<strong>in</strong>d<strong>in</strong>g profile of transcription factorscommonly leads to suppression of gene expression [9]. DNA methylation is catalyzed by threedifferent DNA methyltransferases (DNMTs) encoded by different genes on dist<strong>in</strong>ct chromosomes:DNMT1, DNMT3a, and DNMT3b. De novo methylation is catalyzed by DNMT3a andDNMT3b [10]. In contrast, DNMT1 is responsible for the propagation of DNA methylationpatterns follow<strong>in</strong>g DNA replication dur<strong>in</strong>g mitotic cell division. Mechanisms responsible forthe removal of DNA methylation marks rema<strong>in</strong> poorly understood.20.1.2 Histone AcetylationHistone acetyltransferases (HAT) are enzymes that acetylate conserved lys<strong>in</strong>e am<strong>in</strong>o acids onhistone prote<strong>in</strong>s by transferr<strong>in</strong>g an acetyl group from acetyl CoA to form ε-N-acetyl lys<strong>in</strong>e.Typical HATs are CREB-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> (CBP) and p300, while <strong>in</strong> most cases histone acetylationis l<strong>in</strong>ked to transcriptional activation. HATs hyperacetylate histones that result <strong>in</strong> an<strong>in</strong>compact structure of the DNA, where transcription factors could potentially b<strong>in</strong>d andpromote gene expression. In contrast, HDACs act by remov<strong>in</strong>g the acetyl groups fromhyperacetylated histones and lead to a compact chromat<strong>in</strong> structure and suppression of genes.HATs and HDACs are recruited to gene promoters by DNA-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s that recognizecerta<strong>in</strong> DNA sequences, which results <strong>in</strong> modulation of specific gene expression [11].


CHAPTER 20The Role of <strong>Epigenetics</strong> <strong>in</strong> Cardiovascular <strong>Disease</strong>There are 18 characterized members of human HDACs [12], which can be grouped <strong>in</strong>to fourclasses based on function and DNA similarity (Table 20.1). The class I and class II HDACs areconsidered as the “classical” HDACs, whose activities could be <strong>in</strong>hibited by trichostat<strong>in</strong> A.Class III HDACs represent the silent <strong>in</strong>formation regulator 2 (Sir2) family of NAD þ -dependentHDACs (SIRT1-7) [12], which share structural and functional similarities with yeast Sir2prote<strong>in</strong>. Borradaile et al. [13] have summarized the role of Class III HDACs <strong>in</strong> cardiovasculardisease. F<strong>in</strong>ally, class IV HDAC is the recently discovered HDAC11. Although HDAC11 is mostclosely related to class I HDACs, it cannot be grouped <strong>in</strong>to any of the three exist<strong>in</strong>g classes dueto the low sequence similarities [14].Class I HDACs (HDAC1, 2, 3, 8) are widely expressed, importantly HDAC1, 2, and 8 residenearly exclusively <strong>in</strong> the nucleus, In contrast, HDAC3 is found to shuttle between nucleus andcytoplasm [12]. This <strong>in</strong>dicates that the different localization of HDACs may be related withtheir dist<strong>in</strong>ct function. The members of the class II HDACs (HDAC4, 5, 6, 7, 9, 10) are shownto be <strong>in</strong>volved <strong>in</strong> cell proliferation and cell survival [12]. In a similar manner Class II HDACscan shuttle between the nucleus and cytoplasm, which can be further divided <strong>in</strong>to subclassesIIa (HDAC4, 5, 7, 9) and IIb (HDAC6, 10). Class IIa HDACs dist<strong>in</strong>guish themselves by theirextended N-term<strong>in</strong>al regulatory doma<strong>in</strong> [15], whereas class IIb HDACs conta<strong>in</strong> two catalyticdoma<strong>in</strong>s [14]. Class II HDACs have been shown to possess limited HDAC activity comparedwith Class I HDACs, and they are found to form a complex with corepressors such as N-CoR(Nuclear receptor co-Repressor) and SMRT (Silenc<strong>in</strong>g Mediator for Ret<strong>in</strong>oid and Thyroidreceptors), as well as Class I HDACs such as HDAC3 [16,17].S<strong>in</strong>ce it is generally believed that HDACs could suppress gene expression only when they are <strong>in</strong>the nucleus, modulation of cellular localization is considered to be critical <strong>in</strong> def<strong>in</strong><strong>in</strong>g thefunctions of Class II HDACs. Timothy et al. [18] reported that calcium/calmodul<strong>in</strong>-dependentprote<strong>in</strong> k<strong>in</strong>ase (CaMK) signal<strong>in</strong>g plays a central role <strong>in</strong> regulat<strong>in</strong>g class II HDAC nuclearexport. They found that CaMK could phosphorylate class II HDACs and promote their nuclearexportation, thus provide derepression of HDAC-responsive genes. Ruijter et al. [19]summarized that after phosphorylation by CaMK, class II HDACs are exported to the cytoplasmwith the help of cellular export factor CRM-1. 14-3-3 prote<strong>in</strong>s (a cytoplasmic anchorprote<strong>in</strong> family) then b<strong>in</strong>d to and reta<strong>in</strong> phosphorylated class II HDACs <strong>in</strong> the cytoplasm.397As HDACs have broad functions <strong>in</strong> the cell, various HDAC <strong>in</strong>hibitors are designed target<strong>in</strong>g onthe catalytic sites of HDACs. They could be grouped <strong>in</strong>to four classes based on their structuraldiversity, e.g. hydroxamic acids, short-cha<strong>in</strong> fatty acids, cyclic tetrapeptides, and benzamides[20e43] (Table 20.2). Some <strong>in</strong>hibitors such as trichostat<strong>in</strong> A (TSA) can <strong>in</strong>hibit the HDACactivity of both class I and class II HDACs. However, specific <strong>in</strong>hibitors that could only <strong>in</strong>hibitthe activity of one specific HDAC have been developed. An example is tubac<strong>in</strong>, a smallTABLE 20.1 Classes of HDACsClass <strong>Human</strong> HDAC Subcellular LocationIHDAC1HDAC2HDAC3HDAC8NucleusNucleusNuc/CytNucleusIIaHDAC4Nuc/CytIIbHDAC5HDAC7HDAC9HDAC6HDAC10Nuc/CytNuc/CytNuc/CytMa<strong>in</strong>ly CytNuc/CytIII Sirt1- Sirt7 Nuc/CytIV HDAC11 Nuc/Cyt


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>TABLE 20.2 Structural Classes of HDAC InhibitorsClass Example ReferencesHydroxamic acids Trichostat<strong>in</strong> A(TSA)Suberoyl anilide bishydroxamide (SAHA)PyroxamideM-carboxyc<strong>in</strong>namic acid bishydroxamide (CBHA)Scriptaid[71,61][72,73][74][75,76][77,78]Short-cha<strong>in</strong> fatty acidsCyclic tetrapeptidesValproic acidPhenylbutyrateButyrateTrapox<strong>in</strong>Depsipeptide (FK228)Apicid<strong>in</strong>eChlamydoc<strong>in</strong>[79,80][81,82][83,84]Benzamides MS-275 [92][85,86][87,88][89,90][91]molecule that selectively <strong>in</strong>hibits HDAC6 activity [44]. More <strong>in</strong>formation about the selectiveHDAC <strong>in</strong>hibitors could be found <strong>in</strong> this review [45].20.2 EPIGENETICS AND EC HOMEOSTASISEC proliferation has a major <strong>in</strong>fluence on EC turnover. Reports have shown that high ECturnover correlates with high EC permeability [46,47]. Therefore, EC proliferation plays a corepart <strong>in</strong> regulat<strong>in</strong>g vascular homeostasis and atherosclerosis. DNA methylation and histoneacetylation have been shown to regulate EC proliferation.39820.2.1 DNA Methylation and EC ProliferationJamaludd<strong>in</strong> et al. [48] showed that the cycl<strong>in</strong> A promoter conta<strong>in</strong>s a CpG island for DNAmethyltransferase 1 (DNMT1). Homocyste<strong>in</strong>e <strong>in</strong>hibited DNA methyltransferase 1 (DNMT1)activity by 30%, and reduced the b<strong>in</strong>d<strong>in</strong>g of methyl CpG b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> 2 (MeCP2) and<strong>in</strong>creased the b<strong>in</strong>d<strong>in</strong>gs of acetylated histone H3 and H4 <strong>in</strong> the cycl<strong>in</strong> A promoter. F<strong>in</strong>ally,adenovirus-transduced DNMT1 gene expression reversed the <strong>in</strong>hibitory effect of homocyste<strong>in</strong>eon cycl<strong>in</strong> A expression and EC growth <strong>in</strong>hibition. In conclusion, homocyste<strong>in</strong>e <strong>in</strong>hibits cycl<strong>in</strong> Atranscription and cell growth by <strong>in</strong>hibit<strong>in</strong>g DNA methylation through suppression of DNMT1<strong>in</strong> ECs. These results revealed the potential role of DNA methylation <strong>in</strong> EC proliferation.20.2.2 Histone Acetylation and EC ProliferationOur laboratory has found that HDAC7 controls EC growth through modulation of b-caten<strong>in</strong>translocation [49] (Figure 20.1). We found that overexpression of HDAC7 suppresses humanumbilical ve<strong>in</strong> EC proliferation by prevent<strong>in</strong>g nuclear translocation of b-caten<strong>in</strong> and downregulat<strong>in</strong>gT-cell factor-1/Id2 (<strong>in</strong>hibitor of DNA b<strong>in</strong>d<strong>in</strong>g 2) and cycl<strong>in</strong> D1, lead<strong>in</strong>g to G1phase elongation. Knockdown of HDAC7 by shRNA <strong>in</strong>duced b-caten<strong>in</strong> nuclear translocationbut down-regulated cycl<strong>in</strong> D1, cycl<strong>in</strong> E1, and E2F2, caus<strong>in</strong>g EC hypertrophy. Furtherexperiments showed that HDAC7 could reta<strong>in</strong> b-caten<strong>in</strong> <strong>in</strong> the cytoplasm by direct b<strong>in</strong>d<strong>in</strong>g.We also found that VEGF could <strong>in</strong>duce HDAC7 degradation via PLCg-IP3K signal pathwayand partially rescued HDAC7-mediated suppression of proliferation. Our f<strong>in</strong>d<strong>in</strong>gs demonstratea novel function of HDAC7 <strong>in</strong> the cytoplasm, <strong>in</strong>dicat<strong>in</strong>g that class II HDACs are notonly functional when they are localized <strong>in</strong> the nucleus as it was previously believed. Otherresearchers have also <strong>in</strong>vestigated HDAC7 <strong>in</strong> EC function. Importantly, Chang et al. [50] haveshown that disruption of the HDAC7 gene <strong>in</strong> mice resulted <strong>in</strong> failure of endothelial cellecelladhesion and enlargement of the branchial arteries. The enlargement of the branchialarteries may imply an <strong>in</strong>crease <strong>in</strong> EC number or cell size, which is <strong>in</strong> agreement with our


CHAPTER 20The Role of <strong>Epigenetics</strong> <strong>in</strong> Cardiovascular <strong>Disease</strong>FIGURE 20.1A schematic illustration shows the role of HDAC7 <strong>in</strong> controll<strong>in</strong>g EC growth. HDAC7 acts as a bridge between 14-3-3 prote<strong>in</strong>sand b-caten<strong>in</strong>, which stabilizes b-caten<strong>in</strong> <strong>in</strong> the cytoplasm, result<strong>in</strong>g <strong>in</strong> <strong>in</strong>hibition of EC growth and lead<strong>in</strong>g to G1 phaseelongation. VEGF treatment <strong>in</strong>creases HDAC7 degradation through a VEGF-PLC g-IP3K pathway, releas<strong>in</strong>g b-caten<strong>in</strong> from theHDAC7-b-caten<strong>in</strong>-14-3-3 complex, lead<strong>in</strong>g to b-caten<strong>in</strong> nuclear translocation. The overall effect is an <strong>in</strong>crease <strong>in</strong> b-caten<strong>in</strong>target gene expression and EC growth. This figure is reproduced <strong>in</strong> the color plate section.f<strong>in</strong>d<strong>in</strong>gs that knockdown of HDAC7 by shRNA <strong>in</strong>creased EC size with a concomitant <strong>in</strong>crease<strong>in</strong> cellular metabolism. These f<strong>in</strong>d<strong>in</strong>gs suggest that HDAC7 regulates EC cycle and growth[50]. However, Mottet et al. [51] reported that siRNA-mediated knockdown of HDAC7 doesnot <strong>in</strong>fluence EC proliferation. This discrepancy may result from different culture systems,different effects of gene-knockdown assays, or different methods of measur<strong>in</strong>g cell proliferationand growth.399Injury of the lumen of vessel, e.g. percutaneous coronary <strong>in</strong>tervention, or percutaneoustranslum<strong>in</strong>al coronary angioplasty, could cause endothelial denudation. As mentioned above,endothelial denudation <strong>in</strong>duces local <strong>in</strong>flammation followed by the proliferation andmigration of SMC towards the lesion, lead<strong>in</strong>g to neo<strong>in</strong>tima formation and restenosis. Reendothelialization,which is promoted by EC migration and proliferation, can rescue thisrestenosis process [52]. Several studies have been performed to <strong>in</strong>vestigate the potential effectof epigenetics on EC migration.20.2.3 Histone Acetylation and EC MigrationUrbich et al. [53] revealed that siRNA-mediated knockdown of HDAC5 could promote ECmigration and sprout<strong>in</strong>g, while knockdown of HDAC7 and HDAC9 decreased EC migration.Deletion and mutation study of HDAC5 revealed that the nuclear localization of HDAC5 iscrucial for its function <strong>in</strong> EC migration, while its b<strong>in</strong>d<strong>in</strong>g with MEF2 (myocyte enhancer factor2) and deacetylase activity are dispensable. It seems that HDAC location is more important thanHDAC activity. However, we could not exclude the role of deacetylation <strong>in</strong> this case, becauseclass II HDACs can recruit other HDACs [16,17]. Microarray analysis <strong>in</strong>dicated that HDAC5silenc<strong>in</strong>g <strong>in</strong>creased the expression of secretive prote<strong>in</strong> fibroblast growth factor 2 (FGF2) [17].Inaddition, the conditional medium from ECs transfected with HDAC5 siRNA attracted moremigrated cells compar<strong>in</strong>g with scramble siRNA. Chromat<strong>in</strong> immunoprecipitation assay showedthat HDAC5 bound to the promoter region of FGF2, <strong>in</strong>dicat<strong>in</strong>g HDAC5 functions as a repressorfor FGF2 gene transcription. Their work suggests that HDAC5 is a repressor of EC migration andangiogenesis partially through modulation of FGF2 expression.HDAC7 has also been reported to modulate EC migration. Mottet et al. [51] showed thatsiRNA-mediated knockdown of HDAC7 <strong>in</strong>hibited EC tube formation and migration.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>Moreover, platelet-derived growth factor-B (PDGF-B) and its receptor (PDGFR-b) were themost up-regulated genes follow<strong>in</strong>g HDAC7 silenc<strong>in</strong>g. The <strong>in</strong>creased expression of PDGF-B andPDGFR-b are partially responsible for the <strong>in</strong>hibition of EC migration. Furthermore, treatmentof ECs with phorbol 12-myristate 13-acetate resulted <strong>in</strong> the translocation of HDAC7 out of thenucleus through a prote<strong>in</strong> k<strong>in</strong>ase C/prote<strong>in</strong> k<strong>in</strong>ase D pathway and <strong>in</strong>duced, similarly toHDAC7 silenc<strong>in</strong>g, an <strong>in</strong>crease <strong>in</strong> PDGF-B expression, as well as a partial <strong>in</strong>hibition of ECmigration. Collectively, their data identify HDAC7 as a key modulator of EC migration andhence angiogenesis, at least <strong>in</strong> part by regulat<strong>in</strong>g PDGF-B/PDGFR-b gene expression.Ha et al. [54] demonstrated another way that the VEGF could modulate HDAC7 and ECmigration. They found that VEGF stimulated phosphorylation of HDAC7 at the sites ofSer178, Ser344, and Ser479 <strong>in</strong> a dose- and time-dependent manner, lead<strong>in</strong>g to the cytoplasmicaccumulation of HDAC7. The phosphorylation of HDAC7 has been proved to be mediated byphospholipase Cg/prote<strong>in</strong> k<strong>in</strong>ase C/prote<strong>in</strong> k<strong>in</strong>ase D1 (PKD1)-dependent signal pathway.Infection of ECs with adenoviruses encod<strong>in</strong>g a mutant of HDAC7 specifically deficient <strong>in</strong>PKD1-dependent phosphorylation <strong>in</strong>hibited VEGF-<strong>in</strong>duced primary aortic EC migration.Recent researches have shown that lum<strong>in</strong>al EC apoptosis may be responsible for thrombusformation on eroded plaques without rupture [4,55]. So, the importance of EC apoptosis <strong>in</strong>atherosclerosis could not be neglected. <strong>Epigenetics</strong> have been shown to be <strong>in</strong>volved <strong>in</strong> ECapoptosis.40020.2.4 DNA Methylation and EC ApoptosisMitra et al. [56] found that oxidized-LDL evoked a dose-dependent <strong>in</strong>crease <strong>in</strong> apoptosis <strong>in</strong> thefirst passage ECs that was completely abrogated by LOX-1 (Lect<strong>in</strong>-like oxidized low-densitylipoprote<strong>in</strong>receptor-1) neutraliz<strong>in</strong>g antibody. Oxidized-LDL-<strong>in</strong>duced apoptosis was associatedwith up-regulation of proapoptotic LOX-1, ANXA5, BAX, and CASP3 and <strong>in</strong>hibition ofanti apoptotic BCL2 and cIAP-1 genes accompanied with reciprocal changes <strong>in</strong> the methylationof promoter regions of these genes. Based on these data, they conclude that exposure ofECs to oxidized-LDL <strong>in</strong>duces epigenetic changes lead<strong>in</strong>g to resistance to apoptosis <strong>in</strong> subsequentgenerations, and this effect may be related to a LOX-1-mediated <strong>in</strong>crease <strong>in</strong> DNAmethylation.Rao et al. [57] used methyl-CpG-b<strong>in</strong>d<strong>in</strong>g doma<strong>in</strong> prote<strong>in</strong> 2 (MBD2), an <strong>in</strong>terpreter for DNAmethylome-encoded <strong>in</strong>formation, to dissect the impact of DNA methylation on endothelialfunction. They found that knockdown of MBD2 by siRNA significantly enhanced angiogenesisand provided protection aga<strong>in</strong>st H 2 O 2 -<strong>in</strong>duced apoptosis. Remarkably, Mbd2(e/e) mice wereprotected aga<strong>in</strong>st h<strong>in</strong>dlimb ischemia evidenced by the significant improvement <strong>in</strong> perfusionrecovery, along with <strong>in</strong>creased capillary and arteriole formation. On ischemic <strong>in</strong>sult, keyendothelial genes such as eNOS and vascular endothelial growth factor receptor 2 undergoa DNA methylation turnover, and MBD2 <strong>in</strong>terprets the changes of DNA methylation tosuppress their expressions. Thus, Mbd2 could be a viable epigenetic target for modulat<strong>in</strong>gendothelial apoptosis <strong>in</strong> disease states.20.2.5 Histone Acetylation and EC SurvivalOur group has previously found that HDAC3 plays a crucial role <strong>in</strong> the differentiation of ECsfrom embryonic stem cells [58,59]. Zampetaki et al. [60] found that shRNA-mediated knockdownof HDAC3 resulted <strong>in</strong> an <strong>in</strong>crease of cells show<strong>in</strong>g extensive membrane blebs, reducedcell number and survival, enhanced presence of nucleosomes <strong>in</strong> cytosol, and more Annex<strong>in</strong> Vsta<strong>in</strong><strong>in</strong>g. Ex vivo experiments showed loss of ECs <strong>in</strong> the aortic segments treated with ShRNA ofHDAC3. Coimmunoprecipitation experiments resolved that HDAC3 forms a complex with Akt.Overexpression of HDAC3 resulted <strong>in</strong> <strong>in</strong>creased phosphorylation of Akt and up-regulation ofits k<strong>in</strong>ease activity. Taken together, our f<strong>in</strong>d<strong>in</strong>gs demonstrated that HDAC3 plays a critical role<strong>in</strong> ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g EC survival and prevents arteriosclerosis via Akt activation.


CHAPTER 20The Role of <strong>Epigenetics</strong> <strong>in</strong> Cardiovascular <strong>Disease</strong>A class III HDAC SIRT1 has been shown to prevent EC apoptosis and senescence. Hou et al.[61] showed that EC SIRT1 is vital for the prevention of early membrane apoptotic phosphatidylser<strong>in</strong>eexternalization and subsequent DNA degradation, through a pathway<strong>in</strong>volv<strong>in</strong>g Akt1 and FoxO3a. Zu et al. [62] demonstrated that SIRT1 could promote ECproliferation and prevent senescence by regulat<strong>in</strong>g a ser<strong>in</strong>e/threon<strong>in</strong>e k<strong>in</strong>ase and tumorsuppressor LKB1.HDAC <strong>in</strong>hibitor valproic acid can also affect EC apoptosis. Michaelis et al. [63] showed thatvalproic acid could <strong>in</strong>crease extracellular signal-regulated k<strong>in</strong>ase1/2 (ERK 1/2) phosphorylation<strong>in</strong> ECs. ERK 1/2 phosphorylation leads to phosphorylation of the antiapoptotic prote<strong>in</strong>Bcl-2 and <strong>in</strong>hibits serum starvation-<strong>in</strong>duced EC apoptosis and cytochrome C release from themitochondria. Collectively, their results showed that valproic acid can prevent EC apoptosisthrough the phosphorylation of ERK1/2.Apart from the f<strong>in</strong>d<strong>in</strong>gs of epigenetics <strong>in</strong> EC proliferation, migration, and apoptosis, Banerjeeet al. [64] also found that specific DNA methyltransferase (DNMT) <strong>in</strong>hibitor 5 0 -aza-2 0 -deoxycytid<strong>in</strong>e(aza-dC) could <strong>in</strong>duce embryonic stem cell (ESC) differentiation towards endothelialcell (EC). Significant <strong>in</strong>crease <strong>in</strong> angiogenesis and expression of the mediators of EC differentiationand EC-specific genes was only observed <strong>in</strong> aza-dC-treated cells. The DNMT<strong>in</strong>hibition-mediated <strong>in</strong>crease <strong>in</strong> EC specification and marker gene expression was not associatedwith demethylation of these genes. These studies suggest the crucial role of epigenetics<strong>in</strong> EC differentiation.20.3 EPIGENETICS AND SMC HOMEOSTASISSMC proliferation is a key event <strong>in</strong> neo<strong>in</strong>tima formation or arteriosclerosis. After EC <strong>in</strong>jury andactivation, various growth factors (e.g. PDGF, TGF-b) and cytok<strong>in</strong>es (<strong>in</strong>terferon-g) are releasedby different cell types, <strong>in</strong>clud<strong>in</strong>g EC, platelets, and monocytes [2]. These cytok<strong>in</strong>es and growthfactors promote SMC proliferation, which exacerbates the formation of advanced lesion <strong>in</strong>arteriosclerosis.40120.3.1 DNA Methylation, Atherosclerosis, and SMC Phenotype SwitchSeppo and colleagues [65] have shown that significant genomic hypomethylation developsdur<strong>in</strong>g the first replications of aortic SMCs <strong>in</strong> vitro and that hypomethylation occurs <strong>in</strong> somespecific genes, such as 15-lipoxygenase and extracellular superoxide dismutase. It has also beenshown that regional hypermethylation occurs <strong>in</strong> atherosclerosis. Estrogen receptor-a gene wasfound to have an <strong>in</strong>creased methylation level <strong>in</strong> atheromas compared with normal aorta [66].Estrogen receptor-a gene was also shown to be methylated <strong>in</strong> SMCs <strong>in</strong> vitro dur<strong>in</strong>g thephenotypic switch [67]. These results <strong>in</strong>dicate the methylation status of genes is closely relatedto atherosclerosis.3-Deazaadenos<strong>in</strong>e (c3Ado) is a potent <strong>in</strong>hibitor of S-adenosylhomocyste<strong>in</strong>e hydrolase, whichregulates cellular methyltransferase activity. Sedd<strong>in</strong>g et al. [68] showed that c3Ado dosedependentlyprevented the proliferation and migration of human coronary SMCs <strong>in</strong> vitro.Mechanistically, c3Ado could reduce growth factor-<strong>in</strong>duced extracellular signal-regulated k<strong>in</strong>ase(ERK)1/2 and Akt phosphorylation. For <strong>in</strong> vivo study, the femoral artery of C57BL/6 mice wasdilated and mice were fed a diet conta<strong>in</strong><strong>in</strong>g 150 mg of c3Ado per day. C3Ado prevented dilation<strong>in</strong>ducedRas activation, as well as ERK1/2 and Akt phosphorylation <strong>in</strong> vivo. At day 21, VSMCproliferation, as well as the neo<strong>in</strong>tima/media ratio, was significantly reduced. These f<strong>in</strong>d<strong>in</strong>gshighlight the importance of cellular methyltransferase activity <strong>in</strong> regulat<strong>in</strong>g SMC proliferation.Diabetic patients cont<strong>in</strong>ue to develop <strong>in</strong>flammation and vascular complications even afterachiev<strong>in</strong>g glycemic control. This poorly understood “metabolic memory” phenomenon posesmajor challenges <strong>in</strong> treat<strong>in</strong>g diabetes; Villeneuve et al. [69] showed that <strong>in</strong> SMCs derived fromtype 2 diabetic db/db mice. These cells exhibit a persistent atherogenic and <strong>in</strong>flammatory


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>phenotype even after culture <strong>in</strong> vitro. ChIP assays showed that H3K9me3 levels were significantlydecreased at the promoters of key <strong>in</strong>flammatory genes <strong>in</strong> cultured db/db SMC relative tocontrol db/þ cells. These results <strong>in</strong>dicated the important role of DNA methylation <strong>in</strong> SMCphenotype switch <strong>in</strong> diabetes.20.3.2 Histone Acetylation and SMC ProliferationOkamoto et al. [70] have found that TSA at the concentrations of 0.1, 1, 10 mmol/l could timedependentlysuppress proliferation of primary SMCs isolated from rat thoracic aorta. Theyhave used cell count and [H 3 ]-thymid<strong>in</strong>e <strong>in</strong>corporation methods to measure proliferation.Mechanistic studies revealed that TSA reduced the phosphorylation of Rb prote<strong>in</strong>, and <strong>in</strong>ducedthe expression of p21/WAF1 but not of p16INK4, p27KIP1, or p53. F<strong>in</strong>ally, TSA <strong>in</strong>hibitedHDAC activity of SMCs from p21/WAF1 knockout mice but did not <strong>in</strong>fluence the proliferationof these cells. Their work suggests that TSA <strong>in</strong>hibits SMC proliferation via the <strong>in</strong>duction of p21/WAF1 and subsequent cell-cycle arrest with reduction of the phosphorylation of Rb.Song et al. [71] argued that TSA at the concentration of 0.5 mmol/l could <strong>in</strong>crease PDGF-BBstimulatedproliferation of primary SMC isolated from thoracic and abdom<strong>in</strong>al aorta of rats.They pretreated SMC with TSA, and then stimulated with PDGF-BB, cell viability was measuredand demonstrated that TSA treatment time-dependently decreased thioredox<strong>in</strong> 1 expression <strong>in</strong>rat SMCs at both the mRNA and prote<strong>in</strong> levels. Moreover, siRNA-mediated knockdown ofthioredox<strong>in</strong> 1 could potentiate Akt phosphorylation and enhance SMC proliferation <strong>in</strong>response to PDGF and serum. Collectively, these results <strong>in</strong>dicate that TSA could enhance SMCproliferation by down-regulat<strong>in</strong>g thioredox<strong>in</strong> 1, thus activat<strong>in</strong>g an Akt-dependent pathway.402This discrepancy may be attributed to different methods. Okamoto et al. [70] measured theproliferation of SMC <strong>in</strong> response to serum when treated with TSA, whilst Song et al. [71]measured the proliferation of SMC <strong>in</strong> response to PDGF-BB. Therefore, there may be a possibilitythat TSA activates or <strong>in</strong>hibits a certa<strong>in</strong> pathway that is specifically <strong>in</strong>volved <strong>in</strong> the PDGFstimulation. Also, based on the fact that TSA can <strong>in</strong>hibit both class I and class II HDACs, it mayhave different functions <strong>in</strong> the cell when different concentrations are applied.Recently, studies of our group have revealed that two isoforms of HDAC7 are expressed <strong>in</strong>SMCs through alternative splic<strong>in</strong>g. We sought to exam<strong>in</strong>e the effect of two HDAC7 isoforms onSMC proliferation and the function <strong>in</strong> neo<strong>in</strong>tima formation [72]. We found that overexpressionof the unspliced HDAC7 isoform (HDAC7u) could suppress SMC proliferationthrough down-regulation of cycl<strong>in</strong> D1 and cell cycle arrest, while the spliced HDAC7 isoform(HDAC7s) did not have the same effect. siRNA-mediated knockdown of HDAC7 <strong>in</strong>creasedSMC proliferation and <strong>in</strong>duced b-caten<strong>in</strong> nuclear translocation. Further experiments showedthat only HDAC7u could b<strong>in</strong>d with b-caten<strong>in</strong> and reta<strong>in</strong> it <strong>in</strong> the cytoplasm. Reporter geneassay and reverse transcription PCR revealed less b-caten<strong>in</strong> activity <strong>in</strong> the cells overexpress<strong>in</strong>gHDAC7u, but not HDAC7s. Deletion studies <strong>in</strong>dicate that the C-term<strong>in</strong>al of HDAC7u isresponsible for the b<strong>in</strong>d<strong>in</strong>g with b-caten<strong>in</strong>. However, N-term<strong>in</strong>al additional am<strong>in</strong>o acidsdisrupted the b<strong>in</strong>d<strong>in</strong>g, which gives more strength to the fact that HDAC7s did not b<strong>in</strong>d with b-caten<strong>in</strong>. Growth factor PDGF-BB <strong>in</strong>creased the splic<strong>in</strong>g of HDAC7, decreas<strong>in</strong>g the expressionof HDAC7u. Importantly, <strong>in</strong> an animal model of femoral artery wire <strong>in</strong>jury we demonstratedthat knockdown of HDAC7 by siRNA aggravates neo<strong>in</strong>timal formation <strong>in</strong> comparison withcontrol siRNA. Our f<strong>in</strong>d<strong>in</strong>gs demonstrate that splic<strong>in</strong>g of HDAC7 modulates SMC proliferationand neo<strong>in</strong>tima formation through b-caten<strong>in</strong> translocation, which provide a potentialtherapeutic target <strong>in</strong> vascular disease (Figure 20.2).SMC migrat<strong>in</strong>g from the media to the <strong>in</strong>tima is an important step dur<strong>in</strong>g atherogenesis, whichturns fatty streak <strong>in</strong>to advanced, complicated lesion [2]. Under physiological conditions SMCs aresurrounded by extracellular matrix and kept <strong>in</strong> a low migratory activity. However, dur<strong>in</strong>g thedevelopment of arteriosclerosis, matrix metalloprote<strong>in</strong>ase could be released from various cell


CHAPTER 20The Role of <strong>Epigenetics</strong> <strong>in</strong> Cardiovascular <strong>Disease</strong>FIGURE 20.2A schematic illustration shows the role of HDAC7 splic<strong>in</strong>g <strong>in</strong> controll<strong>in</strong>g SMC differentiation and proliferation. HDAC7 undergoesalternative splic<strong>in</strong>g, produc<strong>in</strong>g spliced HDAC7 (HDAC7s) and unspliced HDAC7 (HDAC7u). HDAC7s, when it is <strong>in</strong> the nucleus,could promote stem cell differentiation towards SMC through HDAC7s-SRF-myocard<strong>in</strong> signal<strong>in</strong>g. HDAC7u could <strong>in</strong>hibit thisdifferentiation process. HDAC7u, on the other hand, could suppress SMC proliferation by sequester<strong>in</strong>g b-caten<strong>in</strong> <strong>in</strong> thecytoplasm and prevent<strong>in</strong>g b-caten<strong>in</strong> nuclear translocation, thus, <strong>in</strong>hibit<strong>in</strong>g the expression of b-caten<strong>in</strong> target genes such ascycl<strong>in</strong> D1. HDAC7s has no effect on SMC proliferation. PDGF-BB could <strong>in</strong>crease the splic<strong>in</strong>g of HDAC7. This figure isreproduced <strong>in</strong> the color plate section.sources (e.g. activated macrophages) to degrade matrix prote<strong>in</strong>s, thus promot<strong>in</strong>g the migration ofSMCs. <strong>Epigenetics</strong> is actively <strong>in</strong>volved <strong>in</strong> the molecular mechanism controll<strong>in</strong>g SMC migration.40320.3.3 Histone Acetylation and SMC MigrationSMCs respond to mechanical stra<strong>in</strong> but the role of HDACs <strong>in</strong> modulat<strong>in</strong>g SMC migration<strong>in</strong>duced by mechanical stra<strong>in</strong> is not well elucidated. Yan et al. [73] stated that cyclic stra<strong>in</strong>could significantly <strong>in</strong>hibit the migration of cultured SMCs. The cyclic stra<strong>in</strong> up-regulated thelevels of acetylased histone H3 and HDAC7 while it down-regulated the level of HDAC3/4 <strong>in</strong>SMCs. Furthermore, the mechanically <strong>in</strong>duced SMC migration was dim<strong>in</strong>ished by treatmentwith tributyr<strong>in</strong>, an HDAC <strong>in</strong>hibitor. They also observed hyperacetylation of histone H3 andreduced expression of HDAC7 upon tributyr<strong>in</strong> treatment. These results provide evidence thatHDACs are <strong>in</strong>volved <strong>in</strong> the migration of SMCs <strong>in</strong>duced by mechanical stra<strong>in</strong>. Similarly, Songet al. [71] found that TSA could enhance SMC migration <strong>in</strong> response to PDGF-BB.By compar<strong>in</strong>g the SMC from normal human coronary arteries and from coronary plaques,Bennett el al. [74] observed higher rates of SMC apoptosis <strong>in</strong> plaque, which may ultimatelycontribute to plaque rupture. It was previously demonstrated that plaque-derived SMCs havereduced <strong>in</strong>sul<strong>in</strong>-like growth factor 1 (IGF1) signal<strong>in</strong>g, result<strong>in</strong>g from a decrease <strong>in</strong> theexpression of IGF1 receptor compared with normal aortic SMCs. Furthermore, they havefound that overexpression of IGF1 receptor could abolish oxidative-stress-<strong>in</strong>duced apoptosis<strong>in</strong> SMCs [75], and oxidative stress repressed IGF1 receptor gene expression <strong>in</strong> turn, whichneeds HDAC1 [76]. These f<strong>in</strong>d<strong>in</strong>gs suggest that HDAC1 is a critical molecule <strong>in</strong> the signal<strong>in</strong>g ofoxidative-stress-<strong>in</strong>duced SMC apoptosis.20.3.4 Histone Acetylation and SMC ApoptosisClass III HDACs have also been <strong>in</strong>volved <strong>in</strong> SMC apoptosis. Veer et al. [77] reported that pre-Bcellcolony-enhanc<strong>in</strong>g factor could reduce SMC apoptosis as revealed by TdT-mediated dUTP


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>Nick-End Label<strong>in</strong>g (TUNEL) analysis. They found that nicot<strong>in</strong>amide aden<strong>in</strong>e d<strong>in</strong>ucleotide(NAD þ )-dependent prote<strong>in</strong> deacetylase activity was required for SMC maturation and thatNAD þ -dependent HDAC activity was augmented by pre-B-cell colony-enhanc<strong>in</strong>g factor. Theseresults provide a novel pathway that class III HDACs can <strong>in</strong>fluence SMC apoptosis andphenotype switch.Traditionally, SMCs <strong>in</strong> the neo<strong>in</strong>tima of atherosclerosis were believed to be orig<strong>in</strong>ated from themedia of <strong>in</strong>jured arteries. These media-orig<strong>in</strong>ated SMCs then proliferate and migrate towardsthe <strong>in</strong>tima <strong>in</strong> response to signals from <strong>in</strong>flammatory cells and ECs, which form neo<strong>in</strong>tima [2].However, recent f<strong>in</strong>d<strong>in</strong>gs from different groups emphasized the importance of stem/progenitorcell-derived SMCs <strong>in</strong> neo<strong>in</strong>tima formation and atherosclerosis [78e84].40420.3.5 DNA Methylation and SMC DifferentiationLockman et al. [85] identified the histone 3 lys<strong>in</strong>e 9 (H3K9)-specific demethylase, Jmjd1abound all three myocard<strong>in</strong> family members, and regulated SMC differentiation marker geneexpression. Overexpression of Jmjd1a <strong>in</strong> multipotential 10T1/2 cells decreased global levels ofdimethyl H3K9, stimulated the SM alpha-act<strong>in</strong> and SM22 promoters, and synergisticallyenhanced MRTF-A- and myocard<strong>in</strong>-dependent transactivation. Us<strong>in</strong>g chromat<strong>in</strong> immunoprecipitationassays, they also demonstrated that TGF-beta-mediated up-regulation of SMCdifferentiation marker gene expression <strong>in</strong> 10T1/2 cells was associated with decreased H3K9dimethylation at the CArG-conta<strong>in</strong><strong>in</strong>g regions of the SMC differentiation marker genepromoters. Importantly, knockdown of Jmjd1a <strong>in</strong> 10T1/2 cells and primary rat aortic SMCs byretroviral delivery of siRNA attenuated TGF-beta-<strong>in</strong>duced up-regulation of endogenous SMmyos<strong>in</strong> heavy-cha<strong>in</strong> expression. These results showed that histone methylation status couldmodulate SMC differentiation. Furthermore, Lee et al. [86] reported that DNA methylationcould <strong>in</strong>fluence the transdifferentiation of myoblasts <strong>in</strong>to smooth muscle cells. The DNAmethylation <strong>in</strong>hibitor, zebular<strong>in</strong>e, <strong>in</strong>duced the morphological transformation of C2C12myoblasts <strong>in</strong>to smooth muscle cells accompanied by de novo synthesis of smooth musclemarkers such as smooth muscle alpha-act<strong>in</strong> and transgel<strong>in</strong>.20.3.6 Histone Acetylation and Smooth Muscle Cell-specific GeneExpressionSp<strong>in</strong> et al. [87] performed a subanalysis exam<strong>in</strong><strong>in</strong>g transcriptional time-course microarray dataobta<strong>in</strong>ed us<strong>in</strong>g the A404 model of SMC differentiation. Ontology analysis <strong>in</strong>dicated a highdegree of p300 <strong>in</strong>volvement <strong>in</strong> SMC differentiation. Knockdown of p300 expression acceleratedSMC differentiation <strong>in</strong> A404 cells and human SMCs, while <strong>in</strong>hibition of p300 HATactivity blunted SMC differentiation. Also Qiu et al. [88] showed that the stimulation of theSM22 promoter by the coactivator CREB-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> (CBP) was dependent on HATactivity. These studies provides evidence that chromat<strong>in</strong> acetylation is <strong>in</strong>volved <strong>in</strong> smoothmuscle cell-specific gene regulation.20.4 EPIGENETICS AND ATHEROSCLEROSISThis section looks at epigenetics and atherosclerosis.20.4.1 DNA Methylation <strong>in</strong> AtherosclerosisRecent results from human and animal studies have shown that DNA methylation correlateswell with atherosclerosis. Sharma et al. [89] observed that the genomic DNA methylation <strong>in</strong>peripheral lymphocytes <strong>in</strong> coronary artery disease (CAD) patients is significantly higher than<strong>in</strong> controls (p < 0.05). Moreover, a significant positive correlation of global DNA methylationwith plasma homocyste<strong>in</strong>e levels was seen <strong>in</strong> CAD patients (p ¼ 0.001). Hiltunen et al. [90]evaluated the methylation status of genomic DNA from peripheral lymphocytes <strong>in</strong> a cohort of287 <strong>in</strong>dividuals: 137 angiographically confirmed CAD patients and 150 controls. They found


CHAPTER 20The Role of <strong>Epigenetics</strong> <strong>in</strong> Cardiovascular <strong>Disease</strong>that (1) genomic hypomethylation occurs dur<strong>in</strong>g atherogenesis <strong>in</strong> human, mouse, and rabbitlesions and that it correlates with <strong>in</strong>creased transcriptional activity; (2) methyltransferase(MTase) is expressed <strong>in</strong> atherosclerotic lesions; and (3) hypomethylation is present <strong>in</strong>advanced lesions at the same level as <strong>in</strong> malignant tumors and may affect cellular proliferationand gene expression <strong>in</strong> atherosclerotic lesions. Kim et al. [91] <strong>in</strong>vestigated DNA methylationchanges <strong>in</strong> cardiovascular atherosclerotic tissues and showed that coronary atherosclerotictissues had higher methylation levels (28.7%) than normal-appear<strong>in</strong>g arterial (6.7e10.1%)and venous tissues (18.2%).For the methylation of specific genes, Post et al. [66] showed estrogen receptor alpha genemethylation appears to be <strong>in</strong>creased <strong>in</strong> coronary atherosclerotic plaques when compared tonormal proximal aorta. Lund et al. [92] showed that DNA methylation profiles, <strong>in</strong>clud<strong>in</strong>g bothhyper- and hypomethylation were present <strong>in</strong> aortas and blood cells of mutant mice with nodetectable atherosclerotic lesion. Castillo et al. [93] carried out a microarray-based survey ofthe methylation status of CpG islands (CGIs) <strong>in</strong> 45 human atherosclerotic arteries and 16controls. Data from 10 367 CGIs revealed that a subset (151 or 1.4%) of these was hypermethylated<strong>in</strong> control arteries. The vast majority (142 or 94%) of this CGI subset was found tobe unmethylated or partially methylated <strong>in</strong> atherosclerotic tissue, while only 17 of thenormally unmethylated CGIs were hypermethylated <strong>in</strong> the diseased tissue. These resultsstrongly <strong>in</strong>dicate that atherosclerosis is closely related to methylation status (Table 20.3).20.4.2 Histone Acetylation <strong>in</strong> ArteriosclerosisAtherosclerosis develops at specific sites of the vasculature that experience disturbed bloodflow [94]. Zampetaki et al. [60] found that HDAC3 expression was up-regulated <strong>in</strong> areas closeto branch open<strong>in</strong>gs where disturbed flow occurs. In aortic isografts of apolipoprote<strong>in</strong> E-knockout mice treated with shHDAC3, a robust atherosclerotic lesion was formed. Surpris<strong>in</strong>gly,three of the eight mice that received shHDAC3-<strong>in</strong>fected grafts died with<strong>in</strong> 2 days after theoperation. Miller sta<strong>in</strong><strong>in</strong>g of the isografts revealed disruption of the basement membrane andrupture of the vessel. These f<strong>in</strong>d<strong>in</strong>gs <strong>in</strong>dicate that HDAC3 <strong>in</strong> the endothelium of artery iscrucial <strong>in</strong> prevent<strong>in</strong>g atherosclerosis. Moreover, Song et al. [71] used a mouse carotid arteryligation model to exam<strong>in</strong>e the effect of TSA on arteriosclerosis. They found that the mice<strong>in</strong>jected with TSA showed 2.2 times more neo<strong>in</strong>tima/media ratio than controls 2 weeks afterligation. They attributed this effect to enhanced PDGF-stimulated SMC proliferation andmigration after TSA treatment.405We recently used a mouse femoral artery wire <strong>in</strong>jury model to <strong>in</strong>vestigate the role of HDAC7 <strong>in</strong>neo<strong>in</strong>tima formation [72]. SiRNA of HDAC7 or control siRNA was perivascularly delivered to theadventitial side of <strong>in</strong>jured femoral arteries. Two weeks later the neo<strong>in</strong>tima area was compared. Wefound that HDAC7 siRNA aggravated neo<strong>in</strong>tima formation compared with control siRNA. Theseresults highlight the crucial role of HDAC7 dur<strong>in</strong>g the pathogenesis of restenosis.Granger et al. [95] showed that utiliz<strong>in</strong>g a standard mur<strong>in</strong>e model of ischemia-reperfusion,chemical HDAC <strong>in</strong>hibitors significantly reduce <strong>in</strong>farct area even when delivered 1 hour afterthe ischemic <strong>in</strong>sult. They demonstrate that HDAC <strong>in</strong>hibitors prevent ischemia-<strong>in</strong>duced activationof gene programs that <strong>in</strong>clude hypoxia <strong>in</strong>ducible factor-1alpha, cell death, and vascularpermeability <strong>in</strong> vivo and <strong>in</strong> vitro. These results reveal that HDAC <strong>in</strong>hibitors can alter theresponse to ischemic <strong>in</strong>jury <strong>in</strong> the heart and reduce <strong>in</strong>farct size.20.5 EPIGENETICS AND HEART FAILURERecent genetic and biochemical analyses <strong>in</strong>dicate that epigenetic changes play a crucial role <strong>in</strong>the development of cardiac hypertrophy and heart failure, with dysregulation <strong>in</strong> histone acetylationstatus. In particular it has been shown to be directly l<strong>in</strong>ked to an impaired contractionability of cardiac myocytes. HATs and HDACs excert their role <strong>in</strong> this process (Table 20.3).


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>TABLE 20.3 <strong>Epigenetics</strong> and Cardiovascular <strong>Disease</strong>406Cell Type Feature Impact Mechanism ReferencesEC Proliferation DNMT1YHDAC7 YKnockdown HDAC7 no effectMigration Knockdown HDAC5 [Knockdown HDAC7 YKnockdown HDAC9 YKnockdown HDAC7 YAapoptosisKnockdown MBD2YKnockdown HDAC3 [VPA YSIRT1YSMC Proliferation C3AdoYTSA YTSA [HDAC7YAtherosclerosisCardiomyocyte/heartfailureCycl<strong>in</strong>Ab-caten<strong>in</strong>Not specifiedFGF2Not specifiedNot specifiedPDGF-B/PDGFR-bbVEGFR2, eNOSAktERK1/2Akt1, FoxO3a, LKB1Differentiation Aza-dC[ VEGF-A, BMP4,EPAS-1, VE-cadher<strong>in</strong>ERK1/2, AktP21 WAF1Trx1b-caten<strong>in</strong>MigrationApoptosisDifferentiationC3AdoYTSA [HDAC1 [ (<strong>in</strong>direct)NADþ HDAC activityJmjd1a[Zebular<strong>in</strong>e[Knockdown p300[Genomic DNA methylation[ <strong>in</strong> CAD patientsGenomic hypomethylation[ <strong>in</strong> CAD patientsCoronary atherosclerotic tissue had [ methylationERa gene methylation [ <strong>in</strong> atherosclerotic plaqueKnockdown HDAC3 [TSA[Knockdown HDAC7[Phenylephr<strong>in</strong>e[ cardiomyocyte hypertrophythrough p300p300[ ES differentiation <strong>in</strong>to cardiomyocytep300[ left ventricular remodel<strong>in</strong>g and myocytehypertrophyp300[ cardiac hyertrophyTSA[ myocardial differentiationHDAC2[ cardiomyocyte hypertrophyHDAC4 phosphorylation[ hypertrophic growthHDAC6 catalytic activity[ by extracellular stimuliHDAC9 knockout mice [ cariomegalySir2a[ size of cardiac myocyteERK1/2, AktTrx1IGF1RNot specifiedSMA, SM22SMA, Transgel<strong>in</strong>Not specifiedNot specifiedNot specifiedNot specifiedNot specifiedNot specifiedNot specifiedNot specifiedGATA4GATA4Not specifiedGATA, MEF2Not specifiedAktNot specifiedNot specifiedNot specifiedNot specified[47][32][50][52][52][52][50][56][59][62][60, 61][63][67][69][70][71][67][70][74][76][84][85][86][88][89][90][65][59][70][70][95][96][97][98][99][101][102][103][104][105]20.5.1 HAT <strong>in</strong> Ventricular Remodel<strong>in</strong>gGATA-4 is a cardiac-specific transcription factor <strong>in</strong> primary cardiac myocytes derived fromneonatal rats. Yanazume et al. [96] found that stimulation with phenylephr<strong>in</strong>e <strong>in</strong>creasedan acetylated form of GATA-4 and its DNA-b<strong>in</strong>d<strong>in</strong>g activity, as well as expression of p300.A dom<strong>in</strong>ant-negative mutant of p300 suppressed phenylephr<strong>in</strong>e-<strong>in</strong>duced nuclear acetylation,activation of GATA-4-dependent endothel<strong>in</strong>-1 promoters, and hypertrophic responses such as<strong>in</strong>crease <strong>in</strong> cell size and sarcomere organization. These f<strong>in</strong>d<strong>in</strong>gs suggest that p300-mediatednuclear acetylation plays a critical role <strong>in</strong> the development of myocyte hypertrophy andrepresents a pathway that leads to decompensated heart failure.


CHAPTER 20The Role of <strong>Epigenetics</strong> <strong>in</strong> Cardiovascular <strong>Disease</strong>In addition, Kawamura et al. [97] found the HAT activity of p300 is required for acetylationand DNA b<strong>in</strong>d<strong>in</strong>g of GATA-4 and its full transcriptional activity as well as for promotion ofa transcriptionally active chromat<strong>in</strong> configuration. However, the roles of HATs and HDACs <strong>in</strong>post-translational modification of GATA-4 dur<strong>in</strong>g the differentiation of ES cells <strong>in</strong>to cardiacmyocytes rema<strong>in</strong> unknown. In an ES cell model of develop<strong>in</strong>g embryoid bodies an acetylatedform of GATA-4 and its DNA b<strong>in</strong>d<strong>in</strong>g <strong>in</strong>creased concomitantly with the expression of p300dur<strong>in</strong>g the stem cell differentiation <strong>in</strong>to cardiac myocytes.Left ventricular remodel<strong>in</strong>g after myocardial <strong>in</strong>farction is associated with hypertrophy ofsurviv<strong>in</strong>g myocytes and represents a major process that leads to heart failure. Miyamoto et al.[98] generated transgenic mice overexpress<strong>in</strong>g <strong>in</strong>tact p300 or mutant p300 <strong>in</strong> the heart. As theresult of its 2-am<strong>in</strong>o acid substitution <strong>in</strong> the p300-histone acetyltransferase doma<strong>in</strong>, thismutant lost its histone acetyltransferase activity and was unable to activate GATA-4-dependenttranscription. The two k<strong>in</strong>ds of transgenic mice and the wild-type mice were subjected tomyocardial <strong>in</strong>farction or sham operation at the age of 12 weeks. Intact p300 transgenic miceshowed significantly more progressive ventricular dilation and dim<strong>in</strong>ished systolic functionafter myocardial <strong>in</strong>farction than wild-type mice, whereas mutant p300 transgenic mice didnot. These f<strong>in</strong>d<strong>in</strong>gs demonstrate that cardiac overexpression of p300 promotes ventricularremodel<strong>in</strong>g after myocardial <strong>in</strong>farction <strong>in</strong> adult mice <strong>in</strong> vivo and that histone acetyltransferaseactivity of p300 is required for these processes.Pressure overload <strong>in</strong>duced by transverse aortic contraction, postnatal physiological growth andhuman heart failure were associated with large <strong>in</strong>creases <strong>in</strong> p300. Wei et al. [99] reported thatm<strong>in</strong>imal transgenic overexpression of p300 (1.5- to 3.5-fold) <strong>in</strong>duced strik<strong>in</strong>g myocyte andcardiac hypertrophy. Heterozygous loss of a s<strong>in</strong>gle p300 allele reduced pressure overload<strong>in</strong>ducedhypertrophy by approximately 50% and rescued the hypertrophic phenotype of p300overexpression. Increased p300 expression enhanced acetylation of the p300 substrateshistone 3 and GATA-4. Interest<strong>in</strong>gly a twofold expression of p300 was associated with thede novo acetylation of MEF2. Consistent with this, genes specifically up-regulated <strong>in</strong> p300transgenic hearts were highly enriched for MEF2 b<strong>in</strong>d<strong>in</strong>g sites. Collectively, these results<strong>in</strong>dicate that p300 has a direct function <strong>in</strong> regulat<strong>in</strong>g cardiomyocyte marker expression andcardiac hypertrophy through transcription factor GATA-4.40720.5.2 HDAC <strong>in</strong> Cardiac HypertrophyHosse<strong>in</strong>khani et al. [100] demonstrated that 24-h stimulation by a histone deacetylase<strong>in</strong>hibitor, TSA facilitated myocardial differentiation of monkey ES cells, <strong>in</strong>dicat<strong>in</strong>g the relevanceof HDAC activity with cardiomyocytes.Kaneda et al. [101] used differential chromat<strong>in</strong> scann<strong>in</strong>g to isolate genomic fragments associatedwith histones subject to differential acetylation. They applied DCS to H9C2 ratembryonic cardiomyocytes <strong>in</strong>cubated with or without TSA, and found that 200 genomicfragments were readily isolated by differential chromat<strong>in</strong> scann<strong>in</strong>g on the basis of the preferentialacetylation of associated histones <strong>in</strong> TSA-treated cells. Their data establish a genomewideprofile of HDAC targets <strong>in</strong> cardiomyocytes, which should provide a basis for further<strong>in</strong>vestigations <strong>in</strong>to the role of epigenetic modification <strong>in</strong> cardiac disorders.Kee et al. [102] showed <strong>in</strong> cardiomyocytes that a forced expression of HDAC2 simulatedhypertrophy <strong>in</strong> an Akt-dependent manner, whereas enzymatically <strong>in</strong>ert HDAC2 H141A didnot. Hypertrophic stimuli <strong>in</strong>duced the expression of heat shock prote<strong>in</strong> (Hsp) 70. The<strong>in</strong>duced Hsp70 physically associated with and activated HDAC2. Hsp70 overexpressionproduced a hypertrophic phenotype, which was blocked either by siHDAC2 or by a dom<strong>in</strong>antnegative Hsp70DeltaABD. These results suggest that the <strong>in</strong>duction of Hsp70 <strong>in</strong> responseto diverse hypertrophic stresses and the ensu<strong>in</strong>g activation of HDAC2 trigger cardiachypertrophy.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>Backs et al. [103] showed that calcium/calmodul<strong>in</strong>-dependent k<strong>in</strong>ase II (CaMKII) signalsspecifically to HDAC4 by b<strong>in</strong>d<strong>in</strong>g to a unique dock<strong>in</strong>g site that is absent <strong>in</strong> other class IIaHDACs. Phosphorylation of HDAC4 by CaMKII promotes nuclear export and prevents nuclearimport of HDAC4, with consequent derepression of HDAC target genes. In cardiomyocytesCaMKII phosphorylation of HDAC4 results <strong>in</strong> hypertrophic growth, which can be blocked bya signal-resistant HDAC4 mutant. These f<strong>in</strong>d<strong>in</strong>gs reveal a central role for CaMKII-HDAC4signal<strong>in</strong>g pathways dur<strong>in</strong>g cardiomyocyte hypertrophy.Lemon et al. [104] developed assays to quantify catalytic activity of dist<strong>in</strong>ct HDAC classes <strong>in</strong>left and right ventricular cardiac tissue from animal models of hypertensive heart disease. ClassI and IIa HDAC activity was elevated <strong>in</strong> some but not all diseased tissues. In contrast, catalyticactivity of the class IIb HDAC, HDAC6, was consistently <strong>in</strong>creased <strong>in</strong> stressed myocardium, butnot <strong>in</strong> a model of physiologic hypertrophy. HDAC6 catalytic activity was also <strong>in</strong>duced bydiverse extracellular stimuli <strong>in</strong> cultured cardiac myocytes and fibroblasts. These f<strong>in</strong>d<strong>in</strong>gssuggest an important role for HDAC6 <strong>in</strong> chronic hypertension.Importantly, Zhang et al. [105] showed that class II HDACs, which repress MEF2 activity, aresubstrates for a stress-responsive k<strong>in</strong>ase specific for conserved ser<strong>in</strong>es that regulate MEF2eHDAC<strong>in</strong>teractions. Signal-resistant HDAC mutants lack<strong>in</strong>g these phosphorylation sites are refractory tohypertrophic signal<strong>in</strong>g and <strong>in</strong>hibit cardiomyocyte hypertrophy. Conversely, mutant micelack<strong>in</strong>g the class II HDAC, HDAC9, are sensitized to hypertrophic signals and exhibit stressdependentcardiomegaly. Thus, class II HDACs act as signal-responsive suppressors of thetranscriptional program govern<strong>in</strong>g cardiac hypertrophy and heart failure.408Alcendor et al. [106] found that overexpression of Sir2 (silent <strong>in</strong>formation regulator 2) alphaprotected cardiac myocytes from apoptosis <strong>in</strong> response to serum starvation and significantly<strong>in</strong>creased the size of cardiac myocytes. Furthermore, Sir2 expression was <strong>in</strong>creased significantly<strong>in</strong> hearts from dogs with heart failure <strong>in</strong>duced by rapid pac<strong>in</strong>g superimposed on stable, severehypertrophy. These results suggest that endogenous Sir2a plays an essential role <strong>in</strong> mediat<strong>in</strong>gcell survival, whereas Sir2a overexpression protects myocytes from apoptosis and causesmodest hypertrophy.20.6 BIOMARKER AND MicroRNAOn the other hand, identify<strong>in</strong>g early-stage cardiovascular disease biomarkers is of unparallelvalue. Peripheral blood leukocytes, which can be easily obta<strong>in</strong>ed from patients, have closerelevance with <strong>in</strong>flammation, atherosclerosis, and cardiovascular disease etiology, could betargets for the development of novel epigenomic biomarkers. For <strong>in</strong>stance, Castro et al. [107]found lower DNA methylation content <strong>in</strong> peripheral blood leukocytes from patients withcardiovascular disease. Results from the Normative Ag<strong>in</strong>g Study have also shown that lowerLINE-1 methylation <strong>in</strong> peripheral blood leukocytes is a predictor of <strong>in</strong>cidence and mortalityfrom ischemic heart disease and stroke [108]. Elevated Alu methylation <strong>in</strong> peripheral bloodleukocytes recently was related to prevalence of cardiovascular disease and obesity <strong>in</strong> Ch<strong>in</strong>ese<strong>in</strong>dividuals [109]. Thus, characteriz<strong>in</strong>g the methylation status of human peripheral bloodleukocytes may be potentially beneficial for the early diagnosis of cardiovascular diseases.MicroRNA is a recently discovered mechanism which plays an important role <strong>in</strong> cardiovasculardisease. Ren et al. [110] performed miRNA arrays analysis to detect the expression pattern ofmiRNAs <strong>in</strong> mur<strong>in</strong>e hearts subjected to ischemia/reperfusion <strong>in</strong> vivo and ex vivo. Surpris<strong>in</strong>gly,they found that only miR-320 expression was significantly decreased <strong>in</strong> the hearts on ischemia/reperfusion <strong>in</strong> vivo and ex vivo. Wang et al. [111] exam<strong>in</strong>ed muscle-enriched miRNAs (miR-1,miR-133a, and miR-499) and cardiac-specific miR-208a <strong>in</strong> circulat<strong>in</strong>g blood. Evaluation of themiRNA levels <strong>in</strong> plasma from patients with heart disease demonstrated that all four miRNAlevels were substantially higher than those from healthy people, patients with non-heart disease,or patients with other cardiovascular diseases. Notably, miR-208a rema<strong>in</strong>ed undetectable <strong>in</strong>


CHAPTER 20The Role of <strong>Epigenetics</strong> <strong>in</strong> Cardiovascular <strong>Disease</strong>non-heart disease patients but it was easily detected <strong>in</strong> 90.9% of heart disease patients and <strong>in</strong>100% patients with<strong>in</strong> 4ah of the onset of symptoms. MiR-208a revealed the higher sensitivityand specificity for diagnosis <strong>in</strong> patients. Small et al. [112] reviewed recent progress of microRNA<strong>in</strong> cardiovascular research, which seems to be a promis<strong>in</strong>g direction <strong>in</strong> future research.20.7 SUMMARY AND FUTURE PERSPECTIVESIn this chapter we summarized the role of epigenetics <strong>in</strong> the pathogenesis of cardiovasculardisease. DNA methylation as well as histone acetylation has crucial functions <strong>in</strong> modulat<strong>in</strong>gSMC and EC homeostasis (proliferation, migration, apoptosis, and differentiation), atherosclerosis,cardiomyocyte hypertrophy, and heart failure. These results have been listed <strong>in</strong> Table20.4, which provides more <strong>in</strong>formation and <strong>in</strong>ternet resources.Epigenetic modifications, which are dynamic and reversible, could represent a way thatorganisms adapt to their environment. Thus understand<strong>in</strong>g the relationship between environmentalconditions and epigenetic changes is of potential value. Future research elucidat<strong>in</strong>gTABLE 20.4 Internet ResourcesName Description AddressNIH EpigenomicsDeposite experiments and samples <strong>in</strong>vestigat<strong>in</strong>gthe function of DNA methylation and histonehttp://www.ncbi.nlm.nih.gov/epigenomicsmodificationNAME21 DNA methylation patterns were analyzed for 190 http://biochem.jacobs-university.<strong>Human</strong> Epigenome AtlasThe <strong>Human</strong> EpigenomeProjectMethDBHHMDGeneimpr<strong>in</strong>tgene promoter regions on chromosome 21The <strong>Human</strong> Epigenome Atlas <strong>in</strong>cludes humanreference epigenomes and the results of their<strong>in</strong>tegrative and comparative analysesIdentifies methylation variable positions (MVPs) <strong>in</strong>the human genome, currently conta<strong>in</strong>s CpGmethylation <strong>in</strong>formation for MHCThe database for DNA methylation andenvironmental epigenetic effectsA comprehensive database for human histonemodifications, which focuses on <strong>in</strong>tegrat<strong>in</strong>g usefulhistone modification <strong>in</strong>formation from experimentaldata that is essential for understand<strong>in</strong>g thesemodifications at a systematic levelImpr<strong>in</strong>ted genes are monnoallelically expressed <strong>in</strong>a parent-of-orig<strong>in</strong>-dependent manner because thesame parental allele is always epigeneticallysilenced. This website is a collection of impr<strong>in</strong>tedgenes by speciesde/name21/http://www.genboree.org/epigenomeatlas/<strong>in</strong>dex.rhtmlhttp://www.epigenome.org/http://www.methdb.de/http://202.97.205.78/hhmd/http://www.geneimpr<strong>in</strong>t.com/site/homeMethprimer Primer design for methylation PCR primers www.urogene.org/methprimer/<strong>in</strong>dex1.htmlMethblastA sequence similarity search program designed toexplore <strong>in</strong> silico bisulfite modified DNA (either or notmethylated at its CpG d<strong>in</strong>ucleotides)http://medgen.ugent.be/methBLAST/MethylatorChromat<strong>in</strong> structure andfunctionEpigenetic MethylationstationAn SVM-based method for DNA methylationpredictionInformation regard<strong>in</strong>g histones, histonemodifications and their biological roles, and relatedl<strong>in</strong>ksWeb source for <strong>in</strong>formation, reference, protocols,methods, techniques and l<strong>in</strong>ks on epigenetics andDNA methylationbio.dfci.harvard.edu/Methylator/http://www.chromat<strong>in</strong>.us/www.epigeneticstation.com/409Adapted from a table of the paper published <strong>in</strong> Circulation Cardiovascular Genetics 2010;3;567e573.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>the mechanisms of how environment <strong>in</strong>fluences epigenetic modifications may help us to bettertreat cardiovascular diseases. Despite the outstand<strong>in</strong>g progress on understand<strong>in</strong>g the role ofHDACs <strong>in</strong> arteriosclerosis, there are still several questions that need to be answered. Onequestion is whether the effects of HDACs are tissue- or cell-type-specific. This is crucial becauseif we want to suppress the migration of SMC dur<strong>in</strong>g atherogenesis, the impact on other celltypes needs to be determ<strong>in</strong>ed. Otherwise, suppression of SMC migration with HDACs (orHDACis) could lead to suppression of EC migration as well. To answer this question, moreexperiments need to be performed on the functions of HDACs <strong>in</strong> various cardiovascular cells.In respect to a cl<strong>in</strong>ical prospect, several cl<strong>in</strong>ical trials of HDAC <strong>in</strong>hibitors have been completed<strong>in</strong> cancer and neurological patients [113,114]. Among these cl<strong>in</strong>ical trials, HDAC <strong>in</strong>hibitorshave shown potent <strong>in</strong>hibition of HDAC activities <strong>in</strong> patients, and only limited side effects havebeen observed. Marks et al. [115] reviewed that HDAC <strong>in</strong>hibition potent to have effect onapoptosis of cancer versus normal human cells. This emphasizes the possibility of a tissuespecificeffect of HDAC <strong>in</strong>hibition, which could expla<strong>in</strong> the fact that HDAC <strong>in</strong>hibition hadlimited side effects <strong>in</strong> humans. Therefore, it seems HDAC <strong>in</strong>hibition could be a promis<strong>in</strong>gtarget <strong>in</strong> treat<strong>in</strong>g cardiovascular diseases. However, detailed attention should be paid to theapplication of such drugs, which may have unexpected effects due to the fact that the majorityof HDAC <strong>in</strong>hibitors are non-specific. S<strong>in</strong>ce different HDACs possess dist<strong>in</strong>ct functions, nonselective<strong>in</strong>hibition of HDACs could trigger side effect responses. Additionally, as an epigeneticmodifier of gene expression a s<strong>in</strong>gle HDAC is usually <strong>in</strong>volved <strong>in</strong> several signal pathways.Thus, development of highly selective and cell-type-specific HDAC <strong>in</strong>hibitors could bea promis<strong>in</strong>g field for the cardiovascular research <strong>in</strong> future. Along with the progress of thefunction of epigenetics <strong>in</strong> atherosclerosis and heart failure, our research on epigenetics willbenefit patients with cardiovascular diseases.410AcknowledgmentsThis work was supported by grants from the British Heart Foundation and the Oak Foundation.References[1] Ordovas JM, Smith CE. <strong>Epigenetics</strong> and cardiovascular disease. Nat Rev Cardiol 2010;7:510e9.[2] Ross R. Atherosclerosisean <strong>in</strong>flammatory disease. N Engl J Med 1999;340:115e26.[3] Wick G, Knoflach M, Xu Q. Autoimmune and <strong>in</strong>flammatory mechanisms <strong>in</strong> atherosclerosis. Annu RevImmunol 2004;22:361e403.[4] Fuster V. Lewis A. Conner Memorial Lecture. Mechanisms lead<strong>in</strong>g to myocardial <strong>in</strong>farction: <strong>in</strong>sights fromstudies of vascular biology. Circulation 1994;90:2126e46.[5] Serpi R, Tolonen AM, Huusko J, Rysa J, Tenhunen O, Yla-Herttuala S, et al. Vascular endothelial growth factor-Bgene transfer prevents angiotens<strong>in</strong> II-<strong>in</strong>duced diastolic dysfunction via proliferation and capillary dilatation <strong>in</strong>rats. Cardiovasc Res 2011;89:204e13.[6] Pons D, de Vries FR, van den Elsen PJ, Heijmans BT, Quax PH, Jukema JW. Epigenetic histone acetylation modifiers<strong>in</strong> vascular remodell<strong>in</strong>g: new targets for therapy <strong>in</strong> cardiovascular disease. Eur Heart J 2009;30:266e77.[7] Bonasio R, Tu S, Re<strong>in</strong>berg D. Molecular signals of epigenetic states. <strong>Science</strong> 2010;330:612e6.[8] Atadja PW. HDAC <strong>in</strong>hibitors and cancer therapy. Prog Drug Res 2011;67:175e95.[9] Hiltunen MO, Yla-Herttuala S. DNA methylation, smooth muscle cells, and atherogenesis. Arterioscler ThrombVasc Biol 2003;23:1750e3.[10] Lei H, Oh SP, Okano M, Juttermann R, Goss KA, Jaenisch R, et al. De novo DNA cytos<strong>in</strong>e methyltransferaseactivities <strong>in</strong> mouse embryonic stem cells. Development 1996;122:3195e205.[11] Zhou B, Margariti A, Zeng L, Xu Q. Role of histone deacetylases <strong>in</strong> vascular cell homeostasis and arteriosclerosis.Cardiovasc Res 2011;90:413e20.[12] Gray SG, Ekstrom TJ. The human histone deacetylase family. Exp Cell Res 2001;262:75e83.[13] Borradaile NM, Picker<strong>in</strong>g JG. NAD(þ), sirtu<strong>in</strong>s, and cardiovascular disease. Curr Pharm Des 2009;15:110e7.[14] Gao L, Cueto MA, Asselbergs F, Atadja P. Clon<strong>in</strong>g and functional characterization of HDAC11, a novel memberof the human histone deacetylase family. J Biol Chem 2002;277:25748e55.


CHAPTER 20The Role of <strong>Epigenetics</strong> <strong>in</strong> Cardiovascular <strong>Disease</strong>[15] Lahm A, Paol<strong>in</strong>i C, Pallaoro M, Nardi MC, Jones P, Neddermann P, et al. Unravel<strong>in</strong>g the hidden catalyticactivity of vertebrate class IIa histone deacetylases. Proc Natl Acad Sci USA 2007;104:17335e40.[16] Fischle W, Dequiedt F, Hendzel MJ, Guenther MG, Lazar MA, Voelter W, et al. Enzymatic activity associatedwith class II HDACs is dependent on a multiprote<strong>in</strong> complex conta<strong>in</strong><strong>in</strong>g HDAC3 and SMRT/N-CoR. Mol Cell2002;9:45e57.[17] Fischle W, Dequiedt F, Fillion M, Hendzel MJ, Voelter W, Verd<strong>in</strong> E. <strong>Human</strong> HDAC7 histone deacetylase activityis associated with HDAC3 <strong>in</strong> vivo. J Biol Chem 2001;276:35826e35.[18] McK<strong>in</strong>sey TA, Zhang CL, Lu J, Olson EN. Signal-dependent nuclear export of a histone deacetylase regulatesmuscle differentiation. Nature 2000;408:106e11.[19] de Ruijter AJ, van Gennip AH, Caron HN, Kemp S, van Kuilenburg AB. Histone deacetylases (HDACs):characterization of the classical HDAC family. Biochem J 2003;370:737e49.[20] Wanczyk M, Roszczenko K, Marc<strong>in</strong>kiewicz K, Bojarczuk K, Kowara M, W<strong>in</strong>iarska M. HDACiego<strong>in</strong>g throughthe mechanisms. Front Biosci 2011;16:340e59.[21] Laherty CD, Yang WM, Sun JM, Davie JR, Seto E, Eisenman RN. Histone deacetylases associated with themS<strong>in</strong>3 corepressor mediate mad transcriptional repression. Cell 1997;89:349e56.[22] Yoshida M, Nomura S, Beppu T. Effects of trichostat<strong>in</strong>s on differentiation of mur<strong>in</strong>e erythroleukemia cells.Cancer Res 1987;47:3688e91.[23] Richon VM, Emiliani S, Verd<strong>in</strong> E, Webb Y, Breslow R, Rifk<strong>in</strong>d RA, et al. A class of hybrid polar <strong>in</strong>ducers oftransformed cell differentiation <strong>in</strong>hibits histone deacetylases. Proc Natl Acad Sci USA 1998;95:3003e7.[24] Richon VM, Sandhoff TW, Rifk<strong>in</strong>d RA, Marks PA. Histone deacetylase <strong>in</strong>hibitor selectively <strong>in</strong>duces p21WAF1expression and gene-associated histone acetylation. Proc Natl Acad Sci USA 2000;97:10014e9.[25] Butler LM, Webb Y, Agus DB, Higg<strong>in</strong>s B, Tolent<strong>in</strong>o TR, Kutko MC, et al. Inhibition of transformed cell growthand <strong>in</strong>duction of cellular differentiation by pyroxamide, an <strong>in</strong>hibitor of histone deacetylase. Cl<strong>in</strong> Cancer Res2001;7:962e70.[26] Glick RD, Swendeman SL, Coffey DC, Rifk<strong>in</strong>d RA, Marks PA, Richon VM, et al. Hybrid polar histonedeacetylase <strong>in</strong>hibitor <strong>in</strong>duces apoptosis and CD95/CD95 ligand expression <strong>in</strong> human neuroblastoma. CancerRes 1999;59:4392e9.[27] Zhang Y, Adachi M, Kawamura R, Imai K. Bmf is a possible mediator <strong>in</strong> histone deacetylase <strong>in</strong>hibitors FK228and CBHA-<strong>in</strong>duced apoptosis. Cell Death Differ 2006;13:129e40.[28] Su GH, Sohn TA, Ryu B, Kern SE. A novel histone deacetylase <strong>in</strong>hibitor identified by high-throughput transcriptionalscreen<strong>in</strong>g of a compound library. Cancer Res 2000;60:3137e42.[29] Cao H, Stamatoyannopoulos G, Jung M. Induction of human gamma glob<strong>in</strong> gene expression by histonedeacetylase <strong>in</strong>hibitors. Blood 2004;103:701e9.[30] Gottlicher M, M<strong>in</strong>ucci S, Zhu P, Kramer OH, Schimpf A, Giavara S, et al. Valproic acid def<strong>in</strong>es a novel class ofHDAC <strong>in</strong>hibitors <strong>in</strong>duc<strong>in</strong>g differentiation of transformed cells. EMBO J 2001;20:6969e78.[31] Williams RS, Cheng L, Mudge AW, Harwood AJ. A common mechanism of action for three mood-stabiliz<strong>in</strong>gdrugs. Nature 2002;417:292e5.[32] Gondcaille C, Depreter M, Fourcade S, Lecca MR, Leclercq S, Mart<strong>in</strong> PG, et al. Phenylbutyrate up-regulates theadrenoleukodystrophy-related gene as a nonclassical peroxisome proliferator. J Cell Biol 2005;169:93e104.[33] Lu Q, Wang DS, Chen CS, Hu YD. Structure-based optimization of phenylbutyrate-derived histone deacetylase<strong>in</strong>hibitors. J Med Chem 2005;48:5530e5.[34] Dangond F, Gullans SR. Differential expression of human histone deacetylase mRNAs <strong>in</strong> response toimmune cell apoptosis <strong>in</strong>duction by trichostat<strong>in</strong> A and butyrate. Biochem Biophys Res Commun1998;247:833e7.[35] Gibson PR. The <strong>in</strong>tracellular target of butyrate’s actions: HDAC or HDON’T? Gut 2000;46:447e8.[36] Sambucetti LC, Fischer DD, Zabludoff S, Kwon PO, Chamberl<strong>in</strong> H, Trogani N, et al. Histone deacetylase<strong>in</strong>hibition selectively alters the activity and expression of cell cycle prote<strong>in</strong>s lead<strong>in</strong>g to specific chromat<strong>in</strong>acetylation and antiproliferative effects. J Biol Chem 1999;274:34940e7.[37] Furumai R, Komatsu Y, Nish<strong>in</strong>o N, Khochb<strong>in</strong> S, Yoshida M, Hor<strong>in</strong>ouchi S. Potent histone deacetylase<strong>in</strong>hibitors built from trichostat<strong>in</strong> A and cyclic tetrapeptide antibiotics <strong>in</strong>clud<strong>in</strong>g trapox<strong>in</strong>. Proc Natl Acad SciUSA 2001;98:87e92.[38] Piekarz RL, Robey R, Sandor V, Bakke S, Wilson WH, Dahmoush L, et al. Inhibitor of histone deacetylation,depsipeptide (FR901228), <strong>in</strong> the treatment of peripheral and cutaneous T-cell lymphoma: a case report. Blood2001;98:2865e8.[39] Furumai R, Matsuyama A, Kobashi N, Lee KH, Nishiyama M, Nakajima H, et al. FK228 (depsipeptide) asa natural prodrug that <strong>in</strong>hibits class I histone deacetylases. Cancer Res 2002;62:4916e21.[40] Kim MS, Son MW, Kim WB, In Park Y, Moon A. Apicid<strong>in</strong>, an <strong>in</strong>hibitor of histone deacetylase, preventsH-ras-<strong>in</strong>duced <strong>in</strong>vasive phenotype. Cancer Lett 2000;157:23e30.411


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>412[41] Han JW, Ahn SH, Park SH, Wang SY, Bae GU, Seo DW, et al. Apicid<strong>in</strong>, a histone deacetylase <strong>in</strong>hibitor, <strong>in</strong>hibitsproliferation of tumor cells via <strong>in</strong>duction of p21WAF1/Cip1 and gelsol<strong>in</strong>. Cancer Res 2000;60:6068e74.[42] De Schepper S, Bruwiere H, Verhulst T, Steller U, Andries L, Wouters W, et al. Inhibition of histone deacetylasesby chlamydoc<strong>in</strong> <strong>in</strong>duces apoptosis and proteasome-mediated degradation of surviv<strong>in</strong>. J Pharmacol Exp Ther2003;304:881e8.[43] Rosato RR, Almenara JA, Grant S. The histone deacetylase <strong>in</strong>hibitor MS-275 promotes differentiation orapoptosis <strong>in</strong> human leukemia cells through a process regulated by generation of reactive oxygen species and<strong>in</strong>duction of p21CIP1/WAF1 1. Cancer Res 2003;63:3637e45.[44] Haggarty SJ, Koeller KM, Wong JC, Groz<strong>in</strong>ger CM, Schreiber SL. Doma<strong>in</strong>-selective small-molecule <strong>in</strong>hibitor ofhistone deacetylase 6 (HDAC6)-mediated tubul<strong>in</strong> deacetylation. Proc Natl Acad Sci USA 2003;100:4389e94.[45] Itoh Y, Suzuki T, Miyata N. Isoform-selective histone deacetylase <strong>in</strong>hibitors. Curr Pharm Des 2008;14:529e44.[46] Caplan BA, Schwartz CJ. Increased endothelial cell turnover <strong>in</strong> areas of <strong>in</strong> vivo Evans Blue uptake <strong>in</strong> the pigaorta. Atherosclerosis 1973;17:401e17.[47] Somer JB, Schwartz CJ. Focal ( 3 H)cholesterol uptake <strong>in</strong> the pig aorta. 2. Distribution of (3 H)cholesterolacross the aortic wall <strong>in</strong> areas of high and low uptake <strong>in</strong> vivo. Atherosclerosis 1972;16:377e88.[48] Jamaludd<strong>in</strong> MD, Chen I, Yang F, Jiang X, Jan M, Liu X, et al. Homocyste<strong>in</strong>e <strong>in</strong>hibits endothelial cell growth viaDNA hypomethylation of the cycl<strong>in</strong> A gene. Blood 2007;110:3648e55.[49] Margariti A, Zampetaki A, Xiao Q, Zhou B, Karamariti E, Mart<strong>in</strong> D, et al. Histone deacetylase 7 controlsendothelial cell growth through modulation of beta-caten<strong>in</strong>. Circ Res 2010;106:1202e11.[50] Chang S, Young BD, Li S, Qi X, Richardson JA, Olson EN. Histone deacetylase 7 ma<strong>in</strong>ta<strong>in</strong>s vascular <strong>in</strong>tegrity byrepress<strong>in</strong>g matrix metalloprote<strong>in</strong>ase 10. Cell 2006;126:321e34.[51] Mottet D, Bellahcene A, Pirotte S, Waltregny D, Deroanne C, Lamour V, et al. Histone deacetylase 7 silenc<strong>in</strong>galters endothelial cell migration, a key step <strong>in</strong> angiogenesis. Circ Res 2007;101:1237e46.[52] Welt FG, Rogers C. Inflammation and restenosis <strong>in</strong> the stent era. Arterioscler Thromb Vasc Biol2002;22:1769e76.[53] Urbich C, Rossig L, Kaluza D, Potente M, Boeckel JN, Knau A, et al. HDAC5 is a repressor of angiogenesis anddeterm<strong>in</strong>es the angiogenic gene expression pattern of endothelial cells. Blood 2009;113:5669e79.[54] Ha CH, Jhun BS, Kao HY, J<strong>in</strong> ZG. VEGF stimulates HDAC7 phosphorylation and cytoplasmic accumulationmodulat<strong>in</strong>g matrix metalloprote<strong>in</strong>ase expression and angiogenesis. Arterioscler Thromb Vasc Biol2008;28:1782e8.[55] Tedgui A, Mallat Z. Apoptosis as a determ<strong>in</strong>ant of atherothrombosis. Thromb Haemost 2001;86:420e6.[56] Mitra S, Khaidakov M, Lu J, Ayyadevara S, Szwedo J, Wang XW, et al. Prior Exposure to Oxidized Low DensityLipoprote<strong>in</strong> Limits Apoptosis <strong>in</strong> Subsequent Generations of Endothelial Cells by Alter<strong>in</strong>g Promoter Methylation.Am J Physiol Heart Circ Physiol 2011.[57] Rao X, Zhong J, Zhang S, Zhang Y, Yu Q, Yang P, et al. Loss of Methyl-CpG-B<strong>in</strong>d<strong>in</strong>g Doma<strong>in</strong> Prote<strong>in</strong> 2 EnhancesEndothelial Angiogenesis and Protects Mice Aga<strong>in</strong>st H<strong>in</strong>d-Limb Ischemic Injury. Circulation 2011;123:2964e74.[58] Xiao Q, Zeng L, Zhang Z, Margariti A, Ali ZA, Channon KM, et al. Sca-1þ progenitors derived from embryonicstem cells differentiate <strong>in</strong>to endothelial cells capable of vascular repair after arterial <strong>in</strong>jury. Arterioscler ThrombVasc Biol 2006;26:2244e51.[59] Zeng L, Xiao Q, Margariti A, Zhang Z, Zampetaki A, Patel S, et al. HDAC3 is crucial <strong>in</strong> shear- and VEGF<strong>in</strong>ducedstem cell differentiation toward endothelial cells. J Cell Biol 2006;174:1059e69.[60] Zampetaki A, Zeng L, Margariti A, Xiao Q, Li H, Zhang Z, et al. Histone deacetylase 3 is critical <strong>in</strong> endothelialsurvival and atherosclerosis development <strong>in</strong> response to disturbed flow. Circulation 2010;121:132e42.[61] Hou J, Chong ZZ, Shang YC, Maiese K. Early apoptotic vascular signal<strong>in</strong>g is determ<strong>in</strong>ed by Sirt1 throughnuclear shuttl<strong>in</strong>g, forkhead traffick<strong>in</strong>g, bad, and mitochondrial caspase activation. Curr Neurovasc Res2010;7:95e112.[62] Zu Y, Liu L, Lee MY, Xu C, Liang Y, Man RY, et al. SIRT1 promotes proliferation and prevents senescencethrough target<strong>in</strong>g LKB1 <strong>in</strong> primary porc<strong>in</strong>e aortic endothelial cells. Circ Res 2010;106:1384e93.[63] Michaelis M, Suhan T, Michaelis UR, Beek K, Rothweiler F, Tausch L, et al. Valproic acid <strong>in</strong>duces extracellularsignal-regulated k<strong>in</strong>ase 1/2 activation and <strong>in</strong>hibits apoptosis <strong>in</strong> endothelial cells. Cell Death Differ2006;13:446e53.[64] Banerjee S, Bacanamwo M. DNA methyltransferase <strong>in</strong>hibition <strong>in</strong>duces mouse embryonic stem cell differentiation<strong>in</strong>to endothelial cells. Exp Cell Res 2010;316:172e80.[65] Laukkanen MO, Mannermaa S, Hiltunen MO, Aittomaki S, Airenne K, Janne J, et al. Local hypomethylation <strong>in</strong>atherosclerosis found <strong>in</strong> rabbit ec-sod gene. Arterioscler Thromb Vasc Biol 1999;19:2171e8.[66] Post WS, Goldschmidt-Clermont PJ, Wilhide CC, Heldman AW, Sussman MS, Ouyang P, et al. Methylation ofthe estrogen receptor gene is associated with ag<strong>in</strong>g and atherosclerosis <strong>in</strong> the cardiovascular system. CardiovascRes 1999;43:985e91.


CHAPTER 20The Role of <strong>Epigenetics</strong> <strong>in</strong> Cardiovascular <strong>Disease</strong>[67] Y<strong>in</strong>g AK, Hassana<strong>in</strong> HH, Roos CM, Smiraglia DJ, Issa JJ, Michler RE, et al. Methylation of the estrogen receptoralphagene promoter is selectively <strong>in</strong>creased <strong>in</strong> proliferat<strong>in</strong>g human aortic smooth muscle cells. Cardiovasc Res2000;46:172e9.[68] Sedd<strong>in</strong>g DG, Trobs M, Reich F, Walker G, F<strong>in</strong>k L, Haberbosch W, et al. 3-Deazaadenos<strong>in</strong>e prevents smooth musclecell proliferation and neo<strong>in</strong>tima formation by <strong>in</strong>terfer<strong>in</strong>g with Ras signal<strong>in</strong>g. Circ Res 2009;104:1192e200.[69] Villeneuve LM, Reddy MA, Lant<strong>in</strong>g LL, Wang M, Meng L, Natarajan R. Epigenetic histone H3 lys<strong>in</strong>e 9methylation <strong>in</strong> metabolic memory and <strong>in</strong>flammatory phenotype of vascular smooth muscle cells <strong>in</strong> diabetes.Proc Natl Acad Sci USA 2008;105:9047e52.[70] Okamoto H, Fujioka Y, Takahashi A, Takahashi T, Taniguchi T, Ishikawa Y, et al. Trichostat<strong>in</strong> A, an <strong>in</strong>hibitor ofhistone deacetylase, <strong>in</strong>hibits smooth muscle cell proliferation via <strong>in</strong>duction of p21(WAF1). J AtherosclerThromb 2006;13:183e91.[71] Song S, Kang SW, Choi C. Trichostat<strong>in</strong> A enhances proliferation and migration of vascular smooth muscle cellsby downregulat<strong>in</strong>g thioredox<strong>in</strong> 1. Cardiovasc Res 2010;85:241e9.[72] Zhou B, Margariti A, Zeng L, Habi O, Xiao Q, Mart<strong>in</strong> D, et al. Splic<strong>in</strong>g of Histone Deacetylase 7 ModulatesSmooth Muscle Cell Proliferation and Neo<strong>in</strong>tima Formation Through Nuclear b-Caten<strong>in</strong> Translocation.Arterioscler Thromb Vasc Biol 2011.[73] Yan ZQ, Yao QP, Zhang ML, Qi YX, Guo ZY, Shen BR, et al. Histone deacetylases modulate vascular smoothmuscle cell migration <strong>in</strong>duced by cyclic mechanical stra<strong>in</strong>. J Biomech 2009;42:945e8.[74] Bennett MR, Evan GI, Schwartz SM. Apoptosis of human vascular smooth muscle cells derived from normalvessels and coronary atherosclerotic plaques. J Cl<strong>in</strong> Invest 1995;95:2266e74.[75] Patel VA, Zhang QJ, Siddle K, Soos MA, Goddard M, Weissberg PL, et al. Defect <strong>in</strong> <strong>in</strong>sul<strong>in</strong>-like growth factor-1survival mechanism <strong>in</strong> atherosclerotic plaque-derived vascular smooth muscle cells is mediated by reducedsurface b<strong>in</strong>d<strong>in</strong>g and signal<strong>in</strong>g. Circ Res 2001;88:895e902.[76] Kavurma MM, Figg N, Bennett MR, Mercer J, Khachigian LM, Littlewood TD. Oxidative stress regulates IGF1Rexpression <strong>in</strong> vascular smooth-muscle cells via p53 and HDAC recruitment. Biochem J 2007;407:79e87.[77] van der Veer E, Nong Z, O’Neil C, Urquhart B, Freeman D, Picker<strong>in</strong>g JG. Pre-B-cell colony-enhanc<strong>in</strong>g factorregulates NADþ-dependent prote<strong>in</strong> deacetylase activity and promotes vascular smooth muscle cell maturation.Circ Res 2005;97:25e34.[78] Han CI, Campbell GR, Campbell JH. Circulat<strong>in</strong>g bone marrow cells can contribute to neo<strong>in</strong>timal formation.J Vasc Res 2001;38:113e9.[79] Li J, Han X, Jiang J, Zhong R, Williams GM, Picker<strong>in</strong>g JG, et al. Vascular smooth muscle cells of recipient orig<strong>in</strong>mediate <strong>in</strong>timal expansion after aortic allotransplantation <strong>in</strong> mice. Am J Pathol 2001;158:1943e7.[80] Sata M, Saiura A, Kunisato A, Tojo A, Okada S, Tokuhisa T, et al. Hematopoietic stem cells differentiate <strong>in</strong>tovascular cells that participate <strong>in</strong> the pathogenesis of atherosclerosis. Nat Med 2002;8:403e9.[81] Hu Y, Davison F, Ludewig B, Erdel M, Mayr M, Url M, et al. Smooth muscle cells <strong>in</strong> transplant atheroscleroticlesions are orig<strong>in</strong>ated from recipients, but not bone marrow progenitor cells. Circulation 2002;106:1834e9.[82] Hu Y, Zhang Z, Torsney E, Afzal AR, Davison F, Metzler B, et al. Abundant progenitor cells <strong>in</strong> the adventitiacontribute to atherosclerosis of ve<strong>in</strong> grafts <strong>in</strong> ApoE-deficient mice. J Cl<strong>in</strong> Invest 2004;113:1258e65.[83] Shimizu K, Sugiyama S, Aikawa M, Fukumoto Y, Rabk<strong>in</strong> E, Libby P, et al. Host bone-marrow cells are a sourceof donor <strong>in</strong>timal smooth- muscle-like cells <strong>in</strong> mur<strong>in</strong>e aortic transplant arteriopathy. Nat Med 2001;7:738e41.[84] Hu Y, Mayr M, Metzler B, Erdel M, Davison F, Xu Q. Both donor and recipient orig<strong>in</strong>s of smooth muscle cells<strong>in</strong> ve<strong>in</strong> graft atherosclerotic lesions. Circ Res 2002;91:e13e20.[85] Lockman K, Taylor JM, Mack CP. The histone demethylase, Jmjd1a, <strong>in</strong>teracts with the myocard<strong>in</strong> factors toregulate SMC differentiation marker gene expression. Circ Res 2007;101:e115e23.[86] Lee WJ, Kim HJ. Inhibition of DNA methylation is <strong>in</strong>volved <strong>in</strong> transdifferentiation of myoblasts <strong>in</strong>to smoothmuscle cells. Mol Cells 2007;24:441e4.[87] Sp<strong>in</strong> JM, Quertermous T, Tsao PS. Chromat<strong>in</strong> remodel<strong>in</strong>g pathways <strong>in</strong> smooth muscle cell differentiation, andevidence for an <strong>in</strong>tegral role for p300. PLoS One 2010;5:e14301.[88] Qiu P, Ritchie RP, Gong XQ, Hamamori Y, Li L. Dynamic changes <strong>in</strong> chromat<strong>in</strong> acetylation and the expressionof histone acetyltransferases and histone deacetylases regulate the SM22alpha transcription <strong>in</strong> response toSmad3-mediated TGFbeta1 signal<strong>in</strong>g. Biochem Biophys Res Commun 2006;348:351e8.[89] Sharma P, Kumar J, Garg G, Kumar A, Patowary A, Karthikeyan G, et al. Detection of altered global DNAmethylation <strong>in</strong> coronary artery disease patients. DNA Cell Biol 2008;27:357e65.[90] Hiltunen MO, Turunen MP, Hakk<strong>in</strong>en TP, Rutanen J, Hedman M, Mak<strong>in</strong>en K, et al. DNA hypomethylation andmethyltransferase expression <strong>in</strong> atherosclerotic lesions. Vasc Med 2002;7:5e11.[91] Kim J, Kim JY, Song KS, Lee YH, Seo JS, Jel<strong>in</strong>ek J, et al. Epigenetic changes <strong>in</strong> estrogen receptor beta gene <strong>in</strong>atherosclerotic cardiovascular tissues and <strong>in</strong>-vitro vascular senescence. Biochim Biophys Acta2007;1772:72e80.413


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>414[92] Lund G, Andersson L, Lauria M, L<strong>in</strong>dholm M, Fraga MF, Villar-Garea A, et al. DNA methylation polymorphismsprecede any histological sign of atherosclerosis <strong>in</strong> mice lack<strong>in</strong>g apolipoprote<strong>in</strong> E. J Biol Chem2004;279:29147e54.[93] Castillo-Diaz SA, Garay-Sevilla ME, Hernandez-Gonzalez MA, Solis-Mart<strong>in</strong>ez MO, Za<strong>in</strong>a S. Extensive demethylationof normally hypermethylated CpG islands occurs <strong>in</strong> human atherosclerotic arteries. Int J MolMed 2010;26:691e700.[94] Davies PF, Polacek DC, Handen JS, Helmke BP, DePaola N. A spatial approach to transcriptional profil<strong>in</strong>g:mechanotransduction and the focal orig<strong>in</strong> of atherosclerosis. Trends Biotechnol 1999;17:347e51.[95] Granger A, Abdullah I, Huebner F, Stout A, Wang T, Huebner T, et al. Histone deacetylase <strong>in</strong>hibition reducesmyocardial ischemia-reperfusion <strong>in</strong>jury <strong>in</strong> mice. FASEB J 2008;22:3549e60.[96] Yanazume T, Hasegawa K, Morimoto T, Kawamura T, Wada H, Matsumori A, et al. Cardiac p300 is <strong>in</strong>volved <strong>in</strong>myocyte growth with decompensated heart failure. Mol Cell Biol 2003;23:3593e606.[97] Kawamura T, Ono K, Morimoto T, Wada H, Hirai M, Hidaka K, et al. Acetylation of GATA-4 is <strong>in</strong>volved <strong>in</strong> thedifferentiation of embryonic stem cells <strong>in</strong>to cardiac myocytes. J Biol Chem 2005;280:19682e8.[98] Miyamoto S, Kawamura T, Morimoto T, Ono K, Wada H, Kawase Y, et al. Histone acetyltransferase activity ofp300 is required for the promotion of left ventricular remodel<strong>in</strong>g after myocardial <strong>in</strong>farction <strong>in</strong> adult mice<strong>in</strong> vivo. Circulation 2006;113:679e90.[99] Wei JQ, Shehadeh LA, Mitrani JM, Pessanha M, Slepak TI, Webster KA, et al. Quantitative control of adaptivecardiac hypertrophy by acetyltransferase p300. Circulation 2008;118:934e46.[100] Hosse<strong>in</strong>khani M, Hasegawa K, Ono K, Kawamura T, Takaya T, Morimoto T, et al. Trichostat<strong>in</strong> A <strong>in</strong>ducesmyocardial differentiation of monkey ES cells. Biochem Biophys Res Commun 2007;356:386e91.[101] Kaneda R, Ueno S, Yamashita Y, Choi YL, Ko<strong>in</strong>uma K, Takada S, et al. Genome-wide screen<strong>in</strong>g for targetregions of histone deacetylases <strong>in</strong> cardiomyocytes. Circ Res 2005;97:210e8.[102] Kee HJ, Eom GH, Joung H, Sh<strong>in</strong> S, Kim JR, Cho YK, et al. Activation of histone deacetylase 2 by <strong>in</strong>ducible heatshock prote<strong>in</strong> 70 <strong>in</strong> cardiac hypertrophy. Circ Res 2008;103:1259e69.[103] Backs J, Song K, Bezprozvannaya S, Chang S, Olson EN. CaM k<strong>in</strong>ase II selectively signals to histone deacetylase4 dur<strong>in</strong>g cardiomyocyte hypertrophy. J Cl<strong>in</strong> Invest 2006;116:1853e64.[104] Lemon DD, Horn TR, Cavas<strong>in</strong> MA, Jeong MY, Haubold KW, Long CS, et al. Cardiac HDAC6 catalytic activityis <strong>in</strong>duced <strong>in</strong> response to chronic hypertension. J Mol Cell Cardiol 2011;51:41e50.[105] Zhang CL, McK<strong>in</strong>sey TA, Chang S, Antos CL, Hill JA, Olson EN. Class II histone deacetylases act as signalresponsiverepressors of cardiac hypertrophy. Cell 2002;110:479e88.[106] Alcendor RR, Kirshenbaum LA, Imai S, Vatner SF, Sadoshima J. Silent <strong>in</strong>formation regulator 2alpha,a longevity factor and class III histone deacetylase, is an essential endogenous apoptosis <strong>in</strong>hibitor <strong>in</strong> cardiacmyocytes. Circ Res 2004;95:971e80.[107] Castro R, Rivera I, Struys EA, Jansen EE, Ravasco P, Camilo ME, et al. Increased homocyste<strong>in</strong>e and S-adenosylhomocyste<strong>in</strong>e concentrations and DNA hypomethylation <strong>in</strong> vascular disease. Cl<strong>in</strong> Chem2003;49:1292e6.[108] Baccarelli A, Wright R, Bollati V, Litonjua A, Zanobetti A, Tarant<strong>in</strong>i L, et al. Ischemic heart disease and stroke<strong>in</strong> relation to blood DNA methylation. Epidemiology 2010;21:819e28.[109] Kim M, Long TI, Arakawa K, Wang R, Yu MC, Laird PW. DNA methylation as a biomarker for cardiovasculardisease risk. PLoS One 2010;5:e9692.[110] Ren XP, Wu J, Wang X, Sartor MA, Qian J, Jones K, et al. MicroRNA-320 is <strong>in</strong>volved <strong>in</strong> the regulation of cardiacischemia/reperfusion <strong>in</strong>jury by target<strong>in</strong>g heat-shock prote<strong>in</strong> 20. Circulation 2009;119:2357e66.[111] Wang GK, Zhu JQ, Zhang JT, Li Q, Li Y, He J, et al. Circulat<strong>in</strong>g microRNA: a novel potential biomarker forearly diagnosis of acute myocardial <strong>in</strong>farction <strong>in</strong> humans. Eur Heart J 2010;31:659e66.[112] Small EM, Frost RJ, Olson EN. MicroRNAs add a new dimension to cardiovascular disease. Circulation2010;121:1022e32.[113] Muller S, Kramer OH. Inhibitors of HDACseeffective drugs aga<strong>in</strong>st cancer? Curr Cancer Drug Targets2010;10:210e28.[114] Hirtz D, Iannaccone S, Heemskerk J, Gw<strong>in</strong>n-Hardy K, Moxley 3rd R, Rowland LP. Challenges and opportunities<strong>in</strong> cl<strong>in</strong>ical trials for sp<strong>in</strong>al muscular atrophy. Neurology 2005;65:1352e7.[115] Marks PA, Xu WS. Histone deacetylase <strong>in</strong>hibitors: Potential <strong>in</strong> cancer therapy. J Cell Biochem2009;107:600e8.


CHAPTER 21<strong>Epigenetics</strong> and <strong>Human</strong>Infectious <strong>Disease</strong>sHans Helmut Niller 1 , Janos M<strong>in</strong>arovits 21 Institute for Medical Microbiology and Hygiene at the University of Regensburg,Regensburg, Germany2 National Center for Epidemiology, Budapest, HungaryCHAPTER OUTLINE21.1 Introduction 41621.2 Epigenetic Modifications Elicited<strong>in</strong> Host Cells Dur<strong>in</strong>g BacterialInfections 41721.2.1 Epigenetic Alterations Inducedby Bacterial Infections<strong>in</strong> Periodontal <strong>Disease</strong> 41721.2.2 Histone modifications Causedby Listeriamonocytogenes 41821.2.3 Histone Dephosphorylation byOspF, the Effector Prote<strong>in</strong>of Shigella flexneri 41821.2.4 Histone Modifications <strong>in</strong>Anaplasma phagocytophilum-Infected Cells 41821.2.5 Epigenetic AlterationsAssociated with Helicobacterpylori Infection 419CpG-Methylation Profiles ofHelicobacter pylori-AssociatedGastric Carc<strong>in</strong>oma 419Epigenetic silenc<strong>in</strong>g of miRNAGenes <strong>in</strong> Helicobacter pylori-Associated Neoplasms 420The Role of Polycomb GroupProte<strong>in</strong>s and Histone Modifications<strong>in</strong> Helicobacter pylori-Associated GastricCancer 42121.2.6 Uropathogenic Escherichia coliInfection Down-RegulatesCDKN2A (p16 INK4A ) 42121.2.7 Chlamydophila spp. EncodedHistone Methyltransferases:Putative Inducers of EpigeneticReprogramm<strong>in</strong>g <strong>in</strong> Host CellNuclei 42121.3 Virus-<strong>in</strong>duced EpigeneticAlterations 42221.3.1 Epigenetic Alterations <strong>in</strong> CellsCarry<strong>in</strong>g LatentGammaherpesvirusGenomes 422Epigenetic Reprogramm<strong>in</strong>g <strong>in</strong>Epste<strong>in</strong>eBarr Virus-AssociatedNeoplasms 422Epigenetic Alterations <strong>in</strong>Neoplasms Carry<strong>in</strong>gKaposi’s Sarcoma-Associated HerpesvirusGenomes 42421.3.2 Interactions of Lytic CycleProte<strong>in</strong>s of <strong>Human</strong>Cytomegalovirus with HistoneDeacetylases 42521.3.3 Epigenetic Dysregulation <strong>in</strong><strong>Human</strong> Retrovirus-InfectedCells 426CpG Island MethylatorPhenotype <strong>in</strong> AdultT-Cell Leukemia/Lymphoma(ATLL): A Putative Role for theHTLV-1 Prote<strong>in</strong> Tax <strong>in</strong> theSilenc<strong>in</strong>g of Key CellularPromoters? 426415T. Tollefsbol (Ed): <strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>. DOI: 10.1016/B978-0-12-388415-2.00021-4Copyright Ó 2012 Elsevier Inc. All rights reserved.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong><strong>Human</strong> ImmunodeficiencyVirus: Epigenetic Silenc<strong>in</strong>g ofLatent Viral Genomes and HostGenes 42621.3.4 Local Hypermethylation andGlobal Hypomethylation of theHost Cell DNA <strong>in</strong> HepatocellularCarc<strong>in</strong>omas Associated withHepatitis B Virus 42821.3.5 Hepatitis C Virus (HCV)-Induced EpigeneticAlterations 42921.3.6 Induction of HistoneMethyltransferase and HistoneDemethylase Enzymes by the<strong>Human</strong> PapillomavirusOncoprote<strong>in</strong> E7 43121.3.7 Epigenetic TranscriptionalSilenc<strong>in</strong>g <strong>in</strong> Merkel CellPolyomavirus-AssociatedCarc<strong>in</strong>oma of the Sk<strong>in</strong> 43121.4 Epigenetic Alterations Elicited <strong>in</strong>the Host Tissue by TrematodeInfections 43221.4.1 Promoter Hypermethylation <strong>in</strong>Cholangiocarc<strong>in</strong>omaAssociated with Opistorchisviverr<strong>in</strong>i (Liver Fluke)Infection 43221.4.2 CpG Island Methylation <strong>in</strong>Schistosoma haematobium-Associated BladderCarc<strong>in</strong>oma 43221.5 Conclusions 432References 43341621.1 INTRODUCTIONPatho-epigenetics is a new discipl<strong>in</strong>e deal<strong>in</strong>g with the pathological consequences of dysregulatedepigenetic processes [1]. In recent years it became more and more obvious that eperhaps follow<strong>in</strong>g the lead of cancer research, where epigenetic and genetic theoriesof neoplastic development complement each other peacefully e epigenetic ideas found theirway to virtually all areas of biomedical research. Look<strong>in</strong>g at the exponentially accumulat<strong>in</strong>gdata one has the impression that epigenetic alterations, <strong>in</strong>duced by certa<strong>in</strong> pathogenic virusesand bacteria <strong>in</strong> the host cells they are <strong>in</strong>teract<strong>in</strong>g with, play an unexpected but most importantrole <strong>in</strong> disease <strong>in</strong>itiation and progression. In this review we focus ma<strong>in</strong>ly on human pathogenselicit<strong>in</strong>g epigenetic changes relevant, or at least potentially relevant, to disease <strong>in</strong>itiation orprogression. Although the idea that microbes do <strong>in</strong>duce epigenetic changes <strong>in</strong> host cells isga<strong>in</strong><strong>in</strong>g more and more support, so far only a m<strong>in</strong>ority of <strong>in</strong>fectious agents was analyzed <strong>in</strong>this respect. Thus, <strong>in</strong> this chapter we do not discuss, due to the lack of available data on theirpotential patho-epigenetic effects, even those pathogens, <strong>in</strong>clud<strong>in</strong>g protozoan parasites andfungi, that are known to use their own sophisticated epigenetic mechanisms to control theexpression of their genomes [2e6]. For the very same reason, we omit alphaherpesviruses <strong>in</strong>spite of the fact that their latent genomes undergo epigenetic changes <strong>in</strong> their host cells [7,8],and oncogenic human adenoviruses that <strong>in</strong>duce malignant tumors <strong>in</strong> experimental animalsand elicit spectacular epigenetic alterations <strong>in</strong> tissue culture [9,10], but lack any associationwith human neoplasms. As to the bacterial pathogens discussed <strong>in</strong> this chapter, the epigeneticaspects of Helicobacter pylori <strong>in</strong>fection were analyzed most <strong>in</strong>tensively, due to its associationwith gastric carc<strong>in</strong>oma. Regard<strong>in</strong>g viruses <strong>in</strong>fect<strong>in</strong>g humans, the epigenotypes of tumorassociatedDNA viruses and proviral DNA copies of retroviral genomes were characterized <strong>in</strong>detail. These data have potential implications as to the therapy of neoplasms and elim<strong>in</strong>ationof latent reservoirs of human immunodeficiency virus and human T-cell lymphotropic virus.Decipher<strong>in</strong>g how viral and other microbial prote<strong>in</strong>s and non-translated RNAs alter the hostcell epigenome may result <strong>in</strong> new tools permitt<strong>in</strong>g early and stage-specific detection of pathologicalalterations. Epigenetic studies may contribute to a better understand<strong>in</strong>g of the diseasescaused by <strong>in</strong>fectious pathogens, first of all microbes, but also macroparasites, <strong>in</strong>clud<strong>in</strong>ghelm<strong>in</strong>ths, fungi, and arthropods.


CHAPTER 21<strong>Epigenetics</strong> and <strong>Human</strong> Infectious <strong>Disease</strong>s21.2 EPIGENETIC MODIFICATIONS ELICITED IN HOST CELLSDURING BACTERIAL INFECTIONSBacteria may affect the epigenetic regulatory mechanisms of their hosts via the metabolicproducts and tox<strong>in</strong>s released <strong>in</strong>to their environment. In addition, certa<strong>in</strong> pathogens synthesizeeffector prote<strong>in</strong>s capable of <strong>in</strong>duc<strong>in</strong>g epigenetic alterations. Such effector prote<strong>in</strong>s are either<strong>in</strong>jected <strong>in</strong>to their target cells or enter the host cell nuclei as the products of bacteria that arecapable of surviv<strong>in</strong>g with<strong>in</strong> the cytoplasm of <strong>in</strong>fected cells. In several cases the exact mechanismof epigenetic changes elicited by bacterial <strong>in</strong>fections rema<strong>in</strong>s to be clarified.21.2.1 Epigenetic Alterations Induced by Bacterial Infections<strong>in</strong> Periodontal <strong>Disease</strong>Pathogenic Gram-negative anaerobic bacteria populat<strong>in</strong>g the tooth-associated biofilm maycause an <strong>in</strong>flammatory reaction (periodontitis). The result<strong>in</strong>g chronic <strong>in</strong>flammation, periodontaldisease, affects the majority (50e90%) of the worldwide population [11]. In additionto its local consequences (occasional pa<strong>in</strong>, impaired mastication, loosen<strong>in</strong>g and loss of teeth),subg<strong>in</strong>gival <strong>in</strong>fection may have a systemic impact <strong>in</strong>clud<strong>in</strong>g adverse pregnancy outcomes,atherosclerosis, rheumatoid arthritis, diabetes, and aspiration pneumonia. A critical step <strong>in</strong>periodontal disease progression is the disruption of the host <strong>in</strong>nate immune system byanaerobic bacteria (reviewed by [12]). The cells of the <strong>in</strong>nate immune system, <strong>in</strong>clud<strong>in</strong>gg<strong>in</strong>gival epithelial cells (GECs), recognize pathogen-associated molecular patterns <strong>in</strong>teract<strong>in</strong>gwith Toll-like receptors (TLRs) and other receptor classes, and respond by the production ofantimicrobial peptides such as human b-defens<strong>in</strong>s and chemok<strong>in</strong>es that activate adaptiveimmune responses as well [13e15]. In addition to Porphyromonas g<strong>in</strong>givalis, Tannerella forsythia,Treponema denticola, and Prevotella <strong>in</strong>termedia, the pathogenic bacteria caus<strong>in</strong>g periodontitis,a series of non-pathogenic bacteria (e.g. Fusobacterium nucleatum) also <strong>in</strong>habit the oral cavity ofperiodontitis patients, form<strong>in</strong>g a microbial community (reviewed by [12]). Similarly to theirpathogenic counterparts, non-pathogenic bacteria may also <strong>in</strong>teract with GECs, modulat<strong>in</strong>gnative immune respones to <strong>in</strong>vad<strong>in</strong>g pathogens.417Porphyromonas g<strong>in</strong>givalis, one of the major etiological agents of periodontal disease, elicitedcomplex changes when <strong>in</strong>cubated with g<strong>in</strong>gival epithelial cells: it suppressed the expression ofhistone deacetylase 1 and 2 (HDAC1 and HDAC2) and DNA methyltransferase 1 (DNMT1)[16]. In parallel, it stimulated human b-defens<strong>in</strong> and CCL20 (CC chemok<strong>in</strong>e ligand 20)expression and <strong>in</strong>creased promoter methylation of six genes, <strong>in</strong>clud<strong>in</strong>g the immune regulatorCD276, elastase 2, TLR2, IL-12A, and two putative tumor-suppressor genes (TSG). The level ofactivat<strong>in</strong>g histone modification H3K4me3 decreased <strong>in</strong> GECs <strong>in</strong>cubated with Porphyromonasg<strong>in</strong>givalis, but not <strong>in</strong> the presence of the non-pathogenic Fusobacterium nucleatum [16]. Shortcha<strong>in</strong>fatty acids, the metabolic by-products of Porphyromonas g<strong>in</strong>givalis, are secreted extracellularlyand may affect local immune responses, contribut<strong>in</strong>g thereby to the development ofperiodontal disease [17e19]. Butyric acid, the major short-cha<strong>in</strong> fatty acid species produced bypathogenic Porphyromonas g<strong>in</strong>givalis stra<strong>in</strong>s, accumulated <strong>in</strong> periodontal pockets [18e20], andit was suggested that as a HDAC <strong>in</strong>hibitor [21,22] (reviewed by [23]), butyric acid may <strong>in</strong>duceacetylation of histone H3 and H4 <strong>in</strong> neighbor<strong>in</strong>g cells. Indeed, supernatants of Porphyromonasg<strong>in</strong>givalis cultures, and butyric acid <strong>in</strong>duced histone acetylation <strong>in</strong> T-cells and macrophagescarry<strong>in</strong>g latent proviral genomes of type 1 human immunodeficiency virus (HIV-1), andcaused HIV-1 reactivation [20]. The role of periodontal disease <strong>in</strong> the progression of acquiredimmunodeficiency syndrome needs further studies.Another bacterium <strong>in</strong>volved <strong>in</strong> periodontal <strong>in</strong>fections, Campylobacter rectus may also <strong>in</strong>duceepigenetic alterations <strong>in</strong> human cells. In experimental mice Campylobacter rectus <strong>in</strong>fectiondown-regulated the expression of the Igf2 (<strong>in</strong>sul<strong>in</strong>-like growth factor 2) gene via hypermethylationof the Igf2 promoter <strong>in</strong> the mur<strong>in</strong>e placenta [24]. This epigenetic change resulted<strong>in</strong> reduced placental growth and fetal growth restriction, suggest<strong>in</strong>g that a similar mechanism


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>may be <strong>in</strong>volved <strong>in</strong> preterm births associated with Campylobacter rectus <strong>in</strong>fection <strong>in</strong> humans[25]. The potential epigenetic consequences of Campylobacter rectuseg<strong>in</strong>gival epithelial cell<strong>in</strong>teractions rema<strong>in</strong> to be established.21.2.2 Histone modifications Caused by Listeria monocytogenesThe Gram-positive bacterium Listeria monocytogenes appears to cause only little <strong>in</strong>flammationdur<strong>in</strong>g <strong>in</strong>fection. L. monocytogenes is associated with foodborne <strong>in</strong>fections, caus<strong>in</strong>g usually selflimit<strong>in</strong>ggastroenteritis, but also sepsis and men<strong>in</strong>gitis <strong>in</strong> immunocompromised patients,per<strong>in</strong>atal and <strong>in</strong>trauter<strong>in</strong>e <strong>in</strong>fection, and abortion <strong>in</strong> pregnant women. It acquired the tools forsurvival with<strong>in</strong> the cytoplasm of macrophages and endothelial cells (reviewed by [26]). L.monocytogenes appears to use its tox<strong>in</strong> listeriolys<strong>in</strong> O (LLO) not only as a pore-form<strong>in</strong>g tox<strong>in</strong> toescape from the phagosome and enter the host cytoplasm, but also <strong>in</strong> an extracellular form,before enter<strong>in</strong>g the host cell, to <strong>in</strong>duce histone modifications <strong>in</strong> the cells to be <strong>in</strong>fected via anunexplored signal<strong>in</strong>g pathway. Extracellular, pathogenic Listeria or purified LLO prote<strong>in</strong><strong>in</strong>duced, at a subset of genes and with<strong>in</strong> 3 hours or even with<strong>in</strong> 20 m<strong>in</strong>utes, dephosphorylationof histone H3 at ser<strong>in</strong>e 10 and deacetylation of histone H4 [27]. Pore formation wasdispensable for the <strong>in</strong>duction of histone H3 dephosphorylation. These changes correlatedwith a decreased expression of the chemoattractant chemok<strong>in</strong>e CXCL2 <strong>in</strong>volved <strong>in</strong> therecruitment of polymorphonuclear cells dur<strong>in</strong>g epithelial cell <strong>in</strong>fection. Expression of thephosphatase DUSP4/MKP2 was also down-regulated, similarly to <strong>in</strong>terferon regulatory factor-3 (IRF-3) and the transcription factor EGR1 (early growth response 1). Thus, L. monocytogenesand its tox<strong>in</strong>, LLO, down-regulates key mediators of <strong>in</strong>nate immunity before <strong>in</strong>vad<strong>in</strong>g the hostcells, by elicit<strong>in</strong>g targeted, local histone modifications at key immune response genes. Thesituation is complex, however, because transcriptome analysis revealed not only 47 repressed,but also 99 <strong>in</strong>duced genes after 20 m<strong>in</strong> of LLO <strong>in</strong>cubation [27].418Pore-form<strong>in</strong>g tox<strong>in</strong>s of other bacteria, belong<strong>in</strong>g to different genera and unrelated to Listeriamonocytogenes, also <strong>in</strong>duced dephosphorylation of histone H3. These cholesterol-dependentcytolys<strong>in</strong>es of the LLO family, perfr<strong>in</strong>golys<strong>in</strong> (PFO) produced by Clostridium perfr<strong>in</strong>gens andpneumolys<strong>in</strong> (PLY) secreted by Streptococcus pneumoniae also exerted their histone-modify<strong>in</strong>geffect <strong>in</strong> a pore-form<strong>in</strong>g-<strong>in</strong>dependent manner [27].21.2.3 Histone Dephosphorylation by OspF, the Effector Prote<strong>in</strong>of Shigella flexneriShigella flexneri, a Gram-negative bacterium, causes diarrhea that is usually self-limit<strong>in</strong>g. Lifethreaten<strong>in</strong>gdisease may develop, however, <strong>in</strong> the absence of adequate medical care or <strong>in</strong>immunocompromised patients [28]. Shigella flexneri abrogated histone H3 phosphorylation atselected cellular promoters by <strong>in</strong>jection of a phosphatase <strong>in</strong>to epithelial cells [29]. The effectorprote<strong>in</strong> OspF entered the nucleus and dephosphorylated the mitogen-activated cellularprote<strong>in</strong> k<strong>in</strong>ases Erk and p38, thereby block<strong>in</strong>g histone H3 phosphorylation at Ser10 [29].These events resulted <strong>in</strong> repression of a narrow set of genes <strong>in</strong>clud<strong>in</strong>g the NF-kB responsive IL8and CCL20. By down-regulat<strong>in</strong>g IL-8 expression, OspF blocked neutrophil recruitment <strong>in</strong> vivo<strong>in</strong> a rabbit model of <strong>in</strong>fection [29].21.2.4 Histone Modifications <strong>in</strong> Anaplasma phagocytophilum-InfectedCellsAnaplasma phagocytophilum, a tick-transmitted rickettsial pathogen causes an acute febriledisease [30]. It is capable of <strong>in</strong>fect<strong>in</strong>g and surviv<strong>in</strong>g <strong>in</strong> granulocytes by block<strong>in</strong>g or delay<strong>in</strong>g keyantimicrobial mechanisms <strong>in</strong>clud<strong>in</strong>g oxidative burst, apoptosis, and phagocytosis, and downregulat<strong>in</strong>gdefense gene expression <strong>in</strong> its host cells [31]. AnkA, the effector prote<strong>in</strong> of Anaplasmaphagocytophilum is translocated <strong>in</strong>to the nucleus of the host where it b<strong>in</strong>ds to the host DNA andnuclear prote<strong>in</strong>s and silences the expression of 19 defense genes arranged <strong>in</strong>to three gene


CHAPTER 21<strong>Epigenetics</strong> and <strong>Human</strong> Infectious <strong>Disease</strong>sclusters. They code for antimicrobial peptides and enzymes or prote<strong>in</strong>s <strong>in</strong>volved <strong>in</strong> thegeneration of reactive oxygen <strong>in</strong>termediates. Silenc<strong>in</strong>g correlated with a transient <strong>in</strong>crease ofHDAC1 expression and a steady <strong>in</strong>crease of HDAC2 mRNA levels <strong>in</strong> Anaplasma phagocytophilum-<strong>in</strong>fectedcells. In addition, at 48 hours follow<strong>in</strong>g <strong>in</strong>fection of the THP-1 acutemonocytic leukemia cell l<strong>in</strong>e, an <strong>in</strong>creased b<strong>in</strong>d<strong>in</strong>g of HDAC1 and a parallel decrease <strong>in</strong>acetylated histone H3 was observed at most of the defense gene promoters, suggest<strong>in</strong>g thatAnaplasma phagocytophilum takes over the epigenetic control of host defense gene clusters [31].21.2.5 Epigenetic Alterations Associated with Helicobacter pyloriInfectionHelicobacter pylori (Hp), a Gram-negative spiral bacterium, colonizes more than half of the world’spopulation <strong>in</strong> early childhood. Primary <strong>in</strong>fection of the stomach regularly triggers an <strong>in</strong>flammation.Infection can lead to gastric or duodenal ulcer disease and different types of chronicgastritis (atrophic gastritis, enlarged fold gastritis, pangastritis). Chronic <strong>in</strong>flammation canprogress to gastric adenocarc<strong>in</strong>oma <strong>in</strong> about 1e2% of the <strong>in</strong>fected patients through the premalignantstages of gastric atrophy, <strong>in</strong>test<strong>in</strong>al metaplasia, and dysplasia. The risk of develop<strong>in</strong>g<strong>in</strong>test<strong>in</strong>al metaplasia or different types of malignant neoplasms (<strong>in</strong>test<strong>in</strong>al type gastric carc<strong>in</strong>oma(GC), sporadic diffuse type GC, MALT lymphoma) is significantly associated with Hp carrierstatus [32] (reviewed by [33,34]). Hp codes for the virulence factors vacuolat<strong>in</strong>g cytotox<strong>in</strong> VacAand cytotoxicity-associated antigen CagA. CagA is encoded with<strong>in</strong> the bacterial cag-pathogenicityisland (cag-PAI). Both prote<strong>in</strong>s are strongly associated with Hp pathogenesis. CagA, a bacterialoncoprote<strong>in</strong> can be <strong>in</strong>jected <strong>in</strong>to gastric epithelial cells by a type IV bacterial secretion system and<strong>in</strong>terfere with multiple cellular signal transduction pathways (reviewed by [35e37]). Throughactivation of the tyros<strong>in</strong>e phosphatase SHP-2, CagA may elicit uncontrolled cell proliferation, and<strong>in</strong>duce chromosomal <strong>in</strong>stability by destabiliz<strong>in</strong>g microtubules dur<strong>in</strong>g mitosis [38].Epigenetic dysregulation is strongly associated with gastric carc<strong>in</strong>ogenesis [39e41].Bothglobalgenomic hypomethylation and CpG island (CGI) hypermethylation correlated with the presenceof Hp <strong>in</strong> gastritis patients, and malignant progression <strong>in</strong> cancer patients [42e47]. Nontumorousgastric tissue of Hp-negative GC patients also showed <strong>in</strong>creased CGI hypermethylation[42,43,48]. Concordantly, Hp eradication led to a significant decl<strong>in</strong>e of CGIhypermethylation <strong>in</strong> gastritis patients, and <strong>in</strong> a small rodent model, but rema<strong>in</strong>ed still higherthan <strong>in</strong> non-<strong>in</strong>fected <strong>in</strong>dividuals [44,49e51] (reviewed by [52]) or animals [53]. With theprogress of Hp-<strong>in</strong>duced chronic gastritis through the advanced stages of atrophy, metaplasia,dysplasia, and GC, Hp was frequently lost from the gastric mucosa [54]. This expla<strong>in</strong>s e throughan epigenetic-based “hit-and-run” scenario e why the hypermethylation profile of gastricmucosa does not regularly correlate with the Hp-carrier status <strong>in</strong> the advanced stages [46].419CPG-METHYLATION PROFILES OF HELICOBACTER PYLORI-ASSOCIATEDGASTRIC CARCINOMATumor-suppressor genes <strong>in</strong> gastric carc<strong>in</strong>ogenesis are more frequently <strong>in</strong>activated by promotermethylation than by mutations [55]. One may wonder, however, whether epigenetic dysregulationis causative for carc<strong>in</strong>ogenesis or a mere epiphenomenon. The study of the E-cadher<strong>in</strong>(CDH1) promoter may help to clarify this question. CDH1 down-regulation decreases cellularadhesion and therefore <strong>in</strong>creases cellular motility and the disposition of cancer cells formetastasis. CDH1 germl<strong>in</strong>e mutation is regularly observed <strong>in</strong> families with hereditary diffuseGC. Thus, CDH1 <strong>in</strong>activation clearly predisposes to GC and is therefore co-causal [56]. SomaticCDH1 mutation is also common <strong>in</strong> sporadic GC, while transcriptional silenc<strong>in</strong>g by promoterCGI hypermethylation represents a second hit for carc<strong>in</strong>ogenesis [57,58]. The follow<strong>in</strong>gobservations support a causative role for epigenetic dysregulation of CDH1 <strong>in</strong> gastric carc<strong>in</strong>ogenesis:(1) CDH1 promoter hypermethylation is especially frequent <strong>in</strong> enlarged fold gastritis,a high-risk factor for GC [59]; (2) <strong>in</strong> the gastric mucosa of Hp-<strong>in</strong>fected patients CDH1 expressionis down-regulated [60], due to promoter methylation [42,49,50,61]; (3) epigenetic silenc<strong>in</strong>g of


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>CDH1 occurs early <strong>in</strong> gastric carc<strong>in</strong>ogenesis [44] (reviewed by [33]). Overall, hypermethylationof CDH1 is clearly co-causal <strong>in</strong> the development of sporadic GC (reviewed by [62]).The patterns of hypermethylated CGIs <strong>in</strong> non-cancerous mucosa from GC patients presenteda rather characteristic f<strong>in</strong>gerpr<strong>in</strong>t of Hp <strong>in</strong>fection [63,64]. Surpris<strong>in</strong>gly, however, and unlike <strong>in</strong>other malignancies, DNMTs were not <strong>in</strong>duced by Hp <strong>in</strong> human gastric mucosa [63], and theywere down-regulated when Hp was cocultured with gastric epithelial cells from gerbils [53].Incontrast, another study found an up-regulation of DNMT1 and DNMT3A through cocultur<strong>in</strong>gHp with two human GC cell l<strong>in</strong>es [65]. However, cancer cell l<strong>in</strong>es may react differently fromprimary cells. Thus, other mechanisms, like altered distribution of DNMTs or <strong>in</strong>flammation<strong>in</strong>duceddisturbance of factors protect<strong>in</strong>g CGIs aga<strong>in</strong>st methylation, may expla<strong>in</strong> HpassociatedCGI methylation [53,66,67]. The role of Hp-encoded 5 0 -CpG-DNMTs [68] rema<strong>in</strong>sto be explored.Ushijima observed a clear association between the methylation levels <strong>in</strong> the histologicallynormal gastric mucosae and the risk of gastric cancer development. He suggested that<strong>in</strong>creased CGI-hypermethylation predisposed to carc<strong>in</strong>ogenesis and proposed that the epigeneticallyaltered mucosal tissue corresponded to an epigenetic field for cancerization [55].Methylation was found at the p16, MLH1, ECAD, DAPK, and MTSS1 loci of the healthymucosa of GC patients with and without Hp-<strong>in</strong>fection [41,45,48,49]. The MLH1 repair-genemethylation was not seen <strong>in</strong> non-neoplastic epithelia from healthy persons, but was frequent<strong>in</strong> non-neoplastic tissue from GC patients [44,48], and it was significantly associated with the<strong>in</strong>test<strong>in</strong>al type of GC [69]. The metastasis suppressor gene MTSS1 is highly expressed <strong>in</strong>normal gastric mucosa and frequently affected by loss of heterozygosity <strong>in</strong> GC, suggest<strong>in</strong>ga physiological role for MTSS1 as a suppressor of gastric carc<strong>in</strong>ogenesis [41].420A series of TSGs and many other genomic loci were methylated at a higher level <strong>in</strong> Hp-positivethan Hp-negative cases [42,44,64,65,70e72]. The degree of methylation at the CDH1, p16,MLH1, APC, and COX2 loci correlated to malignant progression, with CDH1 methylation asan early, and that of MLH1 as a late, event. COX2 hypermethylation was associated with the Hpvirulence factor VacA [69], whereas hypermethylation at p16 and THBD (thrombomodul<strong>in</strong>)correlated significantly with <strong>in</strong>flammatory cell <strong>in</strong>filtration. Thus, chronic <strong>in</strong>flammation waspotentially <strong>in</strong>volved <strong>in</strong> methylation <strong>in</strong>duction [45,51]. Hp-eradication led to a decreasedmethylation at CDH1, p16, APC, FLNc, THBD, MGMT, and to the complete disappearance ofCOX2 (cyclooxygenase 2) methylation [44,51,72]. The specific methylation profiles associatedwith GC and Hp <strong>in</strong>fection may allow the development of new diagnostic tools to detectprecancerous stages, assess the risk of GC development, and estimate its prognosis [64].EPIGENETIC SILENCING OF miRNA GENES IN HELICOBACTER PYLORI-ASSOCIATED NEOPLASMSAndo et al. found that three putative tumor-suppressor miRNA genes, miR-124a1, miR-124a2,miR-124a3, were unmethylated <strong>in</strong> normal mucosa, but hypermethylated <strong>in</strong> tumor cell l<strong>in</strong>es. Inbiopsies, hypermethylation was strongly associated with a positive Hp carrier status [73]. miR-34b and miR-34c were highly methylated <strong>in</strong> GC, but not <strong>in</strong> normal gastric mucosa from Hpnegativepersons. Transfection of these miRNAs suppressed cell growth <strong>in</strong> vitro, suggest<strong>in</strong>ga tumor-suppressor function [74]. The tumor suppressor miR-203 was significantly downregulatedby hypermethylation <strong>in</strong> human low-grade MALT-lymphoma biopsies. Methylationof miR-203 co<strong>in</strong>cided with the <strong>in</strong>creased expression of its direct target, the non-receptortyros<strong>in</strong>e k<strong>in</strong>ase and oncogene ABL1 [75,76]. Overexpression of miR-203 <strong>in</strong> ABL1-express<strong>in</strong>ghuman lymphoma cells <strong>in</strong>hibited proliferation. Further, the ABL1 <strong>in</strong>hibitor imat<strong>in</strong>ib causedtumor regression <strong>in</strong> a MALT lymphoma mouse model based on chronic Helicobacter felis<strong>in</strong>fection [75]. Hypermethylation of the miR-124 and miR-34 groups and of miR-203 maycontribute to the epigenetic field for cancerization and may also serve as a predictive marker forGC risk [55,73,74].


CHAPTER 21<strong>Epigenetics</strong> and <strong>Human</strong> Infectious <strong>Disease</strong>sTHE ROLE OF POLYCOMB GROUP PROTEINS AND HISTONE MODIFICATIONS INHELICOBACTER PYLORI-ASSOCIATED GASTRIC CANCERPolycomb group (PcG) prote<strong>in</strong>s play an important role <strong>in</strong> reversibly repress<strong>in</strong>g transcriptionfactor genes which are <strong>in</strong>volved <strong>in</strong> development and differentiation <strong>in</strong> embryonic stem cells.This reversible PcG-mediated repression utilizes trimethylation on lys<strong>in</strong>e 27 of histone 3(H3K27me3). In cancer cells, but not <strong>in</strong> normal cells, the H3K27me3 chromat<strong>in</strong> markrecruited DNMTs, result<strong>in</strong>g <strong>in</strong> a permanent CpG-methylation [77]. Thus, <strong>in</strong> cancer cells,reversible repression was more frequently replaced by methylation and permanent silenc<strong>in</strong>g atstem-cell PcG-target promoters than at non-PcG target promoters [78] (reviewed by [79]). Aglutathione peroxidase knockout mouse model for <strong>in</strong>flammatory bowel disease and <strong>in</strong>test<strong>in</strong>alcancer confirmed these observations. Healthy wild-type mice mostly kept the H3K27me3chromat<strong>in</strong> mark at PcG target genes, while <strong>in</strong>flammation <strong>in</strong> KO mice led to aberrant methylationmostly of PcG targets which was later also observed <strong>in</strong> cancer tissue [80].Changes <strong>in</strong> global histone modification profiles and HDAC expression patterns correlatedwith the progression of GC <strong>in</strong> general and may serve as prognostic markers (reviewed by [35]).Further, the epigenetic repression through both DNA methylation and histone modification ofspecific genes <strong>in</strong>volved <strong>in</strong> chromat<strong>in</strong> remodel<strong>in</strong>g, cell cycle control and tumor suppression hasbeen l<strong>in</strong>ked with GC as well (reviewed by [35]). Coculture of Hp with mouse macrophagestriggered, through a secreted bacterial factor, NF-kB and MAPK signal<strong>in</strong>g and led to <strong>in</strong>creasedIL6 transcription via phosphorylation of histone H3 at the IL6 promoter [81]. In contrast,coculture of Hp with gastric epithelial cells caused a global decrease of histone H3 phosphorylationand deacetylation of H3 at lys<strong>in</strong>e 23 [82,83]. Histone dephosphorylation wasdependent on the presence of the Hp cag-PAI, but not on the CagA and VacA prote<strong>in</strong>s. Asa consequence of Hp-triggered epigenetic dysregulation, the expression of the c-jun protooncogene<strong>in</strong>creased, and that of hsp-70, cod<strong>in</strong>g for a heat shock prote<strong>in</strong>, decreased [83].21.2.6 Uropathogenic Escherichia coli Infection Down-RegulatesCDKN2A (p16 INK4A )Coculture of uropathogenic E. coli with human uroepithelial cell l<strong>in</strong>es strongly <strong>in</strong>ducedDNMT1 expression <strong>in</strong> comparison with non-pathogenic stra<strong>in</strong>s. In parallel, the tumorsuppressor CDKN2A and the DNA repair gene MGMT were down-regulated, while a set ofother genes (CDH1, MLH1, DAPK1, and TLR4) were not affected. Down-regulation ofCDKN2A correlated to DNA methylation of its promoter. However, the MGMT gene was notmethylated. Frequent UPEC <strong>in</strong>fections might <strong>in</strong>crease the risk for bladder cancer through<strong>in</strong>creas<strong>in</strong>g methylation of TSGs [84].42121.2.7 Chlamydophila spp. Encoded Histone Methyltransferases:Putative Inducers of Epigenetic Reprogramm<strong>in</strong>g <strong>in</strong> Host Cell NucleiChlamydophila pneumoniae, an obligatory <strong>in</strong>tracellular bacterium <strong>in</strong>volved <strong>in</strong> acute respiratorydiseases and implicated <strong>in</strong> chronic <strong>in</strong>flammatory processes, may modify, <strong>in</strong> pr<strong>in</strong>ciple, theepigenotype of its host cells. Similarly to Anaplasma phagocytophilum, C. pneumoniae <strong>in</strong>troducesa putative bacterial effector, <strong>in</strong> this case a SET doma<strong>in</strong> prote<strong>in</strong>, <strong>in</strong>to the host cell nucleus.Although histone-like prote<strong>in</strong>s play an important role <strong>in</strong> the lifecycle of Chlamydophila pneumoniae,its putative effector prote<strong>in</strong>, a histone methyltransferase (HMT), preferentially targetshistone H3 <strong>in</strong> mur<strong>in</strong>e host cells [85] (Table 21.1).Analogously, Chlamydia trachomatis codes for a SET doma<strong>in</strong>-prote<strong>in</strong>, called "nuclear effector"(NUE), which functions as a HMT. Dur<strong>in</strong>g <strong>in</strong>fection, NUE associates with the host cellchromat<strong>in</strong> and methylates histones H2B, H3, and H4 [86]. Modification of host-encodedhistones may reprogram the <strong>in</strong>fected cell, perhaps by silenc<strong>in</strong>g host defense genes, as theAnaplasma phagocytophilum effector AnkA happens to do it.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>TABLE 21.1 Epigenetic Alterations Elicited by Bacterial Infections <strong>in</strong> Host CellsMicrobe Effector Molecule Epigenetic Alteration EffectPorphyromonas Butyric acid Histone acetylation HIV-1 reactivationg<strong>in</strong>givalisListeriamonocytogenesListeriolys<strong>in</strong> O HistonedephosphorylationBlock<strong>in</strong>g <strong>in</strong>nateimmunityHistone deacetylationClostridiumPerfr<strong>in</strong>golys<strong>in</strong> Histone?perfr<strong>in</strong>gensdephosphorylationStreptococcus Pneumolys<strong>in</strong> Histone?pneumoniaedephosphorylationShigella flexneri OspF phosphatase Inhibition of histonephosphorylationBlock<strong>in</strong>g <strong>in</strong>nateimmunityAnaplasmaphagocytophilumAnkA Upregulation of HDAC Block<strong>in</strong>g defencegenesHelicobacter pylori Bacterial DNMTs ? ?? PromoterhypermethylationGastriccarc<strong>in</strong>ogenesis? Histone?dephosphorylationHistone deacetylationCampylobacter rectus ? Methylation of the Igf2 Pre-term birthspromoterUropathogenic E. coli ? PromoterCDKN2A silenc<strong>in</strong>ghypermethylationChlamydophila spp. HistonemethyltransferaseHistone methylation Gene silenc<strong>in</strong>g42221.3 VIRUS-INDUCED EPIGENETIC ALTERATIONSSimilarly to certa<strong>in</strong> bacteria, viruses harbor<strong>in</strong>g either RNA or DNA genomes can also elicitepigenetic changes <strong>in</strong> their host cells. Although most of the studies are related to “tumorviruses” that are associated with neoplasms, epigenetic dysregulation may contribute to othervirus-<strong>in</strong>duced pathological alterations as well.21.3.1 Epigenetic Alterations <strong>in</strong> Cells Carry<strong>in</strong>g LatentGammaherpesvirus GenomesGammaherpesvirus genomes regularly carry viral oncogenes and are associated, consequently,with malignant tumors. Some of the viral oncoprote<strong>in</strong>s encoded by human gammaherpesvirusesturned to be modifiers of the cellular epigenome.EPIGENETIC REPROGRAMMING IN EPSTEINeBARR VIRUS-ASSOCIATEDNEOPLASMSEpste<strong>in</strong>eBarr virus (EBV), a human gammaherpesvirus, is associated with a series of malignantneoplasms <strong>in</strong>clud<strong>in</strong>g lymphomas (Burkitt’s lymphoma, Hodgk<strong>in</strong>’s disease, T/NK-cell lymphoma,post-transplant lymphoproliferative disease, AIDS-associated lymphoma, X-l<strong>in</strong>ked lymphoproliferativesyndrome), carc<strong>in</strong>omas (nasopharyngeal carc<strong>in</strong>oma, gastric carc<strong>in</strong>oma, carc<strong>in</strong>omasof major salivary glands, thymic carc<strong>in</strong>oma, mammary carc<strong>in</strong>oma) and a sarcoma (leiomyosarcoma)(reviewed by [87]). In <strong>in</strong>fected cells, the term<strong>in</strong>al repeats of the l<strong>in</strong>ear double-strandedDNA packaged <strong>in</strong>to the virions fuse with each other. Accord<strong>in</strong>gly, latent EBV genomes persist <strong>in</strong>the tumor cells as circular episomes. They attach to the nuclear matrix <strong>in</strong> <strong>in</strong>terphase nuclei andcoreplicate with the cellular DNA once per cell cycle us<strong>in</strong>g oriP, the latent orig<strong>in</strong> of EBV replication.


CHAPTER 21<strong>Epigenetics</strong> and <strong>Human</strong> Infectious <strong>Disease</strong>sThe expression of latent EBV genes is cell type specific. Epigenetic regulatory mechanisms<strong>in</strong>clud<strong>in</strong>g DNA methylation and histone modifications, and b<strong>in</strong>d<strong>in</strong>g of key cellular regulatoryprote<strong>in</strong>s control the activity of the alternative promoters for transcripts encod<strong>in</strong>g the nuclearantigens EBNA1 to 6. Similar mechanisms affect the activity of promoters for transcriptsencod<strong>in</strong>g transmembrane prote<strong>in</strong>s (LMP1, LMP2A, LMP2B), too. In addition to the genestranscribed by RNA polymerase II, there are also two RNA polIII transcribed genes <strong>in</strong> the EBVgenome (EBER1, EBER2). The 5 0 and <strong>in</strong>ternal regulatory sequences of the EBER1 and EBER2transcription units are <strong>in</strong>variably unmethylated. The highly abundant EBER1 and EBER2 RNAsare not translated to prote<strong>in</strong>. In addition, microRNAs are also generated by RNase-process<strong>in</strong>gof latent EBV transcripts. EBV-derived microRNAs may <strong>in</strong>teract both with cellular and viralmRNAs, thereby modulat<strong>in</strong>g their post-transcriptional level [88].Based on the cell-type-specific epigenetic marks associated with latent EBV genomes one candist<strong>in</strong>guish between unique viral epigenotypes [7]. Although conta<strong>in</strong><strong>in</strong>g identical or nearlyidentical DNA sequences, each viral epigenotype is associated with a different pattern of geneexpression. In addition, alternative conformations adopted by the viral episomes <strong>in</strong> a latencytype-specificmanner may also affect the activity of latent EBV promoters [89].Latent EBV genomes are regularly targeted by epigenetic control mechanisms <strong>in</strong> different cell types.EBV-encoded oncoprote<strong>in</strong>s may, <strong>in</strong> turn, affect the activityof a set of cellular promoters. Thus, their<strong>in</strong>teraction with the cellular epigenetic regulatory mach<strong>in</strong>ery results <strong>in</strong> epigenetic “reprogramm<strong>in</strong>g”of the host cells. The nuclear antigen EBNA2 transactivates both viral and cellularpromoters. EBNA2 <strong>in</strong>teracts with the cellular histone acetyltransferases p300, CBP, and PCAF,whereas the leader prote<strong>in</strong> EBNA-LP (EBNA5) coactivates transcription by displac<strong>in</strong>g histonedeacetylase 4 from EBNA2-bound promoter sites [90,91]. At LMP1p, the EBNA2-associatedhistone acetyltransferases may counteract the silenc<strong>in</strong>g effect of histone deacetylases [90].EBNA3C (EBNA6) was associated both with histone acetylases and deacetylases, although <strong>in</strong>separate complexes [92,94]. EBNA3C and EBNA3A <strong>in</strong>teract with C term<strong>in</strong>al b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>(CtBP) and repress the tumor-suppressor gene p16 INK4A <strong>in</strong> lymphoid cells [95]. These <strong>in</strong>teractionscontribute to the ma<strong>in</strong>tenance of the H3K27me3 repressive chromat<strong>in</strong> mark at the silent p16 INK4Apromoter as well as the susta<strong>in</strong>ed growth of lymphoblastoid cells immortalized by EBV. EBNA3Cand EBNA3A cooperated <strong>in</strong> repression of Bim, a gene cod<strong>in</strong>g for the proapoptotic prote<strong>in</strong> Bcl-2-<strong>in</strong>teract<strong>in</strong>g mediator of cell death [96]. In cells carry<strong>in</strong>g latent EBV genomes the Bim promoter wasenriched <strong>in</strong> the repressive histone mark H3K27me3 and methylated CpGs [97].423LMP1, a transmembrane oncoprote<strong>in</strong>, affected both alternative systems of epigenetic memory,DNA methylation, and Polycomb group complexes. In epithelial cells LMP1 up-regulatedDNMT1, DNMT3A, and DNMT3B, whereas <strong>in</strong> Hodgk<strong>in</strong> lymphoma cells it <strong>in</strong>duced thePolycomb group prote<strong>in</strong> Bmi-1 [98e100]. LMP1 activated the expression of DNMT1 via the c-jun NH(2)-term<strong>in</strong>al k<strong>in</strong>ase/activator prote<strong>in</strong>-1 (JNK-AP-1) signal<strong>in</strong>g pathway [99]. The NF-kBpathway was <strong>in</strong>volved <strong>in</strong> Bmi-1 <strong>in</strong>duction [100]. Dutton et al. suggested that by up-regulat<strong>in</strong>gBmi-1 via the NF-kB pathway, LMP1 may <strong>in</strong>duce the loss of B-cell identity <strong>in</strong> EBV-positiveHodk<strong>in</strong>’s lymphomas via down-regulat<strong>in</strong>g B-cell markers (CD21/MS4A1, BLK, LY9). Inaddition, Bmi-1 may mark a set of promoters for de novo methylation by DNMTs, therebysilenc<strong>in</strong>g tumor-suppressor genes, <strong>in</strong>clud<strong>in</strong>g IGSF4 and ATM. Other LMP1 target genes areactivated by Bmi-1, like STAT, c-MET, and HK, cod<strong>in</strong>g for signal<strong>in</strong>g molecules and hexok<strong>in</strong>ase(reviewed by [101]). Latent EBV <strong>in</strong>fection was associated with hypermethylation of a set ofcellular promoters <strong>in</strong> LMP1-positive tumors <strong>in</strong>clud<strong>in</strong>g nasopharyngeal carc<strong>in</strong>oma [102],Hodgk<strong>in</strong> lymphoma [103e105], and lymphomas develop<strong>in</strong>g <strong>in</strong> AIDS patients [106]. Incontrast, aberrant methylation of p15 and p16 tumor-suppressor genes was <strong>in</strong>frequent <strong>in</strong>iatrogenic lymphomas develop<strong>in</strong>g <strong>in</strong> methotrexate-treated rheumatoid arthritis patients [107].The transmembrane prote<strong>in</strong> LMP2A is also <strong>in</strong>volved <strong>in</strong> promoter silenc<strong>in</strong>g by CpG methylation[108]. LMP2A <strong>in</strong>duces the phosphorylation of STAT3 <strong>in</strong> gastric carc<strong>in</strong>oma cell l<strong>in</strong>es result<strong>in</strong>g <strong>in</strong>


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>the up-regulation of DNMT1 transcription and methylation of the tumor-suppressor genePTEN (phosphatase and tens<strong>in</strong> homolog, deleted on chromosome ten). PTEN functions as a phosphatidyl<strong>in</strong>ositol-3,4,5-triphosphate(PIP 3 ) phosphatase caus<strong>in</strong>g cell cycle arrest and <strong>in</strong>hibitionof cell migration. PTEN contributes to the ma<strong>in</strong>tenance of apicalebasal polarity of epithelialcells, too (reviewed by [109]). Because LMP1 is absent from EBV-associated gastric carc<strong>in</strong>omas,H<strong>in</strong>o et al. suggested that CpG methylation of cellular promoters [110,111], <strong>in</strong>clud<strong>in</strong>g the PTENpromoter, is due to LMP2A expression <strong>in</strong> these neoplasms [108]. One has to add, however, that<strong>in</strong> a significant portion of EBV-associated gastric carc<strong>in</strong>omas LMP2 expression can’t be detected,either [112], suggest<strong>in</strong>g that other latent EBV prote<strong>in</strong>s or RNAs may also contribute to thedevelopment of the CGI methylator phenotype (CIMP).424EPIGENETIC ALTERATIONS IN NEOPLASMS CARRYING KAPOSI’S SARCOMA-ASSOCIATED HERPESVIRUS GENOMES<strong>Human</strong> herpesvirus 8 (HHV-8) or Kaposi’s sarcoma-associated herpesvirus (KSHV) belongs tothe genus rhad<strong>in</strong>ovirus of the human gammaherpesviruses, a subfamily of herpesviruses.HHV8 is causally associated with Kaposi’s sarcoma (KS), a tumor of endothelial cell orig<strong>in</strong>.KSHV is also associated with multicentric Castleman’s disease (MCD) and primary effusionlymphoma (PEL, also called body cavity-based lymphoma) (reviewed by [113]). KSHV exists <strong>in</strong>two replication states, lytic or latent. The key lytic switch and transcriptional transactivatorprote<strong>in</strong> Rta is encoded by the viral immediate early ORF50. Dur<strong>in</strong>g viral latency <strong>in</strong> B-cells, Rtaexpression is down-regulated, together with most other viral prote<strong>in</strong>s. The key regulator ofviral latency, LANA (latency-associated nuclear antigen encoded by ORF73), suppresses bothlytic activation and apoptosis (reviewed by [113]). The KSHV latent orig<strong>in</strong> of DNA replicationis located to the term<strong>in</strong>al repeats (TRs) of the genome where LANA has two b<strong>in</strong>d<strong>in</strong>g sites.LANA recruits cellular replication prote<strong>in</strong>s to the TRs. Histone acetylation at the TRs is high,while histone methylation changes throughout the cell cycle [114]. In addition to replicationfactors, LANA also recruits to the TRs a repressive complex consist<strong>in</strong>g of heterochromat<strong>in</strong>prote<strong>in</strong> (HP) 1 and the HMT SUV39H1 [115].Two recent studies analyzed the chromat<strong>in</strong> structure of entire KSHV genomes <strong>in</strong> latently<strong>in</strong>fected cells. Immunoprecipitation of methylated DNA and of covalently modified histones<strong>in</strong> comb<strong>in</strong>ation with microarrays yielded dist<strong>in</strong>ct profiles of epigenetic modifications forlatency <strong>in</strong> tumor cell l<strong>in</strong>es and <strong>in</strong> newly <strong>in</strong>fected epithelial cells. Characteristic DNAmethylationpatterns were found all over the genome. Latency-specific histone modificationswere rapidly established upon <strong>in</strong>fection. Silenc<strong>in</strong>g of lytic viral promoters was not establishedby removal of the activat<strong>in</strong>g histone marks H3K9ac, H3K14ac, and H3K4me3, but by thedeposition of H3K27me3 across the genome. This “bivalent” modification is transcriptionallyrepressive but enables rapid activation upon the <strong>in</strong>duction of the lytic cycle [116]. A similar“bivalent” chromat<strong>in</strong> structure was found at the IE genes ORF50 and ORF48 <strong>in</strong> the secondstudy. In addition, Enhancer of Zeste Homologue (EZH) 2, the HMT enzyme of PcG prote<strong>in</strong>complexes colocalized with the H3K27me3 marks dur<strong>in</strong>g latency, while the reactivation of thelytic cycle led to the dissociation of EZH2 and H3K27me3 from the viral genome and to theexpression of the lytic prote<strong>in</strong> cascade [117].B<strong>in</strong>d<strong>in</strong>g of the chromat<strong>in</strong> <strong>in</strong>sulator prote<strong>in</strong> CTCF <strong>in</strong> association with cohes<strong>in</strong>s to the KSHVepisome may also <strong>in</strong>fluence viral gene expression by establish<strong>in</strong>g alternative threedimensionalconformations dur<strong>in</strong>g latency and lytic replication [118].In addition to PcG prote<strong>in</strong>s, viral microRNAs also contribute to KSHV-latency. A miRNAdeletion mutant virus elim<strong>in</strong>at<strong>in</strong>g ten of the 12 viral miRNAs from the KSHV genome wasmore lytically active than the wild-type virus. This effect might be expla<strong>in</strong>ed by the fact that twoof the deleted viral miRNAs, miR-K12-5 and miR-K12-4-5p, have a seed sequence target<strong>in</strong>g anddown-regulat<strong>in</strong>g transcripts of the viral transactivator ORF50 and ret<strong>in</strong>oblastoma like prote<strong>in</strong>(Rbl) 2, a repressor of DNMT3A and DNMT3B transcription, respectively [119].


CHAPTER 21<strong>Epigenetics</strong> and <strong>Human</strong> Infectious <strong>Disease</strong>sKSHV <strong>in</strong>fection <strong>in</strong>fluences the epigenetic marks of the host genome. In a LANA-transduced celll<strong>in</strong>e, the promoters for CDH13 (H-Cadher<strong>in</strong>), CCND2 (cycl<strong>in</strong> D2), LDHB (lactate dehydrogenaseB), FOXG1B_FKHL1 (Forkhead box prote<strong>in</strong> G1B_Forkhead-related prote<strong>in</strong> 1) and CREG (cellularrepressor of E1A stimulated genes) were strongly down-regulated. LANA was associated withrepressed cellular promoters and recruited DNMT3A, but also DNMT1 and DNMT3B,result<strong>in</strong>g <strong>in</strong> promoter methylation, as demonstrated <strong>in</strong> the case of the CDH13 promoter [120].At the TGF-b type II receptor (TbetaRII) promoter, deacetylation of promoter-associatedhistones was also demonstrated, <strong>in</strong> addition to promoter methylation and LANA b<strong>in</strong>d<strong>in</strong>g tothe transcriptional corepressor complex mSIN3A [121].In PEL cells carry<strong>in</strong>g both KSHV and EBV genomes, transfection of LANA decreased the activityof the EBV promoters Qp and Cp [122]. Upon transfection <strong>in</strong>to immortalized epithelial celll<strong>in</strong>es, LANA was also associated with the repressive HP1 at the cellular chromat<strong>in</strong> [123].Accord<strong>in</strong>gly, LANA seems to work as an epigenetic modifier of the viral and host genomes.Transfection of LANA <strong>in</strong>to several tumor cell l<strong>in</strong>es led to a fundamental reorganization of thecellular chromat<strong>in</strong>. LANA-<strong>in</strong>duced redistribution of DNA sta<strong>in</strong><strong>in</strong>g co<strong>in</strong>cided with redistributionof the methyl-cytos<strong>in</strong> b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> MeCP2. This effect was attributed to the <strong>in</strong>teractionof LANA with the HMT enzyme SUV39H1 [124]. The <strong>in</strong>teraction of LANA with MeCP2 andnucleosomes would enable LANA to direct regulatory complexes to chromosomal sites andthereby stably reprogram cellular and viral gene expression [125]. Accord<strong>in</strong>gly, p16 INK4A wasfrequently silenced through CpG-methylation <strong>in</strong> PEL cell l<strong>in</strong>es and primary PEL cells [126].Furthermore, CpG-hypermethylation at the MGMT promoter and a loss of the EBV genome <strong>in</strong>cell culture was observed <strong>in</strong> a PEL cell subclone derived from a PEL tumor, which was positivefor KSHV and EBV, but unmethylated at MGMT [127] (Table 21.2).21.3.2 Interactions of Lytic Cycle Prote<strong>in</strong>s of <strong>Human</strong> Cytomegaloviruswith Histone DeacetylasesFollow<strong>in</strong>g primary <strong>in</strong>fection, human cytomegalovirus (HCMV), a betaherpesvirus, persists orestablishes latency <strong>in</strong> cells of the myeloid l<strong>in</strong>eage. Although most <strong>in</strong>fections are asymptomatic,life-threaten<strong>in</strong>g disease can develop <strong>in</strong> immunocompromised patients or if <strong>in</strong>fection occurs <strong>in</strong>utero. Gönczöl et al. observed that HCMV latency and reactivation is connected to theundifferentiated or differentiated nature of the host cell [128]. In undifferentiated cells themajor immediate-early (IE) promoter (MIEP) of HCMV was repressed and located toa hypoacetylated chromat<strong>in</strong> doma<strong>in</strong> associated with HP1 and Ets-2 repressor factor (ERF),a transcriptional repressor recruit<strong>in</strong>g HDAC1 to MIEP [129,130]. Dur<strong>in</strong>g lytic <strong>in</strong>fection,425TABLE 21.2 Epigenetic Alterations Induced by Latent <strong>Human</strong> GammaherpesvirusProte<strong>in</strong>sMicrobe Effector Molecule Epigenetic Alteration EffectEBV EBNA2 Histone acetylation Switch<strong>in</strong>g on promotersEBNALP (EBNA5) HDAC displacement Switch<strong>in</strong>g on promotersEBNA3C (EBNA6) Histone acetylation, histonedeacetylationModulation of promoteractivityLMP1 Up-regulation of DNMTs Promoter silenc<strong>in</strong>gLMP2A Up-regulation of DNMT1 Promoter silenc<strong>in</strong>gKSHV LANA Recruitment of DNMTs to cellular Promoter silenc<strong>in</strong>gpromotersAssociation with mSIN3A, Promoter silenc<strong>in</strong>grecruitment of HDACsAssociation with HP1Promoter silenc<strong>in</strong>gInteraction with SUV39H1,Redistribution of MeCP2Reorganization of thenucleus


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>however, for example when monocytes differentiated to macrophages, MIEP was activated andassociated with hypermethylated histones [129]. In addition, IE1 and IE2, the abundantprote<strong>in</strong> products of the IE genes <strong>in</strong>teracted with histone deacetylases, abolish<strong>in</strong>g theirrepressive effect at the MIEP and the early promoter of the viral polymerase [131,132]. Thus, IEprote<strong>in</strong>s modulate the host epigenetic regulation of viral promoters, thereby facilitat<strong>in</strong>g viralreplication.IE1 and IE2 activated cellular promoters either by direct b<strong>in</strong>d<strong>in</strong>g to CCAAT box b<strong>in</strong>d<strong>in</strong>g factor(CTF1) at TATA-less promoters, or TATA-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> (TBP), at promoters with a TATA motif,respectively [133,134]. In human fibroblasts, activation of the human telomerase reversetranscriptase gene (hTERT) by IE1 was accompanied by <strong>in</strong>creased b<strong>in</strong>d<strong>in</strong>g of the nuclear prote<strong>in</strong>Sp1, acetylation of histone H3, and a reduction of HDAC b<strong>in</strong>d<strong>in</strong>g at the hTERT promoter [135].S<strong>in</strong>ce telomerase activation is crucial for immortalization and malignant transformation ofcells, these data support the po<strong>in</strong>t that HCMV may contribute to tumorigenesis [136].21.3.3 Epigenetic Dysregulation <strong>in</strong> <strong>Human</strong> Retrovirus-Infected CellsThe RNA genomes of human T-cell lymphotropic virus (HTLV) and human immunodeficiencyvirus (HIV) replicate via a DNA <strong>in</strong>termediate that becomes <strong>in</strong>tegrated <strong>in</strong>to the host cellgenome (provirus). Proviral genomes are frequently silenced by epigenetic mechanisms. Inturn, retroviral prote<strong>in</strong>s <strong>in</strong>teract<strong>in</strong>g with the epigenetic regulatory mach<strong>in</strong>ery modulate thegene expression pattern of their host cells.426CPG ISLAND METHYLATOR PHENOTYPE IN ADULT T-CELL LEUKEMIA/LYMPHOMA (ATLL): A PUTATIVE ROLE FOR THE HTLV-1 PROTEIN TAX IN THESILENCING OF KEY CELLULAR PROMOTERS?The proviral genome of type I human T-lymhotropic virus (HTLV-1), a retrovirus associatedwith ATLL and tropical spastic paraparesis, frequently undergoes epigenetic silenc<strong>in</strong>g [137].Therefore, <strong>in</strong> tropical spastic paraparesis patients the activation of viral gene expression us<strong>in</strong>gthe HDAC <strong>in</strong>hibitor valproate seems to be a justified therapeutic approach, because the viruspositivecells may thereby be exposed to the host immune response, result<strong>in</strong>g <strong>in</strong> the collapse ofthe latent viral reservoir [138]. In ATLL patients the CGI methylation frequency of the Srchomology-2-conta<strong>in</strong><strong>in</strong>g prote<strong>in</strong> tyros<strong>in</strong>e phosphatase (Shp1, PTPN6) gene gradually <strong>in</strong>creaseddur<strong>in</strong>g disease progression [139]. In parallel, the number of CGI methylated genes also<strong>in</strong>creased. Because the viral oncoprote<strong>in</strong> Tax <strong>in</strong>duced the dissociation of transcription factorsfrom the Shp1 promoter and subsequent promoter hypermethylation [140], Niller et al.speculated that Tax may act as a “hit-and-run”oncoprote<strong>in</strong> by <strong>in</strong>itiat<strong>in</strong>g the down-regulation ofShp1 expression <strong>in</strong> an early stage of leukemogenesis [141]. This early step may be followed bysilenc<strong>in</strong>g of additional cellular promoters and down-regulation of Tax expression itself, due todeletions of the proviral genome or its epigenetic silenc<strong>in</strong>g.HUMAN IMMUNODEFICIENCY VIRUS: EPIGENETIC SILENCING OF LATENTVIRAL GENOMES AND HOST GENESHIV causes the acquired immunodeficiency syndrome. The virus is transmitted parenterallyand, with the exception of very few long-term non-progressors, ends fatally for the <strong>in</strong>fectedpatients due to the severe course of opportunistic <strong>in</strong>fections which would normally run lessseverely or even subcl<strong>in</strong>ically. Due to the HIV-associated severe immune suppression, HIVpatients have an <strong>in</strong>creased risk to develop virus-associated neoplasms dur<strong>in</strong>g all stages of HIVdisease [142e144].Although highly active antiretroviral therapy (HAART) that comb<strong>in</strong>es several drugs withdifferent viral targets significantly improved the life expectancy of HIV-<strong>in</strong>fected <strong>in</strong>dividuals,there is still no cure for HIV <strong>in</strong>fection. One reason for this failure is the existence of a dormantviral reservoir <strong>in</strong> rest<strong>in</strong>g memory CD4 þ T cells [145]. Upon cessation of HAART and due to


CHAPTER 21<strong>Epigenetics</strong> and <strong>Human</strong> Infectious <strong>Disease</strong>sstimulation of the memory cells, the latent viral genomes start transcription and replicationaga<strong>in</strong>, lead<strong>in</strong>g to viral rebound. The half-life of the latent viral reservoir is approximately 40months. In patients undergo<strong>in</strong>g HAART, occasional short periods of asymptomatic viralreplication, termed “blips”, may allow the virus to replenish the latent reservoir [146].Therefore, viral persistence <strong>in</strong> rest<strong>in</strong>g memory CD4 T cells has been recognized as a majorobstacle aga<strong>in</strong>st curative treatment (reviewed by [147,148]).Mechanisms of HIV Latency: CpG Methylation, Repressive Chromat<strong>in</strong>,Transcriptional Interference, and miRNAsEnzymatic methylation of CpG d<strong>in</strong>ucleotides prevented b<strong>in</strong>d<strong>in</strong>g of transcription factors to theHIV-LTR and led to a transcriptional block <strong>in</strong> transfected cells which could be overcome bydemethylation through 5-aza-C or the presence of the HIV-transactivator Tat. This suggestedthat DNA-methylation might contribute to the silenc<strong>in</strong>g of viral transcription <strong>in</strong> the latent state[149e151]. HIV transcription and chromat<strong>in</strong> structure were exam<strong>in</strong>ed <strong>in</strong> several latently<strong>in</strong>fected cell l<strong>in</strong>es. Induction of viral transcription through the NF-kB stimulator TNFa andHDAC <strong>in</strong>hibitors correlated with an overall <strong>in</strong>crease <strong>in</strong> histone acetylation and the disruptionof nucleosome 1 which blocks the transcriptional start site of the viral 5 0 -LTR [152]. Twotranscriptional repressors, YY1 (Y<strong>in</strong>g Yang 1) and LSF (late simian virus 40 transcriptionfactor), were shown to associate with the HIV LTR, and recruited HDAC1 <strong>in</strong> vitro. YY1-dependent repression was reversed by the HDAC <strong>in</strong>hibitor trichostat<strong>in</strong> A (TSA), suggest<strong>in</strong>g thatrepressive histone marks also contribute to the silenc<strong>in</strong>g of HIV-transcription [153].In rest<strong>in</strong>g CD4 þ T cells isolated from aviremic HIV-<strong>in</strong>fected cell donors on HAART, the HDAC<strong>in</strong>hibitor valproic acid (VPA) led to the acetylation of the <strong>in</strong>tegrated provirus-LTR and to anoutgrowth of HIV from the rest<strong>in</strong>g cells without <strong>in</strong>duc<strong>in</strong>g T-cell activation markers [154]. Thecontribution of different histone methylation marks to LTR-silenc<strong>in</strong>g was exam<strong>in</strong>ed <strong>in</strong> latentlytransfected T-cell l<strong>in</strong>es. EZH2, a component of the polycomb repressive complex (PRC) 2<strong>in</strong>volved <strong>in</strong> H3K27me3 methylation, was enriched at the repressed LTR, while upon LTRactivation it was rapidly displaced. A smaller number of proviruses were silenced throughSUV39H1 which is <strong>in</strong>volved <strong>in</strong> H3K9me3 methylation. Thus, specific <strong>in</strong>activation of PRC2may give a cue to future therapeutic attempts to selectively activate the LTR [155]. The LTRs <strong>in</strong>isolated rest<strong>in</strong>g T cells from aviremic HIV patients and stably transfected HeLa cells wererepressed through a prote<strong>in</strong> complex of transcription factors of c-Myc and Sp1, attract<strong>in</strong>gHDAC1 [156]. CBF1, a transcriptional repressor and effector <strong>in</strong> the Notch signal<strong>in</strong>g pathway,also silenced HIV transcription. CBF1 recruited HDAC1 and the corepressor prote<strong>in</strong>s CIR(CBF1-<strong>in</strong>teract<strong>in</strong>g prote<strong>in</strong>) and mSIN3A to the LTR. Knockdown of CBF1 led to transcriptionalderepression, with a concurrent <strong>in</strong>crease <strong>in</strong> histone acetylation levels, loss of HDAC1 and lossof co-repressor prote<strong>in</strong>s [157].427HIV proviruses preferentially <strong>in</strong>tegrate with<strong>in</strong> actively transcribed genes. Nevertheless, they aretranscriptionally suppressed <strong>in</strong> rest<strong>in</strong>g CD4 T cells from patients under HAART. Proviruses,mostly with<strong>in</strong> <strong>in</strong>trons, were randomly <strong>in</strong>serted as to their transcriptional orientation,suggest<strong>in</strong>g that proviral transcripts may partially end up as rapidly degraded <strong>in</strong>tronic RNAupon transcription of the respective host gene. Thus, <strong>in</strong>tronic <strong>in</strong>tegration represents a dist<strong>in</strong>ctmolecular mechanism of HIV silenc<strong>in</strong>g <strong>in</strong> latency [158,159]. Targeted <strong>in</strong>sertion of a HIVprovirus by homologous recomb<strong>in</strong>ation <strong>in</strong>to the third <strong>in</strong>tron of the HPRT gene was used asa model to study the effect of the transcriptional orientation of host and viral genes. HPRTreadthroughtranscription enhanced HIV transcription, when runn<strong>in</strong>g <strong>in</strong> the same direction,but <strong>in</strong>hibited it, when runn<strong>in</strong>g <strong>in</strong> the opposite direction. In this model, repressive chromat<strong>in</strong>marks did not play a significant role <strong>in</strong> HIV latency [160].Another mechanism of HIV latency <strong>in</strong> memory T cells may rely on miRNAs. A series of CD4 þT-cell l<strong>in</strong>es expressed high levels of Dicer which belongs to the miRNA process<strong>in</strong>g mach<strong>in</strong>ery.In HIV-<strong>in</strong>fected T cells, the viral TAR stem-loop RNA was bound by Dicer and processed to


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>a miRNA. Transfection of a correspond<strong>in</strong>g siRNA-express<strong>in</strong>g construct decreased LTRdependentexpression of a reporter prote<strong>in</strong>. Furthermore, the TAR-siRNA was able to recruitHDAC1 to the LTR, but not to downstream sequences [161]. Cellular miRNAs may alsocontribute to HIV latency. miR-28, miR-125b, miR-150, miR-223, and miR-382 were enriched<strong>in</strong> rest<strong>in</strong>g T cells compared to activated T cells. These miRNAs were recruited to the 3 0 -end ofthe HIV transcript and <strong>in</strong>hibited the translation of HIV prote<strong>in</strong>s. miRNA antisense-constructsreleased the translational block and led to viral replication [162].Silenc<strong>in</strong>g of Host Genes <strong>in</strong> HIV-Infected CellsThe chromat<strong>in</strong> structure <strong>in</strong> host cell genes is <strong>in</strong>fluenced by HIV <strong>in</strong>fection of CD4 þ T cells.DNMT1 levels <strong>in</strong>creased significantly <strong>in</strong> CD4 þ T-cell l<strong>in</strong>es and primary CD4 T cells upon<strong>in</strong>fection with HIV. Acute <strong>in</strong>fection of primary T H 1 cells led to a decrease of IFNg expressionwhich co<strong>in</strong>cided with the de novo methylation of the IFNg promoter. An antisense-mediatedknockdown of DNMT1 led to transcriptional derepression of the IFNg promoter [163]. An<strong>in</strong>crease <strong>in</strong> the level of DNMT1 was also observed <strong>in</strong> a T-cell l<strong>in</strong>e <strong>in</strong>fected with an <strong>in</strong>tegrationandtherefore replication-defective HIV mutant. Thus, the presence of viral nucleic acids wassufficient, but viral replication was not required for the <strong>in</strong>crease of DNMT1 expression. Theoverall methylation level of the cellular DNA <strong>in</strong>creased, the CGI at the p16 INK4A promoterbecame hypermethylated and repressed upon HIV <strong>in</strong>fection, while 5-aza-C treatment led tothe <strong>in</strong>duction of p16 INK4A expression [164].428Another gene down-regulated <strong>in</strong> virus-<strong>in</strong>fected immortalized T cells was UDP-Nacetylglucosam<strong>in</strong>e2-epimerase/N-acetylmannosam<strong>in</strong>e k<strong>in</strong>ase (GNE). GNE produces thesialyl-donor substrate for all cellular sialyl-transferases and thereby profoundly <strong>in</strong>fluences thereceptor and hom<strong>in</strong>g functions of immune cells. In T-cell l<strong>in</strong>es the CGI at the GNE promoterbecame de novo hypermethylated upon acute HIV <strong>in</strong>fection, as demonstrated by bisulfitesequenc<strong>in</strong>g. Treatment with 5-aza-C released both the transcriptional repression andmethylation, and restored sialylation function. GNE hypermethylation may to some degreeexpla<strong>in</strong> the frequently persist<strong>in</strong>g immune disorder of aviremic HIV patients on long-termsuccessful HAART [165].Infection of a T-cell l<strong>in</strong>e or transfection of HeLa cells with an <strong>in</strong>tegration-defective HIV<strong>in</strong>creased DNMT1 expression. The effect was dependent on expression of HIV early prote<strong>in</strong>sTat, Rev, and Nef. The transcription factor AP1 was implicated <strong>in</strong> the HIV-dependent upregulationof DNMT1 [166]. Furthermore, <strong>in</strong> a human neuroblastoma cell l<strong>in</strong>e and <strong>in</strong> humanprimary neuronal cells, Tat was shown to <strong>in</strong>duce HDAC2. This led to the silenc<strong>in</strong>g of the CREBand CaMKIIa genes which contribute to synaptic plasticity and neuronal function. Thus, HIV<strong>in</strong>duceddysregulation of HDAC2 may be <strong>in</strong>volved <strong>in</strong> HIV-associated neurocognitive disorders(HAND) [167] (Table 21.3).21.3.4 Local Hypermethylation and Global Hypomethylation of the HostCell DNA <strong>in</strong> Hepatocellular Carc<strong>in</strong>omas Associated with Hepatitis BVirusHepatocellular carc<strong>in</strong>oma (HCC) is the predom<strong>in</strong>ant form of human liver cancer. HepatitisB virus (HBV) and hepatitis C virus (HCV) <strong>in</strong>fections are the major etiological factors forTABLE 21.3 Epigenetic Alterations Induced by <strong>Human</strong> RetrovirusesMicrobe Effector Molecule Epigenetic Alteration EffectHTLV-I Tax Shp1 promoter methylation Promoter silenc<strong>in</strong>gHIV-1 Tat, rev, nef Upregulation of DNMT1 Promoter silenc<strong>in</strong>gTat Induction of HDAC2 Promoter silenc<strong>in</strong>g


CHAPTER 21<strong>Epigenetics</strong> and <strong>Human</strong> Infectious <strong>Disease</strong>sHCC. HBV-related HCC develops through dist<strong>in</strong>ct stages (reviewed by [168]). Epigeneticalterations <strong>in</strong>clud<strong>in</strong>g aberrant CGI hypermethylation appear already <strong>in</strong> the preneoplasticlesions (cirrhotic nodules) and their frequency gradually <strong>in</strong>creases dur<strong>in</strong>g the early stages ofliver carc<strong>in</strong>ogenesis (low-grade dysplastic nodules, high-grade dysplastic nodules, earlyhepatocellular carc<strong>in</strong>oma), although the methylation level of certa<strong>in</strong> genes may decrease<strong>in</strong> progressed HCCs. Shim et al. found that hypermethylation of the p16 gene starts at anearly stage of hepatocarc<strong>in</strong>ogenesis. Sixty-two percent of cirrhotic nodules (putativepreneoplastic lesions) and 70% of dysplastic nodules, surround<strong>in</strong>g HBV-positive hepatocellularcarc<strong>in</strong>oma lesions, showed p16 hypermethylation [169]. Um et al. described thatAPC, RASSF1A, and SOCS1 were methylated <strong>in</strong> a fraction of cirrhotic nodules but themethylation levels of APC and RASSF1 <strong>in</strong>creased significantly <strong>in</strong> low-grade dysplasticnodules. SOCS1 methylation gradually <strong>in</strong>creased dur<strong>in</strong>g multistep carc<strong>in</strong>ogenesis,peaked <strong>in</strong> early hepatocellular carc<strong>in</strong>oma, and decreased <strong>in</strong> progressed livercarc<strong>in</strong>omas [170].The genome of hepatitis B virus codes for a pleiotropic regulator, called X prote<strong>in</strong> (HBx orpX). pX <strong>in</strong>teracts with multiple key signal<strong>in</strong>g pathways and affects the regulators of cellcycle progression [171e175]. In vitro, pX up-regulates cycl<strong>in</strong> D1 and activates DNMT1expression via the cycl<strong>in</strong> D1-CDK4/6-pRb-E2F1 pathway, result<strong>in</strong>g <strong>in</strong> DNA-methylationmediateddown-regulation of the tumor-suppressor prote<strong>in</strong> p16 INK4a [176,177]. In additionto DNMT1, pX also up-regulated two variants of the de novo DNA methyltransferaseDNMT3A <strong>in</strong> liver cells. DNMT3A1 and DNMT3A2 are translated from differentially splicedtrancripts of the DNMT3A gene and <strong>in</strong>duce regional hypermethylation of specific tumorsuppressorgenes [178]. pXdown-regulated, however, another de novo DNA methyltransferase,DNMT3B, <strong>in</strong> the very same cells. Because satellite 2 repeat sequences aremethylated by DNMT3B, pX-<strong>in</strong>duced down-regulation of the enzyme led to a global hypomethylationof these repeats [178]. This important observation connected the viral oncoprote<strong>in</strong>pX both to global DNA hypomethylation, a phenomenon as widespread <strong>in</strong>neoplastic cells [179], and regional hypermethylation [180], which is another key feature oftumor cells. In hepatocellular carc<strong>in</strong>omas carry<strong>in</strong>g <strong>in</strong>tegrated HBV genomes, CpG-richsequences were also found to be hypomethylated <strong>in</strong> the pericentromeric regions of acrocentricchromosomes [181]. pX was also implicated <strong>in</strong> hypermethylation-mediatedsilenc<strong>in</strong>gof the E-cadher<strong>in</strong> promoter [182e184]. The tumor-suppressor gene RASSF1A (Rasassociation doma<strong>in</strong> family 1A) and the GSTP1 gene, cod<strong>in</strong>g for the p-class glutathioneS-transferase, an enzyme <strong>in</strong>volved <strong>in</strong> protect<strong>in</strong>g aga<strong>in</strong>st electrophilic carc<strong>in</strong>ogens, was alsofrequently <strong>in</strong>activated by CGI methylation <strong>in</strong> HBV-positive hepatocellular carc<strong>in</strong>omas[185,186].429The epigenetic profile of hepatocellular carc<strong>in</strong>oma associated with HBV appears to be differentfrom that of HCV-associated liver cancer. Feng et al. observed that certa<strong>in</strong> genes (HOXA9,RASSF1, SFRP1) were methylated more frequently <strong>in</strong> HBV-positive hepatocellular carc<strong>in</strong>omasthan <strong>in</strong> HCV-positive liver tumors. In contrast, CDKN2A was significantly more frequentlymethylated <strong>in</strong> HCV-positive than <strong>in</strong> HBV-positive liver carc<strong>in</strong>omas [187]. These data supportthe idea that hepatocellular carc<strong>in</strong>omas of different viral etiologies are associated with unique,virus-specific epigenetic signatures.21.3.5 Hepatitis C Virus (HCV)-Induced Epigenetic AlterationsHepatitis C virus belongs to the genus hepacivirus of the flaviviruses. It is transmitted throughbloodeblood contacts and causes <strong>in</strong>flammatory liver disease which turns chronic <strong>in</strong> about80% of the <strong>in</strong>fected patients. Chronic <strong>in</strong>fection may lead to liver cirrhosis and hepatocellularcarc<strong>in</strong>oma <strong>in</strong> the long term. Several large studies described epigenetic down-regulation ofnumerous tumor-suppressor genes <strong>in</strong> HCC tissue samples [187e190] (reviewed by [191]). Theoverall genomic methylation levels, reflected <strong>in</strong> the methylation of LINE1 and SAT2 repeats,


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>TABLE 21.4 Epigenetic Alterations Induced by <strong>Human</strong> Hepatitis VirusesMicrobe Effector Molecule Epigenetic Alteration EffectHBV pX Up-regulation of DNMT1 Promoter silenc<strong>in</strong>gUp-regulation of DNMT3A1and Promoter silenc<strong>in</strong>gDNMT3A2Down-regulation of DNMT3B Satellite 2 repeathypomethylationHCV Core prote<strong>in</strong> Induction of DNMT1, and DNMT3B Promoter silenc<strong>in</strong>gInduction of HDAC (SIRT1)Promoter silenc<strong>in</strong>gInduction of SMYD3, a histonemethyltransferaseSilenc<strong>in</strong>g of RASSF1Adecreased, while methylation at CGIs of TSGs <strong>in</strong>creased with the progression of hepatitis viacirrhosis to HCC [168,192,193] (Table 21.4).430DNA methylation at dist<strong>in</strong>ct tumor-suppressor gene loci is significantly associated with HBV,HCV, or alcoholism, respectively [190] (reviewed by [191]). Methylation at the tumorsuppressorgenes CDKN2A, SOCS-1, GADD45B, STAT1, APC, and p15 is more prevalent <strong>in</strong>HCV-positive than <strong>in</strong> HCV-negative tumors, while HOXA9, RASSF1, and SFRP1 were moremethylated <strong>in</strong> HBV-positive tumors [187,192,194,195] (reviewed by [191]). Methylation ofthe kallikre<strong>in</strong> (KLK) 10 promoter correlated with HCV <strong>in</strong>fection, while methylation at KLK10and GSTP1 <strong>in</strong>versely correlated with HBV <strong>in</strong>fection [190,196]. Another study appliedmethylated-DNA immunoprecipitation on chip and mass spectrometry on HCC tissuesamples to discrim<strong>in</strong>ate between HCV- and HBV-associated HCC. While hypermethylation ofonly the CYP7B1 locus was found <strong>in</strong> HBV-HCC, a multitude of 15 loci was hypermethylated <strong>in</strong>HCV-HCC: age-<strong>in</strong>dependent methylation markers <strong>in</strong>cluded genes <strong>in</strong>hibitory to Ras/Raf/ERKsignal<strong>in</strong>g (NPR1, DUSP4, LOX, and RRAD) and <strong>in</strong>hibitory to Wnt/b-caten<strong>in</strong> signal<strong>in</strong>g (SFRP4and RUNX3) [197].In a transgenic mouse and a cell l<strong>in</strong>e model, epigenetic down-regulation of SOCS1 wasdependent on the expression of the HCV core prote<strong>in</strong> [198]. This observation is <strong>in</strong> accordancewith a cl<strong>in</strong>ical study show<strong>in</strong>g that HCV-<strong>in</strong>fection was associated with SOCS1 methylation,whereas HBV <strong>in</strong>fection was <strong>in</strong>versely correlated with SOCS1 methylation [199]. In HepG2 cellsthe expression of the core prote<strong>in</strong> <strong>in</strong>duced DNMT1 and DNMT3B result<strong>in</strong>g <strong>in</strong> CDH1 promoterhypermethylation and a more aggressive growth behavior [200]. HCV core prote<strong>in</strong> expression<strong>in</strong> the Huh cell l<strong>in</strong>e also led to hypermethylation and silenc<strong>in</strong>g of CDH1. In addition, theHDAC SIRT1 was <strong>in</strong>duced through core prote<strong>in</strong> expression. Inhibit<strong>in</strong>g SIRT1 derepressed theCDH1 promoter [201]. A similar mechanism may silence the promoters of <strong>in</strong>terferonstimulatedgenes, too [202].Alterations of the histone modification mach<strong>in</strong>ery have also been associated with theprogression of HCC (reviewed by [191]). HCC is unusually low <strong>in</strong> H3K4me2, due to deregulationof the trithorax prote<strong>in</strong> Ash2 and histone demethylase LSD1 [203]. Inducible HCVprote<strong>in</strong> expression <strong>in</strong> cell l<strong>in</strong>es led to the overexpression of prote<strong>in</strong> phosphatase 2A (PP2A)which <strong>in</strong>fluenced the histone modification mach<strong>in</strong>ery through b<strong>in</strong>d<strong>in</strong>g to prote<strong>in</strong> arg<strong>in</strong><strong>in</strong>emethyltransferase 1 (PRMT1), and the DNA repair mach<strong>in</strong>ery through dephosphorylat<strong>in</strong>gH2AX. The deregulation of both systems is considered critical <strong>in</strong> HCC development.Accord<strong>in</strong>gly, the expression of a set of <strong>in</strong>dicator genes <strong>in</strong>volved <strong>in</strong> HCC tumorigenesis waschanged concordantly by either the expression of HCV prote<strong>in</strong>s or the overexpression of PP2A<strong>in</strong> cell culture [194] (reviewed by [191]). Transfection of the HCV core prote<strong>in</strong> <strong>in</strong>to cholangiocarc<strong>in</strong>omacell l<strong>in</strong>es <strong>in</strong>duced the SET and MYND doma<strong>in</strong> conta<strong>in</strong><strong>in</strong>g prote<strong>in</strong>, SMYD3,a novel HMT. Overexpression of SMYD3 correlated with hypermethylation of the RASSF1Apromoter [204].


CHAPTER 21<strong>Epigenetics</strong> and <strong>Human</strong> Infectious <strong>Disease</strong>s21.3.6 Induction of Histone Methyltransferase and HistoneDemethylase Enzymes by the <strong>Human</strong> Papillomavirus Oncoprote<strong>in</strong> E7Infection with high-risk human papillomavirus (HPV) stra<strong>in</strong>s (especially type 16 and 18) iscausally related to the development of cervical cancer. Progression of HPV-positive premalignantlesions to <strong>in</strong>vasive cancer is a rare event, however, suggest<strong>in</strong>g that additional steps<strong>in</strong>clud<strong>in</strong>g chromosomal alterations and epigenetic changes are necessary for neoplasticdevelopment [205]. In an <strong>in</strong> vitro model system of cervical carc<strong>in</strong>ogenesis, Henken et al.observed an accumulation of frequent methylation events <strong>in</strong>volv<strong>in</strong>g five tumor-suppressorgenes when primary kerat<strong>in</strong>ocyte cell l<strong>in</strong>es transfected with HPV genomes acquired anchorage<strong>in</strong>dependence [206]. In contrast, no methylation was evident <strong>in</strong> pre-immortal HPV-18transfected cells. Certa<strong>in</strong> cellular genes are also hypermethylated dur<strong>in</strong>g <strong>in</strong> vivo cervicalcarc<strong>in</strong>ogenesis <strong>in</strong> a histological type- or cl<strong>in</strong>ical stage-dependent manner (reviewed by [205]).The E7 oncoprote<strong>in</strong> of HPV-16 was shown to b<strong>in</strong>d to DNMT1 and stimulate its methyltransferaseactivity [207]. This may expla<strong>in</strong> the hypermethylation of selected cellularpromoters <strong>in</strong> HPV-<strong>in</strong>fected cells, and Laurson et al. speculated that E7eDNMT1 <strong>in</strong>teractionmay target DNMT1 to specific sequences, similarly to the recruitment of DNMT3A to specificgenomic regions by the KSHV oncoprote<strong>in</strong> LANA [208].Consistent hypermethylation patterns were described <strong>in</strong> laryngeal papillomas associated withHPV-6 as well [209]. Laryngeal papillomas are usually benign lesions, although they mayprogress to squamous cell carc<strong>in</strong>oma <strong>in</strong> a fraction of cases. Promoter hypermethylationaffect<strong>in</strong>g TIMP3 and CDKN2B occurred early dur<strong>in</strong>g tumor progression and was ma<strong>in</strong>ta<strong>in</strong>edthroughout neoplastic development [209].HPV-16 E7 activated the expresssion of EZH2, a Polycomb prote<strong>in</strong> with histone H3K27methyltransferase activity, too. Cont<strong>in</strong>uous expression of EZH2 was <strong>in</strong>dispensable for theproliferation of HPV-positive tumor cells [210]. Histone H3K27me3 marks may contribute topromoter silenc<strong>in</strong>g either <strong>in</strong> concert with DNMTs or <strong>in</strong>dependently of DNA methylation. E7 isa pleiotropic regulator prote<strong>in</strong> <strong>in</strong>volved both <strong>in</strong> silenc<strong>in</strong>g and activation of certa<strong>in</strong> target genesets. HPV-16 E7 <strong>in</strong>creased histone H3 acetylation at E2F1 and CDC25A promoters <strong>in</strong> humanforesk<strong>in</strong> kerat<strong>in</strong>ocytes [211], and reduced the level of the repressive histone modificationH3K27me3 <strong>in</strong> primary human epithelial cells [212]. The latter change was due to <strong>in</strong>duction ofthe histone demethylases KDM6A and KDM6B target<strong>in</strong>g specifically histone H3K27me3 anddisrupt<strong>in</strong>g Polycomb repressor complexes. E7-mediated reprogramm<strong>in</strong>g resulted <strong>in</strong> an<strong>in</strong>creased expression of KDM6A- and KDM6B-responsive Homeobox genes (HOXC5, HOXC8)that are known to be up-regulated <strong>in</strong> cervical carc<strong>in</strong>omas.43121.3.7 Epigenetic Transcriptional Silenc<strong>in</strong>g <strong>in</strong> Merkel CellPolyomavirus-Associated Carc<strong>in</strong>oma of the Sk<strong>in</strong>Merkel cell carc<strong>in</strong>oma (MCC), a neuroectodermal tumor aris<strong>in</strong>g from mechanoreceptorMerkel cells, frequently carries <strong>in</strong>tegrated genomes of a recently discovered human polyomavirus, called Merkel cell polyomavirus (MCPyV) [213]. MCCs are rare but highly aggressiveneoplasms. Similarly to other polyomaviruses, the dsDNA genome of MCPyV encodestumor antigens (large T antigen and small t antigen) implicated <strong>in</strong> oncogenesis. In MCCsthe large T antigen is regularly truncated render<strong>in</strong>g the virus replication <strong>in</strong>competent [214].The proneural transcription factor Atonal Homologue 1 (Atoh1) is essential for cell fatecommitment of multiple neuronal l<strong>in</strong>eages and acts as a developmental regulator of themechanoreceptive Merkel cells <strong>in</strong> the sk<strong>in</strong>, too [215]. ATOH1 expressionwasfoundtobereduced due to deletion or <strong>in</strong>activation by CpG methylation <strong>in</strong> MCCs as well as colorectalcarc<strong>in</strong>omas [216]. In addition, the promoter of the tumor-suppressor gene RASSF1A wasalsohypermethylated<strong>in</strong>abouthalfoftheMCCsamples[217]. These observations <strong>in</strong>dicatethat epigenetic events may contribute to the pathogenesis of MCPyV-associated Merkelcell carc<strong>in</strong>oma.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>21.4 EPIGENETIC ALTERATIONS ELICITED IN THE HOST TISSUEBY TREMATODE INFECTIONSTrematode <strong>in</strong>fections can lead to epigenetic alterations <strong>in</strong> the host tissue, which we willsubsequently further explore.21.4.1 Promoter Hypermethylation <strong>in</strong> Cholangiocarc<strong>in</strong>oma Associatedwith Opistorchis viverr<strong>in</strong>i (Liver Fluke) InfectionCholangiocarc<strong>in</strong>oma <strong>in</strong> certa<strong>in</strong> geographical areas is related to liver fluke <strong>in</strong>fection. Innortheast Thailand most patients with the malignancy of the bile duct epithelium carry themacroparasite Opistorchis viverr<strong>in</strong>i whereas <strong>in</strong> Cambodia, Ch<strong>in</strong>a, Japan, Korea, Laos, andVietnam Clonorchis s<strong>in</strong>ensis <strong>in</strong>fection is spread<strong>in</strong>g via the consumption of raw freshwater fish,caus<strong>in</strong>g cholangiocarc<strong>in</strong>oma [218,219]. InOpistorchis viverr<strong>in</strong>i-associated cholangiocarc<strong>in</strong>omasdown-regulation of p14ARF expression was predom<strong>in</strong>antly related to <strong>in</strong>activationby DNA methylation whereas p16 INK4A , another tumor-suppressor gene of the 9p21 genecluster was affected by allelic loss. Both mechanisms contributed to down-regulation ofp15 INK4B [220]. Sriraksa et al. analyzed the methylation pattern of 26 CGIs <strong>in</strong> Opistorchisviverr<strong>in</strong>i-associated cholangiocarc<strong>in</strong>oma samples. There was an elevated frequency of hypermethylationat five areas <strong>in</strong>clud<strong>in</strong>g the ovarian tumor-suppressor OPCML (opioid b<strong>in</strong>d<strong>in</strong>gprote<strong>in</strong>/cell adhesion molecule-like gene) and the immunomodulator DcR1 (decoy receptor 1)[221]. Thus, OPCML and DcR1may serve as methylation biomarkers for cholangiocarc<strong>in</strong>oma.43221.4.2 CpG Island Methylation <strong>in</strong> Schistosoma haematobium-Associated Bladder Carc<strong>in</strong>omaSquamous cell carc<strong>in</strong>oma, and to some extent also transitional cell carc<strong>in</strong>oma of the bladder <strong>in</strong>East Africa and the Middle East and other subtropical areas is frequently associated withchronic ur<strong>in</strong>ary Schistosoma haematobium <strong>in</strong>fection. Chronic mechanical irritation by calcifiedeggs deposited by the worms <strong>in</strong> the bladder epithelium and accumulation of carc<strong>in</strong>ogeniccompounds <strong>in</strong> the ur<strong>in</strong>e may be <strong>in</strong>volved <strong>in</strong> the transformation of the uroepithelium.Although the genomic DNA isolated from adult worms of the related Schistosoma mansoni isunmethylated [222], <strong>in</strong> bladder cancer samples of most probably Schistosoma haematobium<strong>in</strong>fectedpatients from Egypt there was a greater degree of CGI methylation than <strong>in</strong> non-Schistosoma-associated tumors [223].21.5 CONCLUSIONSAlthough most of the work related to epigenetic alterations <strong>in</strong>duced by <strong>in</strong>fectious agents <strong>in</strong>host cells focused on tumor-associated microbes and macroparasites implicated <strong>in</strong> tumorigenesis,it is clear from the overview of the literature that other important bacterial and viralpathogens directly not <strong>in</strong>volved <strong>in</strong> the <strong>in</strong>itiation or ma<strong>in</strong>tenance of neoplasia leave theirepigenetic marks on their target cells as well. There is no doubt <strong>in</strong> our m<strong>in</strong>d that <strong>in</strong> addition toviruses and bacteria, other microparasites, i.e. protozoa that do have their own sophisticatedepigenetic regulatory systems, may also cause epigenetic dysregulation <strong>in</strong> their hosts.Furthermore, tumor-associated viruses may have a role <strong>in</strong> other diseases as well, thus theknowledge as to the epigenetic control of their genomes and the epigenetic changes they elicit<strong>in</strong> neoplastic cells may help to decipher the patho-epigenetic mechanisms caus<strong>in</strong>g dysfunctions<strong>in</strong> non-neoplastic cells. In this respect it is worthy to mention that the epigenetic controlof Epste<strong>in</strong>eBarr virus latency appears to be <strong>in</strong>adequate <strong>in</strong> certa<strong>in</strong> autoimmune diseases, andwas implicated <strong>in</strong> trigger<strong>in</strong>g and perpetuat<strong>in</strong>g the pathogenic processes [224]. Other importantresearch topics are also emerg<strong>in</strong>g, <strong>in</strong>clud<strong>in</strong>g the potential role of microbial and other<strong>in</strong>fections <strong>in</strong> the patho-epigenetics of allergic diseases [225] and the modification of hostepigenetic processes by the microbial communities <strong>in</strong>habit<strong>in</strong>g mucosal surfaces and the sk<strong>in</strong>,or even by probiotic bacteria [226]. The epigenetic changes <strong>in</strong>duced by the vast majority of


CHAPTER 21<strong>Epigenetics</strong> and <strong>Human</strong> Infectious <strong>Disease</strong>smacroparasites (helm<strong>in</strong>ths, fungi, arthropods) <strong>in</strong>fect<strong>in</strong>g humans as well as animals rema<strong>in</strong> tobe explored as well.References[1] M<strong>in</strong>arovits J. Microbe-<strong>in</strong>duced epigenetic alterations <strong>in</strong> host cells: the com<strong>in</strong>g era of patho-epigenetics ofmicrobial <strong>in</strong>fections. A review. Acta Microbiol Immunol Hung 2009;56:1e19.[2] Lopez-Rubio JJ, Riviere L, Scherf A. Shared epigenetic mechanisms control virulence factors <strong>in</strong> protozoanparasites. Curr Op<strong>in</strong> Microbiol 2007;10:560e8.[3] Verstrepen KJ, F<strong>in</strong>k GR. Genetic and epigenetic mechanisms underly<strong>in</strong>g cell-surface variability <strong>in</strong> protozoaand fungi. Annu Rev Genet 2009;43:1e24.[4] Dixon SE, Stilger KL, Elias EV, Naguleswaran A, Sullivan WJ. A decade of epigenetic research <strong>in</strong> Toxoplasmagondii. Mol Biochem Parasitol 2010;173:1e9.[5] Merrick CJ, Durais<strong>in</strong>gh MT. <strong>Epigenetics</strong> <strong>in</strong> Plasmodium: what do we really know? Eukaryot Cell 2010;9:1150e8.[6] da Rosa JL, Kaufman PD. Chromat<strong>in</strong>-mediated Candida albicans virulence. Biochim Biophys Acta2012;1819:349e55.[7] M<strong>in</strong>arovits J. Epigenotypes of latent herpesvirus genomes. Curr Top Microbiol Immunol 2006;310:61e80.[8] Bloom DC, Giordani NV, Kwiatkowski DL. Epigenetic regulation of latent HSV-1 gene expression. BiochimBiophys Acta 2010;1799:246e56.[9] Horwitz GA, Zhang K, McBrian MA, Grunste<strong>in</strong> M, Kurdistani SK, Berk AJ. Adenovirus small e1a alters globalpatterns of histone modification. <strong>Science</strong> 2008;321:1084e5.[10] Ferrari R, Pellegr<strong>in</strong>i M, Horwitz GA, Xie W, Berk AJ, Kurdistani SK. Epigenetic reprogramm<strong>in</strong>g by adenoviruse1a. <strong>Science</strong> 2008;321:1086e8.[11] Pihlstrom BL, Michalowicz BS, Johnson NW. Periodontal diseases. Lancet 2005;366:1809e20.[12] Darveau RP. The oral microbial consortium’s <strong>in</strong>teraction with the periodontal <strong>in</strong>nate defense system. DNACell Biol 2009;28:389e95.[13] Pedra JH, Cassel SL, Sutterwala FS. Sens<strong>in</strong>g pathogens and danger signals by the <strong>in</strong>flammasome. Curr Op<strong>in</strong>Immunol 2009;21:10e6.[14] Manavalan B, Basith S, Choi S. Similar Structures but Different Roles - An Updated Perspective on TLRStructures. Front Physiol 2011;2:41.[15] Philb<strong>in</strong> VJ, Levy O. Developmental biology of the <strong>in</strong>nate immune response: implications for neonatal and<strong>in</strong>fant vacc<strong>in</strong>e development. Pediatr Res 2009;65:98Re105R.[16] Y<strong>in</strong> L, Chung WO. Epigenetic regulation of human beta-defens<strong>in</strong> 2 and CC chemok<strong>in</strong>e ligand 20 expression<strong>in</strong> g<strong>in</strong>gival epithelial cells <strong>in</strong> response to oral bacteria. Mucosal Immunol 2011;4:409e19.[17] Kurita-Ochiai T, Fukushima K, Ochiai K. Volatile fatty acids, metabolic by-products of periodontopathicbacteria, <strong>in</strong>hibit lymphocyte proliferation and cytok<strong>in</strong>e production. J Dent Res 1995;74:1367e73.[18] Niederman R, Buyle-Bod<strong>in</strong> Y, Lu BY, Rob<strong>in</strong>son P, Naleway C. Short-cha<strong>in</strong> carboxylic acid concentration <strong>in</strong>human g<strong>in</strong>gival crevicular fluid. J Dent Res 1997;76:575e9.[19] Tonetti M, Eftimiadi C, Damiani G, Buffa P, Buffa D, Botta GA. Short cha<strong>in</strong> fatty acids present <strong>in</strong> periodontalpockets may play a role <strong>in</strong> human periodontal diseases. J Periodontal Res 1987;22:190e1.[20] Imai K, Ochiai K, Okamoto T. Reactivation of latent HIV-1 <strong>in</strong>fection by the periodontopathic bacteriumPorphyromonas g<strong>in</strong>givalis <strong>in</strong>volves histone modification. J Immunol 2009;182:3688e95.[21] Riggs MG, Whittaker RG, Neumann JR, Ingram VM. n-Butyrate causes histone modification <strong>in</strong> HeLa andFriend erythroleukaemia cells. Nature 1977;268:462e4.[22] Sealy L, Chalkley R. The effect of sodium butyrate on histone modification. Cell 1978;14:115e21.[23] Myzak MC, Ho E, Dashwood RH. Dietary agents as histone deacetylase <strong>in</strong>hibitors. Mol Carc<strong>in</strong>og2006;45:443e6.[24] Bobetsis YA, Barros SP, L<strong>in</strong> DM, Weidman JR, Dol<strong>in</strong>oy DC, Jirtle RL, et al. Bacterial <strong>in</strong>fection promotes DNAhypermethylation. J Dent Res 2007;86:169e74.[25] Paschos K, Allday MJ. Epigenetic reprogramm<strong>in</strong>g of host genes <strong>in</strong> viral and microbial pathogenesis. TrendsMicrobiol 2010;18:439e47.[26] Freitag NE, Port GC, M<strong>in</strong>er MD. Listeria monocytogenes - from saprophyte to <strong>in</strong>tracellular pathogen. Nat RevMicrobiol 2009;7:623e8.[27] Hamon MA, Batsche E, Regnault B, Tham TN, Seveau S, Muchardt C, et al. Histone modifications <strong>in</strong>duced bya family of bacterial tox<strong>in</strong>s. Proc Natl Acad Sci USA 2007;104:13467e72.[28] Schroeder GN, Hilbi H. Molecular pathogenesis of Shigella spp.: controll<strong>in</strong>g host cell signal<strong>in</strong>g, <strong>in</strong>vasion, anddeath by type III secretion. Cl<strong>in</strong> Microbiol Rev 2008;21:134e56.433


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>434[29] Arbibe L, Kim DW, Batsche E, Pedron T, Mateescu B, Muchardt C, et al. An <strong>in</strong>jected bacterial effector targetschromat<strong>in</strong> access for transcription factor NF-kappaB to alter transcription of host genes <strong>in</strong>volved <strong>in</strong> immuneresponses. Nat Immunol 2007;8:47e56.[30] Ismail N, Bloch KC, McBride JW. <strong>Human</strong> ehrlichiosis and anaplasmosis. Cl<strong>in</strong> Lab Med 2010;30:261e92.[31] Garcia-Garcia JC, Barat NC, Trembley SJ, Dumler JS. Epigenetic silenc<strong>in</strong>g of host cell defense genes enhances<strong>in</strong>tracellular survival of the rickettsial pathogen Anaplasma phagocytophilum. PLoS Pathog 2009;5:e1000488.[32] Uemura N, Okamoto S, Yamamoto S, Matsumura N, Yamaguchi S, Yamakido M, et al. Helicobacter pylori<strong>in</strong>fection and the development of gastric cancer. N Engl J Med 2001;345:784e9.[33] Nardone G, Compare D, De Colibus P, de Nucci G, Rocco A. Helicobacter pylori and epigenetic mechanismsunderly<strong>in</strong>g gastric carc<strong>in</strong>ogenesis. Dig Dis 2007;25:225e9.[34] Yamamoto E, Suzuki H, Takamaru H, Yamamoto H, Toyota M, Sh<strong>in</strong>omura Y. Role of DNA methylation <strong>in</strong> thedevelopment of diffuse-type gastric cancer. Digestion 2011;83:241e9.[35] D<strong>in</strong>g SZ, Goldberg JB, Hatakeyama M. Helicobacter pylori <strong>in</strong>fection, oncogenic pathways and epigeneticmechanisms <strong>in</strong> gastric carc<strong>in</strong>ogenesis. Future Oncol 2010;6:851e62.[36] Suerbaum S, Michetti P. Helicobacter pylori <strong>in</strong>fection. N Engl J Med 2002;347:1175e86.[37] Hatakeyama M. Oncogenic mechanisms of the Helicobacter pylori CagA prote<strong>in</strong>. Nat Rev Cancer2004;4:688e94.[38] Umeda M, Murata-Kamiya N, Saito Y, Ohba Y, Takahashi M, Hatakeyama M. Helicobacter pylori CagA causesmitotic impairment and <strong>in</strong>duces chromosomal <strong>in</strong>stability. J Biol Chem 2009;284:22166e72.[39] Kang GH, Shim YH, Jung HY, Kim WH, Ro JY, Rhyu MG. CpG island methylation <strong>in</strong> premalignant stages ofgastric carc<strong>in</strong>oma. Cancer Res 2001;61:2847e51.[40] Kang GH, Lee S, Kim JS, Jung HY. Profile of aberrant CpG island methylation along the multistep pathway ofgastric carc<strong>in</strong>ogenesis. Lab Invest 2003;83:635e41.[41] Yamashita S, Tsuj<strong>in</strong>o Y, Moriguchi K, Tatematsu M, Ushijima T. Chemical genomic screen<strong>in</strong>g for methylationsilencedgenes <strong>in</strong> gastric cancer cell l<strong>in</strong>es us<strong>in</strong>g 5-aza-2’-deoxycytid<strong>in</strong>e treatment and oligonucleotidemicroarray. Cancer Sci 2006;97:64e71.[42] Maekita T, Nakazawa K, Mihara M, Nakajima T, Yanaoka K, Iguchi M, et al. High levels of aberrant DNAmethylation <strong>in</strong> Helicobacter pylori-<strong>in</strong>fected gastric mucosae and its possible association with gastric cancerrisk. Cl<strong>in</strong> Cancer Res 2006;12:989e95.[43] Nakajima T, Maekita T, Oda I, Gotoda T, Yamamoto S, Umemura S, et al. Higher methylation levels <strong>in</strong> gastricmucosae significantly correlate with higher risk of gastric cancers. Cancer Epidemiol Biomarkers Prev2006;15:2317e21.[44] Perri F, Cotugno R, Piepoli A, Merla A, Quitadamo M, Gentile A, et al. Aberrant DNA methylation <strong>in</strong> nonneoplasticgastric mucosa of H. Pylori <strong>in</strong>fected patients and effect of eradication. Am J Gastroenterol2007;102:1361e71.[45] Kaise M, Yamasaki T, Yonezawa J, Miwa J, Ohta Y, Tajiri H. CpG island hypermethylation of tumor-suppressorgenes <strong>in</strong> H. pylori-<strong>in</strong>fected non-neoplastic gastric mucosa is l<strong>in</strong>ked with gastric cancer risk. Helicobacter2008;13:35e41.[46] Park SY, Yoo EJ, Cho NY, Kim N, Kang GH. Comparison of CpG island hypermethylation and repetitive DNAhypomethylation <strong>in</strong> premalignant stages of gastric cancer, stratified for Helicobacter pylori <strong>in</strong>fection. J Pathol2009;219:410e6.[47] Compare D, Rocco A, Liguori E, D’Armiento FP, Persico G, Masone S, et al. Global DNA hypomethylation isan early event <strong>in</strong> Helicobacter pylori-related gastric carc<strong>in</strong>ogenesis. J Cl<strong>in</strong> Pathol 2011;64:677e82.[48] Waki T, Tamura G, Tsuchiya T, Sato K, Nishizuka S, Motoyama T. Promoter methylation status of E-cadher<strong>in</strong>,hMLH1, and p16 genes <strong>in</strong> nonneoplastic gastric epithelia. Am J Pathol 2002;161:399e403.[49] Chan AO, Peng JZ, Lam SK, Lai KC, Yuen MF, Cheung HK, et al. Eradication of Helicobacter pylori <strong>in</strong>fectionreverses E-cadher<strong>in</strong> promoter hypermethylation. Gut 2006;55:463e8.[50] Leung WK, Man EP, Yu J, Go MY, To KF, Yamaoka Y, et al. Effects of Helicobacter pylori eradication onmethylation status of E-cadher<strong>in</strong> gene <strong>in</strong> noncancerous stomach. Cl<strong>in</strong> Cancer Res 2006;12:3216e21.[51] Nakajima T, Enomoto S, Yamashita S, Ando T, Nakanishi Y, Nakazawa K, et al. Persistence of a component ofDNA methylation <strong>in</strong> gastric mucosae after Helicobacter pylori eradication. J Gastroenterol 2010;45:37e44.[52] Kabir S. Effect of Helicobacter pylori eradication on <strong>in</strong>cidence of gastric cancer <strong>in</strong> human and animal models:underly<strong>in</strong>g biochemical and molecular events. Helicobacter 2009;14:159e71.[53] Niwa T, Tsukamoto T, Toyoda T, Mori A, Tanaka H, Maekita T, et al. Inflammatory processes triggered byHelicobacter pylori <strong>in</strong>fection cause aberrant DNA methylation <strong>in</strong> gastric epithelial cells. Cancer Res2010;70:1430e40.[54] Kang HY, Kim N, Park YS, Hwang JH, Kim JW, Jeong SH, et al. Progression of atrophic gastritis and <strong>in</strong>test<strong>in</strong>almetaplasia drives Helicobacter pylori out of the gastric mucosa. Dig Dis Sci 2006;51:2310e5.


CHAPTER 21<strong>Epigenetics</strong> and <strong>Human</strong> Infectious <strong>Disease</strong>s[55] Ushijima T. Epigenetic field for cancerization. J Biochem Mol Biol 2007;40:142e50.[56] Guilford P, Hopk<strong>in</strong>s J, Harraway J, McLeod M, McLeod N, Harawira P, et al. E-cadher<strong>in</strong> germl<strong>in</strong>e mutations <strong>in</strong>familial gastric cancer. Nature 1998;392:402e5.[57] Grady WM, Willis J, Guilford PJ, Dunbier AK, Toro TT, Lynch H, et al. Methylation of the CDH1 promoter asthe second genetic hit <strong>in</strong> hereditary diffuse gastric cancer. Nat Genet 2000;26:16e7.[58] Machado JC, Oliveira C, Carvalho R, Soares P, Berx G, Caldas C, et al. E-cadher<strong>in</strong> gene (CDH1) promotermethylation as the second hit <strong>in</strong> sporadic diffuse gastric carc<strong>in</strong>oma. Oncogene 2001;20:1525e8.[59] Miyazaki T, Murayama Y, Sh<strong>in</strong>omura Y, Yamamoto T, Watabe K, Tsutsui S, et al. E-cadher<strong>in</strong> gene promoterhypermethylation <strong>in</strong> H. pylori-<strong>in</strong>duced enlarged fold gastritis. Helicobacter 2007;12:523e31.[60] Terres AM, Pajares JM, O’Toole D, Ahern S, Kelleher D. H pylori <strong>in</strong>fection is associated with downregulationof E-cadher<strong>in</strong>, a molecule <strong>in</strong>volved <strong>in</strong> epithelial cell adhesion and proliferation control. J Cl<strong>in</strong> Pathol1998;51:410e2.[61] Chan AO, Lam SK, Wong BC, Wong WM, Yuen MF, Yeung YH, et al. Promoter methylation of E-cadher<strong>in</strong> gene<strong>in</strong> gastric mucosa associated with Helicobacter pylori <strong>in</strong>fection and <strong>in</strong> gastric cancer. Gut 2003;52:502e6.[62] Yamashita K, Sakuramoto S, Watanabe M. Genomic and epigenetic profiles of gastric cancer: potentialdiagnostic and therapeutic applications. Surg Today 2011;41:24e38.[63] Nakajima T, Yamashita S, Maekita T, Niwa T, Nakazawa K, Ushijima T. The presence of a methylationf<strong>in</strong>gerpr<strong>in</strong>t of Helicobacter pylori <strong>in</strong>fection <strong>in</strong> human gastric mucosae. Int J Cancer 2009;124:905e10.[64] Sh<strong>in</strong> CM, Kim N, Jung Y, Park JH, Kang GH, Park WY, et al. Genome-wide DNA methylation profiles <strong>in</strong>noncancerous gastric mucosae with regard to Helicobacter pylori <strong>in</strong>fection and the presence of gastric cancer.Helicobacter 2011;16:179e88.[65] Yan J, Zhang M, Zhang J, Chen X, Zhang X. Helicobacter pylori <strong>in</strong>fection promotes methylation of WWOXgene <strong>in</strong> human gastric cancer. Biochem Biophys Res Commun 2011;408:99e102.[66] Takeshima H, Yamashita S, Shimazu T, Niwa T, Ushijima T. The presence of RNA polymerase II, active orstalled, predicts epigenetic fate of promoter CpG islands. Genome Res 2009;19:1974e82.[67] Takeshima H, Ushijima T. Methylation dest<strong>in</strong>y: Moira takes account of histones and RNA polymerase II.<strong>Epigenetics</strong> 2010;5:89e95.[68] Vitkute J, Stankevicius K, Tamulaitiene G, Maneliene Z, Tim<strong>in</strong>skas A, Berg DE, et al. Specificities of elevendifferent DNA methyltransferases of Helicobacter pylori stra<strong>in</strong> 26695. J Bacteriol 2001;183:443e50.[69] Alves MK, Ferrasi AC, Lima VP, Ferreira MV, de Moura Campos Pard<strong>in</strong>i MI, Rabenhorst SH. Inactivation ofCOX-2, HMLH1 and CDKN2A Gene by Promoter Methylation <strong>in</strong> Gastric Cancer: Relationship with HistologicalSubtype, Tumor Location and Helicobacter pylori Genotype. Pathobiology 2011;78:266e76.[70] Bussiere FI, Michel V, Memet S, Ave P, Vivas JR, Huerre M, et al. H. pylori-<strong>in</strong>duced promoter hypermethylationdownregulates USF1 and USF2 transcription factor gene expression. Cell Microbiol 2010;12:1124e33.[71] Touati E. When bacteria become mutagenic and carc<strong>in</strong>ogenic: lessons from H. pylori. Mutat Res2010;703:66e70.[72] Sepulveda AR, Yao Y, Yan W, Park DI, Kim JJ, Good<strong>in</strong>g W, et al. CpG methylation and reduced expression ofO6-methylguan<strong>in</strong>e DNA methyltransferase is associated with Helicobacter pylori <strong>in</strong>fection. Gastroenterology2010;138:1836e44.[73] Ando T, Yoshida T, Enomoto S, Asada K, Tatematsu M, Ich<strong>in</strong>ose M, et al. DNA methylation of microRNAgenes <strong>in</strong> gastric mucosae of gastric cancer patients: its possible <strong>in</strong>volvement <strong>in</strong> the formation of epigeneticfield defect. Int J Cancer 2009;124:2367e74.[74] Suzuki H, Yamamoto E, Nojima M, Kai M, Yamano HO, Yoshikawa K, et al. Methylation-associated silenc<strong>in</strong>gof microRNA-34b/c <strong>in</strong> gastric cancer and its <strong>in</strong>volvement <strong>in</strong> an epigenetic field defect. Carc<strong>in</strong>ogenesis2010;31:2066e73.[75] Craig VJ, Cogliatti SB, Rehrauer H, Wundisch T, Muller A. Epigenetic silenc<strong>in</strong>g of microRNA-203 dysregulatesABL1 expression and drives Helicobacter-associated gastric lymphomagenesis. Cancer Res 2011;71:3616e24.[76] Matsushima K, Isomoto H, Inoue N, Nakayama T, Hayashi T, Nakayama M, et al. MicroRNA signatures <strong>in</strong>Helicobacter pylori-<strong>in</strong>fected gastric mucosa. Int J Cancer 2011;128:361e70.[77] Schles<strong>in</strong>ger Y, Straussman R, Keshet I, Farkash S, Hecht M, Zimmerman J, et al. Polycomb-mediated methylationon Lys27 of histone H3 pre-marks genes for de novo methylation <strong>in</strong> cancer. Nat Genet 2007;39:232e6.[78] Widschwendter M, Fiegl H, Egle D, Mueller-Holzner E, Spizzo G, Marth C, et al. Epigenetic stem cell signature<strong>in</strong> cancer. Nat Genet 2007;39:157e8.[79] Ballestar E, Esteller M. Epigenetic gene regulation <strong>in</strong> cancer. Adv Genet 2008;61:247e67.[80] Hahn MA, Hahn T, Lee DH, Esworthy RS, Kim BW, Riggs AD, et al. Methylation of polycomb target genes <strong>in</strong><strong>in</strong>test<strong>in</strong>al cancer is mediated by <strong>in</strong>flammation. Cancer Res 2008;68:10280e9.[81] Pathak SK, Basu S, Bhattacharyya A, Pathak S, Banerjee A, Basu J, et al. TLR4-dependent NF-kappaB activationand mitogen- and stress-activated prote<strong>in</strong> k<strong>in</strong>ase 1-triggered phosphorylation events are central to435


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>436Helicobacter pylori peptidyl prolyl cis-, trans-isomerase (HP0175)-mediated <strong>in</strong>duction of IL-6 release frommacrophages. J Immunol 2006;177:7950e8.[82] Fehri LF, Rechner C, Janssen S, Mak TN, Holland C, Bartfeld S, et al. Helicobacter pylori-<strong>in</strong>duced modificationof the histone H3 phosphorylation status <strong>in</strong> gastric epithelial cells reflects its impact on cell cycle regulation.<strong>Epigenetics</strong> 2009;4:577e86.[83] D<strong>in</strong>g SZ, Fischer W, Kaparakis-Liaskos M, Liechti G, Merrell DS, Grant PA, et al. Helicobacter pylori-<strong>in</strong>ducedhistone modification, associated gene expression <strong>in</strong> gastric epithelial cells, and its implication <strong>in</strong> pathogenesis.PLoS ONE 2010;5:e9875.[84] Tolg C, Sabha N, Cortese R, Panchal T, Ahsan A, Soliman A, et al. Uropathogenic E. coli <strong>in</strong>fection provokesepigenetic downregulation of CDKN2A (p16INK4A) <strong>in</strong> uroepithelial cells. Lab Invest 2011;91:825e36.[85] Murata M, Azuma Y, Miura K, Rahman MA, Matsutani M, Aoyama M, et al. Chlamydial SET doma<strong>in</strong> prote<strong>in</strong>functions as a histone methyltransferase. Microbiology 2007;153:585e92.[86] Penn<strong>in</strong>i ME, Perr<strong>in</strong>et S, Dautry-Varsat A, Subtil A. Histone methylation by NUE, a novel nuclear effector ofthe <strong>in</strong>tracellular pathogen Chlamydia trachomatis. PLoS Pathog 2010;6:e1000995.[87] Niller HH, Wolf H, M<strong>in</strong>arovits J. Epste<strong>in</strong>eBarr Virus. In: M<strong>in</strong>arovits J, Gonczol E, Valyi-Nagy T, editors.Latency Strategies of Herpesviruses. New York: Spr<strong>in</strong>ger; 2007. p. 154e91.[88] Swam<strong>in</strong>athan S. Noncod<strong>in</strong>g RNAs produced by oncogenic human herpesviruses. J Cell Physiol 2008;216:321e6.[89] Tempera I, Klich<strong>in</strong>sky M, Lieberman PM. EBV latency types adopt alternative chromat<strong>in</strong> conformations. PLoSPathog 2011;7:e1002180.[90] Wang L, Grossman SR, Kieff E. Epste<strong>in</strong>eBarr virus nuclear prote<strong>in</strong> 2 <strong>in</strong>teracts with p300, CBP, and PCAFhistone acetyltransferases <strong>in</strong> activation of the LMP1 promoter. Proc Natl Acad Sci USA 2000;97:430e5.[91] Portal D, Rosendorff A, Kieff E. Epste<strong>in</strong>eBarr nuclear antigen leader prote<strong>in</strong> coactivates transcription through<strong>in</strong>teraction with histone deacetylase 4. Proc Natl Acad Sci USA 2006;103:19278e83.[92] Cotter MA, Robertson ES. Modulation of histone acetyltransferase activity through <strong>in</strong>teraction ofEpste<strong>in</strong>eBarr nuclear antigen 3C with prothymos<strong>in</strong> alpha. Mol Cell Biol 2000;20:5722e35.[93] Subramanian C, Hasan S, Rowe M, Hottiger M, Orre R, Robertson ES. Epste<strong>in</strong>eBarr virus nuclear antigen 3Cand prothymos<strong>in</strong> alpha <strong>in</strong>teract with the p300 transcriptional coactivator at the CH1 and CH3/HAT doma<strong>in</strong>sand cooperate <strong>in</strong> regulation of transcription and histone acetylation. J Virol 2002;76:4699e708.[94] Knight JS, Lan K, Subramanian C, Robertson ES. Epste<strong>in</strong>eBarr virus nuclear antigen 3C recruits histonedeacetylase activity and associates with the corepressors mS<strong>in</strong>3A and NCoR <strong>in</strong> human B-cell l<strong>in</strong>es. J Virol2003;77:4261e72.[95] Skalska L, White RE, Franz M, Ruhmann M, Allday MJ. Epigenetic repression of p16(INK4A) by latentEpste<strong>in</strong>eBarr virus requires the <strong>in</strong>teraction of EBNA3A and EBNA3C with CtBP. PLoS Pathog 2010;6:e1000951.[96] Anderton E, Yee J, Smith P, Crook T, White RE, Allday MJ. Two Epste<strong>in</strong>eBarr virus (EBV) oncoprote<strong>in</strong>scooperate to repress expression of the proapoptotic tumour-suppressor Bim: clues to the pathogenesis ofBurkitt’s lymphoma. Oncogene 2008;27:421e33.[97] Paschos K, Smith P, Anderton E, Middeldorp JM, White RE, Allday MJ. Epste<strong>in</strong>eBarr virus latency <strong>in</strong> B cellsleads to epigenetic repression and CpG methylation of the tumour suppressor gene Bim. PLoS Pathog2009;5:e1000492.[98] Tsai CN, Tsai CL, Tse KP, Chang HY, Chang YS. The Epste<strong>in</strong>eBarr virus oncogene product, latent membraneprote<strong>in</strong> 1, <strong>in</strong>duces the downregulation of E-cadher<strong>in</strong> gene expression via activation of DNA methyltransferases.Proc Natl Acad Sci USA 2002;99:10084e9.[99] Tsai CL, Li HP, Lu YJ, Hsueh C, Liang Y, Chen CL, et al. Activation of DNA methyltransferase 1 by EBV LMP1Involves c-Jun NH(2)-term<strong>in</strong>al k<strong>in</strong>ase signal<strong>in</strong>g. Cancer Res 2006;66:11668e76.[100] Dutton A, Woodman CB, Chukwuma MB, Last JI, Wei W, Vockerodt M, et al. Bmi-1 is <strong>in</strong>duced by theEpste<strong>in</strong>eBarr virus oncogene LMP1 and regulates the expression of viral target genes <strong>in</strong> Hodgk<strong>in</strong> lymphomacells. Blood 2007;109:2597e603.[101] Niller HH, Wolf H, M<strong>in</strong>arovits J. Epigenetic dysregulation of the host cell genome <strong>in</strong> Epste<strong>in</strong>eBarr virusassociatedneoplasia. Sem<strong>in</strong> Cancer Biol 2009;19:158e64.[102] Kwong J, Lo KW, To KF, Teo PM, Johnson PJ, Huang DP. Promoter hypermethylation of multiple genes <strong>in</strong>nasopharyngeal carc<strong>in</strong>oma. Cl<strong>in</strong> Cancer Res 2002;8:131e7.[103] Murray PG, Qiu GH, Fu L, Waites ER, Srivastava G, Heys D, et al. Frequent epigenetic <strong>in</strong>activation of theRASSF1A tumor suppressor gene <strong>in</strong> Hodgk<strong>in</strong>’s lymphoma. Oncogene 2004;23:1326e31.[104] Doerr JR, Malone CS, Fike FM, Gordon MS, Soghomonian SV, Thomas RK, et al. Patterned CpG methylationof silenced B cell gene promoters <strong>in</strong> classical Hodgk<strong>in</strong> lymphoma-derived and primary effusion lymphomacell l<strong>in</strong>es. J Mol Biol 2005;350:631e40.[105] Ushmorov A, Leithauser F, Sakk O, We<strong>in</strong>hausel A, Popov SW, Moller P, et al. Epigenetic processes play a majorrole <strong>in</strong> B-cell-specific gene silenc<strong>in</strong>g <strong>in</strong> classical Hodgk<strong>in</strong> lymphoma. Blood 2006;107:2493e500.


CHAPTER 21<strong>Epigenetics</strong> and <strong>Human</strong> Infectious <strong>Disease</strong>s[106] Rossi D, Gaidano G, Glogh<strong>in</strong>i A, Deambrogi C, Franceschetti S, Berra E, et al. Frequent aberrant promoterhypermethylation of O6-methylguan<strong>in</strong>e-DNA methyltransferase and death-associated prote<strong>in</strong> k<strong>in</strong>ase genes <strong>in</strong>immunodeficiency-related lymphomas. Br J Haematol 2003;123:475e8.[107] Au WY, Ma ES, Choy C, Chung LP, Fung TK, Liang R, et al. Therapy-related lymphomas <strong>in</strong> patients with autoimmunediseases after treatment with disease-modify<strong>in</strong>g anti-rheumatic drugs. Am J Hematol 2006;81:5e11.[108] H<strong>in</strong>o R, Uozaki H, Murakami N, Ushiku T, Sh<strong>in</strong>ozaki A, Ishikawa S, et al. Activation of DNA methyltransferase1 by EBV latent membrane prote<strong>in</strong> 2A leads to promoter hypermethylation of PTEN gene <strong>in</strong>gastric carc<strong>in</strong>oma. Cancer Res 2009;69:2766e74.[109] Georgescu MM. PTEN Tumor Suppressor Network <strong>in</strong> PI3K-Akt Pathway Control. Genes Cancer2010;1:1170e7.[110] Kang GH, Lee S, Kim WH, Lee HW, Kim JC, Rhyu MG, et al. Epste<strong>in</strong>eBarr virus-positive gastric carc<strong>in</strong>omademonstrates frequent aberrant methylation of multiple genes and constitutes CpG island methylatorphenotype-positive gastric carc<strong>in</strong>oma. Am J Pathol 2002;160:787e94.[111] Chang MS, Uozaki H, Chong JM, Ushiku T, Sakuma K, Ishikawa S, et al. CpG island methylation status <strong>in</strong>gastric carc<strong>in</strong>oma with and without <strong>in</strong>fection of Epste<strong>in</strong>eBarr virus. Cl<strong>in</strong> Cancer Res 2006;12:2995e3002.[112] Luo B, Wang Y, Wang XF, Liang H, Yan LP, Huang BH, et al. Expression of Epste<strong>in</strong>eBarr virus genes <strong>in</strong> EBVassociatedgastric carc<strong>in</strong>omas. World J Gastroenterol 2005;11:629e33.[113] Pantry SN, Medveczky PG. Epigenetic regulation of Kaposi’s sarcoma-associated herpesvirus replication.Sem<strong>in</strong> Cancer Biol 2009;19:153e7.[114] Stedman W, Deng Z, Lu F, Lieberman PM. ORC, MCM, and histone hyperacetylation at the Kaposi’s sarcomaassociatedherpesvirus latent replication orig<strong>in</strong>. J Virol 2004;78:12566e75.[115] Sakakibara S, Ueda K, Nishimura K, Do E, Ohsaki E, Okuno T, et al. Accumulation of heterochromat<strong>in</strong>components on the term<strong>in</strong>al repeat sequence of Kaposi’s sarcoma-associated herpesvirus mediated by thelatency-associated nuclear antigen. J Virol 2004;78:7299e310.[116] Gunther T, Grundhoff A. The epigenetic landscape of latent Kaposi sarcoma-associated herpesvirus genomes.PLoS Pathog 2010;6:e1000935.[117] Toth Z, Magl<strong>in</strong>te DT, Lee SH, Lee HR, Wong LY, Brulois KF, et al. Epigenetic analysis of KSHV latent and lyticgenomes. PLoS Pathog 2010;6:e1001013.[118] Kang H, Wiedmer A, Yuan Y, Robertson E, Lieberman PM. Coord<strong>in</strong>ation of KSHV latent and lytic gene controlby CTCF-cohes<strong>in</strong> mediated chromosome conformation. PLoS Pathog 2011;7:e1002140.[119] Lu F, Stedman W, Yousef M, Renne R, Lieberman PM. Epigenetic regulation of Kaposi’s sarcoma-associatedherpesvirus latency by virus-encoded microRNAs that target Rta and the cellular Rbl2-DNMT pathway. J Virol2010;84:2697e706.[120] Shamay M, Krithivas A, Zhang J, Hayward SD. Recruitment of the de novo DNA methyltransferase Dnmt3aby Kaposi’s sarcoma-associated herpesvirus LANA. Proc Natl Acad Sci USA 2006;103:14554e9.[121] Di Bartolo DL, Cannon M, Liu YF, Renne R, Chadburn A, et al. KSHV LANA <strong>in</strong>hibits TGF-beta signal<strong>in</strong>gthrough epigenetic silenc<strong>in</strong>g of the TGF-beta type II receptor. Blood 2008;111:4731e40.[122] Krithivas A, Young DB, Liao G, Greene D, Hayward SD. <strong>Human</strong> herpesvirus 8 LANA <strong>in</strong>teracts with prote<strong>in</strong>s ofthe mS<strong>in</strong>3 corepressor complex and negatively regulates Epste<strong>in</strong>eBarr virus gene expression <strong>in</strong> dually<strong>in</strong>fected PEL cells. J Virol 2000;74:9637e45.[123] Lim C, Lee D, Seo T, Choi C, Choe J. Latency-associated nuclear antigen of Kaposi’s sarcoma-associatedherpesvirus functionally <strong>in</strong>teracts with heterochromat<strong>in</strong> prote<strong>in</strong> 1. J Biol Chem 2003;278:7397e405.[124] Stuber G, Mattsson K, Flaberg E, Kati E, Markasz L, Sheldon JA, et al. HHV-8 encoded LANA-1 alters thehigher organization of the cell nucleus. Mol Cancer 2007;6:28.[125] Matsumura S, Persson LM, Wong L, Wilson AC. The latency-associated nuclear antigen <strong>in</strong>teracts with MeCP2and nucleosomes through separate doma<strong>in</strong>s. J Virol 2010;84:2318e30.[126] Platt G, Carbone A, Mittnacht S. p16INK4a loss and sensitivity <strong>in</strong> KSHV associated primary effusionlymphoma. Oncogene 2002;21:1823e31.[127] Carbone A, Cilia AM, Glogh<strong>in</strong>i A, Capello D, Fassone L, Per<strong>in</strong> T, et al. Characterization of a novelHHV-8-positive cell l<strong>in</strong>e reveals implications for the pathogenesis and cell cycle control of primary effusionlymphoma. Leukemia 2000;14:1301e9.[128] Gonczol E, Andrews PW, Plotk<strong>in</strong> SA. Cytomegalovirus replicates <strong>in</strong> differentiated but not <strong>in</strong> undifferentiatedhuman embryonal carc<strong>in</strong>oma cells. <strong>Science</strong> 1984;224:159e61.[129] Murphy JC, Fischle W, Verd<strong>in</strong> E, S<strong>in</strong>clair JH. Control of cytomegalovirus lytic gene expression by histoneacetylation. EMBO J 2002;21:1112e20.[130] Wright E, Ba<strong>in</strong> M, Teague L, Murphy J, S<strong>in</strong>clair J. Ets-2 repressor factor recruits histone deacetylase to silencehuman cytomegalovirus immediate-early gene expression <strong>in</strong> non-permissive cells. J Gen Virol2005;86:535e44.437


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>438[131] Park JJ, Kim YE, Pham HT, Kim ET, Chung YH, Ahn JH. Functional <strong>in</strong>teraction of the human cytomegalovirusIE2 prote<strong>in</strong> with histone deacetylase 2 <strong>in</strong> <strong>in</strong>fected human fibroblasts. J Gen Virol 2007;88:3214e23.[132] Nevels M, Paulus C, Shenk T. <strong>Human</strong> cytomegalovirus immediate-early 1 prote<strong>in</strong> facilitates viral replicationby antagoniz<strong>in</strong>g histone deacetylation. Proc Natl Acad Sci USA 2004;101:17234e9.[133] Hayhurst GP, Bryant LA, Caswell RC, Walker SM, S<strong>in</strong>clair JH. CCAAT box-dependent activation of the TATAlesshuman DNA polymerase alpha promoter by the human cytomegalovirus 72-kilodalton majorimmediate-early prote<strong>in</strong>. J Virol 1995;69:182e8.[134] Caswell R, Bryant L, S<strong>in</strong>clair J. <strong>Human</strong> cytomegalovirus immediate-early 2 (IE2) prote<strong>in</strong> can transactivate thehuman hsp70 promoter by alleviation of Dr1-mediated repression. J Virol 1996;70:4028e37.[135] Straat K, Liu C, Rahbar A, Zhu Q, Liu L, Wolmer-Solberg N, et al. Activation of telomerase by humancytomegalovirus. J Natl Cancer Inst 2009;101:488e97.[136] Michaelis M, Doerr HW, C<strong>in</strong>atl J. The story of human cytomegalovirus and cancer: <strong>in</strong>creas<strong>in</strong>g evidence andopen questions. Neoplasia 2009;11:1e9.[137] Matsuoka M. <strong>Human</strong> T-cell leukemia virus type I (HTLV-I) <strong>in</strong>fection and the onset of adult T-cell leukemia(ATL). Retrovirology 2005;2:27.[138] Lez<strong>in</strong> A, Gillet N, Ol<strong>in</strong>do S, Signate A, Grandvaux N, Verlaeten O, et al. Histone deacetylase mediatedtranscriptional activation reduces proviral loads <strong>in</strong> HTLV-1 associated myelopathy/tropical spastic paraparesispatients. Blood 2007;110:3722e8.[139] Sato H, Oka T, Sh<strong>in</strong>nou Y, Kondo T, Washio K, Takano M, et al. Multi-step aberrant CpG island hyper-methylationis associated with the progression of adult T-cell leukemia/lymphoma. Am J Pathol 2010;176:402e15.[140] Nakase K, Cheng J, Zhu Q, Marasco WA. Mechanisms of SHP-1 P2 promoter regulation <strong>in</strong> hematopoieticcells and its silenc<strong>in</strong>g <strong>in</strong> HTLV-1-transformed T cells. J Leukoc Biol 2009;85:165e74.[141] Niller HH, Wolf H, M<strong>in</strong>arovits J. Viral hit and run-oncogenesis: Genetic and epigenetic scenarios. Cancer Lett2011;305:200e17.[142] Aoki Y, Tosato G. Neoplastic conditions <strong>in</strong> the context of HIV-1 <strong>in</strong>fection. Curr HIV Res 2004;2:343e9.[143] Grulich AE, van Leeuwen MT, Falster MO, Vajdic CM. Incidence of cancers <strong>in</strong> people with HIV/AIDScompared with immunosuppressed transplant recipients: a meta-analysis. Lancet 2007;370:59e67.[144] Elgui de Oliveira D. DNA viruses <strong>in</strong> human cancer: an <strong>in</strong>tegrated overview on fundamental mechanisms ofviral carc<strong>in</strong>ogenesis. Cancer Lett 2007;247:182e96.[145] F<strong>in</strong>zi D, Hermankova M, Pierson T, Carruth LM, Buck C, Chaisson RE, et al. Identification of a reservoir forHIV-1 <strong>in</strong> patients on highly active antiretroviral therapy. <strong>Science</strong> 1997;278:1295e300.[146] Siliciano JD, Kajdas J, F<strong>in</strong>zi D, Qu<strong>in</strong>n TC, Chadwick K, Margolick JB, et al. Long-term follow-up studiesconfirm the stability of the latent reservoir for HIV-1 <strong>in</strong> rest<strong>in</strong>g CD4þ T cells. Nat Med 2003;9:727e8.[147] Richman DD, Margolis DM, Delaney M, Greene WC, Hazuda D, Pomerantz RJ. The challenge of f<strong>in</strong>d<strong>in</strong>ga cure for HIV <strong>in</strong>fection. <strong>Science</strong> 2009;323:1304e7.[148] Margolis DM. Mechanisms of HIV latency: an emerg<strong>in</strong>g picture of complexity. Curr HIV/AIDS Rep2010;7:37e43.[149] Bednarik DP, Mosca JD, Raj NB. Methylation as a modulator of expression of human immunodeficiencyvirus. J Virol 1987;61:1253e7.[150] Bednarik DP, Cook JA, Pitha PM. Inactivation of the HIV LTR by DNA CpG methylation: evidence for a role<strong>in</strong> latency. EMBO J 1990;9:1157e64.[151] Schulze-Forster K, Gotz F, Wagner H, Kroger H, Simon D. Transcription of HIV1 is <strong>in</strong>hibited by DNAmethylation. Biochem Biophys Res Commun 1990;168:141e7.[152] Van L<strong>in</strong>t C, Emiliani S, Ott M, Verd<strong>in</strong> E. Transcriptional activation and chromat<strong>in</strong> remodel<strong>in</strong>g of the HIV-1promoter <strong>in</strong> response to histone acetylation. EMBO J 1996;15:1112e20.[153] Coull JJ, Romerio F, Sun JM, Volker JL, Galv<strong>in</strong> KM, Davie JR, et al. The human factors YY1 and LSF repress thehuman immunodeficiency virus type 1 long term<strong>in</strong>al repeat via recruitment of histone deacetylase 1. J Virol2000;74:6790e9.[154] Ylisastigui L, Arch<strong>in</strong> NM, Lehrman G, Bosch RJ, Margolis DM. Coax<strong>in</strong>g HIV-1 from rest<strong>in</strong>g CD4 T cells:histone deacetylase <strong>in</strong>hibition allows latent viral expression. AIDS 2004;18:1101e8.[155] Friedman J, Cho WK, Chu CK, Keedy KS, Arch<strong>in</strong> NM, Margolis DM, et al. Epigenetic Silenc<strong>in</strong>g of HIV-1 by theHistone H3 Lys<strong>in</strong>e 27 Methyltransferase Enhancer of Zeste 2. J Virol 2011;85:9078e89.[156] Jiang G, Espeseth A, Hazuda DJ, Margolis DM. c-Myc and Sp1 contribute to proviral latency by recruit<strong>in</strong>ghistone deacetylase 1 to the human immunodeficiency virus type 1 promoter. J Virol 2007;81:10914e23.[157] Tyagi M, Karn J. CBF-1 promotes transcriptional silenc<strong>in</strong>g dur<strong>in</strong>g the establishment of HIV-1 latency. EMBO J2007;26:4985e95.[158] Schroder AR, Sh<strong>in</strong>n P, Chen H, Berry C, Ecker JR, Bushman F. HIV-1 <strong>in</strong>tegration <strong>in</strong> the human genome favorsactive genes and local hotspots. Cell 2002;110:521e9.


CHAPTER 21<strong>Epigenetics</strong> and <strong>Human</strong> Infectious <strong>Disease</strong>s[159] Han Y, Lassen K, Monie D, Sedaghat AR, Shimoji S, Liu X, et al. Rest<strong>in</strong>g CD4þ T cells from humanimmunodeficiency virus type 1 (HIV-1)-<strong>in</strong>fected <strong>in</strong>dividuals carry <strong>in</strong>tegrated HIV-1 genomes with<strong>in</strong> activelytranscribed host genes. J Virol 2004;78:6122e33.[160] Han Y, L<strong>in</strong> YB, An W, Xu J, Yang HC, O’Connell K, et al. Orientation-dependent regulation of <strong>in</strong>tegratedHIV-1 expression by host gene transcriptional readthrough. Cell Host Microbe 2008;4:134e46.[161] Klase Z, Kale P, W<strong>in</strong>ograd R, Gupta MV, Heydarian M, Berro R, et al. HIV-1 TAR element is processed by Dicerto yield a viral micro-RNA <strong>in</strong>volved <strong>in</strong> chromat<strong>in</strong> remodel<strong>in</strong>g of the viral LTR. BMC Mol Biol 2007;8:63.[162] Huang J, Wang F, Argyris E, Chen K, Liang Z, Tian H, et al. Cellular microRNAs contribute to HIV-1 latency <strong>in</strong>rest<strong>in</strong>g primary CD4þ T lymphocytes. Nat Med 2007;13:1241e7.[163] Mikovits JA, Young HA, Vert<strong>in</strong>o P, Issa JP, Pitha PM, Turcoski-Corrales S, et al. Infection with humanimmunodeficiency virus type 1 upregulates DNA methyltransferase, result<strong>in</strong>g <strong>in</strong> de novo methylation of thegamma <strong>in</strong>terferon (IFN-gamma) promoter and subsequent downregulation of IFN-gamma production. MolCell Biol 1998;18:5166e77.[164] Fang JY, Mikovits JA, Bagni R, Petrow-Sadowski CL, Ruscetti FW. Infection of lymphoid cells by <strong>in</strong>tegrationdefectivehuman immunodeficiency virus type 1 <strong>in</strong>creases de novo methylation. J Virol 2001;75:9753e61.[165] Giordanengo V, Ollier L, Lanteri M, Lesimple J, March D, Thyss S, et al. Epigenetic reprogramm<strong>in</strong>g of UDP-Nacetylglucosam<strong>in</strong>e2-epimerase/N-acetylmannosam<strong>in</strong>e k<strong>in</strong>ase (GNE) <strong>in</strong> HIV-1-<strong>in</strong>fected CEM T cells. FASEB J2004;18:1961e3.[166] Youngblood B, Reich NO. The early expressed HIV-1 genes regulate DNMT1 expression. <strong>Epigenetics</strong>2008;3:149e56.[167] Saiyed ZM, Gandhi N, Agudelo M, Napuri J, Samikkannu T, Reddy PV, et al. HIV-1 Tat upregulates expressionof histone deacetylase-2 (HDAC2) <strong>in</strong> human neurons: implication for HIV-associated neurocognitivedisorder (HAND). Neurochem Int 2011;58:656e64.[168] Tischoff I, Tannapfel A. DNA methylation <strong>in</strong> hepatocellular carc<strong>in</strong>oma. World J Gastroenterol2008;14:1741e8.[169] Shim YH, Yoon GS, Choi HJ, Chung YH, Yu E. p16 Hypermethylation <strong>in</strong> the early stage of hepatitis B virusassociatedhepatocarc<strong>in</strong>ogenesis. Cancer Lett 2003;190:213e9.[170] Um TH, Kim H, Oh BK, Kim MS, Kim KS, Jung G, et al. Aberrant CpG island hypermethylation <strong>in</strong> dysplasticnodules and early HCC of hepatitis B virus-related human multistep hepatocarc<strong>in</strong>ogenesis. J Hepatol2011;54:939e47.[171] Doria M, Kle<strong>in</strong> N, Lucito R, Schneider RJ. The hepatitis B virus HBx prote<strong>in</strong> is a dual specificity cytoplasmicactivator of Ras and nuclear activator of transcription factors. EMBO J 1995;14:4747e57.[172] Haviv I, Shamay M, Doitsh G, Shaul Y. Hepatitis B virus pX targets TFIIB <strong>in</strong> transcription coactivation. MolCell Biol 1998;18:1562e9.[173] Benn J, Schneider RJ. Hepatitis B virus HBx prote<strong>in</strong> activates Ras-GTP complex formation and establishesa Ras, Raf, MAP k<strong>in</strong>ase signal<strong>in</strong>g cascade. Proc Natl Acad Sci USA 1994;91:10350e4.[174] Choi BH, Choi M, Jeon HY, Rho HM. Hepatitis B viral X prote<strong>in</strong> overcomes <strong>in</strong>hibition of E2F1 activity bypRb on the human Rb gene promoter. DNA Cell Biol 2001;20:75e80.[175] Wang WH, Hull<strong>in</strong>ger RL, Andrisani OM. Hepatitis B virus X prote<strong>in</strong> via the p38MAPK pathway <strong>in</strong>duces E2F1release and ATR k<strong>in</strong>ase activation mediat<strong>in</strong>g p53 apoptosis. J Biol Chem 2008;283:25455e67.[176] Jung JK, Arora P, Pagano JS, Jang KL. Expression of DNA methyltransferase 1 is activated by hepatitis B virus Xprote<strong>in</strong> via a regulatory circuit <strong>in</strong>volv<strong>in</strong>g the p16INK4a-cycl<strong>in</strong> D1-CDK 4/6-pRb-E2F1 pathway. Cancer Res2007;67:5771e8.[177] Zhu YZ, Zhu R, Shi LG, Mao Y, Zheng GJ, Chen Q, et al. Hepatitis B virus X prote<strong>in</strong> promotes hypermethylationof p16(INK4A) promoter through upregulation of DNA methyltransferases <strong>in</strong> hepatocarc<strong>in</strong>ogenesis.Exp Mol Pathol 2010;89:268e75.[178] Park IY, Sohn BH, Yu E, Suh DJ, Chung YH, Lee JH, et al. Aberrant epigenetic modifications <strong>in</strong> hepatocarc<strong>in</strong>ogenesis<strong>in</strong>duced by hepatitis B virus X prote<strong>in</strong>. Gastroenterology 2007;132:1476e94.[179] Ehrlich M. DNA hypomethylation and cancer. In: Ehrlich M, editor. DNA Alterations <strong>in</strong> Cancer. Westborough:Eaton Publish<strong>in</strong>g; 2000. p. 273e91.[180] Bayl<strong>in</strong> SB, Herman JG. <strong>Epigenetics</strong> and loss of gene function <strong>in</strong> cancer. In: Ehrlich M, editor. DNA Alterations<strong>in</strong> Cancer. Westborough: Eaton Publish<strong>in</strong>g; 2000. p. 293e309.[181] Nagai H, Baba M, Konishi N, Kim YS, Nogami M, Okumura K, et al. Isolation of NotI clusters hypomethylated<strong>in</strong> HBV-<strong>in</strong>tegrated hepatocellular carc<strong>in</strong>omas by two-dimensional electrophoresis. DNA Res1999;6:219e25.[182] Wei Y, Van Nhieu JT, Prigent S, Srivatanakul P, Tiollais P, et al. Altered expression of E-cadher<strong>in</strong> <strong>in</strong> hepatocellularcarc<strong>in</strong>oma: correlations with genetic alterations, beta-caten<strong>in</strong> expression, and cl<strong>in</strong>ical features.Hepatology 2002;36:692e701.439


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>440[183] Lee JO, Kwun HJ, Jung JK, Choi KH, M<strong>in</strong> DS, Jang KL. Hepatitis B virus X prote<strong>in</strong> represses E-cadher<strong>in</strong>expression via activation of DNA methyltransferase 1. Oncogene 2005;24:6617e25.[184] Liu J, Lian Z, Han S, Waye MM, Wang H, Wu MC, et al. Downregulation of E-cadher<strong>in</strong> by hepatitis B virus Xantigen <strong>in</strong> hepatocellullar carc<strong>in</strong>oma. Oncogene 2006;25:1008e17.[185] Zhong S, Yeo W, Tang MW, Wong N, Lai PB, Johnson PJ. Intensive hypermethylation of the CpG island of Rasassociation doma<strong>in</strong> family 1A <strong>in</strong> hepatitis B virus-associated hepatocellular carc<strong>in</strong>omas. Cl<strong>in</strong> Cancer Res2003;9:3376e82.[186] Zhong S, Tang MW, Yeo W, Liu C, Lo YM, Johnson PJ. Silenc<strong>in</strong>g of GSTP1 gene by CpG island DNAhypermethylation <strong>in</strong> HBV-associated hepatocellular carc<strong>in</strong>omas. Cl<strong>in</strong> Cancer Res 2002;8:1087e92.[187] Feng Q, Stern JE, Hawes SE, Lu H, Jiang M, Kiviat NB. DNA methylation changes <strong>in</strong> normal liver tissues andhepatocellular carc<strong>in</strong>oma with different viral <strong>in</strong>fection. Exp Mol Pathol 2010;88:287e92.[188] Ahmed R, Salama H, Fouad A, Sabry D, AbdAlah e, Kamal M. Detection of aberrant p16INK4A methylation<strong>in</strong> sera of patients with HCV-related liver diseases: An Egyptian study. Med Sci Monit 2010;16:CR410e5.[189] Formeister EJ, Tsuchiya M, Fujii H, Shpyleva S, Pogribny IP, Rusyn I. Comparative analysis of promotermethylation and gene expression endpo<strong>in</strong>ts between tumorous and non-tumorous tissues from HCV-positivepatients with hepatocellular carc<strong>in</strong>oma. Mutat Res 2010;692:26e33.[190] Lambert MP, Paliwal A, Vaissiere T, Chem<strong>in</strong> I, Zoulim F, Tommas<strong>in</strong>o M, et al. Aberrant DNA methylationdist<strong>in</strong>guishes hepatocellular carc<strong>in</strong>oma associated with HBV and HCV <strong>in</strong>fection and alcohol <strong>in</strong>take. J Hepatol2011;54:705e15.[191] Herceg Z, Paliwal A. Epigenetic mechanisms <strong>in</strong> hepatocellular carc<strong>in</strong>oma: how environmental factors<strong>in</strong>fluence the epigenome. Mutat Res 2011;727:55e61.[192] Yang B, Guo M, Herman JG, Clark DP. Aberrant promoter methylation profiles of tumor suppressor genes <strong>in</strong>hepatocellular carc<strong>in</strong>oma. Am J Pathol 2003;163:1101e7.[193] Archer KJ, Mas VR, Maluf DG, Fisher RA. High-throughput assessment of CpG site methylation for dist<strong>in</strong>guish<strong>in</strong>gbetween HCV-cirrhosis and HCV-associated hepatocellular carc<strong>in</strong>oma. Mol Genet Genomics2010;283:341e9.[194] Duong FH, Christen V, L<strong>in</strong> S, Heim MH. Hepatitis C virus-<strong>in</strong>duced up-regulation of prote<strong>in</strong> phosphatase 2A<strong>in</strong>hibits histone modification and DNA damage repair. Hepatology 2010;51:741e51.[195] Higgs MR, Lerat H, Pawlotsky JM. Downregulation of Gadd45beta expression by hepatitis C virus leads todefective cell cycle arrest. Cancer Res 2010;70:4901e11.[196] Lu CY, Hsieh SY, Lu YJ, Wu CS, Chen LC, Lo SJ, et al. Aberrant DNA methylation profile and frequentmethylation of KLK10 and OXGR1 genes <strong>in</strong> hepatocellular carc<strong>in</strong>oma. Genes Chromosomes Cancer2009;48:1057e68.[197] Deng YB, Nagae G, Midorikawa Y, Yagi K, Tsutsumi S, Yamamoto S, et al. Identification of genes preferentiallymethylated <strong>in</strong> hepatitis C virus-related hepatocellular carc<strong>in</strong>oma. Cancer Sci 2010;101:1501e10.[198] Miyoshi H, Fujie H, Sh<strong>in</strong>tani Y, Tsutsumi T, Sh<strong>in</strong>zawa S, Makuuchi M, et al. Hepatitis C virus core prote<strong>in</strong>exerts an <strong>in</strong>hibitory effect on suppressor of cytok<strong>in</strong>e signal<strong>in</strong>g (SOCS)-1 gene expression. J Hepatol2005;43:757e63.[199] Ko E, Kim SJ, Joh JW, Park CK, Park J, Kim DH. CpG island hypermethylation of SOCS-1 gene is <strong>in</strong>verselyassociated with HBV <strong>in</strong>fection <strong>in</strong> hepatocellular carc<strong>in</strong>oma. Cancer Lett 2008;271:240e50.[200] Arora P, Kim EO, Jung JK, Jang KL. Hepatitis C virus core prote<strong>in</strong> downregulates E-cadher<strong>in</strong> expression viaactivation of DNA methyltransferase 1 and 3b. Cancer Lett 2008;261:244e52.[201] Ripoli M, Barbano R, Balsamo T, Piccoli C, Brunetti V, Coco M, et al. Hypermethylated levels of E-cadher<strong>in</strong>promoter <strong>in</strong> Huh-7 cells express<strong>in</strong>g the HCV core prote<strong>in</strong>. Virus Res 2011;160:74e81.[202] Naka K, Abe K, Takemoto K, Dansako H, Ikeda M, Shimotohno K, et al. Epigenetic silenc<strong>in</strong>g of <strong>in</strong>terferon<strong>in</strong>duciblegenes is implicated <strong>in</strong> <strong>in</strong>terferon resistance of hepatitis C virus replicon-harbor<strong>in</strong>g cells. J Hepatol2006;44:869e78.[203] Magerl C, Ell<strong>in</strong>ger J, Braunschweig T, Kremmer E, Koch LK, Holler T, et al. H3K4 dimethylation <strong>in</strong> hepatocellularcarc<strong>in</strong>oma is rare compared with other hepatobiliary and gastro<strong>in</strong>test<strong>in</strong>al carc<strong>in</strong>omas and correlateswith expression of the methylase Ash2 and the demethylase LSD1. Hum Pathol 2010;41:181e9.[204] Guo N, Chen R, Li Z, Liu Y, Cheng D, Zhou Q, et al. Hepatitis C virus core upregulates the methylation statusof the RASSF1A promoter through regulation of SMYD3 <strong>in</strong> hilar cholangiocarc<strong>in</strong>oma cells. Acta BiochimBiophys S<strong>in</strong> (Shanghai) 2011;43:354e61.[205] Szalmas A, Konya J. Epigenetic alterations <strong>in</strong> cervical carc<strong>in</strong>ogenesis. Sem<strong>in</strong> Cancer Biol 2009;19:144e52.[206] Henken FE, Wilt<strong>in</strong>g SM, Overmeer RM, van Rietschoten JG, Nygren AO, et al. Sequential gene promotermethylation dur<strong>in</strong>g HPV-<strong>in</strong>duced cervical carc<strong>in</strong>ogenesis. Br J Cancer 2007;97:1457e64.[207] Burgers WA, Blanchon L, Pradhan S, de Launoit Y, Kouzarides T, Fuks F. Viral oncoprote<strong>in</strong>s target the DNAmethyltransferases. Oncogene 2007;26:1650e5.


CHAPTER 21<strong>Epigenetics</strong> and <strong>Human</strong> Infectious <strong>Disease</strong>s[208] Laurson J, Khan S, Chung R, Cross K, Raj K. Epigenetic repression of E-cadher<strong>in</strong> by human papillomavirus 16E7 prote<strong>in</strong>. Carc<strong>in</strong>ogenesis 2010;31:918e26.[209] Stephen JK, Chen KM, Shah V, Schweitzer VG, Gardner G, Benn<strong>in</strong>ger MS, et al. Consistent DNA hypermethylationpatterns <strong>in</strong> laryngeal papillomas. Int J Head Neck Surg 2010;1:69e77.[210] Holland D, Hoppe-Seyler K, Schuller B, Lohrey C, Maroldt J, Durst M, et al. Activation of the enhancer ofzeste homologue 2 gene by the human papillomavirus E7 oncoprote<strong>in</strong>. Cancer Res 2008;68:9964e72.[211] Zhang B, Laribee RN, Klemsz MJ, Roman A. <strong>Human</strong> papillomavirus type 16 E7 prote<strong>in</strong> <strong>in</strong>creases acetylationof histone H3 <strong>in</strong> human foresk<strong>in</strong> kerat<strong>in</strong>ocytes. Virology 2004;329:189e98.[212] McLaughl<strong>in</strong>-Drub<strong>in</strong> ME, Crum CP, Munger K. <strong>Human</strong> papillomavirus E7 oncoprote<strong>in</strong> <strong>in</strong>duces KDM6A andKDM6B histone demethylase expression and causes epigenetic reprogramm<strong>in</strong>g. Proc Natl Acad Sci USA2011;108:2130e5.[213] Feng H, Shuda M, Chang Y, Moore PS. Clonal <strong>in</strong>tegration of a polyomavirus <strong>in</strong> human Merkel cell carc<strong>in</strong>oma.<strong>Science</strong> 2008;319:1096e100.[214] Shuda M, Feng H, Kwun HJ, Rosen ST, Gjoerup O, Moore PS, et al. T antigen mutations are a human tumorspecificsignature for Merkel cell polyomavirus. Proc Natl Acad Sci USA 2008;105:16272e7.[215] Ben Arie N, Hassan BA, Berm<strong>in</strong>gham NA, Malicki DM, Armstrong D, Matzuk M, et al. Functional conservationof atonal and Math1 <strong>in</strong> the CNS and PNS. Development 2000;127:1039e48.[216] Bossuyt W, Kazanjian A, De Geest N, Van Kelst S, De Hertogh G, Geboes K, et al. Atonal homolog 1 is a tumorsuppressor gene. PLoS Biol 2009;7:e39.[217] Helmbold P, Lahtz C, Enk A, Herrmann-Trost P, Marsch WC, Kutzner H, et al. Frequent occurrence ofRASSF1A promoter hypermethylation and Merkel cell polyomavirus <strong>in</strong> Merkel cell carc<strong>in</strong>oma. Mol Carc<strong>in</strong>og2009;48:903e9.[218] Vatanasapt V, Uttaravichien T, Mairiang EO, Pairojkul C, Chartbanchachai W, Haswell-Elk<strong>in</strong>s M. Cholangiocarc<strong>in</strong>oma<strong>in</strong> north-east Thailand. Lancet 1990;335:116e7.[219] Sh<strong>in</strong> HR, Oh JK, Masuyer E, Curado MP, Bouvard V, Fang YY, et al. Epidemiology of cholangiocarc<strong>in</strong>oma: anupdate focus<strong>in</strong>g on risk factors. Cancer Sci 2010;101:579e85.[220] Ch<strong>in</strong>nasri P, Pairojkul C, Jearanaikoon P, Sripa B, Bhudhisawasdi V, Tantimavanich S, et al. Preferentiallydifferent mechanisms of <strong>in</strong>activation of 9p21 gene cluster <strong>in</strong> liver fluke-related cholangiocarc<strong>in</strong>oma. HumPathol 2009;40:817e26.[221] Sriraksa R, Zeller C, El Bahrawy MA, Dai W, Daduang J, et al. CpG-island methylation study of liver flukerelatedcholangiocarc<strong>in</strong>oma. Br J Cancer 2011;104:1313e8.[222] Fantappie MR, Gimba ER, Rumjanek FD. Lack of DNA methylation <strong>in</strong> Schistosoma mansoni. Exp Parasitol2001;98:162e6.[223] Gutierrez MI, Siraj AK, Khaled H, Koon N, El Rifai W, Bhatia K. CpG island methylation <strong>in</strong> Schistosoma- andnon-Schistosoma-associated bladder cancer. Mod Pathol 2004;17:1268e74.[224] Niller HH, Wolf H, M<strong>in</strong>arovits J. Regulation and dysregulation of Epste<strong>in</strong>eBarr virus latency: implications forthe development of autoimmune diseases. Autoimmunity 2008;41:298e328.[225] North ML, Ellis AK. The role of epigenetics <strong>in</strong> the developmental orig<strong>in</strong>s of allergic disease. Ann AllergyAsthma Immunol 2011;106:355e61.[226] Licciardi PV, Wong SS, Tang ML, Karagiannis TC. Epigenome target<strong>in</strong>g by probiotic metabolites. Gut Pathog2010;2:24.441


This page <strong>in</strong>tentionally left blank


CHAPTER 22The <strong>Epigenetics</strong> ofEndometriosisSun-Wei GuoFudan University Shanghai College of Medic<strong>in</strong>e, Shanghai, Ch<strong>in</strong>aCHAPTER OUTLINE22.1 Introduction 44322.1.1 Diagnosis andClassification 44422.1.2 Treatment 44422.1.3 Unmet Medical Needs 44522.2 Methods 44522.3 All Roads Lead to<strong>Epigenetics</strong> 44622.4 Evidence <strong>in</strong> Support thatEndometriosis is an Epigenetic<strong>Disease</strong> 44722.4.1 HOXA10Hypermethylation 44722.4.2 PR-B Hypermethylation 44722.4.3 Aberrent Expression of DNMT1,DNMT3A, and DNMT3B 44822.4.4 SF-1 and ERbHypomethylation 44822.4.5 Epigenetic Aberrations <strong>in</strong> OtherGenes 44822.5 Histone Modifications <strong>in</strong>Endometriosis: An UnexploredFrontier 44922.5.1 “Writers” and “Erasers” ofHistone Modifications 450HistoneAcetyltransferases 450HDACs 451HistoneMethyltransferases 451HistoneDemethylases 45222.5.2 “Reader/Effector”Modules 45322.6 Epigenetic Aberration: Cause orConsequence? 45422.7 Therapeutic Implications 45522.7.1 Possible Mechanisms of Action<strong>in</strong> HDACIs as aTherapeutics 45722.7.2 Potential Detrimental Effects ofEpigenetic Therapies andPossible Ways to CircumventThem 45822.8 Diagnostic and PrognosticImplications 46022.9 Conclusions and Future ResearchDirections 460Acknowledgment 461References 46144322.1 INTRODUCTIONEndometriosis, def<strong>in</strong>ed as the presence and growth of functional endometrial-like tissuesoutside the uter<strong>in</strong>e cavity, is a common and benign gynecological disorder with a poorlyunderstood and somewhat enigmatic etiopathogenesis and pathophysiology [1]. Itisa lead<strong>in</strong>g cause of disability <strong>in</strong> women of reproductive age, responsible for dysmenorrhea,pelvic pa<strong>in</strong>, and subfertility [2]. As such, it impacts negatively on patients’ physical, mental,relational, and social wellbe<strong>in</strong>g [3]. It also consumes tremendous healthcare resources. In theT. Tollefsbol (Ed): <strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>. DOI: 10.1016/B978-0-12-388415-2.00022-6Copyright Ó 2012 Elsevier Inc. All rights reserved.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>United States, endometriosis is the third lead<strong>in</strong>g cause of gynecologic hospitalization [4,5].In Ch<strong>in</strong>a, endometriosis-related surgeries constitute about one quarter of all gynecologicalsurgeries.The direct healthcare cost per patient <strong>in</strong> the US was estimated to range from $2801 to $12 644<strong>in</strong> the early 2000s [6e8]. In Italy, more than 65% of patients with endometriosis hadendometriosis-related surgical procedures, <strong>in</strong>clud<strong>in</strong>g hysterectomy, with<strong>in</strong> 1 year of firmdiagnosis [9]. In Belgium, the average non-healthcare costs associated with endometriosis<strong>in</strong>curred dur<strong>in</strong>g the 6 months prior to and follow<strong>in</strong>g surgical treatment are 1514 and 2496Euros, respectively [10]. In Canada, the estimated mean annual societal cost of endometriosiswas $5200 per patient (95% confidence <strong>in</strong>terval: $3700e7100), 78% of which is due to the lostproductivity and lost leisure time [11]. Thus, the economical burden of endometriosis tosociety, due to either healthcare costs or loss of productivity, is enormous because of its highprevalence, costly treatment, and debilitat<strong>in</strong>g nature [4,5,12]. Evidently, endometriosis posesa serious public health problem worldwide.Various theories on the pathogenesis of endometriosis have been proposed, but none has beenunequivocally proven [13]. These theories can be grouped <strong>in</strong>to roughly three themes: <strong>in</strong> situdevelopment (such as coelomic metaplasia or embryonic cell rests), implantation, ora comb<strong>in</strong>ation of <strong>in</strong> situ development and implantation. The implantation theory of Sampson[14], or the retrograde menstruation theory, is the most widely accepted. This theory stipulatesthat viable endometrial cells regurgitate through the fallopian tubes dur<strong>in</strong>g menstruation toimplant and grow <strong>in</strong> peritoneum or other ectopic sites. Indeed, retrograde menstruation isreported to occur <strong>in</strong> over 95% of women of reproductive age with patent fallopian tubes [15].However, far less women are actually <strong>in</strong>flicted with endometriosis. Thus, why there is suchdiscrepancy rema<strong>in</strong>s unresolved.44422.1.1 Diagnosis and ClassificationA def<strong>in</strong>ite diagnosis of endometriosis is based pr<strong>in</strong>cipally on direct visualization throughlaparoscopy and appropriate biopsies <strong>in</strong> conjunction with a thorough medical history,although ultrasound and magnetic resonance imag<strong>in</strong>g may also be useful [16]. Whiletremendous effects have been devoted to the search for non-<strong>in</strong>vasive diagnostic proceduressuch as serum biomarkers, so far no s<strong>in</strong>gle biomarker or group of biomarkers have beenproven to be unequivocally useful cl<strong>in</strong>ically [17].Endometriosis is staged by the classification system of the revised American Fertility Society(rAFS) [18]. Yet the stag<strong>in</strong>g system does not correlate well with either the severity of pa<strong>in</strong> or theextent of <strong>in</strong>fertility, nor does it correlate well with the prognosis [19]. Therefore, the developmentof a better classification system is currently an active research area [20].It has been generally regarded that endometriosis has at least three different subtypes, i.e.ovarian endometriomas, peritoneal endometriosis, and adenomyotic nodules of the rectovag<strong>in</strong>alseptum [21]. This view has been supported by different gene expression patternsbetween ovarian and peritoneal endometriosis based on large-scale gene expression profil<strong>in</strong>gstudies [22].22.1.2 TreatmentIn treat<strong>in</strong>g women with endometriosis, the efficacy has been measured by means of assessmentof pa<strong>in</strong>s and/or pregnancy rate [23]. The current treatment modalities <strong>in</strong>clude medical,surgical, or a comb<strong>in</strong>ation of both, with surgery be<strong>in</strong>g the treatment of choice. However, therecurrence risk after surgery is high: 7e30% of patients reported recurrences 3 years afterlaparoscopic surgery [24]. The risk <strong>in</strong>creases to 40e50% 5 years after surgery [25,26]. S<strong>in</strong>cerepeated surgeries are positively associated with <strong>in</strong>creased morbidity and healthcare costs and,<strong>in</strong> endometriosis, with damage to ovarian reserve [27e31], the risk for reoperation poses


CHAPTER 22The <strong>Epigenetics</strong> of Endometriosisa serious challenge to the effective management of endometriosis. Therefore, non-surgicalmedical therapy, preferably with high safety and cost profiles, is sorely needed.Non-surgical medical therapy is also used as a first-l<strong>in</strong>e therapy for treat<strong>in</strong>g endometriosis, andmay be used <strong>in</strong> conjunction with those patients who undergo surgical therapy for pa<strong>in</strong>. Thecurrent medical treatment for endometriosis has so far focused on the hormonal alteration ofthe menstrual cycle to produce a pseudo-pregnancy, pseudo-menopause, or chronic anovulation,creat<strong>in</strong>g an acyclic, hypoestrogenic environment [23]. This is achieved either byblock<strong>in</strong>g ovarian estrogen production (GnRH agonists, GnRH-a), by <strong>in</strong>duc<strong>in</strong>g pseudopregnancy(progest<strong>in</strong>s), or by locally <strong>in</strong>hibit<strong>in</strong>g estrogenic stimulation of the ectopic endometrium(progest<strong>in</strong>s, androgenic progest<strong>in</strong>s) [16,23,32]. While all hormonal treatments aremore or less equally effective <strong>in</strong> reliev<strong>in</strong>g pa<strong>in</strong>s [33], the relief, however, appears to be relativelyshort term [34]. Given the lack of long-term efficacious medical therapy for endometriosisassociatedpelvic pa<strong>in</strong>s and for m<strong>in</strong>imiz<strong>in</strong>g recurrence risk, and the lack of efficacious medicaltherapy for endometriosis-associated subfertility, there is a clear and press<strong>in</strong>g need for novelmedical therapies with more tolerable side effects and cost profiles [35].22.1.3 Unmet Medical NeedsIn response to this unmet need, numerous encourag<strong>in</strong>g precl<strong>in</strong>ical studies of a vast array ofpotential therapeutics for endometriosis have been reported <strong>in</strong> the last two decades. A handfulof these have undergone phase II/III cl<strong>in</strong>ical trials. Unfortunately, most of these completedtrials were found to be unpublished [36]. For those trials that have been published, the efficacyturns out to be much less impressive than that found <strong>in</strong> precl<strong>in</strong>ical studies [35]. Thus, thereseems to be a bewilder<strong>in</strong>g lost <strong>in</strong> translation <strong>in</strong> the effort to turn discoveries <strong>in</strong> basic research <strong>in</strong>endometriosis <strong>in</strong>to better patient care. In fact, there is a palpable disappo<strong>in</strong>tment over the drugresearch and development (R&D) <strong>in</strong> endometriosis: Vercell<strong>in</strong>i and co-workers recently likenedthe process to the “wait<strong>in</strong>g for Godot” [37].All these unmet medical needs, i.e. a classification system that can better relate symptomologyand/or prognosis, the development of better, more efficacious therapeutics, better non-<strong>in</strong>vasivediagnostic procedures, and possible prevention, stem from the fact that our current understand<strong>in</strong>gof the molecular mechanisms underly<strong>in</strong>g endometriosis pathogenesis is woefully<strong>in</strong>adequate. In this paper, which is an updated version of the previous one [38], I shall reviewour current knowledge of the epigenetic aberrations <strong>in</strong> endometriosis, discuss their implicationsfor del<strong>in</strong>eat<strong>in</strong>g the molecular mechanisms, cl<strong>in</strong>ical diagnosis, therapeutics, and <strong>in</strong>tervention.Aside from genes with known aberrant methylation, the author shall restrict my focuson prote<strong>in</strong>s/enzymes known to be <strong>in</strong>volved <strong>in</strong> DNA methylation and histone modifications esort of “writers” and “erasers” of epigenetic codes or marks e <strong>in</strong> endometriosis. This author shallnot review, however, work on “reader/effector” of histone modifications s<strong>in</strong>ce, as of writ<strong>in</strong>g, nowork has been published <strong>in</strong> the field of endometriosis per se. For the same reason, this authorwill conf<strong>in</strong>e myself on DNA methylation, histone acetylation/deacetylation, histone methylation/demethylation,s<strong>in</strong>ce no work on other types of histone modifications has been published<strong>in</strong> this field as of now. For ease of exposition and also for coherence, the author will leave out thepart on microRNA and its response to steroid hormones <strong>in</strong> endometriosis, which could berightfully subsumed <strong>in</strong>to the realm of endometriosis epigenetics and has received a great deal ofattention recently. The <strong>in</strong>terested readers should consult, for example, Toloubeydokhti et al.[39]. At the end of this review, areas <strong>in</strong> need of future research will be exposed.44522.2 METHODSA systematic and comprehensive search of PUBMED was performed for all studies publishedup to September 30, 2011, us<strong>in</strong>g the follow<strong>in</strong>g search terms: “endometriosis”, “epigenetics”,“histone”, “methylation”, “histone acetylation”, “histone phosphorylation”, “histone ubiquitylation”,“histone sumoylation”, “post-translational modifications”, or a comb<strong>in</strong>ation of


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>these. The studies had to report epigenetic aberrations <strong>in</strong> endometriosis. The search waslimited to publications written <strong>in</strong> English. Evidence for or aga<strong>in</strong>st endometriosis epigeneticswas presented, and its therapeutical, diagnostic, and prognostic implications were discussed.22.3 ALL ROADS LEAD TO EPIGENETICSEndometriosis has been regarded as an ultimate hormonal disease, ow<strong>in</strong>g much to itsestrogen-dependency and aberrations <strong>in</strong> estrogen production and metabolism [40e42]. It alsohas been viewed as an immunological disease due to a myriad immunological aberrations <strong>in</strong>endometriosis [43,44]. In addition, it has been thought of a disease caused by exposure toenvironmental pollution and tox<strong>in</strong>s [45,46] although so far there are no solid human data[47]. F<strong>in</strong>ally, it has been regarded as a genetic disease [48,49], ostensibly due to its reportedfamilial aggregation. Yet even the reported familial aggregation, when exam<strong>in</strong>ed closely, maybe debatable [50]. Incidentally, beyond reported associations with various polymorphisms,there has been little headway made so far <strong>in</strong>to the identification of genetic variants thatpredispose women to endometriosis [50e52].Endometriosis is undoubtedly a hormonal disease and certa<strong>in</strong>ly entails an array of immunologicalaberrations. While so far there is no solid evidence l<strong>in</strong>k<strong>in</strong>g diox<strong>in</strong> exposure to endometriosis,it may still be plausible that diox<strong>in</strong> exposure, at the right time and dosage, might precipitatethe <strong>in</strong>itiation or progression of endometriosis through <strong>in</strong>teraction with estrogen receptors [53] orsuppress<strong>in</strong>g expression of progesterone receptors [54]. So what is the common denom<strong>in</strong>ator fora disease that is hormonal, immunological, and possibly environmental and genetic?446In the last decade, numerous large-scale gene expression profil<strong>in</strong>g studies have demonstrated,unequivocally, that many genes are deregulated <strong>in</strong> endometriosis [22,55e71]. It also has beenshown that a s<strong>in</strong>gle focus of endometriotic lesion orig<strong>in</strong>ates from a s<strong>in</strong>gle progenitor cell [72],form<strong>in</strong>g a cellular l<strong>in</strong>eage. Dur<strong>in</strong>g their development from s<strong>in</strong>gle progenitor cells to endometrioticlesions lead<strong>in</strong>g to various symptoms, endometriotic cells presumably need to makea series of sequential, perhaps dichotomous, and irrevocable cell fate choices. These choices arelikely to be made without any change <strong>in</strong> DNA sequences. This cellular l<strong>in</strong>eage, or identity,<strong>in</strong>evitably requires that cells transcribe, or enable transcription of, specific sets of genes while atthe same time repress<strong>in</strong>g others. To ma<strong>in</strong>ta<strong>in</strong> cellular identity, the gene expression programmust be iterated through cell divisions <strong>in</strong> a heritable fashion by epigenetic processes.Indeed, transcription is regulated, <strong>in</strong> part, by the assembly of a plethora of complexes oftranscription factors on regulatory regions of genes, and can be regulated at various levels:DNA modifications (both chemical and structural), post-transcriptional modifications, andpost-translational modifications. These <strong>in</strong>volve chemical modification of DNA (methylation),histone modification, and various mach<strong>in</strong>eries, such as specific factors, repressors, activators,general transcriptional factors, enhancers, microRNAs (miRNAs) [73,74], and recentlydiscovered, double-stranded, non-cod<strong>in</strong>g RNAs (ncRNAs) [75]. These levels are either part ofthe epigenetic regulation (DNA methylation, histone modifications, miRNA) or closelyrelated. After the DNA is transcribed and mRNA formed, there are extra levels of regulation onhow much the mRNA is translated <strong>in</strong>to prote<strong>in</strong>s. Post-translational modifications of prote<strong>in</strong>products, localization and higher-order <strong>in</strong>teractions with other transcription factors, coactivatorsor corepressors are one set of mechanisms through which transcription can becontrolled at another level.In light of these, epigenetics is very likely to be <strong>in</strong>volved <strong>in</strong> ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g cellular identity <strong>in</strong> ectopicendometrial cells. This is the view that was first expressed <strong>in</strong> 2005 by Wu et al. that “endometriosis,like neoplasia, may also be an epigenetic disease ” [76], after realiz<strong>in</strong>g that epigenetic aberration,as a more general biological phenomenon and possibly one major mechanism for gene deregulation,should not be exclusively restricted <strong>in</strong> cancers or developmental diseases.


CHAPTER 22The <strong>Epigenetics</strong> of Endometriosis22.4 EVIDENCE IN SUPPORT THAT ENDOMETRIOSIS IS ANEPIGENETIC DISEASEEvidence that supports the theory that endometriosis is an epigenetic disease is discussedbelow.22.4.1 HOXA10 HypermethylationThe very first piece of evidence suggest<strong>in</strong>g that endometriosis may be an epigenetic diseasecame from a study show<strong>in</strong>g that the putative promoter of HOXA10 <strong>in</strong> endometrium fromwomen with endometriosis is hypermethylated as compared with that from women withoutendometriosis [76]. HOXA10 is a member of a family of homeobox genes that serve as transcriptionfactors dur<strong>in</strong>g development and has been shown to be important for uter<strong>in</strong>e function.It is expressed <strong>in</strong> human endometrium, and its expression is dramatically <strong>in</strong>creaseddur<strong>in</strong>g the midsecretory phase of the menstrual cycle, correspond<strong>in</strong>g to the time of implantationand <strong>in</strong>crease <strong>in</strong> circulat<strong>in</strong>g progesterone [77]. This suggests that HOXA10 may have animportant function <strong>in</strong> regulat<strong>in</strong>g endometrial development dur<strong>in</strong>g the menstrual cycle and <strong>in</strong>establish<strong>in</strong>g conditions necessary for implantation [78].In endometrium of women with endometriosis, however, HOXA10 gene expression issignificantly reduced, <strong>in</strong>dicat<strong>in</strong>g some defects <strong>in</strong> uter<strong>in</strong>e receptivity [79,80], whichmayberesponsible for reduced fertility <strong>in</strong> women with endometriosis. As promoter hypermethylationis generally associated with gene silenc<strong>in</strong>g, the observed HOXA10 promoterhypermethylation provides a plausible explanation as to why HOXA10 gene expression isreduced <strong>in</strong> endometrium of women with endometriosis [76]. In <strong>in</strong>fertile women withm<strong>in</strong>imal endometriosis, it was recently reported that HOXA10 expression <strong>in</strong> eutopic endometriumis reduced, and bisulfite sequenc<strong>in</strong>g confirmed that the HOXA10 promoter ishypermethylated [81].The HOXA10 promoter hypermethylation also has been demonstrated <strong>in</strong> a baboon model ofendometriosis, which co<strong>in</strong>cides with reduced HOXA10 expression [82]. What is <strong>in</strong>terest<strong>in</strong>g <strong>in</strong>the baboon study is the time course of HOXA10 expression levels, which was reducedprogressively after <strong>in</strong>duction of endometriosis but only became significant 1 year after the<strong>in</strong>duction [82]. In mouse, surgical <strong>in</strong>duction of endometriosis also resulted <strong>in</strong> the downregulationof Hoxa10 as well as hypermethylation [83]. Besides serv<strong>in</strong>g as a validation of thehuman observation, these two experimental studies also challenge the view that endometriosismay orig<strong>in</strong>ate from eutopic endometrium that harbor certa<strong>in</strong>, yet to be identified, molecularaberrations through retrograde menstruation. What is puzzl<strong>in</strong>g and rema<strong>in</strong>s unanswered isjust how endometriotic lesions situated <strong>in</strong> the peritoneal cavity apparently result <strong>in</strong> moleculargenetic changes <strong>in</strong> eutopic endometrium.447Hoxa10 hypermethylation, accompanied by overexpression of Dnmt1 and Dnmt3b, also hasbeen reported recently <strong>in</strong> female mice prenatally exposed to diethylstilbestrol (DES) [84].Interest<strong>in</strong>gly, prenatal exposure to DES also results <strong>in</strong> Hoxa10 down-regulation <strong>in</strong> gubernaculumand <strong>in</strong>hibition of transabdom<strong>in</strong>al testicular descent <strong>in</strong> male rat fetuses [85]. Thisaberrant methylation seems to be a novel mechanism of altered developmental programm<strong>in</strong>g<strong>in</strong>duced by <strong>in</strong> utero DES exposure.22.4.2 PR-B HypermethylationThe second piece of evidence came from the study demonstrat<strong>in</strong>g that the promoter of PR-Bis hypermethylated <strong>in</strong> endometriosis [86]. In addition, the PR-B promoter hypermethylationis concomitant with reduced PR-B gene expression, provid<strong>in</strong>g support for therole of epigenetic aberration <strong>in</strong> PR-B down-regulation. It is well-known that there isa general tendency of progesterone resistance <strong>in</strong> endometriosis [1]. It is also known that PR-B is down-regulated <strong>in</strong> endometriosis [87] and may be responsible for, at least <strong>in</strong> part,


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>progesterone resistance s<strong>in</strong>ce progesterone is mediated through its receptors, <strong>in</strong>clud<strong>in</strong>g PR-B. Yet why there is a persistent PR-B down-regulation was a mystery. PR-B promoterhypermethylation provides a simple yet biologically plausible explanation as to why PR-B ispersistently down-regulated <strong>in</strong> endometriosis. In addition, promoter hypermethylation asa cause for PR-B down-regulation appears to satisfy the Occam’s razor pr<strong>in</strong>ciple, that is, theexplanation of any phenomenon should make as few assumptions as possible, elim<strong>in</strong>at<strong>in</strong>gthose that make no difference <strong>in</strong> the observable predictions of the explanatory hypothesis ortheory.22.4.3 Aberrent Expression of DNMT1, DNMT3A, and DNMT3BPerhaps the most important piece of evidence show<strong>in</strong>g that endometriosis is an epigeneticdisease comes from a study demonstrat<strong>in</strong>g that DNMT1, DNMT3A, and DNMT3B, the threegenes cod<strong>in</strong>g for DNA methyltransferases that are <strong>in</strong>volved <strong>in</strong> genomic DNA methylation, areall overexpressed <strong>in</strong> endometriosis [88]. However, another study reports that DNMT1 andDNMT3B prote<strong>in</strong> expression <strong>in</strong> ectopic endometrium was significantly lower than that <strong>in</strong>control endometrium [89]. The discrepancy is likely due to the use of different materials: theformer study used endometriotic epithelial cells harvested through laser capture microdissectionwhile the latter used tissue culture, which consists of several, mixed cell types.Regardless, the two studies both suggest aberrant expression of DNMTs <strong>in</strong> endometriotictissues. S<strong>in</strong>ce these genes are <strong>in</strong>volved <strong>in</strong> de novo as well as ma<strong>in</strong>tenance methylation, theiraberrant expression suggests that aberrant methylation may be widespread <strong>in</strong> endometriosis.As methylation is closely l<strong>in</strong>ked with chromat<strong>in</strong> remodel<strong>in</strong>g, the aberrant expression of thesegenes may also signal that there are aberrant epigenetic changes, other than DNA methylation,<strong>in</strong> endometriosis.44822.4.4 SF-1 and ERb HypomethylationConsistent with the view that aberrant methylation may be widespread <strong>in</strong> endometriosis,several very recent studies provide further evidence for epigenetic changes <strong>in</strong> endometriosis.Steroidogenic factor-1 (SF-1), a transcriptional factor essential for activation of multiplesteroidogenic genes for estrogen biosynthesis, is usually undetectable <strong>in</strong> normal endometrialstromal cells but is aberrantly expressed <strong>in</strong> endometriotic stromal cells. Xue et al. show that SF-1 promoter has <strong>in</strong>creased methylation <strong>in</strong> endometrial cells yet <strong>in</strong> endometriotic cells it ishypomethylated [90]. They also f<strong>in</strong>d that ERb promoter is hypomethylated <strong>in</strong> endometrioticcells, which accounts for its overexpression [91].22.4.5 Epigenetic Aberrations <strong>in</strong> Other GenesIzawa et al. show that the treatment of endometrial stromal cells, which normally do notexpress aromatase, with a demethylation agent (DMA), 5-aza-deoxycytid<strong>in</strong>e, dramatically<strong>in</strong>creased the aromatase mRNA expression [92]. Further study by the same group found thata stretch of CpG demethylation with<strong>in</strong> a non-promoter CpG island of the aromatase gene <strong>in</strong>endometriotic cells while the same region is heavily methylated and associated with methyl-CpG-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s <strong>in</strong> endometrial cells [93]. Thus, the <strong>in</strong>creased expression of thearomatase gene <strong>in</strong> endometriosis is likely attributable to the epigenetic disorder associatedwith aberrant DNA hypomethylation <strong>in</strong> a non-promoter CpG island [93].Endometriotic cells are found to lack the <strong>in</strong>tercellular adhesion prote<strong>in</strong> E-cadher<strong>in</strong>, a knownmetastasis-suppressor prote<strong>in</strong> <strong>in</strong> epithelial tumor cells whose deregulation also seems to beassociated with <strong>in</strong>vasiveness of endometriotic cells [94,95]. In two immortalized endometrioticcell l<strong>in</strong>es, E-cadher<strong>in</strong> was found to be hypermethylated, and the treatment with trichostat<strong>in</strong>A (TSA) resulted <strong>in</strong> its reactivated expression with concomitant attenuated <strong>in</strong>vasion [96].This seems to suggest that, at least <strong>in</strong> endometriotic cell l<strong>in</strong>es, E-cadher<strong>in</strong> silenced by methylationis associated with <strong>in</strong>vasiveness.


CHAPTER 22The <strong>Epigenetics</strong> of EndometriosisOne <strong>in</strong> silico study based on large-scale gene expression profil<strong>in</strong>g of paired ectopic andeutopic endometrium also suggests a theme of post-translational modification and histonedeacetylation [97], aga<strong>in</strong> support<strong>in</strong>g the role of epigenetics <strong>in</strong> endometriosis. Consistent withthis view, a recent study based on whole-genome scann<strong>in</strong>g of methylation status <strong>in</strong> 25 500promoters compared endometriotic cells <strong>in</strong> three subtypes of endometriosis (superficialendometriosis or SUP, ovarian endometriomas or OMA, and deep <strong>in</strong>filtrat<strong>in</strong>g endometriosisor DIE) with their respective eutopic endometrium, and report that, with a pre-determ<strong>in</strong>edthreshold of 1.5 or 0.66, there are 153 (14), 29 (53), and 19 (20) hypermethylated (hypomethylated)promoters <strong>in</strong> SUP, OMA, and DIE, respectively, as compared with eutopicendometrium [98]. There are 11 hypermethylated and n<strong>in</strong>e hypomethylated chromosomalregions common to all three subtypes of endometriosis. Hypermethylated regions appear tobe located at the ends of chromosomes, while hypomethylated regions are found to berandomly distributed along the chromosomes [98]. While this high-thoughput technologycan identify many aberrant methylations <strong>in</strong> a s<strong>in</strong>gle study, caution should be made. First, notall aberrant methylations are associated with aberrant gene expression, as evidenced by thepoor correlation coefficient between the gene expression <strong>in</strong>duction ratio and methylationratio (r ¼ 0.03, p ¼ 0.90, based on data <strong>in</strong> Table 1 <strong>in</strong> [98]). In fact, among 20 transcriptionfactors for which expression data were also available, the agreement between expressionlevels (<strong>in</strong> direction and <strong>in</strong> terms of statistical significance) and methylation patterns is merely20% (or four out of 20). Second, while the use of paired eutopic and ectopic endometriumcan effectively m<strong>in</strong>imize the between-<strong>in</strong>dividual variation and reveal difference between thetwo tissues, the study design cannot detect aberrant methylations that are shared by the twotissues.Compared with the work on DNA methylation, there has been scanty report on aberranthistone modification <strong>in</strong> endometriosis. Kawano et al. recently reported decreased acetylatedhistone H3 and H4 <strong>in</strong> endometriotic stromal cells as compared with normal endometrialstromal cells [99]. In our lab, we found that immunoreactivity to lys<strong>in</strong>e-specific demethylase 1(LSD1, or KDM1A), which can demethylate mono- and di-methylated lys<strong>in</strong>es, specificallyhistone 3, lys<strong>in</strong>es 4 and 9 (H3K4 and H3K9), is elevated <strong>in</strong> endometriotic lesions as comparedwith normal endometrium [D<strong>in</strong>g et al., unpublished data]. In addition, we also found thatSIRT1, a class III histone deacetylase, and EZH2, a histone methyltransferase that methylatesH3K27, have <strong>in</strong>creased immunoreactivity <strong>in</strong> endometriotic lesions as compared with normalendometrium [D<strong>in</strong>g et al., unpublished data].449Table 22.1 provides a complete list of epigenetic aberrations <strong>in</strong> endometriosis identified so far.22.5 HISTONE MODIFICATIONS IN ENDOMETRIOSIS: ANUNEXPLORED FRONTIERHistones can undergo various k<strong>in</strong>ds of modifications which alter their <strong>in</strong>teraction with DNAand nuclear prote<strong>in</strong>s. The H3 and H4 histones, <strong>in</strong> particular, have long tails protrud<strong>in</strong>g fromthe nucleosome and can be covalently modified post-translationally at various residuals and <strong>in</strong>various ways. Histone modifications, primarily on the N-term<strong>in</strong>al tail, <strong>in</strong>clude acetylation,methylation, phosphorylation, ubiquit<strong>in</strong>ation, sumoylation, citrull<strong>in</strong>ation, and ADPribosylation.The core of the histones H2A and H3 can also be modified. Thus, histonemodifications can take place <strong>in</strong> different histones (e.g. H3 or H4), histone variants (e.g. H3.3),and histone residuals (e.g. arg<strong>in</strong>e, lys<strong>in</strong>e, and ser<strong>in</strong>e). The modification can <strong>in</strong>volve differentchemical groups (e.g. acetyl, methyl, and phosphate), and, for methylation, there can bedifferent degrees (e.g. mono-, di-, and tri-methylation for lys<strong>in</strong>es, mono-, symmetric andassymetric di-methylation for arg<strong>in</strong>es). These kaleidoscopic comb<strong>in</strong>ations of histone modifications<strong>in</strong>fluence the <strong>in</strong>teraction of histones with DNA and nuclear prote<strong>in</strong>s, and actconcertedly <strong>in</strong> gene regulation. Thus, the comb<strong>in</strong>ations of modifications are proposed toconstitute a code for gene expression, the so-called “histone code” [100,101].


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>TABLE 22.1 Epigenetic Aberrations <strong>in</strong> Endometriosis that have been Identified so far.The F<strong>in</strong>d<strong>in</strong>g Reported <strong>in</strong> [98] is not Included.450Yearof the First ReportGeneName Major F<strong>in</strong>d<strong>in</strong>g Reference2005 HOXA10 Hypermethylated <strong>in</strong> eutopicendometrium2006 PR-B Hypermethylated <strong>in</strong> ectopicendometrium2007 Aromatase Endometriotic cells secreted morearomatase than endometrial cells withadded testosterone, yet when treatedwith a demethylation agent, endometrialcells <strong>in</strong>creased the secretion2007 ERb Hypomethylated <strong>in</strong> ectopicendometrium2007 SF-1 Hypomethylated <strong>in</strong> ectopicendometrium2007 E-cadher<strong>in</strong> Methylated and <strong>in</strong>activated <strong>in</strong> anendometriotic epithelial-like cell l<strong>in</strong>e,and can be demethylated andreactivated by the treatment of TSA2007 DNMT1, Overexpressed <strong>in</strong> ectopic endometrioticDNMT3A, epithelial cellsDNMT3B2010 SRC-1 Reduced immunoreactivity <strong>in</strong>endometriotic epithelium2010 HDAC2 Elevated immunoreactivity <strong>in</strong> ectopicendometrium and dorsal root ganglia <strong>in</strong>rats with <strong>in</strong>duced endometriosis2011 Acetylated H3and H42011 DNMT1,DNMT3B,Decreased acetylated H3 and H4 <strong>in</strong>endometriotic stromal cells ascompared with normal endometrialstromal cellsReduced expression <strong>in</strong> ectopicendometrial tissuesMBD1, MBD22011 SRC-1 Elevated immunoreactivity <strong>in</strong> ovarianendometriosis[76,81,82,98][86][92, 93][91][90][96][88][103][106][99][89][104]The <strong>in</strong>vestigation of aberrant histone modifications <strong>in</strong> endometriosis has been scarce. For easeof exposition, we shall review published work, albeit few, <strong>in</strong> “writers” and “erasers” of histoneacetylation and methylation.22.5.1 “Writers” and “Erasers” of Histone ModificationsOne can view histone modifications of various types as some k<strong>in</strong>d of “mark<strong>in</strong>g”, and, as such,the enzymes that make the “mark<strong>in</strong>g” happen can be viewed as “writers”, while those mak<strong>in</strong>gthe “marks” removed as “erasers”.HISTONE ACETYLTRANSFERASESHistone acetyltransferases (HATs) are enzymes that acetylate conserved lys<strong>in</strong>e residuals onhistone prote<strong>in</strong>s by transferr<strong>in</strong>g an acetyl group from acetyl CoA to form ε-N-acetyl lys<strong>in</strong>e. Thismodification neutralizes the positive charge of lys<strong>in</strong>e and may thus disrupt the <strong>in</strong>teractionbetween DNA and histone tails. Acetylated histones are generally associated with euchromat<strong>in</strong>and transcriptional activation. In contrast to histone acetylation, deacetylation restricts DNA


CHAPTER 22The <strong>Epigenetics</strong> of Endometriosisaccessibility through reveal<strong>in</strong>g the positive charge of lys<strong>in</strong>e, permitt<strong>in</strong>g <strong>in</strong>teraction betweenDNA and the histone tail and thus chromat<strong>in</strong> compaction.There are two general categories of HATs: type A and type B. Type A HATs are nuclear andacetylate nucleosomal histones and other chromat<strong>in</strong>-associated prote<strong>in</strong>s, and type B HATs arelocated <strong>in</strong> the cytoplasm, acetylate newly synthesized histones, and have no direct impact ontranscription [102]. Type A HATs can be further categorized <strong>in</strong>to five families: nuclear receptorcoactivators and general transcription factors, p300/CBP (CREB (cAMP-response elementb<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>)-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>), GNAT (GCN5 (general control of nuclear-5)-relatedN-actyl transferases), MYST (MOZ (monocytic leukemia z<strong>in</strong>c-f<strong>in</strong>ger prote<strong>in</strong>), YBF2 (yeastb<strong>in</strong>d<strong>in</strong>g factor 2)/SAS3 (someth<strong>in</strong>g about silenc<strong>in</strong>g 3), and SAS2, TIP60 (Tat <strong>in</strong>teractiveprote<strong>in</strong>-60).In endometriosis, several HATs are <strong>in</strong>vestigated <strong>in</strong> the context of steroid receptor coactivators,s<strong>in</strong>ce many such coactivators are HATs. Suzuki et al. report that SRC-1 immunoreactivity isreduced [103]. Yet <strong>in</strong> ovarian endometriosis, Kumagami et al. report that SRC-1 colocalizeswith ERa and may thus affect the transcriptional activity of ERa [104].HDACSHDACs are a class of enzymes that remove acetyl groups from an ε-N-acetyl lys<strong>in</strong>e am<strong>in</strong>o acidon a histone, which are opposite to the action of HATs. Depend<strong>in</strong>g on sequence identity anddoma<strong>in</strong> organization, HDACs can be classified <strong>in</strong>to four groups: class I consists of HDAC1, 2,3, and 8; class II, HDAC4, 5, 7A, 9, and 10; class III or sirtu<strong>in</strong>s, consists of SIRT1-7; class IV hasone member, HDAC11. Classes I and II are considered “classical” HDACs whose activities canbe <strong>in</strong>hibited by trichostat<strong>in</strong> A (TSA).So far, no report on HDACs activity <strong>in</strong> endometriotic lesions has been published. One recentstudy, however, found that an epithelial-like endometriotic cell l<strong>in</strong>e expresses class I HDAC1,HDAC2, HDAC3, and HADC6, and class II HDAC4 and HDAC5 [105]. Given this result, and<strong>in</strong> view of many encourag<strong>in</strong>g results of HDAC <strong>in</strong>hibitors (HDACIs) on endometroitic cells(detailed below), it is very likely that some HDACs may be aberrantly overexpressed <strong>in</strong>endometriosis. In fact, our study on a rat model of endometriosis <strong>in</strong>dicates that HDAC2 isaberrantly expressed <strong>in</strong> ectopic endometrium [106]. We also found that SIRT1, a class IIIHDAC, has <strong>in</strong>creased immunoreactivity <strong>in</strong> endometriotic lesions as compared with normalendometrium [D<strong>in</strong>g et al., unpublished data]. We also found that class I HDAC1, HDAC2,and HDAC2 immunoreactivity is elevated <strong>in</strong> adenomyosis as compared with normalendometrium [212].451HISTONE METHYLTRANSFERASESHistone methylation occurs on lys<strong>in</strong>es (Ks) and arg<strong>in</strong><strong>in</strong>es (Rs). Hisone lys<strong>in</strong>es can be methylated<strong>in</strong> different forms: mono- (me1), di- (me2), or trimethylated (me3). Histone arg<strong>in</strong><strong>in</strong>emethylation can be monomethylated, symmetrically or asymmetrically dimethylated. Whilehistone methylation has been known s<strong>in</strong>ce the early 1960s, it was generally thought thathistone methylation, unlike acetylation and phorsphorylation, was biochemically stable andirreversible. Yet the identification of the first histone methyltransferases (HMT) <strong>in</strong> 2000 [107],and especially the identification of the first HDM <strong>in</strong> 2004 [108], challenged the notion thathistone methylation is a permanent, irreversible mark. With more HMTs and HDMs be<strong>in</strong>gdiscovered, it is now held that histone methylation can be dynamic [109]. A list of known sitespecificlys<strong>in</strong>e and arg<strong>in</strong><strong>in</strong>e HMTs is shown <strong>in</strong> Table 22.2.Depend<strong>in</strong>g on which residual to catalyze on, there are two types of HMTs: histone lys<strong>in</strong>e N-methyltransferase (KMT) and histone arg<strong>in</strong><strong>in</strong>e N-methyltransferase. HMTs catalyze the transferof one to three methyl groups from S-adenosyl methion<strong>in</strong>e (SAM) to lys<strong>in</strong>e and arg<strong>in</strong><strong>in</strong>e


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>TABLE 22.2 List of Known Site-Specific Lys<strong>in</strong>e and Arg<strong>in</strong><strong>in</strong>e HMTsAdapted from Table 1 <strong>in</strong> Nimura et al. [210] with modifications452Histone Residual Methylation Chromat<strong>in</strong> Status HMT NamesH3K4 me1 Active MLL, SET1A/B, ASH1L, ASH2L,me2 ActiveSmyd1, Smyd3, SET7/9me3 ActiveH3K9 me1 Active Suv39hl, Suv39h2, G9a,GLP,ESET,me2 InactiveRiz1me3 InactiveH3K27 me1 Active EZH2me2 Inactiveme3 InactiveH3K36 me1 Active NSD1, NSD2/WHSC1, Smyd2,me2 NoHYPB/SETD2, NSD3/WHSCI1me3 ActiveH3K79 me1 No DOT1Lme2 Nome3 InactiveH4K20 me1 Active PR-Set7, Suv4-20H1/2, NSD1,me2 UnknownNSD2/WHSC1me3 InactiveH3R2 me1 Unknown PRMT4/CARM1, PRMT6me2(as) Nome2(s) NoH4R3 me1 Unknown PRMT1, PRMT5me2(as) Unknownme2(s) NoH3R17H3R26UnknownPRMT4/CARM1As, asymmetrical; s, symmetrical; Active, correlation of actively transcribed region; Inactive, correlation of <strong>in</strong>actively transcribedregion; No, not significant correlation of transcription.residuals. With few exceptions, HMTs conta<strong>in</strong> a conserved SET (Su(var)3-9, Enhancer of Zeste,Trithorax) doma<strong>in</strong> that methylates specific residues of histone as well as non-histone prote<strong>in</strong>s.As of the time of writ<strong>in</strong>g, there has been no published study on aberrant expression of anyHMT <strong>in</strong> endometriosis. In our lab, we found that immunoreactivity to EZH2, an HMT thatmethylates H3K27, is <strong>in</strong>creased <strong>in</strong> endometriotic lesions as compared with normal endometrium[D<strong>in</strong>g et al., unpublished data].HISTONE DEMETHYLASESSimilar to HDACs, histone demethylases (HDMs) are “eraser” enzymes that site-specificallyremove the methyl group(s) from histone lys<strong>in</strong>e residuals. Two classes of HDMs have beenidentified thus far. One is the KDM1 family HDMs that are flav<strong>in</strong>-aden<strong>in</strong>e d<strong>in</strong>ucleotide (FAD)-dependent am<strong>in</strong>e oxidases, which can act only on mono- and dimethylated lys<strong>in</strong>es. The other ischaracterized by the Jumonji C (JmjC) doma<strong>in</strong>. HDMs <strong>in</strong> the latter class are Fe(II) and2-oxoglutarate-dependent enzymes, and, depend<strong>in</strong>g on sequence homology and the overallarchitecture of associated motifs, can be further classified <strong>in</strong>to different subgroups (Table 22.3).The KDM1 family has two members, KDM1A (LSD1) and KDM1B (LSD2), while the JmjCdoma<strong>in</strong>-conta<strong>in</strong><strong>in</strong>g KDMs have over 30 members identified so far, each with exclusive histoneandresidue-specific demthylat<strong>in</strong>g capabilities. Table 22.3 gives a list of all current KDM families<strong>in</strong> mammals.Histone acetylation, methylation, and phosphorylation are the most <strong>in</strong>vestigated histonemodifications. In general, histone acetylation is associated with transcriptional activation while


CHAPTER 22The <strong>Epigenetics</strong> of EndometriosisTABLE 22.3 The KDM Families <strong>in</strong> Mammals.The Family Names are Arranged Accord<strong>in</strong>g to the Phylogenetic Relationship of DNA Sequences Displayed <strong>in</strong> Figure 2 ofPedersen et al. [211]Family Members Synonyms Specificity RemarksKDM1 KDM1A LSD1/AOF2 H3K4me1/me2, H3K9me1/me2 First discovered KDMKDM1B LSD2/AOF1 H3K4me1/me2KDM2 KDM2A JHDM1A/FBXL11 H3K36me1/me2KDM2B JHDM1B/FBXL10 H3K36me1/me2, H3K4me3KDM3 KDM3A JHDM2A/JMJD1A H3K9me1/me2KDM3BKDM4 KDM4A JHDM3A/JMJD2AKDM4B JHDM3B/JMJD2B H3K36me2/me3KDM4C JHDM3C/JMJD2C H3K9me2/me3KDM4D JHDM3D/JMJD2DKDM5 KDM5A JARID1A/RBP2 H3K4me2/me3KDM5B JARID1B/PLU1KDM5C JARID1C/SMCXKDM5D JARID1D/SMCYKDM6 KDM6A UTX H3K27me2/me3KDM6B JMJD3HIF1ANHSPBAP1JMJD4JMJD5JMJD6 PSR/PTDSR H3R2, H4R3JMJD7JMJD8JHDM1D KIAA1718 H3K9me1/me2, H3K27me1/me2PHF2 JHDM1EPHF8 JHDM1F H3K9me1/me2HRPhylogenetically related with KDM3JMJD1CUTYPhylogenetically related with KDM6JARID2MINANO66H3K4me2/me3, H3K36me2/me3453deacetylation is associated with transcriptional repression [100,110]. In contrast, the effect ofhistone methylation depends on the histone residuals, their positions, and degrees [10,110].As at time of writ<strong>in</strong>g, there has been no published study on aberrant expression of any HDMs<strong>in</strong> endometriosis. In our lab, we found that immunoreactivity to LSD1 (KDM1A) thatdemethylates H3K4me1/me2 and H3K9me1/me2, is <strong>in</strong>creased <strong>in</strong> endometriotic lesions ascompared with normal endometrium [D<strong>in</strong>g et al., unpublished data].22.5.2 “Reader/Effector” ModulesHistone modifications are recognized by “reader/effector” modules, which read and <strong>in</strong>terpretmodification codes or marks and then execute conformational changes <strong>in</strong> chromat<strong>in</strong>s andprovide signals to regulate chromat<strong>in</strong> dynamics. These modules <strong>in</strong>clude Plant Homeo Doma<strong>in</strong>(PHD), chromo (for lys<strong>in</strong>e methylation), bromo (for lys<strong>in</strong>e acetylation), tudor, prol<strong>in</strong>etryptophan-tryptophan-prol<strong>in</strong>e(PWWP), SWI3p-Rsc8p-Moira (SWIRM), SWI3-ADA2-N-CoR-TFIIIB (SANT), and Malignant Bra<strong>in</strong> Tumor (MBT) doma<strong>in</strong>s. By recruit<strong>in</strong>g these reader/effector prote<strong>in</strong>s, histone modifications lead to changes <strong>in</strong> chromat<strong>in</strong> structure as well asdynamics [111]. As at time of writ<strong>in</strong>g, there has been no published account on aberration ofany “reader/effector” modules <strong>in</strong> histone modifications.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>22.6 EPIGENETIC ABERRATION: CAUSE OR CONSEQUENCE?Given the reported epigenetic aberrations <strong>in</strong> endometriosis, one question is whether theseaberrations are the cause or merely the consequence of endometriosis. S<strong>in</strong>ce most, if not all,human studies report<strong>in</strong>g epigenetic aberrations <strong>in</strong> endometriosis are carried out crosssectionally,the reported aberration may be a cause for, but also could be a consequence of,endometriosis. In a l<strong>in</strong>early causal relationship, the cause and consequence can be clearlydef<strong>in</strong>ed, with temporal sequences, and necessary and sufficient cause dist<strong>in</strong>guished. Ina complex system, such as endometriosis which appears to be a system-wide disease [112,113],<strong>in</strong> which there are usually many <strong>in</strong>terconnected parts, l<strong>in</strong>early causal relationship may be rare<strong>in</strong> the first place. In many ways, a complex transcription network often has a highly optimizedtolerance featur<strong>in</strong>g high efficiency, performance, and robustness to designed-for-uncerta<strong>in</strong>tiesyet hypersensitive to design flaws and unanticipated perturbations [114]. In such a system, thedemarcation of cause and consequence could be difficult s<strong>in</strong>ce the removal of one part mayaffect other parts of the system, especially when the system is redundant. Such complexsystems often display emergent properties. Therefore, it may be difficult to prove that <strong>in</strong>endometriosis aberrant methylation is a cause rather than a consequence.454Despite this challenge, it is known that methylation can be <strong>in</strong>duced by various factors. Ag<strong>in</strong>g[115e117], diet [118], chronic <strong>in</strong>flammation [119,120], prolonged transcriptional suppression[121,122], maternal care [123], and prolonged use of <strong>in</strong>trauter<strong>in</strong>e devices [124]. In endometriosis,it has been shown that prolonged stimulation of an endometriotic epithelial-likecell l<strong>in</strong>e by TNFa, which has been shown to have <strong>in</strong>creased production <strong>in</strong> endometriosis,resulted <strong>in</strong> at least partial methylation <strong>in</strong> the PR-B promoter [125]. This provides evidence thatcerta<strong>in</strong> phenotypic changes <strong>in</strong> endometriosis, such as <strong>in</strong>creased production of pro<strong>in</strong>flammatorycytok<strong>in</strong>es, may also cause epigenetic aberrations, which <strong>in</strong> turn result <strong>in</strong> changes<strong>in</strong> gene expression and subsequently other phenotypic changes such as <strong>in</strong>creased cellularproliferation [126] and perhaps some phenotypic changes.In a baboon model of endometriosis, it is reported that the <strong>in</strong>duction of endometriosisresulted <strong>in</strong> progressively decreased expression of HOXA10 and subsequent alteration <strong>in</strong> geneexpression of its downstream target genes <strong>in</strong> eutopic endometrium [82]. More remarkably, thedecreased HOXA10 expression is accompanied by the promoter hypermethylation [82]. Thisf<strong>in</strong>d<strong>in</strong>g has also been replicated <strong>in</strong> a mouse model of endometriosis [83]. In this case, theaberrant methylation at HOXA10 promoter is apparently a consequence of endometriosis.Remarkably, developmental exposures to chemicals can also result <strong>in</strong> aberrant methylation.Mice neonataly exposed to diethylstilbestrol (DES) are reported to have demethylation ofestrogen-responsive gene lactoferr<strong>in</strong> <strong>in</strong> their uteri, along with uter<strong>in</strong>e tumor [127,128].Neonatal exposure to DES can also lead to the hypomethylation nucleosomal b<strong>in</strong>d<strong>in</strong>gprote<strong>in</strong> 1 (Nsbp1) <strong>in</strong> mice [129]. In mice exposed to DES <strong>in</strong> utero, Hoxa10 hypermethylationhas been reported very recently [84]. Nutritional factors and stress have alsobeen reported to alter DNA methylation dur<strong>in</strong>g early life [123,130e133]. While it is stillunclear as to how much nutritional factors, stress, and exposure to certa<strong>in</strong> chemicals <strong>in</strong>early life and thus aberrant epigenetic changes that they may cause contribute to the risk ofendometriosis, it should be noted that the conceptof“fetalorig<strong>in</strong>sofadult-onsetdiseases”is fairly new, and the human research <strong>in</strong> this area can be quite challeng<strong>in</strong>g for obviousreasons. Nevertheless, the developmental orig<strong>in</strong>s of many chronic diseases such as type 2diabetes have now been demonstrated epidemiologically [134]. Incidentally, Missmer et al.reported that <strong>in</strong> utero exposure to DES nearly doubles the risk of develop<strong>in</strong>g endometriosis<strong>in</strong> women while low birthweight <strong>in</strong>creases the risk by 30% [135]. Further research <strong>in</strong> thisarea is sorely needed, not just for the sake of understand<strong>in</strong>g of endometriosis pathogenesisbut also because proper nutritional <strong>in</strong>tervention may reverse the aberrant epigeneticchanges [136,137].


CHAPTER 22The <strong>Epigenetics</strong> of Endometriosis22.7 THERAPEUTIC IMPLICATIONSUnlike DNA mutations or copy number changes, DNA methylation, histone and prote<strong>in</strong>modifications are reversible. Hence, enzymes that regulate the epigenetic changes could beideal targets for <strong>in</strong>tervention by pharmacological means. Given the accumulat<strong>in</strong>g evidence thatendometriosis may be an epigenetic disease, naturally one may wonder as to whether endometriosiscan be treated by correct<strong>in</strong>g epigenetic aberrations through pharmacological means.Indeed, encourag<strong>in</strong>g <strong>in</strong> vitro and <strong>in</strong> vivo results on the use of HDAC <strong>in</strong>hibitors (HDACIs) asa potential therapeutics for endometriosis have been reported.Treatment of an endometrial stromal cell l<strong>in</strong>e with trichostat<strong>in</strong> A (TSA) resulted <strong>in</strong> decreasedproliferation [138]. Treatment with TSA or valproic acid (VPA) resulted <strong>in</strong> cell cycle arrest and<strong>in</strong>duction of p21, a cell-cycle related gene [139]. The effect is likely through, perhaps <strong>in</strong> part, theup-regulation of PR-B by TSA [138], possibly through <strong>in</strong>creased acetylation of histones <strong>in</strong> chromat<strong>in</strong>s.These results have been replicated recently by Kawano et al. <strong>in</strong> primary endometrioticstromal cells us<strong>in</strong>g three different classes of HDACIs (VPA, suberoyl anilide bishydroxam<strong>in</strong>e orSAHA, and apicid<strong>in</strong>) [99]. Kawano et al. also show that treatment with HDACIs <strong>in</strong>ducedexpression of cell-cycle-related prote<strong>in</strong>s such as p21, p16, p27, and chk2 as well as apoptosisrelatedprote<strong>in</strong>s such as cleaved caspase 9 and Bcl-X L , and also elevated acetylation levels <strong>in</strong> thepromoter region of p21, p16, p27, and chk2 as well as acetylated H3 and H4 <strong>in</strong> endometrioticstromal cells [99]. What is remarkable is that, when it comes to <strong>in</strong>hibition of proliferation,endometriotic cells are more sensitive to treatment with HDACs than normal endometrialstromal cells [99,140], a fact that may be further exploited when consider<strong>in</strong>g dos<strong>in</strong>g.Treatment of TSA also <strong>in</strong>hibited IL-1b-<strong>in</strong>duced COX-2 expression [141]. This is significant,s<strong>in</strong>ce COX-2 overexpression has been observed <strong>in</strong> ectopic endometrium [142], found tocorrelate with endometriosis-associated pa<strong>in</strong> [143,144], and reported to be a biomarker forrecurrence [145]. TSA treatment up-regulated PPARg expression <strong>in</strong> endometrial stromal cells[146]. PPARg agonists have been reported to <strong>in</strong>hibit VEGF expression and angiogenesis <strong>in</strong>endometrial cells [147], <strong>in</strong>hibit TNF-<strong>in</strong>duced IL-8 production <strong>in</strong> endometriotic cells [148],and repress ectopic implants <strong>in</strong> animal models of endometriosis [149e151]. TSA treatmentcan also attenuate constitutive and TNFa-<strong>in</strong>duced NF-kB activation <strong>in</strong> endometriotic cells[140]. S<strong>in</strong>ce NF-kB plays pivotal roles <strong>in</strong> <strong>in</strong>flammation, proliferation, and angiogenesis [152]and is known to be constitutively activated <strong>in</strong> endometriosis [153,154], its attenuation by TSAstrongly suggests that HDACIs may be a promis<strong>in</strong>g therapeutic for endometriosis.455In two endometriotic cell l<strong>in</strong>es, TSA treatment resulted <strong>in</strong> attenuated <strong>in</strong>vasion and reactivatedE-cadher<strong>in</strong> expression [96]. This appears to suggest that some cellular phenotypes of endometrioticcells, such as <strong>in</strong>vasiveness, may be mediated epigenetically and, as such, could betamed by epigenetic reprogramm<strong>in</strong>g through pharmaceutical means.In an endometriotic epithelial-like cell l<strong>in</strong>e, another HDACI, romideps<strong>in</strong>, also known as FK-228 and depsipeptide that was orig<strong>in</strong>ally isolated from a broth culture of Chromobacteriumviolaceum, has been shown to reduce HDAC activity, <strong>in</strong>duce acetylation of H2A, H2B, H3, andH4, <strong>in</strong>hibit proliferation, and activate apoptosis through <strong>in</strong>duction of p21, caspase 3, caspase9, and PARP-1 as well as reduction of Cycl<strong>in</strong> B1 and Cycl<strong>in</strong> D1 [105]. Romideps<strong>in</strong> also <strong>in</strong>hibitsthe transcription, expression, and secretion of VEGF, a known and major factor <strong>in</strong>volved <strong>in</strong>angiogenesis <strong>in</strong> endometriosis [155].In a prelim<strong>in</strong>ary study, TSA has been found to <strong>in</strong>hibit the expression of SLIT2 [Zhao et al.,unpublished data], a member of the SLIT family of secretory glycoprote<strong>in</strong>s that was recently foundto attract vascular endothelial cells <strong>in</strong> vitro and promote tumor-<strong>in</strong>duced angiogenesis [156],and,more recently, found to be a constituent biomarker for recurrence of endometriosis [157].There are <strong>in</strong>dications that show HDACIs may be analgesic when treat<strong>in</strong>g endometriosis. Thefirst such <strong>in</strong>dication comes from the report that three HDACIs, TSA, suberic bishydroxamate,


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>and VPA, suppress spontaneous and oxytox<strong>in</strong>-<strong>in</strong>duced uter<strong>in</strong>e contractility [158]. It has beenshown that women with endometriosis have aberrant uter<strong>in</strong>e contractility dur<strong>in</strong>g menses with<strong>in</strong>creased frequency, amplitude, and basal pressure tone as compared with those without[159]. There is a sign that <strong>in</strong> the uterus of women with dysmenorrhea there is a lack ofsynchronization <strong>in</strong> fundal-cervical contraction [160]. Incidentally, progesterone, a traditionaldrug for treat<strong>in</strong>g endometriosis-associated dysmenorrhea, can also <strong>in</strong>hibit myometrialcontraction [161].Animal studies also show the protential of HDACIs <strong>in</strong> treat<strong>in</strong>g endometriosis. In mice withsurgically <strong>in</strong>duced endometriosis, treatment with TSA significantly reduced the averagesize of ectopic implants as compared with the controls [162]. Thisf<strong>in</strong>d<strong>in</strong>ghasbeenreplicated <strong>in</strong> rats treated with VPA [163]. More remarkably, it was found that <strong>in</strong>ducedendometriosis resulted <strong>in</strong> hyperalgesia or “central sensitization”, while TSA or VPA treatmentsignificantly improved mice’s or rats’ perception of pa<strong>in</strong> <strong>in</strong>duced by noxious stimuli[162,163]. It should be added that the improvement <strong>in</strong> pa<strong>in</strong> behavior <strong>in</strong> rats with <strong>in</strong>ducedendometriosis is endometriosis-specific, not due to the general analgesic property that VPAmay have [163].456Tak<strong>in</strong>g advantage of an exist<strong>in</strong>g drug, VPA, that is an HDACI with known pharmacology, andthe advantage that adenomyosis, once called endometriosis <strong>in</strong>terna, can be diagnosed quiteaccurately by non-<strong>in</strong>vasive imag<strong>in</strong>g techniques and that adenomyosis shares with endometriosismany similarities, Liu and Guo tested VPA on three patients as a new therapeutic andfound that it was well tolerated and, after 2 months of use, the pa<strong>in</strong> symptoms weredramatically reduced [164]. In addition, the uterus size was reduced by an average of onethird. Results from more patients show that VPA can effectively alleviate adenomyosisassociatedpa<strong>in</strong> and reduce uterus size [165]. These cl<strong>in</strong>ical observations corroborate wellwith the <strong>in</strong> vitro data that TSA treatment suppresses proliferation and cell cycle progression<strong>in</strong> ectopic endometrium <strong>in</strong> adenomyosis [166]. They are also consistent with the <strong>in</strong> vivo datathat VPA treatment results <strong>in</strong> reduction <strong>in</strong> myometrial <strong>in</strong>filtration, uter<strong>in</strong>e contractility, andcontractile irregularity [167], along with alleviation of adenomyosis-associated pa<strong>in</strong>[167,168].Chronic adm<strong>in</strong>istration of VPA has been shown to reduce bra<strong>in</strong> N-methyl-D-asparate(NMDA) signal<strong>in</strong>g <strong>in</strong> rats [169]. NMDA receptors (NMDARs), along with calciton<strong>in</strong> generelatedpeptide (CGRP), c-Fos, acid-sens<strong>in</strong>g ion channel 3 (ASIC-3), are known to beexpressed <strong>in</strong> sensory neurons <strong>in</strong> dorsal root ganglion (DRG) <strong>in</strong> the presence of centralsensitization [170e173]. In particular NMDARs and CGRP are known to be synaptic triggersof central sensitization [174]. NGF and its high-aff<strong>in</strong>ity receptor, TrkA, are mediators of<strong>in</strong>flammatory pa<strong>in</strong> [175]. NMDAR blockade and anti-NGF therapy have been shown to beeffective <strong>in</strong> reduc<strong>in</strong>g central sensitization [170,176]. In rats with <strong>in</strong>duced endometriosis,VPA treatment results <strong>in</strong> significantly decreased immunoreactivity of NMDAR1, c-Fos, ASIC3,TrkA, and CGRP <strong>in</strong> DRG, along with improved thermal latency [106], demonstrat<strong>in</strong>g thatVPA, and perhaps other HDACIs as well, may be efficacious <strong>in</strong> reduc<strong>in</strong>g central sensitization<strong>in</strong>duced by endometriosis and possibly <strong>in</strong> alleviat<strong>in</strong>g endometriosis-associated pa<strong>in</strong> <strong>in</strong>humans.On the surface, there may seem to be a mismatch between the reported aberrant methylation<strong>in</strong> endometriosis and the focus of current therapeutic approach, which has been so farconf<strong>in</strong>ed to HDACIs. Yet this mismatch is merely a trompe l’oeil, s<strong>in</strong>ce these exists a cross-talkbetween DNA methylation and histone modifications and they work <strong>in</strong> concert to controlgene expression [177,178], although it is unclear as to whether DNA methylation or histonemodification is the primary signal by which gene expression is determ<strong>in</strong>ed. Hence the change<strong>in</strong> histone modification may result <strong>in</strong> change <strong>in</strong> DNA methylation, and vice versa. The <strong>in</strong>hibitionof histone deacetylation can result <strong>in</strong> DNA demethylation, as evidenced by the demethylationof E-cadher<strong>in</strong> as a result of HDACI treatment [96].


CHAPTER 22The <strong>Epigenetics</strong> of Endometriosis22.7.1 Possible Mechanisms of Action <strong>in</strong> HDACIs as a TherapeuticsAs alluded to above, quite extensive <strong>in</strong> vitro and <strong>in</strong> vivo studies have shown that certa<strong>in</strong>HDACIs such as VPA and TSA can promote apoptosis, h<strong>in</strong>der cell cycle progression, <strong>in</strong>hibitproliferation, reduce <strong>in</strong>flammation and angiogenesis, and attenuate <strong>in</strong>vasiveness <strong>in</strong> endometriosis.In other cell types, VPA has been shown to reduce basal and FSH-stimulatedestrodiol secretion and FSH-<strong>in</strong>duced aromatase activity <strong>in</strong> human ovarian follicular cells [179]and <strong>in</strong> forskol<strong>in</strong>-stimulated H295R cells [180]. Thus, VPA may potentially <strong>in</strong>terfere withsteroidogenesis <strong>in</strong> endometriosis.TABLE 22.4 Summary of the Activities of HDACIs <strong>in</strong> Endometriotic Lesions and Other Cell/TissuesName of HDACIGene/Prote<strong>in</strong>Name Effect Cell/Tissue Type ReferenceTSA PR-B [ NESCL [138]TSA, VPA, SAHA, apicid<strong>in</strong> p21 [ NESCL, EESC, EECL [99,105,139]Romideps<strong>in</strong>VPA, SAHA, apicid<strong>in</strong> p16 [ EESC [99]VPA, SAHA, apicid<strong>in</strong> p27 [ EESC [99]VPA, SAHA, apicid<strong>in</strong> chk2 [ EESC [99]Romideps<strong>in</strong> Cycl<strong>in</strong> B1 Y EECL [105]Romideps<strong>in</strong> Cycl<strong>in</strong> D1 Y EECL [105]Romideps<strong>in</strong> Caspase 3 [ EECL [105]Romideps<strong>in</strong> Caspase 9 [ EECL [105]Romideps<strong>in</strong> PARP-1 [ EECL [105]VPA, SAHA, apicid<strong>in</strong> Bcl-XL [ EESC [99]TSA COX-2 Y NESCL [141]TSA, VPA NF-kB Y EECL, also <strong>in</strong> a rat model of[106,140]endometriosisTSA PPARg [ NESCL [146]TSA E-cadher<strong>in</strong> [ EECL, ESCL [96]Romideps<strong>in</strong> VEGF Y EECL [155]Romideps<strong>in</strong> HIF-1a Y EECL [105]VPA aromatase Y Forskol<strong>in</strong>-stimulated human adrenalcarc<strong>in</strong>oma cell l<strong>in</strong>e (H295R)VPA c-Fos Y DRG, <strong>in</strong> a rat model of endometriosis [106]VPA CGRP Y DRG, <strong>in</strong> a rat model of endometriosis [106]VPA TrkA Y DRG, <strong>in</strong> a rat model of endometriosis [106]VPA ASIC3 Y DRG, <strong>in</strong> a rat model of endometriosis [106]VPA HDAC2 Y DRG, <strong>in</strong> a rat model of endometriosis [106]VPA NMDAR1 Y DRG, <strong>in</strong> a rat model of endometriosis [106]TSA TRPV1 Y Eutopic endometrium, <strong>in</strong> a mouse [162]model of endometriosisTSA PKCε Y Ectopic endometrium, <strong>in</strong> a mouse [162]model of endometriosisTSA PGP9.5 Y Vag<strong>in</strong>a, <strong>in</strong> a mouse model of[162]endometriosisVPA HDAC2 Y Ectopic endometrium, <strong>in</strong> a rat model [106]of endometriosisVPA TrkA Y Ectopic endometrium, <strong>in</strong> a rat model [106]of endometriosisVPA CGRP Y Ectopic endometrium, <strong>in</strong> a rat model [106]of endometriosisVPA OTR Y Primary myometrial smooth musclecells[Guo et al.unpublished data]457NESCL, normal endometrial stromal cell l<strong>in</strong>e; EESC, endometriotic stromal cells; EECL, epithelial-like endometriotic cell l<strong>in</strong>e; ESCL, stromal-like endometriotic celll<strong>in</strong>e; DRG, dorsal root ganglia.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>FIGURE 22.1Schematic illustration of potential therapeutic effects of HDACIs <strong>in</strong> endometriosis. This figure is reproduced <strong>in</strong> the color plate section.458In human myometrial cells, it has been shown that long-term treatment resulted <strong>in</strong> repressionof NF-kB DNA b<strong>in</strong>d<strong>in</strong>g and <strong>in</strong>hibition of the expression of pro<strong>in</strong>flammatory genes such asCOX-2, IL-8, IL-6, and RANTES [181]. HDACIs have been reported to suppress TNFa-<strong>in</strong>ducedtissue factor expression [182] and also suppress the transcription, expression, and secretion ofvascular endothelial growth factor (VEGF) <strong>in</strong> endometriotic cells [155] and other cell types[183]. Both tissue factor and VEGF (and its receptors) are known to be key players <strong>in</strong>volved <strong>in</strong>angiogenesis <strong>in</strong> endometriosis [184e186]. Our unpublished data also <strong>in</strong>dicate that VPA can<strong>in</strong>hibit the expression of oxytoc<strong>in</strong> receptor (OTR) <strong>in</strong> primary myometrial smooth muscle cells[Guo et al., unpublished data]. The OTR expression has been found to be positively correlatedwith the amplitude of uter<strong>in</strong>e contractility and also with the the severity of dysmenorrhea <strong>in</strong>women with adenomyosis [Guo et al., unpublished data].Table 22.4 summarizes various effects of HDACIs on expression of some important genes thatare known to be <strong>in</strong>volved <strong>in</strong> endometriosis. Figure 22.1 depicts possible mechanisms of actionas to how HDACIs such as VPA can be therapeutically valuable.There is an <strong>in</strong>dication that HDACIs appears to synergize with demethylation agents (DMAs),result<strong>in</strong>g <strong>in</strong> more potent antiproliferative effects than either used alone and more robust reexpressionof methylation-silenced genes [166], as <strong>in</strong> cancer cells [187]. Clearly, futureresearch should illum<strong>in</strong>ate this further.22.7.2 Potential Detrimental Effects of Epigenetic Therapies andPossible Ways to Circumvent ThemS<strong>in</strong>ce global hypomethylation is a notable feature of cancer and is reported to cause genomic<strong>in</strong>stability [188,189], there may be a legitimate concern as whether the use of demethylationagents and/or HDACIs <strong>in</strong> treat<strong>in</strong>g endometriosis would <strong>in</strong>crease the risk for cancer. After all,endometriosis is not a fatal disease even if left untreated, hence the demand for better safetyand side effect profiles is higher than anticancer drugs [35].


CHAPTER 22The <strong>Epigenetics</strong> of EndometriosisSeveral studies have shown that only a small percentage (0.2e3%) of silenced genes are upregulatedby DMA treatment <strong>in</strong> cancer cells [190,191] and <strong>in</strong> normal fibroblast cell l<strong>in</strong>es thenumber of genes affected is even lower (0.4%) [192]. Similarly, HDACIs also up-regulatea small subset of genes (0.4e2%) and are quite specific <strong>in</strong> their activation and repression ofdist<strong>in</strong>ct genes [191,193]. While the percentage of affected genes is generally small, it is stillpossible that these affected genes may be important enough <strong>in</strong> caus<strong>in</strong>g unacceptable sideeffects, even though such data are lack<strong>in</strong>g as of now. In addition, it has been shown that thewithdrawal of methyltransferase <strong>in</strong>hibitors (such as DMAs) is followed by a rapid return ofmethylation [194], suggest<strong>in</strong>g that achiev<strong>in</strong>g long-last<strong>in</strong>g epigenetic reprogramm<strong>in</strong>g mayrequire cont<strong>in</strong>ued drug treatment.Even with these concerns, it should be noted that two DMAs, 5-azacitid<strong>in</strong>e and 5-aza-2 0 -deoxycitid<strong>in</strong>e (decitab<strong>in</strong>e), appear to be well-tolerated, and no significant demethylation ofrepetitive elements or any <strong>in</strong>dication of secondary malignancies was found [195]. In fact,chromosomal abnormalities were even found to be reversed <strong>in</strong> 31% of patients with myelodysplasticsyndrome (MDS) who took decitab<strong>in</strong>e [196]. Therefore, the two drugs have nowbeen approved <strong>in</strong> the US for treat<strong>in</strong>g MDS and the only agents known to improve the naturalhistory of MDS [197].There is also a concern that VPA use may <strong>in</strong>crease the risk of polycystic ovarian syndrome(PCOS) [198]. Yet from the very same study based on which the concern was raised, theauthors actually stated explicitly that “[n]one of the tested AEDs <strong>in</strong>fluenced 3bHSDII orP450c17 activities at concentrations normally used <strong>in</strong> AED therapy” [199] (AEDs stands forantiepileptic drugs, <strong>in</strong>clud<strong>in</strong>g VPA; author’s italics). As Paracelsus, considered to be the fatherof modern toxicology, said, Sola dosis facit venenum (only dose makes the poison). Hence theextrapolation of the observation under the high dosage to the situation of low dosage shouldbe made with extreme caution.Just as the dose, the duration of medication, perhaps to a lesser degree, can also makea difference. The two studies based on which the concern was raised actually exam<strong>in</strong>ed womentak<strong>in</strong>g VPA for a period of time much longer than 3 months as we used. The women <strong>in</strong> onestudy [200] had taken VPA for 2 years while <strong>in</strong> the other [201] the average duration of tak<strong>in</strong>gVPA was 28 months. Even if VPA is proved to have an unfavorable risk to benefit ratio, it is stillpremature to throw the baby out with the bathwater, s<strong>in</strong>ce VPA is just one of many HDACIsand some novel HDACIs may still hold the promise of be<strong>in</strong>g more efficacious while hav<strong>in</strong>g lessside effects.Besides dose and treatment duration, one promis<strong>in</strong>g way to improve drug safety and tom<strong>in</strong>imize side effects is to use the local route for adm<strong>in</strong>istration, namely the drug-conta<strong>in</strong><strong>in</strong>g<strong>in</strong>trauter<strong>in</strong>e system (IUS). There is an <strong>in</strong>dication that levonorgestrel-releas<strong>in</strong>g IUS (LNG-IUS)appears to be efficacious <strong>in</strong> treat<strong>in</strong>g endometriosis, adenomyosis, and their associated pa<strong>in</strong>[102], but little work has been done <strong>in</strong> this area.Regardless, the long-term safety of HDACIs and/or DMAs, when used to treat endometriosis,should be carefully evaluated even when they prove to be efficacious. Fortunately, such datamay come from cancer cl<strong>in</strong>ical trials.One f<strong>in</strong>al po<strong>in</strong>t is that many natural products conta<strong>in</strong> weak HDACI activities. In particular,royal jelly, the material responsible for the mak<strong>in</strong>g of honey bee queens, has weak HDACIactivities [203], and may be responsible for the epigenetic changes lead<strong>in</strong>g to strik<strong>in</strong>glydifferent phenotypes <strong>in</strong> the queen as compared with her workers who both share identicalgenomes [204]. The divergent developmental pathways of the queen and workers are associatedwith changes <strong>in</strong> subtle gene expression patterns <strong>in</strong> a particular group of genes encod<strong>in</strong>gconserved physiometabolic prote<strong>in</strong>s [204,205]. Thus, reduced methylation, or the change ofepigenome, <strong>in</strong> young larvae can mimic the effects of royal jelly. This example demonstrates thatHDACI activity, at least <strong>in</strong> weak form, does not necessarily cause detrimental health effect.459


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>22.8 DIAGNOSTIC AND PROGNOSTIC IMPLICATIONSBesides provid<strong>in</strong>g novel targets for drug therapy, epigenetic aberrations, once identified, mayalso provide promis<strong>in</strong>g prospects for diagnostic and/or prognostic purposes. One attractiveapproach is the identification of DNA methylation markers, which can be used for manyspecimens, such as menstrual blood.Any biomarker, <strong>in</strong> order to be cl<strong>in</strong>ically useful, should ideally have high specificity and sensitivity.In addition, it should be easily detectable <strong>in</strong> specimens procured through a m<strong>in</strong>imally<strong>in</strong>vasive manner. DNA methylation biomarkers appear to fit the latter requirement quite well.S<strong>in</strong>ce menstrual blood conta<strong>in</strong>s the same DNA (and thus methylation status) as that fromendometrial cells, and s<strong>in</strong>ce the endometrium from women with endometriosis is somewhatdifferent from that of women without [113], menstrual blood could be a valuable, abundant,non-<strong>in</strong>vasive, and convenient source for detection of methylation changes, as reported [206].A recent prelim<strong>in</strong>ary study us<strong>in</strong>g menstrual blood provides the evidence that the frequency ofERb hypermethylation <strong>in</strong> women with endometriosis is significantly lower than that <strong>in</strong> womenwithout endometriosis [Shen et al. unpublished data]. This seems to echo the result by Xueet al. that ERb is hypomethylated <strong>in</strong> endometriosis [91].Of course, it is unclear as of now as to whether the DNA methylation markers based onmenstrual blood are of any use for early diagnosis of endometriosis. It is also unclear as towhether they would be of value for the differential diagnosis of endometriosis, which could bemore challeng<strong>in</strong>g. Much more work is warranted.460DNA methylation markers may also prove to be useful for prognostic purposes. Theprelim<strong>in</strong>ary results <strong>in</strong> our lab seem to suggest that PR-B promoter hypermethylation found <strong>in</strong>tissue samples harvested at the time of surgery may be a biomarker for recurrence [Shen et al.unpublished data], which is consistent with the published f<strong>in</strong>d<strong>in</strong>gs [86,207]. In any case, verylittle has been published <strong>in</strong> this area, even though it is an area that is likely to be cl<strong>in</strong>icallymost useful and could br<strong>in</strong>g tangible results to better patient care. The identification ofpatients with high risk of recurrence should accord for further <strong>in</strong>tervention. On the otherhand, patients with low risk of recurrence may be advised not to take any medication, whichoften have side effects.22.9 CONCLUSIONS AND FUTURE RESEARCH DIRECTIONSGrow<strong>in</strong>g evidence now suggests that endometriosis is an epigenetic disorder, <strong>in</strong> the sense thatepigenetics plays a def<strong>in</strong>ite role <strong>in</strong> the pathogenesis and pathophysiology of endometriosis.This is characterized, at least <strong>in</strong> part, by aberrant methylation, dysregulated microRNA, andvery recently by deregulation of some ill-behaved epigenetic “writers” and “erasors” <strong>in</strong> eutopicas well as ectopic endometrium. Quite extensive data also have shown that HDACIs have manydesirable effects and, as such, have great potential as a therapeutic for endometriosis. Inaddition, DNA methylation-based as well as miRNA-based biomarkers may hold potential <strong>in</strong>the diagnosis of endometriosis and predict<strong>in</strong>g recurrence risks after surgery.Despite these advances, however, our current knowledge on the epigenetics of endometriosis,and its pathophysiological significance is still <strong>in</strong> its <strong>in</strong>fancy. This can be seen from the vast gapbetween a myriad of enzymes/prote<strong>in</strong>s <strong>in</strong>volved <strong>in</strong> DNA methylation and histone modificationsand just a handful of them that just have been evaluated <strong>in</strong> endometriosis. Indeed, so farwe have merely scratched the surface of the epigenetics of endometriosis.While a complete understand<strong>in</strong>g of the epigenetics of endometriosis holds keys to a fullknowledge of how genes are dysregulated and coord<strong>in</strong>ated <strong>in</strong> the genesis and development ofendometriosis and the manifest of variable symptoms, such an enormous task is quite challeng<strong>in</strong>g,and somewhat daunt<strong>in</strong>g. This is because, first of all, epigenetics itself is evolv<strong>in</strong>g andrapidly develop<strong>in</strong>g field. Many issues, such as the existance of DNA demethylase(s) or not, are


CHAPTER 22The <strong>Epigenetics</strong> of Endometriosisstill unresolved. In addition, while aberrant DNA methylation <strong>in</strong> endometriosis has begun tobe <strong>in</strong>vestigated, few histone modifications have, if any, been <strong>in</strong>vestigated <strong>in</strong> endometriosis.There are 17 HDACs, over 50 HMTs [208], and possibly a similar number of HDMs, yet few ofthem have been evaluated <strong>in</strong> endometriosis. Besides DNMTs, methyl-CpG b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s,HDACs, HATs, HMT, HDMs, and microRNAs, there are E3 ubiquit<strong>in</strong> ligases, k<strong>in</strong>ases, smallubiquit<strong>in</strong>-related modifier (SUMO)-conjugated enzymes, and ADP-ribosyl transferases(ADPRTs) and other enzymes that may also play important roles <strong>in</strong> endometriosis epigenetics.Even for histone methylation, there is an added layer of complexity of hav<strong>in</strong>g three forms oflys<strong>in</strong>e methylation and two forms of arg<strong>in</strong><strong>in</strong>e methylation. These complexities are furthercompounded by nearly an astronomical number of comb<strong>in</strong>atorial assortment of varioushistone modifications, by crosstalks between and among different DNA and histone modifications,and by possible temporal and spatial dynamics of these modifications. Moreover, theepigenetic aberrations may exist not only <strong>in</strong> endometriotic cells, but also <strong>in</strong> other types of cells,such as endothelial cells and macrophages, that are with<strong>in</strong> or surround the endometrioticlesions which are also <strong>in</strong>timately <strong>in</strong>volved <strong>in</strong> the pathogenesis of endometriosis. As of the timeof writ<strong>in</strong>g, no work <strong>in</strong> this area has been published.Regardless, it is now clear that chromat<strong>in</strong>, once considered just a structural scaffold allow<strong>in</strong>gthe packag<strong>in</strong>g of DNA, is actually a dynamic and key regulatory element of gene expressionand actively participates <strong>in</strong> several cellular processes such as mitosis and differentiation [209].The epigenetics of endometriosis is a rapidly grow<strong>in</strong>g field, and may likely transform ourunderstand<strong>in</strong>g of the pathogenesis and pathophysiology of endometriosis, open<strong>in</strong>g newavenues for diagnosis, treatment, and prognostic prediction. So far we have only scratched itssurface. With more research, we may come closer to the full understand<strong>in</strong>g of the etiopathogenesisof endometriosis and will be <strong>in</strong> a better position to treat or perhaps prevent thisunrelent<strong>in</strong>gly pa<strong>in</strong>ful and dreadful disease that is endometriosis.AcknowledgmentThis work was supported by grant 30872759 from the National <strong>Science</strong> Foundation of Ch<strong>in</strong>a, grants 09PJD015 and10410700200 from the Shanghai <strong>Science</strong> and Technology Commission (SWG), and grant 09-11 from the State KeyLaboratory of Medical Neurobiology of Fudan University, and f<strong>in</strong>ancial support from Shanghai Key Laboratory ofFemale Reproductive Endocr<strong>in</strong>e-Related <strong>Disease</strong>s and from the Key Specialty Project of the M<strong>in</strong>istry of Health, Ch<strong>in</strong>a.461References[1] Giudice LC, Kao LC. Endometriosis. Lancet 2004;364:1789e99.[2] Farquhar CM. Extracts from the cl<strong>in</strong>ical evidence. Endometriosis. Bmj 2000;320:1449e52.[3] Fourquet J, Gao X, Zavala D, Orengo JC, Abac S, Ruiz A, Laboy J, Flores I. Patients’ report on how endometriosisaffects health, work, and daily life. Fertil Steril 93:2424e8.[4] Bol<strong>in</strong>g RO, Abbasi R, Ackerman G, Schipul Jr AH, Chaney SA. Disability from endometriosis <strong>in</strong> the UnitedStates Army. J Reprod Med 1988;33:49e52.[5] Kjerulff KH, Erickson BA, Langenberg PW. Chronic gynecological conditions reported by US women: f<strong>in</strong>d<strong>in</strong>gsfrom the National Health Interview Survey, 1984 to 1992. Am J Public Health 1996;86:195e9.[6] Simoens S, Hummelshoj L, D’Hooghe T. Endometriosis: cost estimates and methodological perspective. HumReprod Update 2007;13:395e404.[7] Zhao SZ, Wong JM, Davis MB, Gersh GE, Johnson KE. The cost of <strong>in</strong>patient endometriosis treatment: ananalysis based on the Healthcare Cost and Utilization Project Nationwide Inpatient Sample. Am J Manag Care1998;4:1127e34.[8] Gao X, Outley J, Botteman M, Spald<strong>in</strong>g J, Simon JA, Pashos CL. Economic burden of endometriosis. FertilSteril 2006;86:1561e72.[9] Fuldeore M, Chwalisz K, Marx S, Wu N, Boulanger L, Ma L, Lamothe K. Surgical procedures and their costestimates among women with newly diagnosed endometriosis: a US database study. J Med Econ14:115e23.[10] Simoens S, Meuleman C, D’Hooghe T. Non-health-care costs associated with endometriosis. Hum Reprod26:2363e7.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>462[11] Levy AR, Osenenko KM, Lozano-Ortega G, Sambrook R, Jeddi M, Belisle S, Reid RL. Economic burden ofsurgically confirmed endometriosis <strong>in</strong> Canada. J Obstet Gynaecol Can 33:830e7.[12] Luisi S, Lazzeri L, Ciani V, Petraglia F. Endometriosis <strong>in</strong> Italy: from cost estimates to new medical treatment.Gynecol Endocr<strong>in</strong>ol 2009;25:734e40.[13] V<strong>in</strong>atier D, Orazi G, Cosson M, Dufour P. Theories of endometriosis. Eur J Obstet Gynecol Reprod Biol2001;96:21e34.[14] Sampson J. Peritoneal endometriosis due to the menstrual dissem<strong>in</strong>ation of endometrial tissue <strong>in</strong>to theperitoneal cavity. Am J Obstet Gynecol 1927;14:422e69.[15] Halme J, Hammond MG, Hulka JF, Raj SG, Talbert LM. Retrograde menstruation <strong>in</strong> healthy women and <strong>in</strong>patients with endometriosis. Obstet Gynecol 1984;64:151e4.[16] Valle RF, Sciarra JJ. Endometriosis: treatment strategies. Ann N Y Acad Sci 2003;997:229e39.[17] May KE, Conduit-Hulbert SA, Villar J, Kirtley S, Kennedy SH, Becker CM. Peripheral biomarkers of endometriosis:a systematic review. Hum Reprod Update 16:651e74.[18] Revised American Society for Reproductive Medic<strong>in</strong>e classification of endometriosis: 1996. Fertil Steril1997;67:817e21.[19] Canis M, Bouquet De Jol<strong>in</strong>ieres J, Wattiez A, Pouly JL, Mage G, Manhes H, et al. Classification of endometriosis.Baillieres Cl<strong>in</strong> Obstet Gynaecol 1993;7:759e74.[20] Adamson GD. Endometriosis classification: an update. Curr Op<strong>in</strong> Obstet Gynecol 23:213e20.[21] Nisolle M, Donnez J. Peritoneal endometriosis, ovarian endometriosis, and adenomyotic nodules of therectovag<strong>in</strong>al septum are three different entities. Fertil Steril 1997;68:585e96.[22] Wu Y, Kajdacsy-Balla A, Strawn E, Basir Z, Halverson G, Jailwala P, et al. Transcriptional characterizations ofdifferences between eutopic and ectopic endometrium. Endocr<strong>in</strong>ology 2006;147:232e46.[23] Olive DL, Pritts EA. Treatment of endometriosis. N Engl J Med 2001;345:266e75.[24] Weedon MN, Lango H, L<strong>in</strong>dgren CM, Wallace C, Evans DM, Mang<strong>in</strong>o M, et al. Genome-wide associationanalysis identifies 20 loci that <strong>in</strong>fluence adult height. Nat Genet 2008;40:575e83.[25] Guo SW. Recurrence of endometriosis and its control. Hum Reprod Update 2009.[26] Wheeler JM, Mal<strong>in</strong>ak LR. Recurrent endometriosis: <strong>in</strong>cidence, management, and prognosis. Am J ObstetGynecol 1983;146:247e53.[27] Ragni G, Somigliana E, Benedetti F, Paffoni A, Vegetti W, Restelli L, et al. Damage to ovarian reserve associatedwith laparoscopic excision of endometriomas: a quantitative rather than a qualitative <strong>in</strong>jury. Am J ObstetGynecol 2005;193:1908e14.[28] Hachisuga T, Kawarabayashi T. Histopathological analysis of laparoscopically treated ovarian endometrioticcysts with special reference to loss of follicles. Hum Reprod 2002;17:432e5.[29] Somigliana E, Ragni G, Benedetti F, Borroni R, Vegetti W, Crosignani PG. Does laparoscopic excision ofendometriotic ovarian cysts significantly affect ovarian reserve? Insights from IVF cycles. Hum Reprod2003;18:2450e3.[30] Somigliana E, Ragni G, Infant<strong>in</strong>o M, Benedetti F, Arnoldi M, Crosignani PG. Does laparoscopic removal ofnonendometriotic benign ovarian cysts affect ovarian reserve? Acta Obstet Gynecol Scand 2006;85:74e7.[31] Candiani M, Barbieri M, Bottani B, Bertulessi C, Vignali M, Agnoli B, et al. Ovarian recovery after laparoscopicenucleation of ovarian cysts: <strong>in</strong>sights from echographic short-term postsurgical follow-up. J M<strong>in</strong>im InvasiveGynecol 2005;12:409e14.[32] Lessey BA. Medical management of endometriosis and <strong>in</strong>fertility. Fertil Steril 2000;73:1089e96.[33] Kennedy S, Bergqvist A, Chapron C, D’Hooghe T, Dunselman G, Greb R, et al. ESHRE guidel<strong>in</strong>e for thediagnosis and treatment of endometriosis. Hum Reprod 2005;20:2698e704.[34] Waller KG, Shaw RW. Gonadotrop<strong>in</strong>-releas<strong>in</strong>g hormone analogues for the treatment of endometriosis: longtermfollow-up. Fertil Steril 1993;59:511e5.[35] Guo SW. Emerg<strong>in</strong>g drugs for endometriosis. Expert Op<strong>in</strong> Emerg<strong>in</strong>g Drugs 2008;13:547e71.[36] Guo SW, Hummelshoj L, Olive DL, Bulun SE, D’Hooghe TM, Evers JL. A call for more transparency ofregistered cl<strong>in</strong>ical trials on endometriosis. Hum Reprod 2009;24:1247e54.[37] Vercell<strong>in</strong>i P, Crosignani P, Somigliana E, Vigano P, Frattaruolo MP, Fedele L. ’Wait<strong>in</strong>g for Godot’: a commonsenseapproach to the medical treatment of endometriosis. Hum Reprod 26:3e13.[38] Guo SW. <strong>Epigenetics</strong> of endometriosis. Mol Hum Reprod 2009;15:587e607.[39] Toloubeydokhti T, Pan Q, Luo X, Bukulmez O, Cheg<strong>in</strong>i N. The expression and ovarian steroid regulation ofendometrial micro-RNAs. Reprod Sci 2008;15:993e1001.[40] Bulun SE, Yang S, Fang Z, Gurates B, Tamura M, Sebastian S. Estrogen production and metabolism <strong>in</strong>endometriosis. Ann N Y Acad Sci 2002;955:75e85. discussion 86e8, 396e406.[41] Gurates B, Bulun SE. Endometriosis: the ultimate hormonal disease. Sem<strong>in</strong> Reprod Med 2003;21:125e34.


CHAPTER 22The <strong>Epigenetics</strong> of Endometriosis[42] Kitawaki J, Kado N, Ishihara H, Koshiba H, Kitaoka Y, Honjo H. Endometriosis: the pathophysiology as anestrogen-dependent disease. J Steroid Biochem Mol Biol 2002;83:149e55.[43] Paul Dmowski W, Braun DP. Immunology of endometriosis. Best Pract Res Cl<strong>in</strong> Obstet Gynaecol2004;18:245e63.[44] Ulukus M, Arici A. Immunology of endometriosis. M<strong>in</strong>erva G<strong>in</strong>ecol 2005;57:237e48.[45] Rier SE, Mart<strong>in</strong> DC, Bowman RE, Dmowski WP, Becker JL. Endometriosis <strong>in</strong> rhesus monkeys (Macaca mulatta)follow<strong>in</strong>g chronic exposure to 2,3,7,8-tetrachlorodibenzo-p-diox<strong>in</strong>. Fundam Appl Toxicol 1993;21:433e41.[46] Rier SE. The potential role of exposure to environmental toxicants <strong>in</strong> the pathophysiology of endometriosis.Ann N Y Acad Sci 2002;955:201e12. discussion 230-2, 396-406.[47] Guo SW. The L<strong>in</strong>k between Exposure to Diox<strong>in</strong> and Endometriosis: A Critical Reappraisal of Primate Data.Gynecol Obstet Invest 2004;57:157e73.[48] Simpson JL, Bischoff FZ, Kamat A, Buster JE, Carson SA. Genetics of endometriosis. Obstet Gynecol Cl<strong>in</strong> NorthAm 2003;30:21e40. vii.[49] Barlow DH, Kennedy S. Endometriosis: new genetic approaches and therapy. Annu Rev Med 2005;56:345e56.[50] Di W, Guo SW. The search for genetic variants predispos<strong>in</strong>g women to endometriosis. Curr Op<strong>in</strong> ObstetGynecol 2007;19:395e401.[51] Falconer H, D’Hooghe T, Fried G. Endometriosis and genetic polymorphisms. Obstet Gynecol Surv2007;62:616e28.[52] Guo SW. The relevance of genetics <strong>in</strong> endometriosis. In: Garcia-Velasco JA, Rizk B, editors. Endometriosis:Current management and future trends, Vol. aypee Medical Publishers; 2009. In press.[53] Ohtake F, Takeyama K, Matsumoto T, Kitagawa H, Yamamoto Y, Nohara K, et al. Modulation of oestrogenreceptor signall<strong>in</strong>g by association with the activated diox<strong>in</strong> receptor. Nature 2003;423:545e50.[54] Igarashi TM, Bruner-Tran KL, Yeaman GR, Lessey BA, Edwards DP, Eisenberg E, et al. Reduced expression ofprogesterone receptor-B <strong>in</strong> the endometrium of women with endometriosis and <strong>in</strong> cocultures of endometrialcells exposed to 2,3,7,8-tetrachlorodibenzo-p-diox<strong>in</strong>. Fertil Steril 2005;84:67e74.[55] Carson DD, Lagow E, Thathiah A, Al-Shami R, Farach-Carson MC, Vernon M, et al. Changes <strong>in</strong> gene expressiondur<strong>in</strong>g the early to mid-luteal (receptive phase) transition <strong>in</strong> human endometrium detected by high-densitymicroarray screen<strong>in</strong>g. Mol Hum Reprod 2002;8:871e9.[56] Arimoto T, Katagiri T, Oda K, Tsunoda T, Yasugi T, Osuga Y, et al. Genome-wide cDNA microarray analysis ofgene-expression profiles <strong>in</strong>volved <strong>in</strong> ovarian endometriosis. Int J Oncol 2003;22:551e60.[57] Kao LC, Germeyer A, Tulac S, Lobo S, Yang JP, Taylor RN, et al. Expression profil<strong>in</strong>g of endometrium fromwomen with endometriosis reveals candidate genes for disease-based implantation failure and <strong>in</strong>fertility.Endocr<strong>in</strong>ology 2003;144:2870e81.[58] Matsuzaki S, Canis M, Vaurs-Barriere C, Boespflug-Tanguy O, Dastugue B, Mage G. DNA microarray analysis ofgene expression <strong>in</strong> eutopic endometrium from patients with deep endometriosis us<strong>in</strong>g laser capture microdissection.Fertil Steril 2005;84(Suppl. 2):1180e90.[59] Burney RO, Talbi S, Hamilton AE, Vo KC, Nyegaard M, Nezhat CR, et al. Gene expression analysis of endometriumreveals progesterone resistance and candidate susceptibility genes <strong>in</strong> women with endometriosis.Endocr<strong>in</strong>ology 2007;148:3814e26.[60] Eyster KM, Kl<strong>in</strong>kova O, Kennedy V, Hansen KA. Whole genome deoxyribonucleic acid microarray analysis ofgene expression <strong>in</strong> ectopic versus eutopic endometrium. Fertil Steril 2007;88:1505e33.[61] Flores I, Rivera E, Ruiz LA, Santiago OI, Vernon MW, Appleyard CB. Molecular profil<strong>in</strong>g of experimentalendometriosis identified gene expression patterns <strong>in</strong> common with human disease. Fertil Steril2007;87:1180e99.[62] Hever A, Roth RB, Hevezi P, Mar<strong>in</strong> ME, Acosta JA, Acosta H, et al. <strong>Human</strong> endometriosis is associated withplasma cells and overexpression of B lymphocyte stimulator. Proc Natl Acad Sci USA 2007;104:12451e6.[63] Konno R, Fujiwara H, Netsu S, Odagiri K, Shimane M, Nomura H, et al. Gene expression profil<strong>in</strong>g of the ratendometriosis model. Am J Reprod Immunol 2007;58:330e43.[64] Mettler L, Salmassi A, Schollmeyer T, Schmutzler AG, Pungel F, Jonat W. Comparison of c-DNA microarrayanalysis of gene expression between eutopic endometrium and ectopic endometrium (endometriosis). J AssistReprod Genet 2007;24:249e58.[65] Chand AL, Murray AS, Jones RL, Hannan NJ, Salamonsen LA, Rombauts L. Laser capture microdissection andcDNA array analysis of endometrium identify CCL16 and CCL21 as epithelial-derived <strong>in</strong>flammatory mediatorsassociated with endometriosis. Reprod Biol Endocr<strong>in</strong>ol 2007;5:18.[66] Van Langendonckt A, Punyadeera C, Kamps R, Dunselman G, Kle<strong>in</strong>-Hitpass L, Schurgers LJ, et al. Identificationof novel antigens <strong>in</strong> blood vessels <strong>in</strong> rectovag<strong>in</strong>al endometriosis. Mol Hum Reprod 2007;13:875e86.[67] Hull ML, Escareno CR, Godsland JM, Doig JR, Johnson CM, Phillips SC, et al. Endometrial-peritoneal<strong>in</strong>teractions dur<strong>in</strong>g endometriotic lesion establishment. Am J Pathol 2008;173:700e15.463


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>464[68] Sherw<strong>in</strong> JR, Sharkey AM, Mihalyi A, Simsa P, Catalano RD, D’Hooghe TM. Global gene analysis of latesecretory phase, eutopic endometrium does not provide the basis for a m<strong>in</strong>imally <strong>in</strong>vasive test of endometriosis.Hum Reprod 2008;23:1063e8.[69] Zafrakas M, Tarlatzis BC, Streichert T, Pournaropoulos F, Wolfle U, Smeets SJ, et al. Genome-wide microarraygene expression, array-CGH analysis, and telomerase activity <strong>in</strong> advanced ovarian endometriosis: a high degreeof differentiation rather than malignant potential. Int J Mol Med 2008;21:335e44.[70] Pelch KE, Schroder AL, Kimball PA, Sharpe-Timms KL, Wade Davis J, Nagel SC. Aberrant gene expressionprofile <strong>in</strong> a mouse model of endometriosis mirrors that observed <strong>in</strong> women. Fertil Steril 2010;93:1615e27.[71] Umezawa M, Tanaka N, Ta<strong>in</strong>aka H, Takeda K, Ihara T, Sugamata M. Microarray analysis provides <strong>in</strong>sight <strong>in</strong>tothe early steps of pathophysiology of mouse endometriosis model <strong>in</strong>duced by autotransplantation of endometrium.Life Sci 2009;84:832e7.[72] Wu Y, Basir Z, Kajdacsy-Balla A, Strawn E, Macias V, Montgomery K, et al. Resolution of clonal orig<strong>in</strong>s forendometriotic lesions us<strong>in</strong>g laser capture microdissection and the human androgen receptor (HUMARA)assay. Fertil Steril 2003;79(Suppl. 1):710e7.[73] Ambros V. The functions of animal microRNAs. Nature 2004;431:350e5.[74] Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 2004;116:281e97.[75] Kurokawa R, Rosenfeld MG, Glass CK. Transcriptional regulation through noncod<strong>in</strong>g RNAs and epigeneticmodifications. RNA Biol 2009;6.[76] Wu Y, Halverson G, Basir Z, Strawn E, Yan P, Guo SW. Aberrant methylation at HOXA10 may be responsible for itsaberrant expression <strong>in</strong> the endometrium of patients with endometriosis. Am J Obstet Gynecol 2005;193:371e80.[77] Troiano RN, Taylor KJ. Sonographically guided therapeutic aspiration of benign-appear<strong>in</strong>g ovarian cysts andendometriomas. AJR Am J Roentgenol 1998;171:1601e5.[78] Taylor HS, Arici A, Olive D, Igarashi P. HOXA10 is expressed <strong>in</strong> response to sex steroids at the time ofimplantation <strong>in</strong> the human endometrium. J Cl<strong>in</strong> Invest 1998;101:1379e84.[79] Taylor HS, Bagot C, Kardana A, Olive D, Arici A. HOX gene expression is altered <strong>in</strong> the endometrium ofwomen with endometriosis. Hum Reprod 1999;14:1328e31.[80] Gui Y, Zhang J, Yuan L, Lessey BA. Regulation of HOXA-10 and its expression <strong>in</strong> normal and abnormalendometrium. Mol Hum Reprod 1999;5:866e73.[81] Szczepanska M, Wirstle<strong>in</strong> P, Luczak M, Jagodz<strong>in</strong>ski PP, Skrzypczak J. Reduced expression of HOXA10 <strong>in</strong> themidluteal endometrium from <strong>in</strong>fertile women with m<strong>in</strong>imal endometriosis. Biomed Pharmacother2010;64:697e705.[82] Kim JJ, Taylor HS, Lu Z, Ladhani O, Hast<strong>in</strong>gs JM, Jackson KS, et al. Altered expression of HOXA10 <strong>in</strong> endometriosis:potential role <strong>in</strong> decidualization. Mol Hum Reprod 2007;13:323e32.[83] Lee B, Du H, Taylor HS. Experimental mur<strong>in</strong>e endometriosis <strong>in</strong>duces DNA methylation and altered geneexpression <strong>in</strong> eutopic endometrium. Biol Reprod 2009;80:79e85.[84] Bromer JG, Wu J, Zhou Y, Taylor HS. Hypermethylation of HOXA10 by <strong>in</strong> utero diethylstilbestrol exposure: anepigenetic mechanism for altered developmental programm<strong>in</strong>g. Endocr<strong>in</strong>ology 2009;150:3376e82.[85] Zhang L, Zheng XM, Hubert J, Zheng H, Yang ZW, Li SW. Prenatal exposure to diaethylstilbestrol <strong>in</strong> the rat<strong>in</strong>hibits transabdom<strong>in</strong>al testicular descent with <strong>in</strong>volvement of the INSL3/LGR8 system and HOXA10. Ch<strong>in</strong>Med J (Engl) 2009;122:967e71.[86] Wu Y, Strawn E, Basir Z, Halverson G, Guo SW. Promoter hypermethylation of progesterone receptor isoformB (PR-B) <strong>in</strong> endometriosis. <strong>Epigenetics</strong> 2006;1:106e11.[87] Attia GR, Zeitoun K, Edwards D, Johns A, Carr BR, Bulun SE. Progesterone receptor isoform A but not B isexpressed <strong>in</strong> endometriosis. J Cl<strong>in</strong> Endocr<strong>in</strong>ol Metab 2000;85:2897e902.[88] Wu Y, Strawn E, Basir Z, Halverson G, Guo SW. Aberrant expression of deoxyribonucleic acid methyltransferasesDNMT1, DNMT3A, and DNMT3B <strong>in</strong> women with endometriosis. Fertil Steril 2007;87:24e32.[89] van Kaam KJ, Delvoux B, Romano A, D’Hooghe T, Dunselman GA, Groothuis PG. Deoxyribonucleic acidmethyltransferases and methyl-CpG-b<strong>in</strong>d<strong>in</strong>g doma<strong>in</strong> prote<strong>in</strong>s <strong>in</strong> human endometrium and endometriosis.Fertil Steril 2011;95:1421e7.[90] Xue Q, L<strong>in</strong> Z, Y<strong>in</strong> P, Milad MP, Cheng YH, Conf<strong>in</strong>o E, et al. Transcriptional activation of steroidogenic factor-1by hypomethylation of the 5’ CpG island <strong>in</strong> endometriosis. J Cl<strong>in</strong> Endocr<strong>in</strong>ol Metab 2007;92:3261e7.[91] Xue Q, L<strong>in</strong> Z, Cheng YH, Huang CC, Marsh E, Y<strong>in</strong> P, et al. Promoter methylation regulates estrogen receptor 2<strong>in</strong> human endometrium and endometriosis. Biol Reprod 2007;77:681e7.[92] Izawa M, Harada T, Taniguchi F, Ohama Y, Takenaka Y, Terakawa N. An epigenetic disorder may cause aberrantexpression of aromatase gene <strong>in</strong> endometriotic stromal cells. Fertil Steril 2008;89:1390e6.[93] Izawa M, Taniguchi F, Uegaki T, Takai E, Iwabe T, Terakawa N, Harada T. Demethylation of a nonpromotercytos<strong>in</strong>e-phosphate-guan<strong>in</strong>e island <strong>in</strong> the aromatase gene may cause the aberrant up-regulation <strong>in</strong> endometriotictissues. Fertil Steril 95:33e9.


CHAPTER 22The <strong>Epigenetics</strong> of Endometriosis[94] Starz<strong>in</strong>ski-Powitz A, Gaetje R, Zeitvogel A, Kotzian S, Handrow-Metzmacher H, Herrmann G, et al. Trac<strong>in</strong>gcellular and molecular mechanisms <strong>in</strong>volved <strong>in</strong> endometriosis. Hum Reprod Update 1998;4:724e9.[95] Starz<strong>in</strong>ski-Powitz A, Zeitvogel A, Schre<strong>in</strong>er A, Baumann R. In search of pathogenic mechanisms <strong>in</strong> endometriosis:the challenge for molecular cell biology. Curr Mol Med 2001;1:655e64.[96] Wu Y, Starz<strong>in</strong>ski-Powitz A, Guo SW. Trichostat<strong>in</strong> A, a histone deacetylase <strong>in</strong>hibitor, attenuates <strong>in</strong>vasivenessand reactivates E-cadher<strong>in</strong> expression <strong>in</strong> immortalized endometriotic cells. Reprod Sci 2007;14:374e82.[97] Wren JD, Wu Y, Guo SW. A system-wide analysis of differentially expressed genes <strong>in</strong> ectopic and eutopicendometrium. Hum Reprod 2007;22:2093e102.[98] Borghese B, Barbaux S, Mondon F, Santulli P, Pierre G, V<strong>in</strong>ci G, Chapron C, Vaiman D. Research resource:genome-wide profil<strong>in</strong>g of methylated promoters <strong>in</strong> endometriosis reveals a subtelomeric location ofhypermethylation. Mol Endocr<strong>in</strong>ol 24:1872e85.[99] Kawano Y, Nasu K, Li H, Tsuno A, Abe W, Takai N, Narahara H. Application of the histone deacetylase<strong>in</strong>hibitors for the treatment of endometriosis: histone modifications as pathogenesis and novel therapeutictarget. Hum Reprod 26:2486e98.[100] Jenuwe<strong>in</strong> T, Allis CD. Translat<strong>in</strong>g the histone code. <strong>Science</strong> 2001;293:1074e80.[101] Strahl BD, Allis CD. The language of covalent histone modifications. Nature 2000;403:41e5.[102] Keppler BR, Archer TK. Chromat<strong>in</strong>-modify<strong>in</strong>g enzymes as therapeutic targets e Part 1. Expert Op<strong>in</strong> TherTargets 2008;12:1301e12.[103] Suzuki A, Horiuchi A, Oka K, Miyamoto T, Kashima H, Shiozawa T. Immunohistochemical detection ofsteroid receptor cofactors <strong>in</strong> ovarian endometriosis: <strong>in</strong>volvement of down-regulated SRC-1 expression <strong>in</strong> thelimited growth activity of the endometriotic epithelium. Virchows Arch 456:433e41.[104] Kumagami A, Ito A, Yoshida-Komiya H, Fujimori K, Sato A. Expression patterns of the steroid receptorcoactivator family <strong>in</strong> human ovarian endometriosis. J Obstet Gynaecol Res 2011;37:1269e76.[105] Imesch P, F<strong>in</strong>k D, Fedier A. Romideps<strong>in</strong> reduces histone deacetylase activity, <strong>in</strong>duces acetylation of histones,<strong>in</strong>hibits proliferation, and activates apoptosis <strong>in</strong> immortalized epithelial endometriotic cells. Fertil Steril94:2838e42.[106] Zhao T, Liu X, Zhen X, Guo SW. Levo-tetrahydropalmat<strong>in</strong>e retards the growth of ectopic endometrialimplants and alleviates generalized hyperalgesia <strong>in</strong> experimentally <strong>in</strong>duced endometriosis <strong>in</strong> rats. Reprod Sci18:28e45.[107] Rea S, Eisenhaber F, O’Carroll D, Strahl BD, Sun ZW, Schmid M, et al. Regulation of chromat<strong>in</strong> structure bysite-specific histone H3 methyltransferases. Nature 2000;406:593e9.[108] Shi Y, Lan F, Matson C, Mulligan P, Whetst<strong>in</strong>e JR, Cole PA, et al. Histone demethylation mediated by thenuclear am<strong>in</strong>e oxidase homolog LSD1. Cell 2004;119:941e53.[109] Pedersen MT, Hel<strong>in</strong> K. Histone demethylases <strong>in</strong> development and disease. Trends Cell Biol 2010;20:662e71.[110] Bernste<strong>in</strong> BE, Meissner A, Lander ES. The mammalian epigenome. Cell 2007;128:669e81.[111] Klose RJ, Zhang Y. Regulation of histone methylation by demethylim<strong>in</strong>ation and demethylation. Nat Rev MolCell Biol 2007;8:307e18.[112] Leyendecker G. Redef<strong>in</strong><strong>in</strong>g endometriosis: endometriosis is an entity with extreme pleiomorphism. HumReprod 2000;15:4e7.[113] V<strong>in</strong>atier D, Cosson M, Dufour P. Is endometriosis an endometrial disease? Eur J Obstet Gynecol Reprod Biol2000;91:113e25.[114] Carlson JM, Doyle J. Highly optimized tolerance: robustness and design <strong>in</strong> complex systems. Phys Rev Lett2000;84:2529e32.[115] Richardson BC. Role of DNA methylation <strong>in</strong> the regulation of cell function: autoimmunity, ag<strong>in</strong>g and cancer.J Nutr 2002;132:2401Se5S.[116] Wilson VL, Jones PA. DNA methylation decreases <strong>in</strong> ag<strong>in</strong>g but not <strong>in</strong> immortal cells. <strong>Science</strong>1983;220:1055e7.[117] Toyota M, Issa JP. CpG island methylator phenotypes <strong>in</strong> ag<strong>in</strong>g and cancer. Sem<strong>in</strong> Cancer Biol 1999;9:349e57.[118] Jacob RA, Gretz DM, Taylor PC, James SJ, Pogribny IP, Miller BJ, et al. Moderate folate depletion <strong>in</strong>creasesplasma homocyste<strong>in</strong>e and decreases lymphocyte DNA methylation <strong>in</strong> postmenopausal women. J Nutr1998;128:1204e12.[119] Hsieh CJ, Klump B, Holzmann K, Borchard F, Gregor M, Porschen R. Hypermethylation of the p16INK4apromoter <strong>in</strong> colectomy specimens of patients with long-stand<strong>in</strong>g and extensive ulcerative colitis. Cancer Res1998;58:3942e5.[120] Issa JP, Ahuja N, Toyota M, Bronner MP, Brentnall TA. Accelerated age-related CpG island methylation <strong>in</strong>ulcerative colitis. Cancer Res 2001;61:3573e7.[121] Song JZ, Stirzaker C, Harrison J, Melki JR, Clark SJ. Hypermethylation trigger of the glutathione-S-transferasegene (GSTP1) <strong>in</strong> prostate cancer cells. Oncogene 2002;21:1048e61.465


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>466[122] Stirzaker C, Song JZ, Davidson B, Clark SJ. Transcriptional gene silenc<strong>in</strong>g promotes DNA hypermethylationthrough a sequential change <strong>in</strong> chromat<strong>in</strong> modifications <strong>in</strong> cancer cells. Cancer Res 2004;64:3871e7.[123] Champagne FA, Weaver IC, Diorio J, Dymov S, Szyf M, Meaney MJ. Maternal care associated with methylationof the estrogen receptor-alpha1b promoter and estrogen receptor-alpha expression <strong>in</strong> the medialpreoptic area of female offspr<strong>in</strong>g. Endocr<strong>in</strong>ology 2006;147:2909e15.[124] Lu Y, Nie J, Liu X, Guo SW. Reduced expression and concomitant promoter hypermethylation of HOXA10 <strong>in</strong>endometrium from women wear<strong>in</strong>g <strong>in</strong>trauter<strong>in</strong>e devices. Fertil Steril 94:1583e8.[125] Wu Y, Starz<strong>in</strong>ski-Powitz A, Guo SW. Prolonged stimulation with tumor necrosis factor-alpha <strong>in</strong>duced partialmethylation at PR-B promoter <strong>in</strong> immortalized epithelial-like endometriotic cells. Fertil Steril2008;90:234e7.[126] Wu Y, Shi X, Guo SW. The knockdown of progesterone receptor isoform B (PR-B) promotes proliferation <strong>in</strong>immortalized endometrial stromal cells. Fertil Steril 2008;90:1320e3.[127] Li S, Washburn KA, Moore R, Uno T, Teng C, Newbold RR, et al. Developmental exposure to diethylstilbestrolelicits demethylation of estrogen-responsive lactoferr<strong>in</strong> gene <strong>in</strong> mouse uterus. Cancer Res 1997;57:4356e9.[128] McLachlan JA, Simpson E, Mart<strong>in</strong> M. Endocr<strong>in</strong>e disrupters and female reproductive health. Best Pract ResCl<strong>in</strong> Endocr<strong>in</strong>ol Metab 2006;20:63e75.[129] Tang WY, Newbold R, Mardilovich K, Jefferson W, Cheng RY, Medvedovic M, et al. Persistent hypomethylation<strong>in</strong> the promoter of nucleosomal b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> 1 (Nsbp1) correlates with overexpression ofNsbp1 <strong>in</strong> mouse uteri neonatally exposed to diethylstilbestrol or geniste<strong>in</strong>. Endocr<strong>in</strong>ology2008;149:5922e31.[130] Li S, Hurst<strong>in</strong>g SD, Davis BJ, McLachlan JA, Barrett JC. Environmental exposure, DNA methylation, and generegulation: lessons from diethylstilbesterol-<strong>in</strong>duced cancers. Ann N Y Acad Sci 2003;983:161e9.[131] McGowan PO, Sasaki A, D’Alessio AC, Dymov S, Labonte B, Szyf M, et al. Epigenetic regulation of theglucocorticoid receptor <strong>in</strong> human bra<strong>in</strong> associates with childhood abuse. Nat Neurosci 2009;12:342e8.[132] Weaver IC, Cervoni N, Champagne FA, D’Alessio AC, Sharma S, Seckl JR, et al. Epigenetic programm<strong>in</strong>g bymaternal behavior. Nat Neurosci 2004;7:847e54.[133] Waterland RA, Jirtle RL. Early nutrition, epigenetic changes at transposons and impr<strong>in</strong>ted genes, andenhanced susceptibility to adult chronic diseases. Nutrition 2004;20:63e8.[134] Barker DJ. The developmental orig<strong>in</strong>s of adult disease. Eur J Epidemiol 2003;18:733e6.[135] Missmer SA, Hank<strong>in</strong>son SE, Spiegelman D, Barbieri RL, Michels KB, Hunter DJ. In utero exposures and the<strong>in</strong>cidence of endometriosis. Fertil Steril 2004;82:1501e8.[136] Dol<strong>in</strong>oy DC, Huang D, Jirtle RL. Maternal nutrient supplementation counteracts bisphenol A-<strong>in</strong>duced DNAhypomethylation <strong>in</strong> early development. Proc Natl Acad Sci USA 2007;104:13056e61.[137] Dol<strong>in</strong>oy DC, Weidman JR, Waterland RA, Jirtle RL. Maternal geniste<strong>in</strong> alters coat color and protects Avymouse offspr<strong>in</strong>g from obesity by modify<strong>in</strong>g the fetal epigenome. Environ Health Perspect 2006;114:567e72.[138] Wu Y, Guo SW. Inhibition of proliferation of endometrial stromal cells by trichostat<strong>in</strong> A, RU486, CDB-2914,N-acetylcyste<strong>in</strong>e, and ICI 182780. Gynecol Obstet Invest 2006;62:193e205.[139] Wu Y, Guo SW. Histone deacetylase <strong>in</strong>hibitors trichostat<strong>in</strong> A and valproic acid <strong>in</strong>duce cell cycle arrest and p21expression <strong>in</strong> immortalized human endometrial stromal cells. Eur J Obstet Gynecol Reprod Biol2008;137:198e203.[140] Wu Y, Starz<strong>in</strong>ski-Powitz A, Guo SW. Constitutive and tumor necrosis factor-alpha-stimulated activation ofnuclear factor-kappaB <strong>in</strong> immortalized endometriotic cells and their suppression by trichostat<strong>in</strong> A. GynecolObstet Invest 70:23e33.[141] Wu Y, Guo SW. Suppression of IL-1beta-<strong>in</strong>duced COX-2 expression by trichostat<strong>in</strong> A (TSA) <strong>in</strong> humanendometrial stromal cells. Eur J Obstet Gynecol Reprod Biol 2007;135:88e93.[142] Ota H, Igarashi S, Sasaki M, Tanaka T. Distribution of cyclooxygenase-2 <strong>in</strong> eutopic and ectopic endometrium<strong>in</strong> endometriosis and adenomyosis. Hum Reprod 2001;16:561e6.[143] Matsuzaki S, Canis M, Pouly JL, Wattiez A, Okamura K, Mage G. Cyclooxygenase-2 expression <strong>in</strong> deependometriosis and matched eutopic endometrium. Fertil Steril 2004;82:1309e15.[144] Buchweitz O, Staebler A, Wulf<strong>in</strong>g P, Hauzman E, Greb R, Kiesel L. COX-2 overexpression <strong>in</strong> peritoneal lesionsis correlated with nonmenstrual chronic pelvic pa<strong>in</strong>. Eur J Obstet Gynecol Reprod Biol 2006;124:216e21.[145] Yuan L, Shen F, Lu Y, Liu X, Guo SW. Cyclooxygenase-2 overexpression <strong>in</strong> ovarian endometriomas is associatedwith higher risk of recurrence. Fertil Steril 2008;91:1303e6.[146] Wu Y, Guo SW. Peroxisome proliferator-activated receptor-gamma and ret<strong>in</strong>oid X receptor agonists synergisticallysuppress proliferation of immortalized endometrial stromal cells. Fertil Steril 2009;91:2142e7.[147] Peeters LL, Vigne JL, Tee MK, Zhao D, Waite LL, Taylor RN. PPARgamma represses VEGF expression <strong>in</strong> humanendometrial cells: implications for uter<strong>in</strong>e angiogenesis. Angiogenesis 2005;8:373e9.


CHAPTER 22The <strong>Epigenetics</strong> of Endometriosis[148] Ohama Y, Harada T, Iwabe T, Taniguchi F, Takenaka Y, Terakawa N. Peroxisome proliferator-activatedreceptor-gamma ligand reduced tumor necrosis factor-alpha-<strong>in</strong>duced <strong>in</strong>terleuk<strong>in</strong>-8 production and growth <strong>in</strong>endometriotic stromal cells. Fertil Steril 2008;89:311e7.[149] Aytan H, Caliskan AC, Demirturk F, Aytan P, Koseoglu DR. Peroxisome proliferator-activated receptor-gammaagonist rosiglitazone reduces the size of experimental endometriosis <strong>in</strong> the rat model. Aust N Z J ObstetGynaecol 2007;47:321e5.[150] Lebovic DI, Kir M, Casey CL. Peroxisome proliferator-activated receptor-gamma <strong>in</strong>ducesregression of endometrial explants <strong>in</strong> a rat model of endometriosis. Fertil Steril 2004;82(Suppl. 3):1008e13.[151] Lebovic DI, Mwenda JM, Chai DC, Mueller MD, Santi A, Fisseha S, et al. PPAR-gamma receptor ligand<strong>in</strong>duces regression of endometrial explants <strong>in</strong> baboons: a prospective, randomized, placebo- and drugcontrolledstudy. Fertil Steril 2007;88:1108e19.[152] Guo SW. Nuclear Factor-kappaB (NF-kappaB): An Unsuspected Major Culprit <strong>in</strong> the Pathogenesis ofEndometriosis That is Still At Large? Gynecol Obestet Invest 2006;63:71e97.[153] Gonzalez-Ramos R, Donnez J, Defrere S, Leclercq I, Squifflet J, Lousse JC, et al. Nuclear factor-kappa B isconstitutively activated <strong>in</strong> peritoneal endometriosis. Mol Hum Reprod 2007;13:503e9.[154] Gonzalez-Ramos R, Van Langendonckt A, Defrere S, Lousse JC, Colette S, Devoto L, et al. Involvement of thenuclear factor-kappaB pathway <strong>in</strong> the pathogenesis of endometriosis. Fertil Steril 2010;94:1985e94.[155] Imesch P, Samartzis P, Schneider M, F<strong>in</strong>k D, Fedier A. Inhibition of transcription, expression, and secretion ofthe vascular epithelial growth factor <strong>in</strong> human epithelial endometriotic cells by romideps<strong>in</strong>. Fertil Steril95:1579e83.[156] Wang B, Xiao Y, D<strong>in</strong>g BB, Zhang N, Yuan X, Gui L, et al. Induction of tumor angiogenesis by Slit-Robosignal<strong>in</strong>g and <strong>in</strong>hibition of cancer growth by block<strong>in</strong>g Robo activity. Cancer Cell 2003;4:19e29.[157] Shen FH, Liu XS, Geng JG, Guo SW. Increased immunoreactivity to SLIT/ROBO1 <strong>in</strong> ovarian endometriomasand as a likely constituent biomarker for recurrence. Amer J Pathol 2009;175:479e88.[158] Moynihan AT, Hehir MP, Sharkey AM, Robson SC, Europe-F<strong>in</strong>ner GN, Morrison JJ. Histone deacetylase<strong>in</strong>hibitors and a functional potent <strong>in</strong>hibitory effect on human uter<strong>in</strong>e contractility. Am J Obstet Gynecol2008;199(167):e1e7.[159] Bulletti C, D, D. E. Z., Setti PL, Cic<strong>in</strong>elli E, Polli V, Flamigni C. The patterns of uter<strong>in</strong>e contractility <strong>in</strong> normalmenstruat<strong>in</strong>g women: from physiology to pathology. Ann N Y Acad Sci 2004;1034:64e83.[160] Kitlas A, Oczeretko E, Swiatecka J, Borowska M, Laudanski T. Uter<strong>in</strong>e contraction signalseapplication of thel<strong>in</strong>ear synchronization measures. Eur J Obstet Gynecol Reprod Biol 2009;144(Suppl. 1):S61e4.[161] Ruddock NK, Shi SQ, Ja<strong>in</strong> S, Moore G, Hank<strong>in</strong>s GD, Romero R, et al. Progesterone, but not 17-alphahydroxyprogesteronecaproate, <strong>in</strong>hibits human myometrial contractions. Am J Obstet Gynecol2008;199(391):e1e7.[162] Lu Y, Nie J, Liu X, Zheng Y, Guo SW. Trichostat<strong>in</strong> A, a histone deacetylase <strong>in</strong>hibitor, reduces lesion growth andhyperalgesia <strong>in</strong> experimentally <strong>in</strong>duced endometriosis <strong>in</strong> mice. Hum Reprod 25:1014e25.[163] Liu M, Liu X, Zhang Y, Guo SW. Valproic acid and progest<strong>in</strong> <strong>in</strong>hibit lesion growth and reduce hyperalgesia <strong>in</strong>experimentally <strong>in</strong>duced endometriosis <strong>in</strong> rats. Reprod Sci 2012; Feb 16. [Epub ahead of pr<strong>in</strong>t].[164] Liu X, Guo SW. A pilot study on the off-label use of valproic acid to treat adenomyosis. Fertil Steril2008;89:246e50.[165] Liu XS, Guo SW. Valproic acid alleviates generalized hyperalgesia <strong>in</strong> mice with <strong>in</strong>duced adenomyosis.J Gynaecol Obstet Res 2011;37:696e708.[166] Jichan N, Xishi L, Guo SW. Promoter hypermethylation of progesterone receptor isoform B (PR-B) <strong>in</strong>adenomyosis and its rectification by a histone deacetylase <strong>in</strong>hibitor and a demethylation agent. Reprod Sci17:995e1005.[167] Mao X, Wang Y, Carter AV, Zhen X, Guo SW. The Retardation of Myometrial Infiltration, Reduction of Uter<strong>in</strong>eContractility, and Alleviation of Generalized Hyperalgesia <strong>in</strong> Mice With Induced Adenomyosis by Levo-Tetrahydropalmat<strong>in</strong>e (l-thp) and Andrographolide. Reprod Sci 2011;18:1025e37.[168] Liu X, Guo SW. Valproic acid alleviates generalized hyperalgesia <strong>in</strong> mice with <strong>in</strong>duced adenomyosis. J ObstetGynaecol Res 37:696e708.[169] Bassel<strong>in</strong> M, Chang L, Chen M, Bell JM, Rapoport SI. Chronic adm<strong>in</strong>istration of valproic acid reduces bra<strong>in</strong>NMDA signal<strong>in</strong>g via arachidonic acid <strong>in</strong> unanesthetized rats. Neurochem Res 2008;33:2229e40.[170] Woolf CJ, Thompson SW. The <strong>in</strong>duction and ma<strong>in</strong>tenance of central sensitization is dependent on N-methyl-D-aspartic acid receptor activation; implications for the treatment of post-<strong>in</strong>jury pa<strong>in</strong> hypersensitivity states.Pa<strong>in</strong> 1991;44:293e9.[171] Gao YJ, Ji RR. c-Fos and pERK, which is a better marker for neuronal activation and central sensitization afternoxious stimulation and tissue <strong>in</strong>jury? Open Pa<strong>in</strong> J 2009;2:11e7.467


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>468[172] Sun RQ, Lawand NB, Willis WD. The role of calciton<strong>in</strong> gene-related peptide (CGRP) <strong>in</strong> the generation andma<strong>in</strong>tenance of mechanical allodynia and hyperalgesia <strong>in</strong> rats after <strong>in</strong>tradermal <strong>in</strong>jection of capsaic<strong>in</strong>. Pa<strong>in</strong>2003;104:201e8.[173] Ikeuchi M, Kolker SJ, Burnes LA, Walder RY, Sluka KA. Role of ASIC3 <strong>in</strong> the primary and secondary hyperalgesiaproduced by jo<strong>in</strong>t <strong>in</strong>flammation <strong>in</strong> mice. Pa<strong>in</strong> 2008;137:662e9.[174] Latremoliere A, Woolf CJ. Central sensitization: a generator of pa<strong>in</strong> hypersensitivity by central neural plasticity.J Pa<strong>in</strong> 2009;10:895e926.[175] McMahon SB. NGF as a mediator of <strong>in</strong>flammatory pa<strong>in</strong>. Philos Trans R Soc Lond B Biol Sci1996;351:431e40.[176] Sevcik MA, Ghilardi JR, Peters CM, L<strong>in</strong>dsay TH, Halvorson KG, Jonas BM, et al. Anti-NGF therapy profoundlyreduces bone cancer pa<strong>in</strong> and the accompany<strong>in</strong>g <strong>in</strong>crease <strong>in</strong> markers of peripheral and central sensitization.Pa<strong>in</strong> 2005;115:128e41.[177] Fuks F. DNA methylation and histone modifications: team<strong>in</strong>g up to silence genes. Curr Op<strong>in</strong> Genet Dev2005;15:490e5.[178] Fuks F, Burgers WA, Brehm A, Hughes-Davies L, Kouzarides T. DNA methyltransferase Dnmt1 associates withhistone deacetylase activity. Nat Genet 2000;24:88e91.[179] Tauboll E, Gregoraszczuk EL, Wojtowicz AK, Milewicz T. Effects of levetiracetam and valproate on reproductiveendocr<strong>in</strong>e function studied <strong>in</strong> human ovarian follicular cells. Epilepsia 2009;50:1868e74.[180] von Krogh K, Harjen H, Almas C, Zimmer KE, Dahl E, Olsaker I, Tauboll E, Ropstad E, Verhaegen S. Theeffect of valproate and levetiracetam on steroidogenesis <strong>in</strong> forskol<strong>in</strong>-stimulated H295R cells. Epilepsia51:2280e8.[181] L<strong>in</strong>dstrom TM, Mohan AR, Johnson MR, Bennett PR. Histone deacetylase <strong>in</strong>hibitors exert time-dependenteffects on nuclear factor-kappaB but consistently suppress the expression of pro<strong>in</strong>flammatory genes <strong>in</strong> humanmyometrial cells. Mol Pharmacol 2008;74:109e21.[182] Wang J, Mahmud SA, Bitterman PB, Huo Y, Slungaard A. Histone deacetylase <strong>in</strong>hibitors suppress TF-kappaBdependentagonist-driven tissue factor expression <strong>in</strong> endothelial cells and monocytes. J Biol Chem2007;282:28408e18.[183] Dong XF, Song Q, Li LZ, Zhao CL, Wang LQ. Histone deacetylase <strong>in</strong>hibitor valproic acid <strong>in</strong>hibits proliferationand <strong>in</strong>duces apoptosis <strong>in</strong> KM3 cells via downregulat<strong>in</strong>g VEGF receptor. Neuro Endocr<strong>in</strong>ol Lett2007;28:775e80.[184] Krikun G, Hu Z, Osteen K, Bruner-Tran KL, Schatz F, Taylor HS, Toti P, Arcuri F, Konigsberg W, Garen A,Booth CJ, Lockwood CJ. The immunoconjugate “icon” targets aberrantly expressed endothelial tissue factorcaus<strong>in</strong>g regression of endometriosis. Am J Pathol 176:1050e6.[185] Krikun G, Schatz F, Taylor H, Lockwood CJ. Endometriosis and tissue factor. Ann N YAcad Sci 2008;1127:101e5.[186] Rogers PA, Donoghue JF, Walter LM, Girl<strong>in</strong>g JE. Endometrial angiogenesis, vascular maturation, andlymphangiogenesis. Reprod Sci 2009;16:147e51.[187] Cameron EE, Bachman KE, Myohanen S, Herman JG, Bayl<strong>in</strong> SB. Synergy of demethylation and histonedeacetylase <strong>in</strong>hibition <strong>in</strong> the re-expression of genes silenced <strong>in</strong> cancer. Nat Genet 1999;21:103e7.[188] Eden A, Gaudet F, Waghmare A, Jaenisch R. Chromosomal <strong>in</strong>stability and tumors promoted by DNAhypomethylation. <strong>Science</strong> 2003;300:455.[189] Gaudet F, Hodgson JG, Eden A, Jackson-Grusby L, Dausman J, Gray JW, et al. Induction of tumors <strong>in</strong> mice bygenomic hypomethylation. <strong>Science</strong> 2003;300:489e92.[190] Suzuki H, Gabrielson E, Chen W, Anbazhagan R, van Engeland M, Weijenberg MP, et al. A genomic screen forgenes upregulated by demethylation and histone deacetylase <strong>in</strong>hibition <strong>in</strong> human colorectal cancer. NatGenet 2002;31:141e9.[191] Heller G, Schmidt WM, Ziegler B, Holzer S, Mullauer L, Bilban M, et al. Genome-wide transcriptionalresponse to 5-aza-2’-deoxycytid<strong>in</strong>e and trichostat<strong>in</strong> a <strong>in</strong> multiple myeloma cells. Cancer Res 2008;68:44e54.[192] Liang G, Gonzales FA, Jones PA, Orntoft TF, Thykjaer T. Analysis of gene <strong>in</strong>duction <strong>in</strong> human fibroblastsand bladder cancer cells exposed to the methylation <strong>in</strong>hibitor 5-aza-2’-deoxycytid<strong>in</strong>e. Cancer Res 2002;62:961e6.[193] Van L<strong>in</strong>t C, Emiliani S, Verd<strong>in</strong> E. The expression of a small fraction of cellular genes is changed <strong>in</strong> response tohistone hyperacetylation. Gene Expr 1996;5:245e53.[194] Bodden-Heidrich R, Hilber<strong>in</strong>k M, Frommer J, Stratkotter A, Rechenberger I, Bender HG, et al. [Qualitativeresearch on psychosomatic aspects of endometriosis]. Z Psychosom Med Psychother 1999;45:372e89.[195] Yang AS, Estecio MR, Garcia-Manero G, Kantarjian HM, Issa JP. Comment on “Chromosomal <strong>in</strong>stability andtumors promoted by DNA hypomethylation” and “Induction of tumors <strong>in</strong> nice by genomic hypomethylation”.<strong>Science</strong> 2003;302:1153. author reply 1153.


CHAPTER 22The <strong>Epigenetics</strong> of Endometriosis[196] Lubbert M, Wijermans P, Kunzmann R, Verhoef G, Bosly A, Ravoet C, et al. Cytogenetic responses <strong>in</strong> high-riskmyelodysplastic syndrome follow<strong>in</strong>g low-dose treatment with the DNA methylation <strong>in</strong>hibitor 5-aza-2’-deoxycytid<strong>in</strong>e. Br J Haematol 2001;114:349e57.[197] Garcia-Manero M, Santana GT, Alcazar JL. Relationship between Microvascular Density and Expression ofVascular Endothelial Growth Factor <strong>in</strong> Patients with Ovarian Endometriosis. J Womens Health (Larchmt)2008;17:777e82.[198] Chandrareddy A, Muneyyirci-Delale O. Risks versus benefits of valproic acid? Fertil Steril 2008;90:238.author reply 238e9.[199] Fluck CE, Yaworsky DC, Miller WL. Effects of anticonvulsants on human p450c17 (17alpha-hydroxylase/17,20 lyase) and 3beta-hydroxysteroid dehydrogenase type 2. Epilepsia 2005;46:444e8.[200] Bof<strong>in</strong>ger DP, Feng L, Chi LH, Love J, Stephen FD, Sutter TR, et al. Effect of TCDD exposure on CYP1A1 andCYP1B1 expression <strong>in</strong> explant cultures of human endometrium. Toxicol Sci 2001;62:299e314.[201] McIntyre RS, Manc<strong>in</strong>i DA, McCann S, Sr<strong>in</strong>ivasan J, Kennedy SH. Valproate, bipolar disorder and polycysticovarian syndrome. Bipolar Disord 2003;5:28e35.[202] Bahamondes L, Petta CA, Fernandes A, Monteiro I. Use of the levonorgestrel-releas<strong>in</strong>g <strong>in</strong>trauter<strong>in</strong>e system <strong>in</strong>women with endometriosis, chronic pelvic pa<strong>in</strong> and dysmenorrhea. Contraception 2007;75:S134e9.[203] Spannhoff A, Kim YK, Raynal NJ, Gharibyan V, Su MB, Zhou YY, et al. Histone deacetylase <strong>in</strong>hibitor activity<strong>in</strong> royal jelly might facilitate caste switch<strong>in</strong>g <strong>in</strong> bees. EMBO Rep 2011;12:238e43.[204] Kucharski R, Maleszka J, Foret S, Maleszka R. Nutritional control of reproductive status <strong>in</strong> honeybees viaDNA methylation. <strong>Science</strong> 2008;319:1827e30.[205] Barchuk AR, Crist<strong>in</strong>o AS, Kucharski R, Costa LF, Simoes ZL, Maleszka R. Molecular determ<strong>in</strong>ants of castedifferentiation <strong>in</strong> the highly eusocial honeybee Apis mellifera. BMC Dev Biol 2007;7:70.[206] Fiegl H, Gattr<strong>in</strong>ger C, Widschwendter A, Schneitter A, Ramoni A, Sarlay D, et al. Methylated DNA collected bytamponsea new tool to detect endometrial cancer. Cancer Epidemiol Biomarkers Prev 2004;13:882e8.[207] Shen F, Wang Y, Lu Y, Yuan L, Liu X, Guo SW. Immunoreactivity of progesterone receptor isoform B andnuclear factor kappa-B as biomarkers for recurrence of ovarian endometriomas. Am J Obstet Gynecol2008;199:e1e486.e10.[208] Jenuwe<strong>in</strong> T. The epigenetic magic of histone lys<strong>in</strong>e methylation. FEBS J 2006;273:3121e35.[209] Wolffe AP. Chromat<strong>in</strong> remodel<strong>in</strong>g: why it is important <strong>in</strong> cancer. Oncogene 2001;20:2988e90.[210] Nimura K, Ura K, Kaneda Y. Histone methyltransferases: regulation of transcription and contribution tohuman disease. J Mol Med 88:1213e20.[211] Pedersen MT, Hel<strong>in</strong> K. Histone demethylases <strong>in</strong> development and disease. Trends Cell Biol 20:662e71.[212] Liu XS, Nie J, Guo SW. Elevated immunoreactivity aga<strong>in</strong>st class I histone deacetylases (HDACs) <strong>in</strong>adenomyosis. Gynecol Obstet Invest 2012. <strong>in</strong> press.469


This page <strong>in</strong>tentionally left blank


CHAPTER 23Aberrant DNA Methylation<strong>in</strong> Endometrial CancerKenta Masuda, Kouji Banno, Megumi Yanokura, Kosuke Tsuji, Iori Kisu, Arisa Ueki,Yusuke Kobayashi, Hiroyuki Nomura, Akira Hirasawa, Nobuyuki Susumu, Daisuke AokiKeio University School of Medic<strong>in</strong>e, Tokyo, JapanCHAPTER OUTLINE23.1 Introduction 47123.2 Epigenetic DNAHypermethylation <strong>in</strong> CancerCells 47223.3 Aberrant DNA Methylation <strong>in</strong>Endometrial Cancer 47323.4 Methylation of microRNA<strong>in</strong> EndometrialCancer 47423.5 Application of Aberrant DNAHypermethylationto Diagnostics 47523.6 Application of Aberrant DNAHypermethylationto Treatment 47623.7 Future Directions andConclusion 478References 47847123.1 INTRODUCTION<strong>Epigenetics</strong> refers to the <strong>in</strong>formation stored after somatic cell division that is not conta<strong>in</strong>edwith<strong>in</strong> the DNA base sequence. Recent f<strong>in</strong>d<strong>in</strong>gs have shown that epigenetic changes e selectiveabnormalities <strong>in</strong> gene function that are not due to DNA base sequence abnormalities e playa significant role <strong>in</strong> carc<strong>in</strong>ogenesis <strong>in</strong> various organs [1,2]. In particular, the relationship betweencancer and aberrant hypermethylation of specific genome regions has attracted attention.A completely new model for the mechanism of carc<strong>in</strong>ogenesis has been proposed <strong>in</strong> whichhypermethylation of unmethylated CpG islands <strong>in</strong> the promoter regions of cancer-related genes<strong>in</strong> normal cells silences these genes and leads to the cell becom<strong>in</strong>g cancerous (Figure 23.1). Bothgenetic and epigenetic changes are <strong>in</strong>tricately <strong>in</strong>volved <strong>in</strong> the process through which cellsbecome cancerous, and hypermethylation of cancer-related genes such as p16,APC,andhMLH1has been associated with several types of cancer [3,4]. The ma<strong>in</strong> difference between epigeneticabnormalities and genetic abnormalities, such as gene mutations, is that epigenetic changes arereversible and do not <strong>in</strong>volve changes <strong>in</strong> base sequence, which suggests that gene re-expression ispossible and that epigenetic data may lead to important molecular targets for treatment.Attempts have begun to detect aberrant DNA methylation of cancer cells present <strong>in</strong> m<strong>in</strong>utequantities <strong>in</strong> biological samples and to apply the results to cancer diagnosis, prediction of therisk of carc<strong>in</strong>ogenesis, and def<strong>in</strong>ition of the properties of a particular cancer [5].In Japan, the number of women with endometrial cancer and the prevalence and mortality rateof this cancer cont<strong>in</strong>ue to <strong>in</strong>crease due to westernization of lifestyles and environmentalT. Tollefsbol (Ed): <strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>. DOI: 10.1016/B978-0-12-388415-2.00023-8Copyright Ó 2012 Elsevier Inc. All rights reserved.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>FIGURE 23.1Mechanism of <strong>in</strong>activation of cancer-related genes.472changes. Endometrial cancer is positively associated with higher BMI and obesity at age 20 andweight ga<strong>in</strong> dur<strong>in</strong>g adulthood among Japanese women [6,7]. Endometrial cancer currentlyaccounts for approximately 40% of all cancers of the uterus and an <strong>in</strong>crease <strong>in</strong> the totalnumber of patients and the number of young women with this condition has been forecast.Elucidation of the pathogenesis and establishment of effective treatment for endometrialcancer are significant challenges <strong>in</strong> gynecological oncology, but many aspects of the carc<strong>in</strong>ogenicmechanism are still poorly understood. The conventional explanation of the mechanism<strong>in</strong>volv<strong>in</strong>g genetic changes e mutations of cancer-related genes e is <strong>in</strong>adequate and epigeneticchanges <strong>in</strong> endometrial cancer are now be<strong>in</strong>g exam<strong>in</strong>ed. In particular, aberrant DNA methylationis thought to play a key role <strong>in</strong> endometrial carc<strong>in</strong>ogenesis. Breakdown of the DNAmismatch repair mechanism plays a particularly important role <strong>in</strong> the development of type Iendometrial cancer, and <strong>in</strong>hibition of hMLH1 expression due to DNA methylation maycontribute significantly to this mechanism. Therefore, an understand<strong>in</strong>g of the epigenetics ofDNA methylation may shed light on the mechanism of carc<strong>in</strong>ogenesis and improve diagnosis,risk evaluation, treatment, and prevention of endometrial cancer.23.2 EPIGENETIC DNA HYPERMETHYLATION IN CANCER CELLSMechanisms <strong>in</strong>volved <strong>in</strong> epigenetic regulation of gene expression <strong>in</strong>volve DNA methylation,histone modification, and polycomb-group prote<strong>in</strong>s [8]. DNA methylation patterns arefaithfully stored after cell division and DNA methylation is one of the most common and beststudiedepigenetic modifications <strong>in</strong> mammals. Genomic DNA methylation <strong>in</strong> vertebratesoccurs at the cytos<strong>in</strong>e <strong>in</strong> CpG sites; that is, where a cytos<strong>in</strong>e is directly followed by a guan<strong>in</strong>e <strong>in</strong>the DNA sequence. Transfer of the methyl group from S-adenosyl-L-methion<strong>in</strong>e is catalyzed byDNA methyltransferase enzymes <strong>in</strong> two dist<strong>in</strong>ct processes referred to as ma<strong>in</strong>tenance methylationand de novo methylation. Ma<strong>in</strong>tenance methylation takes place after DNA replication,which produces hemimethylated DNA <strong>in</strong> which only one of the strands is methylated. CpGsites on the daughter strand are then methylated <strong>in</strong> exactly the same way as the parent strand.In ma<strong>in</strong>tenance methylation, the methylation pattern of the parent strand is copied onto thedaughter strand produced by DNA replication dur<strong>in</strong>g cell division, thus allow<strong>in</strong>g ma<strong>in</strong>tenanceof the pattern. De novo methylation <strong>in</strong>volves methylation of a completely unmethylated CpG,allow<strong>in</strong>g new methylation <strong>in</strong> the course of the generation or differentiation of cells, ag<strong>in</strong>g, orneoplastic transformation. The known DNA methyltransferases Dnmt1, Dnmt2, Dnmt3a,


CHAPTER 23Aberrant DNA Methylation <strong>in</strong> Endometrial CancerDnmt3b, and Dnmt3L are classified accord<strong>in</strong>g to whether they catalyze ma<strong>in</strong>tenance or denovo methylation. Dnmt1 ma<strong>in</strong>ta<strong>in</strong>s attachment of methyl groups to hemimethylated DNAdur<strong>in</strong>g replication, whereas Dnmt3a and Dnmt3b can catalyze de novo methylation of DNA.DNA methylation <strong>in</strong> a region with a dense concentration of CpG sites (CpG islands) upstreamfrom the transcription <strong>in</strong>itiation site has a critical effect on gene expression [9]. It has also beenshown that a region of unmethylated DNA tends not to form nucleosomes, allow<strong>in</strong>g transcriptionto occur, whereas methylated DNA <strong>in</strong>duces nucleosome formation that renderstranscription impossible [10,11].Tumor suppressor genes such as CDKN2A, CDH1 (E-cadher<strong>in</strong>), and hMLH1 are silenced asa result of aberrant DNA methylation of CpG islands <strong>in</strong> their promoter regions. Aberrant DNAmethylation may play a significant role <strong>in</strong> carc<strong>in</strong>ogenesis, as for gene mutations [12].Decreased DNA methylation (hypomethylation) is an early event <strong>in</strong> carc<strong>in</strong>ogenesis, and one ofthe first epigenetic alterations [13]. It is associated with early-stage genetic <strong>in</strong>stability and upregulationof gene expression [14].23.3 ABERRANT DNA METHYLATION IN ENDOMETRIAL CANCEREndometrial cancer is classified <strong>in</strong>to types I and II accord<strong>in</strong>g to cl<strong>in</strong>icopathological characteristics.Type I endometrial cancer ma<strong>in</strong>ly occurs <strong>in</strong> pre- or perimenopausal women; isestrogen-dependent and positive for both estrogen and progesterone receptors; and developsfrom endometrial hyperplasia. Pathologically, type I endometrial cancer is a well-differentiatedendometrioid adenocarc<strong>in</strong>oma with a low <strong>in</strong>cidence of lymph node metastasis and myometrial<strong>in</strong>vasion. The prognosis is usually comparatively favorable. Type II endometrial cancerma<strong>in</strong>ly occurs <strong>in</strong> postmenopausal women; is estrogen-<strong>in</strong>dependent; and is thought to developfrom a normal endometrium directly or from unspecified precancerous lesions, but not fromendometrial hyperplasia. Histologically, it is an unusual type with poorly differentiatedendometrioid adenocarc<strong>in</strong>oma or serous adenocarc<strong>in</strong>oma, and the prognosis is usually poor.Different molecular mechanisms are thought to be <strong>in</strong>volved <strong>in</strong> the development of the twotypes of endometrial cancer. An epigenetic mechanism has been proposed for development oftype I endometrial cancer.473Aberrant DNA methylation is common <strong>in</strong> type I but not type II endometrial cancer [15]. Thisdifference <strong>in</strong> the methylation levels between types I and II cancers could be expla<strong>in</strong>ed by the<strong>in</strong>creased expression levels <strong>in</strong> DNA methyltransferase1 (DNMT1) and DNMT3B <strong>in</strong> type Iendometrial cancer <strong>in</strong> contrast to the reduced DNMT1 and DNMT3B expression levels <strong>in</strong> typeII endometrial cancer [16].In type I endometrial cancer, DNA mismatch repair (MMR) deficiency is a typical geneticdefect. The DNA mismatch repair system corrects errors <strong>in</strong> bases that arise when genes arereplicated dur<strong>in</strong>g cell division and silenc<strong>in</strong>g of DNA mismatch repair genes reduces theability to repair gene mutations. This results <strong>in</strong> an accumulation of cancer-related genemutations, lead<strong>in</strong>g to carc<strong>in</strong>ogenesis. The MMR gene hMLH1 is a typical gene that is silencedby promoter DNA methylation. In endometrial cancer, hMLH1 silenc<strong>in</strong>g is found <strong>in</strong>approximately 40% of cases and is an important step <strong>in</strong> the early stages of carc<strong>in</strong>ogenesis,with the loss of DNA mismatch repair function proposed to lead to mutation of genes such asPTEN [17,18].Microsatellite <strong>in</strong>stability (MSI) occurs when the mismatch repair system is damaged. Microsatellitesare DNA sequences of repeat<strong>in</strong>g units of between one and five base-pairs. Abnormalities<strong>in</strong> the mismatch repair system may cause replication errors <strong>in</strong> the repeat<strong>in</strong>g unit,lead<strong>in</strong>g to changes <strong>in</strong> length that are referred to as microsatellite <strong>in</strong>stability. MSI is observed <strong>in</strong>certa<strong>in</strong> types of cancer, <strong>in</strong>clud<strong>in</strong>g approximately 20e30% of cases of endometrial cancer [19].These results suggest that MMR gene abnormalities occur frequently <strong>in</strong> endometrial cancer.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>FIGURE 23.2Frequencies of aberrant methylation ofcancer-related genes <strong>in</strong> specimens fromnormal endometria, atypical endometrialhyperplasia and endometrial cancer. NE,normal endometrium; AEH, atypicalendometrial hyperplasia; EC,endometrial cancer. This figure isreproduced <strong>in</strong> the color plate section.Source: Reproduced from [17], withpermission.474In patients with endometrial cancer, aberrant DNA hypermethylation was found <strong>in</strong> thepromoter CpG islands of hMLH1, APC, E-cadher<strong>in</strong>, and CHFR. The frequencies of aberranthypermethylation were 40.4% <strong>in</strong> hMLH1, 22.0% <strong>in</strong> APC, 14.0% <strong>in</strong> E-cadher<strong>in</strong>, and 13.3% <strong>in</strong>CHFR, respectively. A significant decrease <strong>in</strong> prote<strong>in</strong> expression was found <strong>in</strong> patients withaberrant methylation of hMLH1 (p < 0.01) and E-cadher<strong>in</strong> (p < 0.05), and aberrant methylationof hMLH1 was also found <strong>in</strong> 14.3% of patients with atypical endometrial hyperplasia.However, no aberrant methylation of the four cancer-related genes was found <strong>in</strong> patients witha normal endometrium. These results <strong>in</strong>dicate that aberrant methylation of specific genesassociated with carc<strong>in</strong>ogenesis <strong>in</strong> endometrial cancer does not occur <strong>in</strong> a normal endometrium,with aberrant methylation of the hMLH1 gene be<strong>in</strong>g most frequent [17]. The aberrantmethylation of hMLH1 <strong>in</strong> atypical endometrial hyperplasia, which is found <strong>in</strong> the first stage ofendometrial cancer, supports the hypothesis that hMLH1 aberrant methylation is an importantevent <strong>in</strong> carc<strong>in</strong>ogenesis <strong>in</strong> endometrial cancer [17] (Figure 23.2).23.4 METHYLATION OF microRNA IN ENDOMETRIAL CANCERMicroRNAs (miRNAs) are small non-cod<strong>in</strong>g RNAs of approximately 22 base-pairs that regulatethe expression of genes by target<strong>in</strong>g mRNA with complementarity with the miRNA basesequence. Regulation of gene expression by miRNAs is important <strong>in</strong> cellular development anddifferentiation, and recent studies suggest a relationship between human diseases and thebreakdown of gene silenc<strong>in</strong>g mechanisms <strong>in</strong>duced by miRNA abnormalities. In particular,abnormal miRNA expression has been detected <strong>in</strong> various cancers and the target genes havebeen identified.The first evidence of a correlation between miRNAs and cancer was reported by Cal<strong>in</strong> et al.,who observed knockdown or knockout of miR-15a and miR-16-1 <strong>in</strong> approximatively 69% ofCLL patients [20]. It is now known that many miRNAs have actions that make them equivalentto oncogenes or tumor-suppressor genes <strong>in</strong> cancer development and progression.MiRNA expression can be regulated by several mechanisms, chromosomal abnormalities,mutations, polymorphisms (SNPs) [21,22]. In addition, also epigenetic mechanisms, such aspromoter methylation or histone acetylation, can modulate miRNA expression, and anaberrant regulation at this level is found <strong>in</strong> different diseases, <strong>in</strong>clud<strong>in</strong>g cancer. Several


CHAPTER 23Aberrant DNA Methylation <strong>in</strong> Endometrial Cancerevidences have <strong>in</strong>deed proved that an altered methylation status can be responsible for thederegulated expression of miRNAs <strong>in</strong> cancer. In endometrial cancer, Huang et al. reported thatSOX4 oncogene was overexpressed, and miRNA, miR-129-2, was validated to be an upstreamregulator of SOX4. The hypermethylation of the miR-129-2 CpG island, which was observed <strong>in</strong>endometrial cancer cell l<strong>in</strong>es and primary tumors, silences miR-129-2 and depresses itsoncogenic target, SOX4 [23]. Tsuruta et al. found miR-152 was a tumor-suppressor miRNAgene <strong>in</strong> endometrial cancer, and miR-152 was silenced by DNA hypermethylation. They alsoidentified E2F3, MET, and Rictor as candidate targets of miR-152 [24]. MiRNAs themselves canregulate the expression of components of the epigenetic mach<strong>in</strong>ery, creat<strong>in</strong>g a highlycontrolled feedback mechanism.An aberrant expression of these miRNAs, called “epi-miRNAs”, has been often related todevelopment or progression of human cancer. The first evidence of the existence of epimiRNAswas reported <strong>in</strong> lung cancer, where miR-29 family has been shown to directly targetthe de novo DNA methyltransferases DNMT-3A and -3B, and more recently the ma<strong>in</strong>tenanceDNA methyl transferase DNMT [25].23.5 APPLICATION OF ABERRANT DNA HYPERMETHYLATIONTO DIAGNOSTICSAberrant DNA methylation can be analyzed us<strong>in</strong>g the polymerase cha<strong>in</strong> reaction (PCR), whichshows a high degree of sensitivity for m<strong>in</strong>ute quantities of DNA <strong>in</strong> biological samples.However, it is important to ensure that the DNA hypermethylation is specific to cancer cells,s<strong>in</strong>ce aberrant DNA hypermethylation can also occur <strong>in</strong> non-cancerous cells. Specific aberrantDNA hypermethylation may be applicable to cancer diagnosis, but use of this method forcancer screen<strong>in</strong>g requires detection of cancer-cell genes with aberrant hypermethylation <strong>in</strong>cl<strong>in</strong>ical samples that also conta<strong>in</strong> normal cells. Methylation-specific PCR (MSP), whichcomb<strong>in</strong>es bisulfite sequenc<strong>in</strong>g and PCR, can detect aberrant hypermethylation with a highdegree of sensitivity us<strong>in</strong>g small quantities of DNA. In bisulfite sequenc<strong>in</strong>g, cytos<strong>in</strong>e isconverted to uracil, but methylated cytos<strong>in</strong>e is not converted. The PCR is thus set up withprimers for sequences conta<strong>in</strong><strong>in</strong>g cytos<strong>in</strong>e or uracil and amplification is performed to detectaberrant hypermethylation (Figure 23.3). In several types of cancer, this procedure can be usedwith various biological samples <strong>in</strong>clud<strong>in</strong>g sputum, plasma, and ur<strong>in</strong>e. In endometrial cancer,MSP has been used with endometrial cell samples to detect aberrant methylation of cancerassociatedgenes [17].475The methylation status of common tumor-suppressor genes could be useful to dist<strong>in</strong>guishbetween histological subtypes of endometrial cancer. Seeber et al. compared methylationFIGURE 23.3Pr<strong>in</strong>ciples of bisulfite treatment and methylation-specific PCR.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>status of a set of common tumor-suppressor genes, previously studied by Joensuu et al. [26],<strong>in</strong>endometrioid endometrial carc<strong>in</strong>oma and uter<strong>in</strong>e serous carc<strong>in</strong>oma. Promoter methylation ofCDH13 (Cadher<strong>in</strong> 13, H-cadher<strong>in</strong>) and MLH1 was more frequently present <strong>in</strong> endometrioidcarc<strong>in</strong>oma, while CDKN2B and TP73 were more frequently methylated <strong>in</strong> serous carc<strong>in</strong>oma.Almost 90% of endometrioid carc<strong>in</strong>oma and 70% of serous carc<strong>in</strong>oma could be predicted byCDH13 and TP73 [15].Aberrant DNA hypermethylation has been reported to affect several genes <strong>in</strong> endometrialcancer, <strong>in</strong> addition to hMLH1. Such genes <strong>in</strong>clude CASP8 (27), an apoptosis-related gene; TGFbRII(28), a TGF-b receptor with a tumor-suppressor effect; p73 (27), a tumor-suppressor gene;HOXA11 (29), which is important <strong>in</strong> uter<strong>in</strong>e development; and COMT (30), which codes forthe catechol-O-methyltransferase that metabolizes catechol, an estrogen metabolite that playsa role <strong>in</strong> carc<strong>in</strong>ogenesis. Methylation of each of these genes results <strong>in</strong> a loss of prote<strong>in</strong>expression that promotes cancer, and the degree of methylation of the genes differs significantlybetween normal tissues and endometrial cancer tissues [31].Aberrant DNA methylation of some tumor-suppressor genes was evident before endometrialcarc<strong>in</strong>oma diagnosis <strong>in</strong> women with the DNA mismatch repair gene mutation. That means thetim<strong>in</strong>g and molecular alterations of the critical events <strong>in</strong> endometrial carc<strong>in</strong>ogenesis may beuseful to identify DNA methylation profile for early detection of endometrial cancer [32].476The properties of cancer cells can be significantly affected by aberrant DNA hypermethylation.For example, <strong>in</strong> colorectal cancer, methylation of CHFR is observed <strong>in</strong> 40% of cases and isclosely related to sensitivity to the microtubule <strong>in</strong>hibitor docetaxel [33]. Other examples ofsimilar effects <strong>in</strong>clude the relationship between aberrant hypermethylation of the DNA repairenzyme MGMT and alkylat<strong>in</strong>g agent sensitivity [34], the l<strong>in</strong>k between simultaneous hypermethylationof CDKN2A and FHIT and recurrence of lung cancer [35], and the l<strong>in</strong>k betweenmethylation of ER-a and prognosis dur<strong>in</strong>g treatment with tamoxifen [36]. This last example<strong>in</strong>dicates that detection of aberrant hypermethylation can be used for prognostic evaluation.Aberrant DNA hypermethylation is sometimes seen <strong>in</strong> the elderly and <strong>in</strong> non-cancerousregions <strong>in</strong> cancer patients [37e39]. In particular, it has been shown that exposure to H. pylori,a carc<strong>in</strong>ogenic factor for gastric cancer, <strong>in</strong>duces aberrant DNA hypermethylation and that thedegree of aberration is correlated with carc<strong>in</strong>ogenic risk. This leads to the concept of an“epigenetic field for cancerization”, with similar results reported for colorectal and breastcancer [39]. Accumulation of aberrant methylation <strong>in</strong> normal tissue may trigger carc<strong>in</strong>ogenesisand such <strong>in</strong>formation may be useful to evaluate carc<strong>in</strong>ogenic risk.To analyze DNA methylation patterns on a genome-wide scale, several techniques have beendeveloped. Immunoprecipitation approach by an antibody aga<strong>in</strong>st 5-methylcytos<strong>in</strong>ecomb<strong>in</strong>ed with DNA microarrays was developed [40]. As a similar method, the MIRA(methylated-CpG island recovery assay) microarray approach was devised. It makes use of thehigh aff<strong>in</strong>ity of the MBD (methyl-CpG b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s) complex for methylated DNA [41].One of quantitative approaches currently used as standard methods employs base-specificcleavage and MALDI-TOF MS (matrix-assisted laser desorption ionization time-of-flight massspectrometry) analysis [42,43].23.6 APPLICATION OF ABERRANT DNA HYPERMETHYLATIONTO TREATMENTUnlike irreversible genetic changes, epigenetic patterns observed <strong>in</strong> cancer can be partly or fullyreversed pharmacologically. Indeed, this is the ma<strong>in</strong> reason for development of anticancerdrugs for epigenetic modification. Treatment of cancer us<strong>in</strong>g demethylat<strong>in</strong>g agents to restoreexpression of cancer-suppressor genes silenced through methylation has been attempted forsome time and use of methylation <strong>in</strong>hibitors to treat cancer has a long history. Development ofan antileukemic agent based on this pr<strong>in</strong>ciple was performed <strong>in</strong> the 1960s, but the drug could


CHAPTER 23Aberrant DNA Methylation <strong>in</strong> Endometrial Cancernot be used cl<strong>in</strong>ically due to toxicity problems at high concentrations. More recent reports haveshown antitumor effects at lower drug concentrations with a lower <strong>in</strong>cidence of adverse drugreactions and concomitant use with other chemotherapeutic agents may further improveefficacy. Lubbert et al. reported a response rate of 60% <strong>in</strong> patients with myelodysplasticsyndrome (MDS) follow<strong>in</strong>g adm<strong>in</strong>istration of the methylation <strong>in</strong>hibitor 5-aza-2 0 -deoxycytid<strong>in</strong>e(5-aza-dC) [44], with <strong>in</strong>duction of expression of p15INK4A follow<strong>in</strong>g demethylation<strong>in</strong> patients who were responsive to 5-aza-dC. A group from MD Anderson Cancer Centerreported that repeated adm<strong>in</strong>istration of low-concentration 5-aza-dC produced a response rateof approximately 60% <strong>in</strong> patients with acute myeloid leukemia [45]. However, the disadvantageof this methylation <strong>in</strong>hibitor is that it is not sequence-specific, which may lead to adverseeffects through demethylation of physiologically important genes and reactivation of cancergenes silenced by methylation. Therefore, development of sequence-specific demethylat<strong>in</strong>gagents based on b<strong>in</strong>d<strong>in</strong>g sequence of transcription factors is a current area of research.Epigenetic abnormalities have also been exam<strong>in</strong>ed as markers of anticancer drug sensitivity.Esteller et al. discovered that the DNA repair enzyme MGMT gene is silenced by methylationand showed that tumor cell l<strong>in</strong>es <strong>in</strong> which MGMT is methylated are highly responsive toalkylat<strong>in</strong>g agents, with a greater antitumor effect of BNCU observed <strong>in</strong> patients <strong>in</strong> whomMGMT methylation was detected <strong>in</strong> the tumor compared to those with no MGMT methylation[46]. Sato et al. reported that aberrant hypermethylation of CHFR, a mitotic checkpo<strong>in</strong>t gene,is strongly correlated with responsiveness to taxanes, which are microtubule <strong>in</strong>hibitors [47](Figure 23.4). These f<strong>in</strong>d<strong>in</strong>gs suggest that it may be possible to select a treatment based onmethylation as an <strong>in</strong>dicator of the biological characteristics of tumor cells. CHFR methylationmay be a particularly sensitive marker <strong>in</strong> endometrial cancer and analysis of this gene may playan important role <strong>in</strong> treatment of this type of cancer. CHFR is an M-phase checkpo<strong>in</strong>t genewith folk-head associated (FHA) and r<strong>in</strong>g f<strong>in</strong>ger doma<strong>in</strong>s that was first identified as a yeastDMA 1 gene homolog [48]. If the cell is subjected to mitotic stress <strong>in</strong> the M-phase, CHFRdelays progression from the start of prophase to the later part of prophase <strong>in</strong> mitosis. CHFR isan ubiquit<strong>in</strong> ligase that <strong>in</strong>cludes Aurora-A and PLK1 among its substrates, and degradation ofthese prote<strong>in</strong>s is thought to stop the cell cycle follow<strong>in</strong>g CHFR activation by microtubularstress [49,50]. The FHA doma<strong>in</strong> of CHFR is <strong>in</strong>volved <strong>in</strong> b<strong>in</strong>d<strong>in</strong>g of phosphorylated prote<strong>in</strong>sand is important <strong>in</strong> the checkpo<strong>in</strong>t function, but further studies are required to identifymolecules that <strong>in</strong>teract with CHFR.477CHFR methylation is thought to occur frequently <strong>in</strong> cancer [33,47] and is related to the mitotic<strong>in</strong>dex as follows. In normally function<strong>in</strong>g cells with no CHFR methylation, cell-cycle arrestoccurs dur<strong>in</strong>g the G2/M phase follow<strong>in</strong>g adm<strong>in</strong>istration of docetaxel and the mitotic <strong>in</strong>dex islow. Conversely, <strong>in</strong> cells <strong>in</strong> which CHFR hypermethylation has reduced expression of the gene,FIGURE 23.4Hypothesis for the relationship of expression of CHFR and sensitivity of cancer cells to taxanes.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>the mitotic <strong>in</strong>dex is high follow<strong>in</strong>g adm<strong>in</strong>istration of docetaxel. These results show that theexpression level of CHFR is negatively correlated with the mitotic <strong>in</strong>dex [33]. In cells <strong>in</strong> whichthe CHFR checkpo<strong>in</strong>t does not function, translocation of cycl<strong>in</strong> B1 to the nucleus dur<strong>in</strong>gmitotic stress cannot be prevented. Treatment of cells with CHFR hypermethylation with themethylation <strong>in</strong>hibitor 5-aza-dC restores the checkpo<strong>in</strong>t function and decreases the mitotic<strong>in</strong>dex. CHFR methylation and microtubule <strong>in</strong>hibitor sensitivity are useful molecular markers<strong>in</strong> gastric and cervical cancer, as well as <strong>in</strong> endometrial cancer, and methylation may be a usefulpredictor of anticancer drug responsiveness [51e53].23.7 FUTURE DIRECTIONS AND CONCLUSIONProgress made <strong>in</strong> epigenetics <strong>in</strong> recent years has suggested that aberrant DNA hypermethylationplays a role <strong>in</strong> carc<strong>in</strong>ogenesis <strong>in</strong> several types of cancer. Various genes are silencedas a result of aberrant hypermethylation, <strong>in</strong>clud<strong>in</strong>g cell cycle regulatory genes, apoptosisrelatedgenes, and DNA repair enzymes.478Epigenetic research <strong>in</strong> endometrial cancer suggests that damage to the mismatch repair systemplays a significant role <strong>in</strong> development of type I endometrial cancer and that hMLH1 hypermethylationis important <strong>in</strong> this mechanism. Such research has potential for prevention,diagnosis, risk assessment, and treatment of endometrial cancer. Cancer-specific DNA methylationmay be useful for diagnosis us<strong>in</strong>g methods such as MSP for detection of such abnormalities.Aberrant DNA hypermethylation can be detected with a high level of sensitivity andcancer cells can be detected <strong>in</strong> m<strong>in</strong>ute quantities of endometrial samples. Treatment withmethylation <strong>in</strong>hibitors such as 5-aza-dC may also be effective, s<strong>in</strong>ce a low concentration of thisdrug has an antitumor effect with a reduced <strong>in</strong>cidence of adverse drug reactions, andconcomitant use with other chemotherapy drugs may show even greater efficacy. Attempts arealso be<strong>in</strong>g made to use epigenetic abnormalities as <strong>in</strong>dicators of anticancer drug sensitivity,which may allow selection of the most appropriate treatment based on the biological characteristicsof tumor cells. Aberrant CHFR hypermethylation is strongly correlated with sensitivityto microtubule <strong>in</strong>hibitors and these f<strong>in</strong>d<strong>in</strong>gs may be applicable <strong>in</strong> treatment ofendometrial cancer. The ma<strong>in</strong> objective of epigenetics <strong>in</strong> oncology research is to identifyaberrant gene hypermethylation associated with carc<strong>in</strong>ogenesis. These f<strong>in</strong>d<strong>in</strong>gs may lead tonew methods of diagnosis and treatment based on control of methylation, <strong>in</strong>clud<strong>in</strong>g newapproaches to treatment of endometrial cancer.References[1] Chan MW, Chan LW, Tang NL, Tong JH, Lo KW, Lee TL, et al. Hypermethylation of multiple genes <strong>in</strong> tumortissues and voided ur<strong>in</strong>e <strong>in</strong> ur<strong>in</strong>ary bladder cancer patients. Cl<strong>in</strong> Cancer Res 2002;8(2):464e70.[2] Palmisano WA, Div<strong>in</strong>e KK, Saccomanno G, Gilliland FD, Bayl<strong>in</strong> SB, Herman JG, et al. Predict<strong>in</strong>g lung cancer bydetect<strong>in</strong>g aberrant promoter methylation <strong>in</strong> sputum. Cancer Res 2000;60(21):5954e8.[3] Bayl<strong>in</strong> SB, Herman JG. DNA hypermethylation <strong>in</strong> tumorigenesis: epigenetics jo<strong>in</strong>s genetics. Trends Genet2000;16:168e74.[4] Esteller M, Corn PG, Bayl<strong>in</strong> SB, Herman JG. A gene hypermethylation profile of human cancer. Cancer Res2001;61:3225e9.[5] Fiegl H, Gattr<strong>in</strong>ger C, Widschwendter A, Schneitter A, Ramoni A, Sarlay D, et al. Methylated DNA collected bytamponsdA new tool to detect endometrial cancer. Cancer Epidemiol. Biomarkers Prev 2004;13(5):882e8.[6] Kuriyama S, Tsubono Y, Hozawa A, Shimazu T, Suzuki Y, Koizumi Y, et al. Obesity and risk of cancer <strong>in</strong> Japan.Inter J Cancer 2005;113:148e57.[7] Satoyo H, Keitaro M, Kaoru H, Hidemi I, Takeshi S, Takakazu K, et al. Weight ga<strong>in</strong> dur<strong>in</strong>g adulthood and bodyweight at age 20 are associated with the risk of endometrial cancer <strong>in</strong> Japanese Women. J Epidemiol2011;21(6):466e73.[8] Goldberg AD, Allis CD, Bernste<strong>in</strong> E. <strong>Epigenetics</strong>: a landscape takes shape. Cell 2007;128(4):635e8.[9] Ushijima T. Detection and <strong>in</strong>terpretation of altered methylation patterns <strong>in</strong> cancer cells. Nat Rev Cancer2005;5:223e31.


CHAPTER 23Aberrant DNA Methylation <strong>in</strong> Endometrial Cancer[10] Gal-Yam EN, Jeong S, Tanay A, Egger G. Lee AS and Jones PA: Constitutive nucleosome depletion and orderedfactor assembly at the GRP78 promoter revealed by s<strong>in</strong>gle molecule footpr<strong>in</strong>t<strong>in</strong>g. PLoS Genet 2006;2:160.[11] Appanah R, Dickerson DR, Goyal P, Groud<strong>in</strong>e M, Lor<strong>in</strong>cz MC. An unmethylated 3’ promoter-proximal regionis required for efficient transcription <strong>in</strong>itiation. PLoS Genet 2007;3:27.[12] Jones PA, Bayl<strong>in</strong> SB. The epigenomics of cancer. Cell 2007;128:683e92.[13] Fe<strong>in</strong>berg AP, Vogelste<strong>in</strong> B. Hypomethylation dist<strong>in</strong>guishes genes of some human cancers from their normalcounterparts. Nature 1983;301:89e92.[14] Wilson AS, Power BE, Molloy PL. DNA hypomethylation and human diseases. Biochim Biophs Acta2007;1775:138e62.[15] Seeber LM, Zweemer RP, Marchionni L, Massuger LF, Smit VT, van Baal WM, et al. Methylation profiles ofendometrioid and serous endometrial cancers. Endocr Relat Cancer 2010;17(3):663e73.[16] Zhou XC, Dowdy SC, Podratz KC, Jiang SW. Epigenetic considerations for endometrial cancer prevention,diagnosis and treatment. Gynecol Oncol 2007;107:143e53.[17] Banno K, Yanokura M, Susumu N, Kawaguchi M, Hirao N, Hirasawa A, et al. Relationship of aberrant DNAhypermethylation of cancer-related genes with carc<strong>in</strong>ogenesis of endometrial cancer. Oncol Rep2006;16:1189e96.[18] Kanaya T, Kyo S, Sakaguchi J, Maida Y, Nakamura M, Takakura M, et al. Association of mismatch repairdeficiency with PTEN frameshift mutations <strong>in</strong> endometrial cancers and the precursors <strong>in</strong> a Japanese population.Am J Cl<strong>in</strong> Pathol 2005;124:89e96.[19] Kanaya T, Kyo S, Maida Y, Yatabe N, Tanaka M, Nakamura M, et al. Frequent hypermethylation of MLH1promoter <strong>in</strong> normal endometrium of patients with endometrial cancers. Oncogene 2003;22:2352e60.[20] Cal<strong>in</strong> GA, Dumitru CD, Shimizu M, Bichi R, Zupo S, Noch E, et al. Frequent deletions and down-regulation ofmicroRNA genes miR15 and miR16 at 13q14 <strong>in</strong> chronic lymphocytic leukemia. Proc Natl Acad Sci USA2002;99:15524e9.[21] Zhang L, Huang J, Yang N, Greshock J, Megraw MS, Giannakakis A, et al. MicroRNAs exhibit high frequencygenomic alterations <strong>in</strong> human cancer. Proc Natl Acad Sci USA 2006;103:9136e41.[22] Hu Z, Chen J, Tian T, Zhou X, Gu H, Xu L, et al. Genetic variants of miRNA sequences and non-small cell lungcancer survival. J Cl<strong>in</strong> Invest 2008;118:2600e8.[23] Huang YW, Liu JC, Deatherage DE, Luo J, Mutch DG, Goodfellow PJ, et al. Epigenetic repression of microRNA-129-2 leads to overexpression of SOX4 oncogene <strong>in</strong> endometrial cancer. Cancer Res 2009;69:9038e46.[24] Tsuruta T, Kozaki KI, Uesugi A, Furuta M, Hirasawa A, Imoto I, et al. miR-152 is a tumor suppressor microRNAthat is silenced by DNA hypermethylation <strong>in</strong> endometrial cancer. Cancer Res 2011;71(20):6450e62.[25] Fabbri M, Garzon R, Cimm<strong>in</strong>o A, Liu Z, Zanesi N, Callegari E, et al. MicroRNA-29 family reverts aberrantmethylation <strong>in</strong> lung cancer by target<strong>in</strong>g DNA methyltransferases3A and 3B. Proc Natl Acad Sci USA2007;104:15805e10.[26] Joensuu EI, Abdel-Rahman WM, Ollika<strong>in</strong>en M, Ruosaari S, Knuutila S, Peltomaki P. Epigenetic signatures offamilial cancer are characteristic of tumor type and family category. Cancer Res 2008;68:4597e605.[27] Yang HJ, Liu VW, Wang Y, Tsang PC, Ngan HY. Differential DNA methylation profiles <strong>in</strong> gynecological cancersand correlation with cl<strong>in</strong>ico-pathological data. BMC Cancer 2006;6:212.[28] Sakaguchi J, Kyo S, Kanaya T, Maida Y, Hashimoto M, Nakamura M, et al. Aberrant expression and mutationsof TGF-beta receptor type II gene <strong>in</strong> endometrial cancer. Gyneol Oncol 2005;98:427e33.[29] Whitcomb BP, Mutch DG, Herzog TJ, Rader JS, Gibb RK, Goodfellow PJ. Frequent HOXA11 and THBS2promoter methylation, and a methylator phenotype <strong>in</strong> endometrial adenocarc<strong>in</strong>oma. Cl<strong>in</strong> Cancer Res2003;9:2277e87.[30] Sasaki M, Kaneuchi M, Sakuragi N, Dahiya R. Multiple promoters of catechol-O-methyltransferase gene areselectively <strong>in</strong>activated by CpG hypermethylation <strong>in</strong> endometrial cancer. Cancer Res 2003;63:3101e6.[31] Fiegl H, Gattr<strong>in</strong>ger C, Widschwendter A, Schneitter A, Ramoni A, Sarlay D, et al. Methylated DNA collected bytamponsea new tool to detect endometrial cancer. Cancer Epidemiol Biomarkers Prev 2004;13:882e8.[32] Niem<strong>in</strong>en TT, Gyll<strong>in</strong>g A, Abdel-Rahman WM, Nuorva K, Aarnio M, Renkonen-S<strong>in</strong>isalo L, et al. Molecularanalysis of endometrial tumorigenesis: importance of complex hyperplasia regardless of atypia. Cl<strong>in</strong> CancerRes 2009;83(15):5772e83.[33] Toyota M, Sasaki Y, Satoh A, Ogi K, Kikuchi T, Suzuki H, et al. Epigenetic <strong>in</strong>activation of CHFR <strong>in</strong> humantumors. Proc Natl Acad Sci USA 2003;100:7818e23.[34] Esteller M, Herman JG. Generat<strong>in</strong>g mutations but provid<strong>in</strong>g chemosensitivity: the role of O6-methylguan<strong>in</strong>eDNA methyltransferase <strong>in</strong> human cancer. Oncogene 2004;23:1e8.[35] Kim JS, Kim JW, Han J, Shim YM, Park J, Kim DH. Cohypermethylation of p16 and FHIT promoters asa prognostic factor of recurrence <strong>in</strong> surgically resected stage I non-small cell lung cancer. Cancer Res2006;66:4049e54.479


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>480[36] Mori T, Mart<strong>in</strong>ez SR, O’Day SJ, Morton DL, Umetani N, Kitago M, et al. Estrogen receptor-alpha methylationpredicts melanoma progression. Cancer Res 2006;66:6692e8.[37] Waki T, Tamura G, Sato M, Motoyama T. Age-related methylation of tumor suppressor and tumor-relatedgenes: an analysis of autopsy samples. Oncogene 2003;22:4128e33.[38] Maekita T, Nakazawa K, Mihara M, Nakajima T, Yanaoka K, Iguchi M, et al. High levels of aberrant DNAmethylation <strong>in</strong> Helicobacter pylori-<strong>in</strong>fected gastric mucosae and its possible association with gastric cancerrisk. Cl<strong>in</strong> Cancer Res 2006;12:989e95.[39] Ushijima T. Epigenetic field for cancerization. J Biochem Mol Biol 2007;40:142e50.[40] Weber M, Davies JJ, Wittig D, Oakeley EJ, Haase M, Lam WL, et al. Chromosome-wide and promoter-specificanalyses identify sites of differential DNA methylation <strong>in</strong> normal and transformed human cells. Nat Genet2005;37(8):853e62.[41] Rauch T, Li H, Wu X, Pfeifer GP. MIRA-assisted microarray analysis, a new technology for the determ<strong>in</strong>ation ofDNA methylation patterns, identifies frequent methylation of homeodoma<strong>in</strong>-conta<strong>in</strong><strong>in</strong>g genes <strong>in</strong> lung cancercells. Cancer Res 2006;66(16):7939e47.[42] Ehrich M, Nelson MR, Stanssens P, Zabeau M, Liloglou T, X<strong>in</strong>arianos G, et al. Quantitative high-throughputanalysis of DNA methylation patterns by base-specific cleavage and mass spectrometry. Proc Natl Acad Sci USA2005;102(44):15785e90.[43] Schatz P, Distler J, Berl<strong>in</strong> K, Schuster M. Novel method for high throughput DNA methylation marker evaluationus<strong>in</strong>g PNA-probe library hybridization and MALDI-TOF detection. Nucleic Acids Res 2006;34(8):e59.[44] Daskalakis M, Nguyen TT, Nguyen C, Guldberg P, Köhler G, Wijermans P, et al. Demethylation of a hypermethylatedP15/INK4B gene <strong>in</strong> patients with myelodysplastic syndrome by 5-Aza-2’-deoxycytid<strong>in</strong>e (decitab<strong>in</strong>e)treatment. Blood 2002;100:2957e64.[45] Issa JP, Garcia-Manero G, Giles FJ, Mannari R, Thomas D, Faderl S, et al. Phase 1 study of low-dose prolongedexposure schedules of the hypomethylat<strong>in</strong>g agent 5-aza-2’-deoxycytid<strong>in</strong>e (decitab<strong>in</strong>e) <strong>in</strong> hematopoieticmalignancies. Blood 2003;103:1635e40.[46] Esteller M, Garcia-Foncillas J, Andion E, Goodman SN, Hidalgo OF, Vanaclocha V, et al. Inactivation of theDNA-repair gene MGMT and the cl<strong>in</strong>ical response of gliomas to alkylat<strong>in</strong>g agents. N Engl J Med2000;343:1350e4.[47] Satoh A, Toyota M, Itoh F, Sasaki Y, Suzuki H, Ogi K, et al. Epigenetic <strong>in</strong>activation of CHFR and sensitivity tomicrotubule <strong>in</strong>hibitors <strong>in</strong> gastric cancer. Cancer Res 2003;63:8606e13.[48] Scolnick DM, Halazonetis TD. CHFR def<strong>in</strong>es a mitotic stress checkpo<strong>in</strong>t that delays entry <strong>in</strong>to metaphase.Nature 2000;406:430e5.[49] Kang D, Chen J, Wong J, Fang G. The checkpo<strong>in</strong>t prote<strong>in</strong> CHFR is a ligase that ubiquit<strong>in</strong>ates Plk1 and <strong>in</strong>hibitsCdc2 at the G2 to M transition. J Cell Biol 2002;156:249e59.[50] Yu X, M<strong>in</strong>ter-Dykhouse K, Malureanu L, Zhao WM, Zhang D, Merkle CJ, et al. CHFR is required for tumorsuppression and Aurora A regulation. Nat Genet 2005;37:401e6.[51] Ogi K, Toyota M, Mita H, Satoh A, Kashima L, Sasaki Y, et al. Small <strong>in</strong>terfer<strong>in</strong>g RNA-<strong>in</strong>duced CHFR silenc<strong>in</strong>gsensitizes oral squamous cell cancer cells to microtubule <strong>in</strong>hibitors. Cancer Biol Ther 2005;4:773e80.[52] Banno K, Yanokura M, Kawaguchi M, Kuwabara Y, Akiyoshi J, Kobayashi Y, et al. Epigenetic <strong>in</strong>activation of theCHFR gene <strong>in</strong> cervical cancer contributes to sensitivity to taxanes. Int J Oncol 2007;31:713e20.[53] Koga Y, Kitajima Y, Miyoshi A, Sato K, Sato S, Miyazaki K. The significance of aberrant CHFR methylation forcl<strong>in</strong>ical response to microtubule <strong>in</strong>hibitors <strong>in</strong> gastric cancer. J Gastroenterol 2006;41:133e9.


CHAPTER 24Stem Cell <strong>Epigenetics</strong>and <strong>Human</strong> <strong>Disease</strong>Mehdi Shafa, Derrick E. RancourtUniversity of Calgary, Calgary, AB, CanadaCHAPTER OUTLINE24.1 Introduction 48124.2 <strong>Epigenetics</strong> 48224.3 Stem Cell <strong>Epigenetics</strong> 48224.4 Histone Variantsand Exchangeof Histones 48524.5 Chromat<strong>in</strong> Bivalency<strong>in</strong> ESCs 48624.6 Chang<strong>in</strong>g the EpigeneticLandscape Dur<strong>in</strong>g CellularReprogramm<strong>in</strong>g 48724.7 Stem Cell <strong>Epigenetics</strong> and<strong>Human</strong> <strong>Disease</strong> 49124.8 Model<strong>in</strong>g of <strong>Human</strong> EpigeneticDisorders Us<strong>in</strong>g iPSCs 49324.9 Future Studies 495References 49548124.1 INTRODUCTIONRecent analysis of the mechanisms by which chromat<strong>in</strong> remodel<strong>in</strong>g regulates embryonic andadult stem cell pluripotency/multipotency and differentiation have provided important<strong>in</strong>sights <strong>in</strong>to our understand<strong>in</strong>g of human and mouse developmental biology. There is nowa grow<strong>in</strong>g body of <strong>in</strong>formation, which shows the crucial key role of epigenetic changes andchromat<strong>in</strong> organization <strong>in</strong> the activation or repression of genes dur<strong>in</strong>g embryogenesis and <strong>in</strong>ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g pluripotency <strong>in</strong> stem cells. Epigenetic changes <strong>in</strong>clude histone acetylation,methylation and phosphorylation, and DNA methylation/demethylation. Higher-orderchromat<strong>in</strong> architecture <strong>in</strong>tegrity is also crucial for proper gene activity <strong>in</strong> stem cells. Pluripotentstem cells, <strong>in</strong>clud<strong>in</strong>g embryonic stem cells (ESCs) and primordial germ cells, have thepotential to differentiate <strong>in</strong>to any cell type <strong>in</strong> an organism, whereas multipotent or unipotentstem cells have limited differentiation capacity, giv<strong>in</strong>g rise to def<strong>in</strong>ed progenies. Normally,progression from a stem state to a more differentiated cell l<strong>in</strong>eage needs dist<strong>in</strong>guished changes<strong>in</strong> cell function, gene expression patterns, and morphology. Recent evidence reveals the role ofepigenetic modifications dur<strong>in</strong>g a stem cell’s ma<strong>in</strong>tenance of self-renewal, as well as dur<strong>in</strong>gdifferentiation. Alterations <strong>in</strong> the epigenetic status of cells are associated with different types ofhuman cancer and congenital disease. Induced pluripotent stem cells (iPSCs) have recentlybeen developed by reprogramm<strong>in</strong>g of mouse and human dermal fibroblasts follow<strong>in</strong>g thetransduction of def<strong>in</strong>ed transcription factors us<strong>in</strong>g retroviral vectors. The reprogramm<strong>in</strong>g ofsomatic cells through this technology is a valuable tool to identify the mechanisms of pluripotency.It also provides the potential of model<strong>in</strong>g human degenerative disorders andproduc<strong>in</strong>g patient-specific pluripotent stem cells. In this chapter, the fundamental impact ofT. Tollefsbol (Ed): <strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>. DOI: 10.1016/B978-0-12-388415-2.00024-XCopyright Ó 2012 Elsevier Inc. All rights reserved.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>chromat<strong>in</strong> dynamics <strong>in</strong> stem cells, as well as the critical role of epigenetic changes <strong>in</strong> thegeneration of human diseases, will be discussed. Furthermore, the relationship between stemcell epigenetics and human diseases such as cancer and the application of iPSCs <strong>in</strong> model<strong>in</strong>ghuman epigenetic disorders will be discussed.24.2 EPIGENETICSThe functional properties of a specific cell are not only dependent on DNA sequence, but alsoon the comb<strong>in</strong>ation of active and silent genes <strong>in</strong> a particular time dur<strong>in</strong>g development.Although all of the cells <strong>in</strong> the body have the same identical DNA sequence, every cell has itsown exclusive phenotype and gene expression pattern, which shows that the DNA is morecomplex and dynamic than has previously been known. There are other important mechanismsof gene regulation that rely mostly on the cell’s epigenetic status, which controls thetim<strong>in</strong>g and degree of gene expression <strong>in</strong> a particular time. In eukaryotes, genomic DNA isorganized <strong>in</strong>to DNA/prote<strong>in</strong> complexes known as chromat<strong>in</strong>. The central unit of chromat<strong>in</strong> isthe nucleosome, which is comprised of a family of small, basic prote<strong>in</strong>s called histones. Thenucleosome consists of two copies of each histone prote<strong>in</strong> termed H2A, H2B, H3, and H4 witha w146 base-pair of DNA wound around its surface [1]. These bead-like core prote<strong>in</strong>s are oftenassociated with a 15e55-bp sequence of l<strong>in</strong>ker DNA and a fifth histone, H1, which is believedto be responsible for organiz<strong>in</strong>g higher-order structure between the beads.482<strong>Epigenetics</strong> is a term <strong>in</strong>itially used by Wadd<strong>in</strong>gton <strong>in</strong> 1942 describ<strong>in</strong>g the relationship betweengenotype and phenotype through different gene <strong>in</strong>teractions [2]. The term epigenetic is usedtoday to describe transcriptional memory without affect<strong>in</strong>g DNA sequence and is mediatedmostly by variation <strong>in</strong> DNA methylation and chromat<strong>in</strong> structure. For example, a chromat<strong>in</strong>change <strong>in</strong> the bra<strong>in</strong>, <strong>in</strong>fluenc<strong>in</strong>g gene expression, can be “epigenetic” if it lasts regardless of celldivision.Specific residues <strong>in</strong> the N-term<strong>in</strong>al ends of histones, which protrude from the nucleosomesurface, are susceptible to numerous reversible post-translational modifications <strong>in</strong>clud<strong>in</strong>gmethylation, acetylation, and phosphorylation [3]. These alterations are obta<strong>in</strong>ed via variouschromat<strong>in</strong>-modify<strong>in</strong>g enzyme complexes with different antagonized functions, which areaccountable for the dynamic performance of chromat<strong>in</strong>. The “histone code” theory is assignedto the two different states of chromat<strong>in</strong> acquired by these modify<strong>in</strong>g complexes [4]. Generally,regions of transcriptional activity are dist<strong>in</strong>guished by lys<strong>in</strong>e acetylation alleviated by histoneacetyltranferases (HATs). Alternatively, histone deacetylases (HDACs), which mediate lys<strong>in</strong>edeacetylation, are related with regions of transcriptional quiescence.Higher-order chromat<strong>in</strong> organization is dist<strong>in</strong>guished by highly packed heterochromat<strong>in</strong> andrelatively broadened euchromat<strong>in</strong> regions with<strong>in</strong> the genome [5]. It has long been assumed thatheterochromat<strong>in</strong> is transcriptionally <strong>in</strong>ert compared to euchromat<strong>in</strong>. However, many recentstudies have questioned this model of transcriptionally silent heterochromat<strong>in</strong>. Some studieshave shown that transcription of heterochromat<strong>in</strong> is <strong>in</strong>dispensable for its own repression [6].Furthermore, a few prote<strong>in</strong>-cod<strong>in</strong>g genes <strong>in</strong> heterochromatic doma<strong>in</strong> [7,8] as well as RNAsexpressed <strong>in</strong> telomeric and pericentric regions have been also reported <strong>in</strong> different species [9,10].The crucial role of epigenetics <strong>in</strong> the regulation of stem cells and the etiology of human disordersis <strong>in</strong>creas<strong>in</strong>gly acknowledged and will be discussed <strong>in</strong> detail <strong>in</strong> the follow<strong>in</strong>g sections.24.3 STEM CELL EPIGENETICSThe important characteristics of stem cells (SCs), <strong>in</strong>clud<strong>in</strong>g self-renewal, ma<strong>in</strong>tenance ofpluripotency and potency e the capacity to differentiate along specialized cell l<strong>in</strong>eages erequire the presence of specific molecular mechanisms dur<strong>in</strong>g early development. Themechanism by which SCs select between self-renewal and differentiation seems to not onlyrely on specific molecular cues, but also more importantly on prom<strong>in</strong>ent modifications <strong>in</strong>


CHAPTER 24Stem Cell <strong>Epigenetics</strong> and <strong>Human</strong> <strong>Disease</strong>their chromat<strong>in</strong> state. Progression from the pluripotent state to a differentiated phenotype istypically highlighted by dist<strong>in</strong>guished changes <strong>in</strong> cellular function, which are predeterm<strong>in</strong>edby global gene expression patterns dur<strong>in</strong>g early development. Recent understand<strong>in</strong>g of themechanisms by which chromat<strong>in</strong> remodel<strong>in</strong>g controls stem cell pluripotency and differentiationhave had significant <strong>in</strong>fluence on our knowledge about developmental biology. There iscurrently a grow<strong>in</strong>g body of evidence, which reveals the decisive role of epigenetic variationsand chromat<strong>in</strong> reshap<strong>in</strong>g <strong>in</strong> the repression or activation of genes dur<strong>in</strong>g early developmentand also ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g the identity and self-renewal of stem cells (Figure 24.1).The regulatory mechanisms that regulate stem cell self-renewal are yet to be understood;however, the important function of transcription factors like Oct4, Sox2, and Nanog has beenelucidated. Several recent l<strong>in</strong>es of evidence underscore that stem cell differentiation and earlymammalian development largely depend on the elasticity of epigenetic alterations [11]. Themechanism of gene regulation dur<strong>in</strong>g early development does not only rely upon the <strong>in</strong>teractionsamong different transcription factors and signal<strong>in</strong>g pathways, but also on epigeneticmodifications, such as the ATP-dependent chromat<strong>in</strong> remodel<strong>in</strong>g [12], covalent alterations ofhistones [13], exchange of histones and histone variant [14], DNA methylation at CpG islands[15], RNA-mediated gene regulation <strong>in</strong>clud<strong>in</strong>g RNAi pathways and non-prote<strong>in</strong>-cod<strong>in</strong>g RNAs(ncRNA) [16,17]. All of these have been found to play critical roles <strong>in</strong> ma<strong>in</strong>tenance of stem cellpluripotency/multipotency, block<strong>in</strong>g differentiation <strong>in</strong> stem cells, and controll<strong>in</strong>g the<strong>in</strong>herited characteristics of cellular memories dur<strong>in</strong>g early development.One of the significant determ<strong>in</strong><strong>in</strong>g factors of gene expression <strong>in</strong> stem cells is DNA and histonemethylation. DNA methylation occurs mostly at the 5 0 end of the cytos<strong>in</strong>e nucleotide of CpGsd<strong>in</strong>ucleotide islands. However, non-CpG methylation like <strong>in</strong> CpT and CpA motifs can also takeplace <strong>in</strong> ESCs mediated by methytransferase 3a [18]. Particularly, unmethylated clusters ofCpG islands are located at the promoters of tissue-specific and housekeep<strong>in</strong>g genes, which arerequired to be expressed for ma<strong>in</strong>tenance purposes. These unmodified CpG pairs recruit othertranscription factors to start transcription. Conversely, methylated CpGs are normally locatedat the promoters of silent genes. Although DNA methylation at promoter regions converselyassociates with gene activity, this <strong>in</strong>verse relationship is reliant upon the amount of CpGmotifs with<strong>in</strong> a specific promoter. Additionally, there is no apparent relationship between geneexpression and methylation <strong>in</strong> promoters without dist<strong>in</strong>ct CpG content [19]. Methylated CpGislands are detected with<strong>in</strong> promoters of def<strong>in</strong>ite tissue-specific genes [20], but they are absent<strong>in</strong> other regions of the genome.483DNA methylation patterns are directed and conserved by the DNA methyltransferase (DNMT)family of prote<strong>in</strong>s, whereas the effects of DNA methylation are mediated by methyl-CpGb<strong>in</strong>d<strong>in</strong>gdoma<strong>in</strong> (MBD) familys, which dist<strong>in</strong>guish DNA sequences with high CpG content [21].There are three ma<strong>in</strong> functional DNA methytransferases <strong>in</strong> mammals <strong>in</strong>clud<strong>in</strong>g Dnmt1,Dnmt3a, Dnmt3b, and Dnmt3L. Dnmt1 is ubiquitously expressed <strong>in</strong> divid<strong>in</strong>g somatic cellsthroughout mammalian development. Dnmt1 was the first DNA methyltransferase enzyme tobe cloned [22] and shows a preference toward hemimethylated DNA over unmethylated DNAstrands [23]. Dnmt3a and Dnmt3b are recognized as de novo methyltransferases and have theresponsibility of establish<strong>in</strong>g methylation pattern dur<strong>in</strong>g mammalian early development and <strong>in</strong>germ cells. Both of the enzymes are highly expressed <strong>in</strong> ESCs and down-regulated upondifferentiation. Dnmt3L is enzymaticaly <strong>in</strong>active and has some regulatory functions <strong>in</strong> germcells. It has been shown that the DNA methylation pattern <strong>in</strong> differentiated cells is persistent andcan be <strong>in</strong>herited [24]. Cells achieve this pattern progressively as they move towards their ultimatespecific cell l<strong>in</strong>eage dur<strong>in</strong>g development. As the zygote divides, methylation of paternal andmaternal alleles are removed through down-regulation of Dnmt1 gene expression with<strong>in</strong> thenucleus [25]. This phenomenon is followed by reacquirement of a new DNA methylationpattern dur<strong>in</strong>g embryogenesis and germ layer formation. The epigenomic status of ESCs <strong>in</strong>sidethe blastocyst’s <strong>in</strong>ner cell mass is restored dur<strong>in</strong>g this period, to re-establish pluripotency.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>484FIGURE 24.1The pluripotency of ES cells and their differentiation rely largely on transcription factor circuitry and chromat<strong>in</strong> modifications. In this model, there isa multidirectional <strong>in</strong>teraction between transcription factors and the epigenetic signature of the cell. With<strong>in</strong> this scenario, the extracellular matrix (ECM) of EScells play an important role by provid<strong>in</strong>g necessary environmental signals. This figure is reproduced <strong>in</strong> the color plate section. Source: Adapted from [132]


CHAPTER 24Stem Cell <strong>Epigenetics</strong> and <strong>Human</strong> <strong>Disease</strong>The role of various DNMTs <strong>in</strong> mouse ESCs has been expla<strong>in</strong>ed us<strong>in</strong>g homozygous mutants.Dnmt1 e/e ESCs can multiply when cultured as undifferentiated cells, suggest<strong>in</strong>g that thehypomethylated state can preserve self-renewal. The <strong>in</strong>duction of apoptosis <strong>in</strong> these ESCsupon differentiation is highly correlated to their <strong>in</strong>ability to suppress pluripotency factors likeOct4 and Nanog by DNA methylation [26]. Recent advances suggest that the CpG motif with<strong>in</strong>the Oct4 promoter is hypomethylated <strong>in</strong> pluripotent ESCs yet is highly methylated <strong>in</strong> differentk<strong>in</strong>ds of somatic cells. In a study on a neuronally committed human teratocarc<strong>in</strong>oma cell l<strong>in</strong>e(NT2), down-regulation of Oct4 and Nanog expression dur<strong>in</strong>g differentiation is corelated tomethylation of 5 0 -flank<strong>in</strong>g regions of both these genes [27]. ESCs have the potential capabilityto impose their pluripotent epigenetic status to differentiated cells upon fusion. For <strong>in</strong>stance,when term<strong>in</strong>ally differentiated cells are fused with ESCs, result<strong>in</strong>g heterokaryons acquire thepluripotent state [28]. It has been believed that this capacity is largely restricted to earlyembryonic cells <strong>in</strong>clud<strong>in</strong>g ESCs and that l<strong>in</strong>eage-restricted cells loose this capacity to revert theepigenetic status of other nuclei.Global chromat<strong>in</strong> dynamism is also of <strong>in</strong>terest <strong>in</strong> ESCs, as recent studies stress its fundamentalrole <strong>in</strong> the preservation of pluripotency and regulation of gene expression [29]. Several studiespo<strong>in</strong>t out that ESCs are del<strong>in</strong>eated by less-condensed chromat<strong>in</strong> and higher transcriptionactivity compared to differentiated cells. Dur<strong>in</strong>g the advancement of stem cell differentiation,active chromat<strong>in</strong> is substituted with a repressed and <strong>in</strong>active state [30]. For example, ESCsaccumulate highly compacted heterochromat<strong>in</strong> <strong>in</strong> the pericentric doma<strong>in</strong> of some chromosomesdur<strong>in</strong>g differentiation, which is not detected <strong>in</strong> the pluripotent state [31,32]. Likewise,the number, size, and distribution of highly compact heterochromat<strong>in</strong> foci are changed dur<strong>in</strong>gESC neural differentiation [33,34].Epigenetic mechanisms have profound effects on mammalian stem cell regulation and theirdysfunction can give rise to several human diseases such as neurodegenerative disorders andcancer. Nevertheless, the extents to which these modifications can determ<strong>in</strong>e stem cell fate arestill ma<strong>in</strong>ly unrecognized and many questions have to be answered.48524.4 HISTONE VARIANTS AND EXCHANGE OF HISTONESSeveral other replacement variants for H2A (H2A.X, H2A.Z, H2A-Bbd, MacroH2A) and H3(H3.2, H3.3, CenpA) have been identified <strong>in</strong> addition to the four conserved histone prote<strong>in</strong>s(H2A, H2B, H3, and H4). Unlike core prote<strong>in</strong>s, these histone variants are transcribed froma polyA mRNA and their assembly <strong>in</strong>to chromat<strong>in</strong> is controlled separately from DNA replication.The vast majority of the histone variants are found at specific chromat<strong>in</strong> conformationsand at def<strong>in</strong>ed developmental stages. For example, MacroH2A is found at the <strong>in</strong>active X-chromosome [35] and acts like a suppressor variant [36], whereas H2A.Z chaperons euchromat<strong>in</strong>from the ectopic distribution of <strong>in</strong>active heterochromat<strong>in</strong> by mediat<strong>in</strong>g stabilization ofthe nucleosome [37]. Conversely, Histone H2A-Bbd has the opposite effect of destabiliz<strong>in</strong>g thenucleosome components.The role of histone variants <strong>in</strong> the regulation of ESC differentiation has been documented <strong>in</strong>several studies. For <strong>in</strong>stance, H2A.Z is highly expressed <strong>in</strong> human embryonic carc<strong>in</strong>oma celll<strong>in</strong>es, but its expression is dim<strong>in</strong>ished upon differentiation [38]. Tip60-p400, a chromat<strong>in</strong>remodel<strong>in</strong>g prote<strong>in</strong>, behaves as a transcriptional enhancer by <strong>in</strong>clud<strong>in</strong>g the H2A.Z to targetpromoters <strong>in</strong> ESCs [39]. It can be hypothesized that H2A.Z helps ma<strong>in</strong>ta<strong>in</strong> ESC’s openchromat<strong>in</strong> state, which is <strong>in</strong>dispensable for pluripotency. It is important to verify thehypothesis by understand<strong>in</strong>g how different histone variants like H2A.Z target specific genes <strong>in</strong>ESCs and how they may correlate to the suppression or expression of particular genes dur<strong>in</strong>gESCs self-renewal and differentiation.To better understand how the histone variants contribute to pluripotency <strong>in</strong> ESCs, Meshoreret al. looked for the exchange rate of different chromat<strong>in</strong> prote<strong>in</strong>s <strong>in</strong> mouse ESCs and their


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>differentiated cell progeny us<strong>in</strong>g fluorescent recovery after photobleach<strong>in</strong>g (FRAP) [34]. Theysuggested that the histone structure <strong>in</strong> ESCs is highly dynamic and various histone variants<strong>in</strong>teract with chromat<strong>in</strong> rapidly and transiently, rang<strong>in</strong>g from a few seconds to a few m<strong>in</strong>utes.These f<strong>in</strong>d<strong>in</strong>gs demonstrated that a large number of specific histone variants (25%) attachloosely to chromat<strong>in</strong> <strong>in</strong> ESC, as compared to differentiated cells.Recently, Ng and Gurdon showed the significant role of histone H3.3 variants <strong>in</strong> cellularmemory <strong>in</strong> Xenopus [40]. They provide evidence that the epigenetic memory of a transcriptionallyactive state relies upon histone H3.3 <strong>in</strong> chromat<strong>in</strong>. So, DNA methylation is not thesole mechanism <strong>in</strong>volved <strong>in</strong> cellular memory. This new discovery may def<strong>in</strong>e how ESCs canpass their identity through their daughter cells after division, thereby ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g self-renewal.These f<strong>in</strong>d<strong>in</strong>gs have resulted <strong>in</strong> the hyperdynamic (“breath<strong>in</strong>g”) theory of chromat<strong>in</strong> architecture<strong>in</strong> ESCs. Chromat<strong>in</strong> hyperdynamics is an exclusive characteristic of ESCs and isimportant <strong>in</strong> ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g ESC self-renewal and pluripotency, as differentiated and nonpluripotentcells do not show this property [34]. This poised chromat<strong>in</strong> structure, which ispartly dependent upon histone variants, gives an opportunity to ESCs to rapidly differentiate<strong>in</strong>to any l<strong>in</strong>eage-specific cell.48624.5 CHROMATIN BIVALENCY IN ESCSRecent studies propose that several types of prote<strong>in</strong> complexes mediate the histone methylation<strong>in</strong> eukaryotes. Several studies <strong>in</strong> Drosophila have emphasized the crucial role of Polycomb(PcG) and Thrithorax multiprote<strong>in</strong> complexes <strong>in</strong> regulat<strong>in</strong>g l<strong>in</strong>eage-specific gene expression[41]. Polycomb prote<strong>in</strong>s (PcG) repress gene activity by regulat<strong>in</strong>g trimethylation of lys<strong>in</strong>e 27 athistone H3 (H3K27me3) and impart<strong>in</strong>g a silent state of gene expression. PcG prote<strong>in</strong>s downregulatemany signal<strong>in</strong>g and developmental genes by <strong>in</strong>duc<strong>in</strong>g methylation of H3K27. PRC1and PRC2, as two major PcG complexes, have been found to mediate gene silenc<strong>in</strong>g <strong>in</strong>mammals. PRC2 mediates both H3K27me3 and H3K27me2 methylation through its Ezh2,Suz12, and EeD subunits [41,42]. PRC2 has been suggested to be responsible for genesilenc<strong>in</strong>g by ubiquit<strong>in</strong>ation of H2A at Lys<strong>in</strong>e-119, which accelerate the <strong>in</strong>duction of chromat<strong>in</strong>compaction [43]. Thrithorax prote<strong>in</strong>s have the opposite effect by act<strong>in</strong>g to control geneexpression through regulation of chromat<strong>in</strong> structure by catalyz<strong>in</strong>g or <strong>in</strong>teract<strong>in</strong>g with H3K4methylation (H3K4me3). Other members of this family can provoke ATP-dependent chromat<strong>in</strong>remodel<strong>in</strong>g.In 2006, two studies suggested that PcG prote<strong>in</strong> complexes play a regulatory function <strong>in</strong>ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g ESC pluripotency [44e46]. Mouse development and ESC self-renewal aredependent upon the expression of PcG genes, as no ESC l<strong>in</strong>es can be derived from Ezh2-deficient mice [47]. It should be noted that PRC2 was found to be dispensable for ESCspluripotency [48]. Some of the developmentally important genes <strong>in</strong> ESCs are regulated by PcGrepressor complex [49,50]. Interest<strong>in</strong>gly, a significant number of these sites are also occupiedby Oct4, Nanog, and Sox2 [49]. This is <strong>in</strong>terest<strong>in</strong>g s<strong>in</strong>ce it appears that the promoters of someof the developmental regulatory genes are controlled simultaneously by activators andrepressors. This may po<strong>in</strong>t out that ESCs modulate their chromat<strong>in</strong> organization by an unusualand complex mechanism compared to somatic cells.Recent studies <strong>in</strong>dicate that ESCs engage an uncommon and novel mechanism for l<strong>in</strong>eagespecificgene regulation. These genes are normally silent <strong>in</strong> pluripotent stem cells, but may beexpressed upon differentiation. Recently, a new “histone bivalent” model (active and <strong>in</strong>activehistone modifications) has been suggested to expla<strong>in</strong> the mechanism of gene expressionregulation <strong>in</strong> ESCs. Accord<strong>in</strong>g to this model, some l<strong>in</strong>eage-specific genes are marked with bothrepressive and active modifications at the same time. By apply<strong>in</strong>g a quantitative sequentialchromat<strong>in</strong> immune-precipitation (ChIP) approach, Azuara et al. found that several important<strong>in</strong>active developmental genes <strong>in</strong> mouse ESCs such as Pax3, Irx3, Sox1, and Nkx2.2 were


CHAPTER 24Stem Cell <strong>Epigenetics</strong> and <strong>Human</strong> <strong>Disease</strong>simultaneously enriched by repressive modifications (H3K27met3) and activat<strong>in</strong>g marks(H3K4me3 and H3K9ac3) <strong>in</strong> their promoters [44]. Their results also suggested that ESCs,which are defective <strong>in</strong> the Eed-dependent methytransferase enzyme, expressed l<strong>in</strong>eage-specificgenes. They concluded that H3K27 methylation <strong>in</strong> ESCs assists to ma<strong>in</strong>ta<strong>in</strong> pluripotency andsuppress developmental gene expression.Bernste<strong>in</strong> et al. also supported this bivalent model by demonstrat<strong>in</strong>g that promoter regions ofl<strong>in</strong>eage-control genes <strong>in</strong>clud<strong>in</strong>g POU, Pax, Sox, and Hox are dist<strong>in</strong>guished by both suppressive(H3K27me3) and activat<strong>in</strong>g (H3K4me3) marks [45]. They found that these opposite histonemodifications take place at the same chromosomal locations with<strong>in</strong> the same ESC population.Most of these uncommon bivalent patterns of histone changes are erased upon ESC differentiation<strong>in</strong>to neural progenitor cells. Specifically, some of the neural-specific genes cont<strong>in</strong>ueto be expressed (H3K4me3), but lose their repress<strong>in</strong>g (H3K27me) histone modifications. Incontrast, <strong>in</strong>active pluripotent genes preserved the repressor mark (H3K27me3), while los<strong>in</strong>gthe activator (H3K4me3) changes. These f<strong>in</strong>d<strong>in</strong>gs suggest that key developmental regulat<strong>in</strong>ggenes exhibit a “primed or poised status” <strong>in</strong> ESCs as def<strong>in</strong>ed by opposite comb<strong>in</strong>ations ofhistone modifications.Other studies revealed that this <strong>in</strong>terest<strong>in</strong>g histone mark can also be found at later stages <strong>in</strong>development, <strong>in</strong>dicat<strong>in</strong>g that differentiated cells have the capacity to ga<strong>in</strong> some characteristicsof ESCs [51,52]. Fewer bivalent doma<strong>in</strong>s are observed <strong>in</strong> differentiated cells, whereas thenumber of these doma<strong>in</strong>s <strong>in</strong> ESCs is about 2000e3000 genes [53e55]. These studies po<strong>in</strong>tedout that this chromat<strong>in</strong> mark might, at least <strong>in</strong> part, be responsible for the pluripotency ofESCs, s<strong>in</strong>ce these changes down-regulate the expression of genes <strong>in</strong> ESCs due to the dom<strong>in</strong>ant<strong>in</strong>fluence of H3K27me3 over H3K4me3. L<strong>in</strong>eage-specific genes are ma<strong>in</strong>ta<strong>in</strong>ed <strong>in</strong>active bychromat<strong>in</strong> modifications, but may be poised for subsequent <strong>in</strong>duction as ESCs select todifferentiate to three embryonic l<strong>in</strong>eages. This bivalent model supports the idea that ESCpluripotency and self-renewal is preserved by the expression of differentiation genes, which are<strong>in</strong> a dormant but poised state.487Histone demethylases may also play an important role <strong>in</strong> ESC pluripotency and epigenetics.The exact mechanism of histone demethylation <strong>in</strong> ESCs has only recently been revealed[3,56,57]. Several studies have implicated specific H3K27 trimethylation removal by twoenzyme families: UTX and jumonji (JMJD) [51,58,59]. These two groups have the oppositefunctions <strong>in</strong> modify<strong>in</strong>g gene expression with<strong>in</strong> PcG complexes and are crucial <strong>in</strong> earlydevelopment and differentiation. Oct4 can up-regulate Jmjd1a and Jmjd2c genes, which areresponsible for H3K9me2 and H3K9me3 demethylation. While a correspond<strong>in</strong>g decrease <strong>in</strong>l<strong>in</strong>eage-specific gene expression occurs upon depletion of either Jmjd1a or Jmjd2c, an <strong>in</strong>crease<strong>in</strong> the expression of ESC-specific genes happens. Up-regulation of other key pluripotencyfactors, such as Nanog, is also <strong>in</strong>duced by Jmjd2a, whereas Jmjd1a affects the demethylation ofactivat<strong>in</strong>g mark H3K9me2 at the promoter region of Tc11, Zfp57, and Tcfcp211, among others[60]. Predom<strong>in</strong>antly, histone demethylases play a prom<strong>in</strong>ent role <strong>in</strong> self-renewal and pluripotency.They are an <strong>in</strong>separable portion of the ESC transcriptome network regulat<strong>in</strong>g thetranscription circuitry to chromat<strong>in</strong> structure dur<strong>in</strong>g early development, and later with<strong>in</strong>tissue-specific differentiation.24.6 CHANGING THE EPIGENETIC LANDSCAPE DURING CELLULARREPROGRAMMINGIn 2006, a milestone was achieved by artificially convert<strong>in</strong>g mouse embryonic and adultfibroblasts to <strong>in</strong>duced pluripotent stem cells (iPSCs) us<strong>in</strong>g Oct4, Klf4, c-myc, and Sox2 transcriptionfactors [61]. In the subsequent year and <strong>in</strong> two <strong>in</strong>dependent studies led by Thomsonand Yamanaka, the same process reverted adult human somatic cells to ES-like cells by viraltransduction of four def<strong>in</strong>ed pluripotency transcription factor genes [62,63]. Yamanaka’s group


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>used the same cocktail <strong>in</strong> mouse, while Thomson’s group replaced c-Myc and Klf4 with Nanogand L<strong>in</strong>28. These iPSCs are morphologically very similar to that of ESCs; they express cell surfacemarkers characteristic of ESCs, demonstrate multil<strong>in</strong>eage differentiation both <strong>in</strong> vivo and<strong>in</strong> vitro and give rise to live chimeras. S<strong>in</strong>ce 2006, hundreds of studies have tried to derive iPSCsfrom different mouse and human sources us<strong>in</strong>g various approaches [64e71].Although some of the molecular highlights that happen dur<strong>in</strong>g the course of iPSCs derivationhave been elucidated, the mechanism and dynamics of the process by which ectopic expressionof a few transcription factors may change cell dest<strong>in</strong>y rema<strong>in</strong>s to be largely unknown.Several l<strong>in</strong>es of evidence po<strong>in</strong>t out to the role of epigenetic marks dur<strong>in</strong>g iPSC generation andthe concomitant overturn<strong>in</strong>g of cell state [72,73]. It has been suggested that cell fate can bereset through modification of l<strong>in</strong>eage-specific epigenetic marks such as DNA methylation,histone methylation/acetylation, and nucleosome spac<strong>in</strong>g. Experimental data propose thatmouse-derived iPSCs have the ability to ga<strong>in</strong> the chromat<strong>in</strong> signature of pluripotent ESCs andcan transmit these epigenetic patterns to their progenies through the germl<strong>in</strong>e [74].488There are several important questions to be answered: what k<strong>in</strong>ds of chromat<strong>in</strong> remodel<strong>in</strong>g arereally tak<strong>in</strong>g place dur<strong>in</strong>g the immediate expression of reprogramm<strong>in</strong>g factors until theappearance of the first pluripotency markers such as E-Cadher<strong>in</strong>, SSEA1 (mouse), or SSEA3(human) and how do the reprogramm<strong>in</strong>g factors <strong>in</strong>duce the reversion of epigenetic marks?The role of each reprogramm<strong>in</strong>g factor has been <strong>in</strong>vestigated <strong>in</strong> more detail s<strong>in</strong>ce the <strong>in</strong>itialiPSC studies. In differentiated cells, Oct4 and Sox2 cannot f<strong>in</strong>d their target genes, possibly dueto large differences <strong>in</strong> epigenetic status between ESCs and their somatic cell progenies.Although, the application of Oct4 and Sox2 as part of an autoregulatory loop are <strong>in</strong>dispensiblefor iPSC generation, c-Myc and Klf4 are not necessary for reprogramm<strong>in</strong>g, but they significantly<strong>in</strong>crease the efficiency, possibly through <strong>in</strong>teraction with the chromat<strong>in</strong> [75] orsuppression of genes related to differentiation [76]. It has been postulated that c-Myc and Klf4modify the organization of chromat<strong>in</strong> enabl<strong>in</strong>g Oct4 and Sox2 access to their targets, thereby<strong>in</strong>creas<strong>in</strong>g the expression of downstream genes [61]. For <strong>in</strong>stance, c-Myc is a well-characterizedoncogene transcription activator and a modulator of DNA replication. It has been proposedthat c-Myc <strong>in</strong>duces the up-regulation of histone acetyltransferase gene (Gcn5), which is a keyfactor <strong>in</strong> histone structure, there by improv<strong>in</strong>g the accessibility of target genes to Oct4. Klf4 isalso acetylated by p300 (acetyl transferase prote<strong>in</strong>) and has the capacity to control genetranscription through regulation of histone acetylation [77]. These results are <strong>in</strong> agreementwith the open chromat<strong>in</strong> theory of ESC self-renewal and pluripotency.Us<strong>in</strong>g ChIP and cDNA microarray approaches, it is well established that Oct4 regulates theexpression of over 350 genes <strong>in</strong> ESCs <strong>in</strong>clud<strong>in</strong>g several epigenetic regulators [78]. Asmentioned earlier, two histone demethylases, Jmjd1a and Jmjd2c have been identified to bepart of the groups of the genes regulated by Oct4 [60]. This study also demonstrated thatJmdj2c is recruited to the Nanog promoter (an important regulator of the ESC self renewalmach<strong>in</strong>ery) and found that upon depletion of Jmdj2c, subsequent differentiation could onlybe rescued by ectopic expression of Nanog. These results clearly demonstrate that Oct4 bothdirectly and <strong>in</strong>directly regulates the genes necessary for ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g the accessible and openchromat<strong>in</strong> state needed for ESC self-renewal and pluripotency. A positive feedback loopbetween transcriptional circuitry and epigenetic modification has also been found. In this way,ESC transcription factors control the expression of chromat<strong>in</strong> remodel<strong>in</strong>g genes and, <strong>in</strong> turn,assist to open up chromat<strong>in</strong> structure <strong>in</strong> the promoter regions of target genes allow<strong>in</strong>g for selfregulationof the epigenetic network.In 1957, Wadd<strong>in</strong>gton proposed his famous “epigenetic landscape” model by compar<strong>in</strong>g theearly developmental differentiation with a ball travell<strong>in</strong>g downward a canal. This journey startsfrom a fertilized totipotent embryo and ends up as different l<strong>in</strong>eage-committed cells.Accord<strong>in</strong>g to this developmental model, cells <strong>in</strong>side the canal move through various one-waybranched valleys and select their f<strong>in</strong>al irreversible cellular fates dur<strong>in</strong>g this trip [79]. As they


CHAPTER 24Stem Cell <strong>Epigenetics</strong> and <strong>Human</strong> <strong>Disease</strong>reach the end of each valley (cell l<strong>in</strong>eage), they are urged to stay <strong>in</strong> that valley and cannot jumpover boundaries <strong>in</strong>to other branches or return to their start<strong>in</strong>g po<strong>in</strong>t. With the discovery ofcellular reprogramm<strong>in</strong>g, now this trip is not irreversible. The cells can now move back with<strong>in</strong>the valley from somatic cell to pluripotent state or even transdifferentiate from one l<strong>in</strong>eage tothe other without return<strong>in</strong>g back to the pluripotent state [80]. With regard to iPSC generation,it has been postulated that the four reprogramm<strong>in</strong>g transcription factors push cells backward<strong>in</strong> this canal by remov<strong>in</strong>g specific epigenetic barriers, which stabilize cells <strong>in</strong> their differentiatedstatus/valley under normal conditions [81].Recently, based on Wadd<strong>in</strong>gton’s epigenetic model, a stochastic model for iPSC generation hasbeen proposed [81]. This model is built on an opposite concept suggested by Wadd<strong>in</strong>gton.Specially, reprogrammed cells undergo four various events. As they beg<strong>in</strong> their journeybackwards along the slope toward pluripotency, some epigenetic obstacles h<strong>in</strong>der the cellsfrom roll<strong>in</strong>g back and consequently they will achieve self-renewal capability. The second groupof cells will be partially reprogrammed and lose their pluripotency state and differentiate <strong>in</strong>tospecific cell l<strong>in</strong>eage without constant expression of reprogramm<strong>in</strong>g factors. The third categorymay trans-differentiate as a result of <strong>in</strong>sufficient and improper expression of ectopic factors.The fourth group doesn’t even beg<strong>in</strong> the reprogramm<strong>in</strong>g and experience apoptosis or cellularsenescence (Figure 24.2). In agreement with these series of events and based on a stochasticmodel of ESCs, any gene or molecule, with the ability to enhance this movement up the slopeand avert the cell from return<strong>in</strong>g back would facilitate the derivation of iPSCs.The endogenous loci of all reprogramm<strong>in</strong>g factors are excessively methylated <strong>in</strong> somatic cells,though their loci are hypomethylated <strong>in</strong> ESCs and iPSCs [82]. For iPSC derivation, all of thesepromoters require to be reactivated by enzymes responsible for DNA demethylation. Thisevent requires other downstream activated epigenetic modifiers, s<strong>in</strong>ce the factors used <strong>in</strong> directreprogramm<strong>in</strong>g do not have known demethylat<strong>in</strong>g activity. Based on CpG methylationdifferences between l<strong>in</strong>eage-specific cells and ESCs, one group demonstrated an essential rolefor CpG methylation as an epigenetic modifier <strong>in</strong> iPSC generation. Doi et al. found that CpG489FIGURE 24.2The stochastic model proposed by Yamanaka for iPS cell generation is based on the Wadd<strong>in</strong>gton model. The reprogrammedcells may encounter four possible scenarios. One group of cells is blocked by epigenetic barriers and beg<strong>in</strong>s self-renewal(1). Other cells are trapped <strong>in</strong>side a semi-reprogrammed state due to <strong>in</strong>efficient epigenetic modification and travel backdown their valley <strong>in</strong> the absence of ectopic expression of pluripotency factors (2). Some of the cells may move to otherneighbor<strong>in</strong>g valleys and transdifferentiate <strong>in</strong>to other cell types due to <strong>in</strong>efficient expression of reprogramm<strong>in</strong>g factors (3). Thefourth group of the cells experiences apoptosis or cellular senescence (4). This figure is reproduced <strong>in</strong> the color platesection. Source: Adapted from [133]


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>methylation can dist<strong>in</strong>guish iPSCs, ESCs, and fibroblasts. They proposed that epigeneticreprogramm<strong>in</strong>g <strong>in</strong>corporates the same differentially methylated regions (DMRs) of CpGisland “shores” that mark normal differentiation [83]. Specific loci <strong>in</strong> iPSCs rema<strong>in</strong> semireprogrammed,which means that the methylation pattern of iPSCs differs from ESCs.Another obstacle toward iPSC generation is the requirement for histone modification andremodel<strong>in</strong>g dur<strong>in</strong>g reprogramm<strong>in</strong>g. Although, it has been shown that histone H4 with<strong>in</strong> thepromoters of reprogramm<strong>in</strong>g genes is deacetylated <strong>in</strong> somatic cells, it is hyperacetylated <strong>in</strong>iPSCs and ESCs. Several studies <strong>in</strong>dicate that the H3 and H4 histones with<strong>in</strong> the Oct4 andNanog promoters are hyperacetylated [84,85]. Among the four factors, only c-Myc has theability to modify chromat<strong>in</strong>. It mediates chromat<strong>in</strong> modification by <strong>in</strong>creas<strong>in</strong>g the expressionof Gcn5 recruit<strong>in</strong>g it for the modification of target genes [86].Another factor <strong>in</strong> the derivation of iPSCs is the state of histone methylation. ESCs and iPSCsare tagged by activator histone modifications, which are marked by H3K4me3 and demethylationof lys<strong>in</strong>e 9 with<strong>in</strong> pluripotency genes. These two histone modifications show thebivalent chromat<strong>in</strong> characteristics of pluripotent genes, which are accomplished by simultaneousmethylation at H3K27me3 and H3K4me3. For efficient generation of iPSCs, it isrequired that this histone modification be achieved either through genetic approaches or withthe aid of some small molecules.490Reflect<strong>in</strong>g on the role of chromat<strong>in</strong> remodel<strong>in</strong>g dur<strong>in</strong>g reprogramm<strong>in</strong>g, researchers haverecently applied small molecules to circumvent these epigenetic blocks and enhance thegeneration of iPSCs. Several different chemical <strong>in</strong>hibitors for histone deacetylases, as well asDNA and histone methyltransferases, have been used <strong>in</strong> comb<strong>in</strong>ation with genetic factors[87,88] (Table 24.1). Kubicek et al. found a small-molecule <strong>in</strong>hibitor of G9a histone methyltransferase,BIX-01294, could enhance the <strong>in</strong>duction of reprogramm<strong>in</strong>g <strong>in</strong> neural stem cellswhile replac<strong>in</strong>g Oct4 [89]. S<strong>in</strong>ce G9a is a down-regulator of Oct4 dur<strong>in</strong>g early development,they suggested that BIX-01294 enhances iPSC formation by <strong>in</strong>hibit<strong>in</strong>g G9a and subsequentlyreleas<strong>in</strong>g Oct4 from negative regulation. This group also generated iPSCs from mouseembryonic fibroblasts (MEFs) with only two factors; Oct4 and Klf4 <strong>in</strong> the presence of BIX-TABLE 24.1 Chemicals with Epigenetic Modification Properties Used to Enhance iPSCGenerationName Function Effects ReferenceRG-108BIX-01294VPASAHATrichostat<strong>in</strong>-A5-azacytid<strong>in</strong>eDNAmethyltransferase<strong>in</strong>hibitorG9 histonemethyltransferase<strong>in</strong>hibitorHistone deacetylase<strong>in</strong>hibitorHistone deacetylase<strong>in</strong>hibitorHistone deacetylase<strong>in</strong>hibitorMethyltransferase<strong>in</strong>hibitorPromote MEFsreprogramm<strong>in</strong>gReprogramm<strong>in</strong>g ofneural progenitor cellsand MEFsEnhancedreprogramm<strong>in</strong>g ofmouse and humanfibroblastsEnhancesreprogramm<strong>in</strong>g ofmouse fibroblastsEnhancesreprogramm<strong>in</strong>g ofmouse fibroblastsEnhancedreprogramm<strong>in</strong>g ofmouse fibroblasts[134][134,135][134,135][135][135][135,136]


CHAPTER 24Stem Cell <strong>Epigenetics</strong> and <strong>Human</strong> <strong>Disease</strong>01294. Interest<strong>in</strong>gly, valproic acid (VPA) <strong>in</strong>creased the efficiency and k<strong>in</strong>etics of reprogramm<strong>in</strong>gby 100-fold <strong>in</strong> a four-factor system. VPA could also replace either Klf4 or c-Myc <strong>in</strong>reprogramm<strong>in</strong>g and enabled iPSC generation with only Oct4 and Sox2 from human fibroblasts[90]. These results imply that DNA methylation, histone methylation, and histonedeacetylation contribute to epigenetic hurdles, which have to be overcome for successful iPSCgeneration.24.7 STEM CELL EPIGENETICS AND HUMAN DISEASEStudies <strong>in</strong> the past two decades have revealed a great deal about the basis of known epigeneticdefects caus<strong>in</strong>g disease. There are two major categories of disease-related epigenetic defects:histone modification and DNA methylation. Enzymes such as histone deacetylases (HDACs),histone methyltransferases, histone acetylases, DNA methyltransferases, and methyl-CpGb<strong>in</strong>d<strong>in</strong>g doma<strong>in</strong> prote<strong>in</strong> (MECP2) are among the best-known epigenetic modifiers prone toabnormality. Any variation <strong>in</strong> these epigenetic apparatus may alter gene expression, which <strong>in</strong>turn has enormous and critical cl<strong>in</strong>ical consequences. One of the best-known epigenetic causesof disease is the loss of DNA methylation patterns. Alterations <strong>in</strong> DNA methylation have beenobserved <strong>in</strong> various disorders <strong>in</strong>volv<strong>in</strong>g genomic impr<strong>in</strong>t<strong>in</strong>g, X chromosome <strong>in</strong>activation, andcancer.Cancer comprises a multistep process where<strong>in</strong> both genetic and epigenetic abnormalities worktogether to transform a normal cell <strong>in</strong>to an abnormal malignant tumor cell. Cancer is generallycharacterized by global hypomethylation and gene-specific hypermethylation [91]. ModifiedDNA methylation <strong>in</strong> a cell alters the expression of cancer-related genes. DNA hypermethylationcommences the down-regulation of tumor-suppressor genes, whereas DNAhypomethylation activates proto-oncogenes and promotes cell <strong>in</strong>stability via chromosomalaberrations [92]. DNA methylation patterns are created and preserved by a group of enzymescalled DNMTs, which are important for appropriate gene expression. Knock<strong>in</strong>g out of DNMTgenes <strong>in</strong> animal models is detrimental and the overexpression of these enzymes <strong>in</strong> human maylead to various k<strong>in</strong>ds of cancers. In 1983, hypomethylation patterns were found to dist<strong>in</strong>guishhuman cancer cells from their normal counterparts [93] and now there is a grow<strong>in</strong>g body ofevidence that hypomethylation of certa<strong>in</strong> proto-oncogenes, such as HRAS and cycl<strong>in</strong> D2, causetumor formation. To the contrary, it has been revealed that hypermethylation of promoterregions of tumor suppressors such as RB, P16, VHL, APC, and E-Cadher<strong>in</strong> <strong>in</strong>duce malignancyand are mostly found <strong>in</strong> sporadic cancer [91,94e96].491Recent studies have shed light over the role of stem cell chromat<strong>in</strong> marks <strong>in</strong> cancer development.They report that there is an epigenetic stem cell signature <strong>in</strong> cancer and this stem-cell-likechromat<strong>in</strong> pattern makes the tumor-suppressor genes prone to DNA hypermethylation andsubsequent silenc<strong>in</strong>g [97,98]. It is <strong>in</strong>terest<strong>in</strong>g that several numbers of these tumor-related genesare associated with the bivalent chromat<strong>in</strong> state <strong>in</strong> ESCs. For example, two suppressive marks<strong>in</strong>clud<strong>in</strong>g H3K9me3 and H3K9me2 are l<strong>in</strong>ked to DNA hypermethylation <strong>in</strong> cancer [98]. Also,methylation of H3K27 by polycomb prote<strong>in</strong>, along with the previously mentioned repressivemarks, targets some genes for de novo methylation by methyltransferases <strong>in</strong> cancer [99].Understand<strong>in</strong>g the exact epigenetic mechanism govern<strong>in</strong>g cancer cells can have significanttherapeutic consequences. For <strong>in</strong>stance, it has been revealed that any modification <strong>in</strong> chromat<strong>in</strong>organization can affect normal development and cellular tumorigenic transformation[100]. These observations have resulted <strong>in</strong> the development of new drugs such as HDAC andDNMT <strong>in</strong>hibitors for the treatment of different cancers [101,102]. Elucidation of chromat<strong>in</strong>defects, which transform a normal cell to a malignant tumor, may lead to the development ofnew designs for cancer treatment as well as its early diagnosis.Recent studies have evaluated the role of epigenetic defects <strong>in</strong> the onset of various pediatric andadult neurodegenerative disorders [103] (Table 24.2). Some mood disorders such as


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>TABLE 24.2 Relationship Between <strong>Human</strong> <strong>Disease</strong>s and Epigenetic Modifications<strong>Disease</strong> Gene Function ReferenceNeurodegenerative DisordersSchizophrenia RELN Hypermethylation [137]Bipolar disorders, memory RELN Hypermethylation [138]formationPsychosis DNMT1 Overexpression [137]Alzheimer’s diseases Presenil<strong>in</strong> I Hypomethylation [139]Impr<strong>in</strong>t<strong>in</strong>g <strong>Disease</strong>sBWS 11p15 Impr<strong>in</strong>t<strong>in</strong>g [140]ICF syndrome DNMT3B Mutation [141]Rett syndrome MECP2 Mutation [142]ATRX ATRX Chromat<strong>in</strong> conformation [143]PWS/AS 15q11-q13 Impr<strong>in</strong>t<strong>in</strong>g [144]FraX FMR1 Gene suppression [145]492CancerBra<strong>in</strong> MGMT Hypermethylation [416]Colon Different genes Hypermethylation [147]Esophagus CDH1 Hypermethylation [147]Kidney TIMP-3 Hypermethylation [147]Ovary SAT2 Hypomethylation [148]Pancreas APC Hypermethylation [147]Prostate BRCA2 Hypermethylation [149]Uterus hMLH1 Hypermethylation [147]Ag<strong>in</strong>gDNA and Chromat<strong>in</strong> Hypo-,[150]hypermethylationImmunity and Related DisordersLupus LFAr Hypomethylation [151]schizophrenia have been reported to be associated with DNA methyltransferase gene mutations.For example, the overexpression of DNMTI has been found <strong>in</strong> the gamma-am<strong>in</strong>obutyricacid (GABA)-ergic neurons of schizophrenic <strong>in</strong>dividuals. This overexpression <strong>in</strong>hibits theactivity of Reel<strong>in</strong> prote<strong>in</strong> <strong>in</strong> patients with bipolar disorders and psychosis, which is normallyneeded for proper memory composition and normal neurotransmission [104,105]. Furthermore,it has been suggested that valproic acid, an HDAC <strong>in</strong>hibitor, corrects the schizophrenialikebehavior of a mouse model of schizophrenia by prevent<strong>in</strong>g hypermethylation of Reel<strong>in</strong>promoter [106].New f<strong>in</strong>d<strong>in</strong>gs <strong>in</strong> Alzheimer’s disease (AD) have shed light over the connection betweenmethylation pattern <strong>in</strong> specific genes and neural survival, as well as memory loss. Some studiesrevealed the connection between the hypomethylation of presenil<strong>in</strong> I gene and its upregulationand beta-amylois production. Also, a role for folate-mediated methylation <strong>in</strong>Alzheimer’s disease has been suggested [107,108].Recently, a connection between genetic/epigenetic changes and autism has been proposed[109]. Autism is a disease of neural development <strong>in</strong> the bra<strong>in</strong> which impairs an <strong>in</strong>dividual’snormal development of social and communication <strong>in</strong>teractions. The ma<strong>in</strong> defect <strong>in</strong>volved <strong>in</strong>the onset of autism is known to be mutation or changes <strong>in</strong> the activity of “methyl CpG b<strong>in</strong>d<strong>in</strong>gprote<strong>in</strong> 2” (MECP2), which is believed to be essential for normal bra<strong>in</strong> development. MECP2is an X-l<strong>in</strong>ked gene and can b<strong>in</strong>d specifically to the region of methylated DNA. It is also


CHAPTER 24Stem Cell <strong>Epigenetics</strong> and <strong>Human</strong> <strong>Disease</strong>capable of repress<strong>in</strong>g transcription from methylated gene promoters. Once bound, MeCP2forms a complex with histone deacetylases (HDAC) to condense chromat<strong>in</strong> structure. If thegene undergoes any defect, cells do not have enough MeCP2 prote<strong>in</strong> for silenc<strong>in</strong>g other genes.Interest<strong>in</strong>gly, mutations <strong>in</strong> the MECP2 gene cause other neurodevelopmental disorders<strong>in</strong>clud<strong>in</strong>g Rett syndrome, some cases of X-l<strong>in</strong>ked mental retardation, and systemic lupuserythematosus. Based upon all of these f<strong>in</strong>d<strong>in</strong>gs, there may be a potential future role forepigenetics <strong>in</strong> the diagnosis and treatment of neurological diseases.24.8 MODELING OF HUMAN EPIGENETIC DISORDERSUSING IPSCSStudies on the mechanism and pathophysiology of human diseases have been impeded by thelack of suitable <strong>in</strong> vitro practical models and the <strong>in</strong>ability to obta<strong>in</strong> sufficient numbers ofprimary cells. iPSCs may provide potent models of human genetic and multifactoral diseasesdue to their enormous similarities to ESCs. Reprogrammed cells can be differentiated todesired cell types us<strong>in</strong>g established protocols for hESCs. With this tool, the molecularmechanism of disease, as well as drug and therapeutic screen<strong>in</strong>g, can be performed us<strong>in</strong>gderived affected cells. So far, iPSCs have been generated from patients with different types ofdiseases <strong>in</strong>clud<strong>in</strong>g neurological and neurodegenerative disorders (HD, PD, AD, ALS, SMA, Rettsyndrome), muscular dystrophy (DMD, BMD), hematopoietic disorders (beta-thalassemia,sickle cell anemia, Fanconi anemia), cardiovascular diseases (long QT, LEOPARD syndrome),diabetes, Down syndrome, and dyskeratosis congenita (DC) [110]. In this review, we will onlyfocus on diseases with an epigenetic component.Autism spectrum disorders (ASDs) are complex neurodevelopmental disorders where<strong>in</strong> thephenotype of patients is the result of various comb<strong>in</strong>ations of genetic mutations. Rettsyndrome (RTT) is an X-l<strong>in</strong>ked neurodevelopment disorder caused by a heterozygous mutation<strong>in</strong> the MECP2 gene, which can arise sporadically or from germl<strong>in</strong>e mutations. It affectsfemales more than males. In about 10% of cases, mutation <strong>in</strong> the genes FOXG1 or CDKL5 havealso been found to cause the syndrome. The mutation affects postnatal neural developmentand results <strong>in</strong> communication defects and mental retardation <strong>in</strong> affected children. Asmentioned earlier, with the mutant gene, cells do not have enough MeCP2 prote<strong>in</strong> for transcriptionalsilenc<strong>in</strong>g or activat<strong>in</strong>g of other genes.493For the first time, fibroblasts of an 8-year-old girl with R306C missense mutation <strong>in</strong> MECP2have been reprogrammed us<strong>in</strong>g four retroviral factors <strong>in</strong>clud<strong>in</strong>g Oct4, Sox2, Klf4, and c-Myc[111]. The pluripotency of derived cells was confirmed by <strong>in</strong> vivo teratoma assay and <strong>in</strong> vitrodifferentiation through EB formation. The authors could differentiate Rett-derived iPSCs toneurons, but further characterization was not reported. In the other study, iPSCs generatedfrom a Rett syndrome patient <strong>in</strong>duced neural differentiation. Differentiated cells demonstratedabnormalities <strong>in</strong> neural characteristics <strong>in</strong>clud<strong>in</strong>g smaller size, modified calcium signal<strong>in</strong>g,dim<strong>in</strong>ished synapse, and electrophysiological abnormalities [112].Recently, MECP2-null Rett syndrome hiPSCs and isogenic controls have been derived throughX-chromosome <strong>in</strong>activation. Phenotypic evaluation of mutant RTT-hiPSC-derived neuronsrevealed a reduction <strong>in</strong> soma size compared with the isogenic control neurons from the samepatient [113]. Such state-of-the-art cell characterization is go<strong>in</strong>g to open a new era <strong>in</strong> molecularmedic<strong>in</strong>e for reveal<strong>in</strong>g mechanisms of disease and new approaches for drug screen<strong>in</strong>g.iPSCs have also been recently derived from the genomic impr<strong>in</strong>t<strong>in</strong>g diseases PradereWilli andAngelman syndromes. Genomic impr<strong>in</strong>t<strong>in</strong>g is a genetic event by which particular gene locibecome transcribed <strong>in</strong> a parent-of-orig<strong>in</strong>-determ<strong>in</strong>ed way. This means that the phenotypetriggered from a certa<strong>in</strong> locus is differentially altered by the sex of the parent provid<strong>in</strong>g thatspecific allele. Genomic impr<strong>in</strong>t<strong>in</strong>g is an epigenetic event that applies histone and DNAmethylation patterns <strong>in</strong> order to acquire monoallelic gene transcription without alter<strong>in</strong>g the


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>DNA sequence. These epigenetic hallmarks are placed dur<strong>in</strong>g germl<strong>in</strong>e establishment and arepreserved throughout the lifetime of an organism. Proper expression of impr<strong>in</strong>ted genes iscrucial for normal development. Multiple genetic diseases are associated with defects <strong>in</strong>impr<strong>in</strong>t<strong>in</strong>g loci such as Angelman and PradereWilli syndromes, BeckwitheWiedemannsyndrome and SilvereRussell syndrome [114]. Among these disorders, iPSCs have been onlyderived from dermal fibroblasts of patients with Angelman syndrome (AS) and PradereWillisyndrome (PWS) [115,116]. AS is a complex neurogenetic disorder which <strong>in</strong>itially affects thenervous system. It is characterized by speech impairment, frequent seizures, <strong>in</strong>tellectualdisability, and ataxia and affected children have typically a happy and excitable demeanor.PWS is characterized by speech delay, obesity, small stature, and behavior problems. AS isusually caused by deletion or <strong>in</strong>activation of genes on chromosome 15q11-q13 region; thisregion is <strong>in</strong>herited from the mother while the paternal allele is impr<strong>in</strong>ted and hence silenced.Many symptoms of AS result from the loss of function of E3 ubiquit<strong>in</strong> ligase UBE3. PWS iscaused by the deletion of paternally expressed genes on the chromosome 15q11-q13, but itsgenetic basis is largely unknown [117,118].Fibroblasts from AS and PWS patients were reprogrammed us<strong>in</strong>g retroviral vectors encod<strong>in</strong>gOct4, Sox2, Klf4, c-Myc, and LIN28 transcription factors. iPSCs derived from AS and PWSpatients were <strong>in</strong>duced to differentiate to functional neurons [115]. It is <strong>in</strong>terest<strong>in</strong>g that theiPSCs showed no sign of DNA methylation removal at the impr<strong>in</strong>t<strong>in</strong>g center (IC) of chromosome15. Furthermore, <strong>in</strong> normal bra<strong>in</strong> neurons, the impr<strong>in</strong>t<strong>in</strong>g of UBE3A is establisheddur<strong>in</strong>g neural differentiation of AS iPSCs. This study highlights the importance of the iPSCmodel of epigenetic disorders <strong>in</strong> elucidat<strong>in</strong>g the disease mechanism and developmentaltim<strong>in</strong>g of gene activation or silenc<strong>in</strong>g <strong>in</strong> the affected cells of patients.494There are grow<strong>in</strong>g bodies of evidence that show a relationship between premature ag<strong>in</strong>g andadult stem cell malfunction. Ag<strong>in</strong>g is an exceed<strong>in</strong>gly complex trend whose molecular mechanismis still ma<strong>in</strong>ly unknown. Del<strong>in</strong>eation of many molecular aspects of ag<strong>in</strong>g has beenfacilitated by <strong>in</strong>vestigations on premature ag<strong>in</strong>g syndromes [119]. The premature-age<strong>in</strong>gdisease Hutch<strong>in</strong>soneGilford progeria syndrome (HGPS) is one of the rare fatal human geneticdisorders that the role of both epigenetic changes and stem cell misregulation have beenelucidated. HGPS is caused by a s<strong>in</strong>gle po<strong>in</strong>t mutation <strong>in</strong> the LMNA gene result<strong>in</strong>g <strong>in</strong>constitutive production of proger<strong>in</strong>, a truncated splic<strong>in</strong>g mutant form of the nuclear structuralprote<strong>in</strong> lam<strong>in</strong> A [120]. Accumulation of proger<strong>in</strong> <strong>in</strong> several tissues leads to diverse ag<strong>in</strong>grelatednuclear defects such as structural disorganization of nuclear lam<strong>in</strong>a and function of thenucleus and chromat<strong>in</strong> [121]. It also causes epigenetic changes by histone modifications and<strong>in</strong>creased DNA damage [122].Interest<strong>in</strong>gly, recent studies demonstrated that Lam<strong>in</strong> A-dependent dysfunction of adult stemcells is associated with accelerated ag<strong>in</strong>g <strong>in</strong> humans [123]. The authors could provide evidencethat the <strong>in</strong>duction of proger<strong>in</strong> h<strong>in</strong>ders the proper function of human mesenchymal stem cells(hMSCs) by activat<strong>in</strong>g downstream effector prote<strong>in</strong>s of the Notch signal<strong>in</strong>g pathway andmodified cellular characteristics and differentiation capacity. These f<strong>in</strong>d<strong>in</strong>gs are <strong>in</strong>terest<strong>in</strong>gs<strong>in</strong>ce the Notch signal<strong>in</strong>g pathway regulates stem cell differentiation [124] and many of theaffected tissues <strong>in</strong> HGPS patients are of mesenchymal orig<strong>in</strong> [125]. Alternatively, there isa possibility that the Notch effector genes are regulated by their direct association with lam<strong>in</strong>aor misregulation of the pathway is due to the epigenetic alterations normally observed <strong>in</strong>normal ag<strong>in</strong>g and specifically <strong>in</strong> HGPS patients [126]. Malfunction of adult stem cells may alsohave implications for the normal ag<strong>in</strong>g process s<strong>in</strong>ce the proger<strong>in</strong> is present at very low levels<strong>in</strong> cells from normal <strong>in</strong>dividuals [126].In another study, Zmpste24-null progeroid mice (with nuclear lam<strong>in</strong>a defects and acceleratedag<strong>in</strong>g) were evaluated for the number and functional capacity of stem cells [127]. The authorsused telogen hair follicles, which conta<strong>in</strong> multipotent stem cells of both epidermal and neuralorig<strong>in</strong>. They showed that Zmpste24 deficiency causes a change <strong>in</strong> the number and proliferative


CHAPTER 24Stem Cell <strong>Epigenetics</strong> and <strong>Human</strong> <strong>Disease</strong>capacity of epidermal stem cells characteristic with nuclear architecture disorganization and<strong>in</strong>creased apoptosis. These changes are associated with some signal<strong>in</strong>g pathways such as Wntand microphthalmia transcription factor. These results confirmed the existence of a relationshipbetween stem cell misregulation and age-related nuclear envelope deformity.Recently, several studies <strong>in</strong>vestigated the possibility of reprogramm<strong>in</strong>g cells from prematureag<strong>in</strong>g Hutch<strong>in</strong>soneGilford progeria syndrome to the iPSC state [128,129]. Adult dermalfibroblasts from HGPS patients were transfected with Oct4, Sox2, Klf4, and c-Myc. HGPSiPSCsshow absence of proger<strong>in</strong>, and lack the nuclear envelope and epigenetic alterations thatare normally associated with premature ag<strong>in</strong>g. Furthermore, HGPS-iPSCs revealed vascularsmooth muscle and mesenchymal stem cell defects. These studies clearly provide an <strong>in</strong> vitroiPSC-based model to study the pathogenesis of human premature and physiological vascularag<strong>in</strong>g.24.9 FUTURE STUDIESEarly development and ESC differentiation necessitate complex <strong>in</strong>teractions between varioustranscription factors and epigenetic regulators such as histone-modify<strong>in</strong>g enzymes andchromat<strong>in</strong>-remodel<strong>in</strong>g prote<strong>in</strong>s. Based on what is currently known about this <strong>in</strong>teraction, itseems that chromat<strong>in</strong> dynamics is one of the most critical determ<strong>in</strong>ants of ESC pluripotencyand self-renewal. iPSC derivation has triggered a paradigm shift <strong>in</strong> developmental biology.However, this revolutionary discovery has raised several discussions regard<strong>in</strong>g the exactmechanism of reprogramm<strong>in</strong>g and the function of epigenetic changes. At the present time,there is uncerta<strong>in</strong>ty about the epigenetic status of cells dur<strong>in</strong>g the iPSC procedure. For <strong>in</strong>stance,do the iPSCs have any ‘‘memory’’ of their orig<strong>in</strong>al epigenetic state? If so, is this memoryfavorable or disadvantageous for their future cl<strong>in</strong>ical applications? Recently, several studieshave demonstrated the existence of an epigenetic memory dur<strong>in</strong>g iPSC generation. It wasrevealed that reprogramm<strong>in</strong>g leaves an epigenetic memory of the tissue of orig<strong>in</strong> which mayaffect their differentiation and application <strong>in</strong> disease model<strong>in</strong>g [130,131].495Advancements <strong>in</strong> understand<strong>in</strong>g the role of epigenetic obstacles will def<strong>in</strong>itely move this fieldforward, establish<strong>in</strong>g straightforward and more efficient methods. Furthermore, iPSCs mayprovide an important model of human genetic and multifactorial disorders. Specially, with thelack of a proper <strong>in</strong> vitro cellular models of epigenetic diseases to study the mechanism, thefield will move toward the establishment of iPSCs from different epigenetic disorders such asBeckwitheWiedemann syndrome, SilvereRussell syndrome, Albright hereditary osteodystrophy,pseudohypoparathyroidism type IA (PHPIA), immunodeficiency/centromeric <strong>in</strong>stability/facialanomalies (ICF), alpha-thalassemia/mental retardation, X-l<strong>in</strong>ked (ATRX),Rub<strong>in</strong>ste<strong>in</strong>-Taybi syndrome, asthma, multiple sclerosis, obesity, and different types of cancercells. A common characteristic of these disorders is that mutations <strong>in</strong> the components ofchromat<strong>in</strong> regulators and epigenetic mach<strong>in</strong>ery cause the pathophysiological symptoms. Asthe mechanisms of human epigenetic disorders are recognized (for example through diseasederivediPSCs model<strong>in</strong>g), there will be even more developments <strong>in</strong> epigenetic therapies.Because epigenetic changes are the key factors <strong>in</strong> human health and disease, there is hope thatunderstand<strong>in</strong>g the mechanism of epigenome regulation will aid <strong>in</strong> the treatment of humansickness that may ultimately be beneficial for the health and wellbe<strong>in</strong>g of mank<strong>in</strong>d. F<strong>in</strong>ally, ifwe can modify the epigenetic status of adult somatic cells toward pluripotency without<strong>in</strong>terven<strong>in</strong>g <strong>in</strong> their genetic <strong>in</strong>tegrity, we will be one step closer toward the cl<strong>in</strong>ical applicationof these cells <strong>in</strong> the near future.References[1] Luger K, Mader AW, Richmond RK, Sargent DF, Richmond TJ. Crystal structure of the nucleosome core particleat 2.8 A resolution. Nature 1997;389:251e60.[2] Wadd<strong>in</strong>gton CH. L’epigenotype. Endeavour 1942;1:18e20.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>496[3] Kouzarides T. Chromat<strong>in</strong> modifications and their function. Cell 2007;128:693e705.[4] Jenuwe<strong>in</strong> T, Allis CD. Translat<strong>in</strong>g the histone code. <strong>Science</strong> 2001;293:1074e80.[5] Horn PJ, Peterson CL. Heterochromat<strong>in</strong> assembly: a new twist on an old model. Chromosome Res2006;14:83e94.[6] Grewal SI, Jia S. Heterochromat<strong>in</strong> revisited. Nat Rev Genet 2007;8:35e46.[7] Yasuhara JC, Wakimoto BT. Oxymoron no more: the expand<strong>in</strong>g world of heterochromatic genes. Trends Genet2006;22:330e8.[8] Smith CD, Shu S, Mungall CJ, Karpen GH. The Release 5.1 annotation of Drosophila melanogaster heterochromat<strong>in</strong>.<strong>Science</strong> 2007;316:1586e91.[9] Pezer Z, Ugarkovic D. RNA Pol II promotes transcription of centromeric satellite DNA <strong>in</strong> beetles. PLoS One2008:3:e1594.[10] Li F, Sonbuchner L, Kyes SA, Epp C, Deitsch KW. Nuclear non-cod<strong>in</strong>g RNAs are transcribed from thecentromeres of Plasmodium falciparum and are associated with centromeric chromat<strong>in</strong>. J Biol Chem2008;283:5692e8.[11] Reik W. Stability and flexibility of epigenetic gene regulation <strong>in</strong> mammalian development. Nature2007;447:425e32.[12] Lusser A, Kadonaga JT. Chromat<strong>in</strong> remodel<strong>in</strong>g by ATP-dependent molecular mach<strong>in</strong>es. Bioessays2003;25:1192e200.[13] Iizuka M, Smith MM. Functional consequences of histone modifications. Curr Op<strong>in</strong> Genet Dev2003;13:154e60.[14] Ahmad K, Henikoff S. Histone H3 variants specify modes of chromat<strong>in</strong> assembly. Proc Natl Acad Sci USA2002;99(Suppl. 4):16477e84.[15] Bird AP, Wolffe AP. Methylation-<strong>in</strong>duced repressionebelts, braces, and chromat<strong>in</strong>. Cell 1999;99:451e4.[16] Almeida R, Allshire RC. RNA silenc<strong>in</strong>g and genome regulation. Trends Cell Biol 2005;15:251e8.[17] Bartel DP. MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell 2004;116:281e97.[18] Ramsahoye BH, B<strong>in</strong>iszkiewicz D, Lyko F, Clark V, Bird AP, Jaenisch R. Non-CpG methylation is prevalent <strong>in</strong>embryonic stem cells and may be mediated by DNA methyltransferase 3a. Proc Natl Acad Sci USA2000;97:5237e42.[19] Weber M, Hellmann I, Stadler MB, Ramos L, Paabo S, Rebhan M, et al. Distribution, silenc<strong>in</strong>g potential andevolutionary impact of promoter DNA methylation <strong>in</strong> the human genome. Nat Genet 2007;39:457e66.[20] Suzuki Y, Tsunoda T, Sese J, Taira H, Mizushima-Sugano J, Hata H, et al. Identification and characterization ofthe potential promoter regions of 1031 k<strong>in</strong>ds of human genes. Genome Res 2001;11:677e84.[21] Hendrich B, Tweedie S. The methyl-CpG b<strong>in</strong>d<strong>in</strong>g doma<strong>in</strong> and the evolv<strong>in</strong>g role of DNA methylation <strong>in</strong>animals. Trends Genet 2003;19:269e77.[22] Fatemi M, Hermann A, Pradhan S, Jeltsch A. The activity of the mur<strong>in</strong>e DNA methyltransferase Dnmt1 iscontrolled by <strong>in</strong>teraction of the catalytic doma<strong>in</strong> with the N-term<strong>in</strong>al part of the enzyme lead<strong>in</strong>g to anallosteric activation of the enzyme after b<strong>in</strong>d<strong>in</strong>g to methylated DNA. J Mol Biol 2001;309:1189e99.[23] Bestor T, Laudano A, Mattaliano R, Ingram V. Clon<strong>in</strong>g and sequenc<strong>in</strong>g of a cDNA encod<strong>in</strong>g DNA methyltransferaseof mouse cells. The carboxyl-term<strong>in</strong>al doma<strong>in</strong> of the mammalian enzymes is related to bacterialrestriction methyltransferases. J Mol Biol 1988;203:971e83.[24] Probst AV, Dunleavy E, Almouzni G. Epigenetic <strong>in</strong>heritance dur<strong>in</strong>g the cell cycle. Nat Rev Mol Cell Bio2009;10:192e206.[25] Reik W, Dean W, Walter J. Epigenetic reprogramm<strong>in</strong>g <strong>in</strong> mammalian development. <strong>Science</strong> 2001;293:1089e93.[26] Jackson M, Krassowska A, Gilbert N, Chevassut T, Forrester L, Ansell J, et al. Severe global DNA hypomethylationblocks differentiation and <strong>in</strong>duces histone hyperacetylation <strong>in</strong> embryonic stem cells. Mol CellBiol 2004;24:8862e71.[27] Deb-R<strong>in</strong>ker P, Ly D, Jezierski A, Sikorska M, Walker PR. Sequential DNA methylation of the Nanog and Oct-4upstream regions <strong>in</strong> human NT2 cells dur<strong>in</strong>g neuronal differentiation. J Biol Chem 2005;280:6257e60.[28] Do JT, Han DW, Scholer HR. Reprogramm<strong>in</strong>g somatic gene activity by fusion with pluripotent cells. Stem CellRev 2006;2:257e64.[29] Boyer LA, Mathur D, Jaenisch R. Molecular control of pluripotency. Curr Op<strong>in</strong> Genet Dev 2006;16:455e62.[30] Kashyap V, Rezende NC, Scotland KB, Shaffer SM, Persson JL, Gudas LJ, et al. Regulation of stem cell pluripotencyand differentiation <strong>in</strong>volves a mutual regulatory circuit of the NANOG, OCT4, and SOX2 pluripotencytranscription factors with polycomb repressive complexes and stem cell microRNAs. Stem Cells Dev2009;18:1093e108.[31] Wibl<strong>in</strong> AE, Cui W, Clark AJ, Bickmore WA. Dist<strong>in</strong>ctive nuclear organisation of centromeres and regions<strong>in</strong>volved <strong>in</strong> pluripotency <strong>in</strong> human embryonic stem cells. J Cell Sci 2005;118:3861e8.


CHAPTER 24Stem Cell <strong>Epigenetics</strong> and <strong>Human</strong> <strong>Disease</strong>[32] Williams RR, Azuara V, Perry P, Sauer S, Dvork<strong>in</strong>a M, Jorgensen H, et al. Neural <strong>in</strong>duction promotes large-scalechromat<strong>in</strong> reorganisation of the Mash1 locus. J Cell Sci 2006;119:132e40.[33] Meshorer E, Misteli T. Chromat<strong>in</strong> <strong>in</strong> pluripotent embryonic stem cells and differentiation. Nat Rev Mol CellBiol 2006;7:540e6.[34] Meshorer E, Yellajoshula D, George E, Scambler PJ, Brown DT, Misteli T. Hyperdynamic plasticity of chromat<strong>in</strong>prote<strong>in</strong>s <strong>in</strong> pluripotent embryonic stem cells. Dev Cell 2006;10:105e16.[35] Csankovszki G, Pann<strong>in</strong>g B, Bates B, Pehrson JR, Jaenisch R. Conditional deletion of Xist disrupts histonemacroH2A localization but not ma<strong>in</strong>tenance of X <strong>in</strong>activation. Nat Genet 1999;22:323e4.[36] Angelov D, Molla A, Perche PY, Hans F, Cote J, Khochb<strong>in</strong> S, et al. The histone variant macroH2A <strong>in</strong>terferes withtranscription factor b<strong>in</strong>d<strong>in</strong>g and SWI/SNF nucleosome remodel<strong>in</strong>g. Mol Cell 2003;11:1033e41.[37] Menegh<strong>in</strong>i MD, Wu M, Madhani HD. Conserved histone variant H2A.Z protects euchromat<strong>in</strong> from the ectopicspread of silent heterochromat<strong>in</strong>. Cell 2003;112:725e36.[38] Hatch CL, Bonner WM. An upstream region of the H2AZ gene promoter modulates promoter activity <strong>in</strong>different cell types. Biochim Biophys Acta 1996;1305:59e62.[39] Squatrito M, Gorr<strong>in</strong>i C, Amati B. Tip60 <strong>in</strong> DNA damage response and growth control: many tricks <strong>in</strong> one HAT.Trends Cell Biol 2006;16:433e42.[40] Ng RK, Gurdon JB. Epigenetic memory of an active gene state depends on histone H3.3 <strong>in</strong>corporation <strong>in</strong>tochromat<strong>in</strong> <strong>in</strong> the absence of transcription. Nat Cell Biol 2008;10:102e9.[41] Schuettengruber B, Chourrout D, Vervoort M, Leblanc B, Cavalli G. Genome regulation by polycomb andtrithorax prote<strong>in</strong>s. Cell 2007;128:735e45.[42] Kirmizis A, Bartley SM, Kuzmichev A, Margueron R, Re<strong>in</strong>berg D, Green R, et al. Silenc<strong>in</strong>g of human polycombtarget genes is associated with methylation of histone H3 Lys 27. Genes Dev 2004;18:1592e605.[43] Wang H, Wang L, Erdjument-Bromage H, Vidal M, Tempst P, Jones RS, et al. Role of histone H2A ubiquit<strong>in</strong>ation<strong>in</strong> Polycomb silenc<strong>in</strong>g. Nature 2004;431:873e8.[44] Azuara V, Perry P, Sauer S, Spivakov M, Jorgensen HF, John RM, et al. Chromat<strong>in</strong> signatures of pluripotent celll<strong>in</strong>es. Nat Cell Biol 2006;8:532e8.[45] Bernste<strong>in</strong> BE, Mikkelsen TS, Xie X, Kamal M, Huebert DJ, Cuff J, et al. A bivalent chromat<strong>in</strong> structure marks keydevelopmental genes <strong>in</strong> embryonic stem cells. Cell 2006;125:315e26.[46] Jorgensen HF, Giadrossi S, Casanova M, Endoh M, Koseki H, Brockdorff N, et al. Stem cells primed for action:polycomb repressive complexes restra<strong>in</strong> the expression of l<strong>in</strong>eage-specific regulators <strong>in</strong> embryonic stem cells.Cell Cycle 2006;5:1411e4.[47] O’Carroll D, Erhardt S, Pagani M, Barton SC, Surani MA, Jenuwe<strong>in</strong> T. The polycomb-group gene Ezh2 isrequired for early mouse development. Mol Cell Biol 2001;21:4330e6.[48] Chamberla<strong>in</strong> SJ, Yee D, Magnuson T. Polycomb repressive complex 2 is dispensable for ma<strong>in</strong>tenance ofembryonic stem cell pluripotency. Stem Cells 2008;26:1496e505.[49] Lee TI, Jenner RG, Boyer LA, Guenther MG, Lev<strong>in</strong>e SS, Kumar RM, et al. Control of developmental regulatorsby Polycomb <strong>in</strong> human embryonic stem cells. Cell 2006;125:301e13.[50] Boyer LA, Plath K, Zeitl<strong>in</strong>ger J, Brambr<strong>in</strong>k T, Medeiros LA, Lee TI, et al. Polycomb complexes repress developmentalregulators <strong>in</strong> mur<strong>in</strong>e embryonic stem cells. Nature 2006;441:349e53.[51] De Santa F, Totaro MG, Prosper<strong>in</strong>i E, Notarbartolo S, Testa G, Natoli G. The histone H3 lys<strong>in</strong>e-27 demethylaseJmjd3 l<strong>in</strong>ks <strong>in</strong>flammation to <strong>in</strong>hibition of polycomb-mediated gene silenc<strong>in</strong>g. Cell 2007;130:1083e94.[52] Roh TY, Cuddapah S, Cui K, Zhao K. The genomic landscape of histone modifications <strong>in</strong> human T cells. ProcNatl Acad Sci USA 2006;103:15782e7.[53] Mikkelsen TS, Ku M, Jaffe DB, Issac B, Lieberman E, Giannoukos G, et al. Genome-wide maps of chromat<strong>in</strong>state <strong>in</strong> pluripotent and l<strong>in</strong>eage-committed cells. Nature 2007;448:553e60.[54] Pan G, Tian S, Nie J, Yang C, Ruotti V, Wei H, et al. Whole-genome analysis of histone H3 lys<strong>in</strong>e 4 and lys<strong>in</strong>e27 methylation <strong>in</strong> human embryonic stem cells. Cell Stem Cell 2007;1:299e312.[55] Barski A, Cuddapah S, Cui K, Roh TY, Schones DE, Wang Z, et al. High-resolution profil<strong>in</strong>g of histonemethylations <strong>in</strong> the human genome. Cell 2007;129:823e37.[56] Shi Y, Whetst<strong>in</strong>e JR. Dynamic regulation of histone lys<strong>in</strong>e methylation by demethylases. Mol Cell2007;25:1e14.[57] Seward DJ, Cubberley G, Kim S, Schonewald M, Zhang L, Tripet B, et al. Demethylation of trimethylatedhistone H3 Lys4 <strong>in</strong> vivo by JARID1 JmjC prote<strong>in</strong>s. Nat Struct Mol Biol 2007;14:240e2.[58] Agger K, Cloos PA, Christensen J, Pas<strong>in</strong>i D, Rose S, Rappsilber J, et al. UTX and JMJD3 are histone H3K27demethylases <strong>in</strong>volved <strong>in</strong> HOX gene regulation and development. Nature 2007;449:731e4.[59] Lee MG, Villa R, Trojer P, Norman J, Yan KP, Re<strong>in</strong>berg D, et al. Demethylation of H3K27 regulates polycombrecruitment and H2A ubiquit<strong>in</strong>ation. <strong>Science</strong> 2007;318:447e50.497


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>498[60] Loh YH, Zhang W, Chen X, George J, Ng HH. Jmjd1a and Jmjd2c histone H3 Lys 9 demethylases regulate selfrenewal<strong>in</strong> embryonic stem cells. Genes Dev 2007;21:2545e57.[61] Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblastcultures by def<strong>in</strong>ed factors. Cell 2006;126:663e76.[62] Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, et al. Induction of pluripotent stem cellsfrom adult human fibroblasts by def<strong>in</strong>ed factors. Cell 2007;131:861e72.[63] Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J, Frane JL, Tian S, et al. Induced pluripotent stem celll<strong>in</strong>es derived from human somatic cells. <strong>Science</strong> 2007;318:1917e20.[64] Stadtfeld M, Nagaya M, Utikal J, Weir G, Hochedl<strong>in</strong>ger K. Induced pluripotent stem cells generated withoutviral <strong>in</strong>tegration. <strong>Science</strong> 2008;322:945e9.[65] Okita K, Nakagawa M, Hyenjong H, Ichisaka T, Yamanaka S. Generation of mouse <strong>in</strong>duced pluripotent stemcells without viral vectors. <strong>Science</strong> 2008;322:949e53.[66] Soldner F, Hockemeyer D, Beard C, Gao Q, Bell GW, Cook EG, et al. Park<strong>in</strong>son’s disease patient-derived<strong>in</strong>duced pluripotent stem cells free of viral reprogramm<strong>in</strong>g factors. Cell 2009;136:964e77.[67] Kaji K, Norrby K, Paca A, Mileikovsky M, Mohseni P, Woltjen K. Virus-free <strong>in</strong>duction of pluripotency andsubsequent excision of reprogramm<strong>in</strong>g factors. Nature 2009;458:771e5.[68] Woltjen K, Michael IP, Mohseni P, Desai R, Mileikovsky M, Hamala<strong>in</strong>en R, et al. piggyBac transpositionreprograms fibroblasts to <strong>in</strong>duced pluripotent stem cells. Nature 2009;458:766e70.[69] Zhou H, Wu S, Joo JY, Zhu S, Han DW, L<strong>in</strong> T, et al. Generation of <strong>in</strong>duced pluripotent stem cells us<strong>in</strong>grecomb<strong>in</strong>ant prote<strong>in</strong>s. Cell Stem Cell 2009;4:381e4.[70] Yakubov E, Rechavi G, Rozenblatt S, Givol D. Reprogramm<strong>in</strong>g of human fibroblasts to pluripotent stem cellsus<strong>in</strong>g mRNA of four transcription factors. Biochem Biophys Res Commun 394:189e193[71] Miyoshi N, Ishii H, Nagano H, Haraguchi N, Dewi DL, Kano Y, et al. Reprogramm<strong>in</strong>g of Mouse and <strong>Human</strong>Cells to Pluripotency Us<strong>in</strong>g Mature MicroRNAs. Cell Stem Cell 2011;8:633e8.[72] Zaehres H, Scholer HR. Induction of pluripotency: from mouse to human. Cell 2007;131:834e5.[73] Rodriguez RT, Velkey JM, Lutzko C, Seerke R, Kohn DB, O’Shea KS, et al. Manipulation of OCT4 levels <strong>in</strong>human embryonic stem cells results <strong>in</strong> <strong>in</strong>duction of differential cell types. Exp Biol Med (Maywood)2007;232:1368e80.[74] Okita K, Ichisaka T, Yamanaka S. Generation of germl<strong>in</strong>e-competent <strong>in</strong>duced pluripotent stem cells. Nature2007;448:313e7.[75] Knoepfler PS. Why myc? An unexpected <strong>in</strong>gredient <strong>in</strong> the stem cell cocktail. Cell Stem Cell 2008;2:18e21.[76] Sridharan R, Tchieu J, Mason MJ, Yachechko R, Kuoy E, Horvath S, et al. Role of the Mur<strong>in</strong>e Reprogramm<strong>in</strong>gFactors <strong>in</strong> the Induction of Pluripotency. Cell 2009;136:364e77.[77] Evans PM, Zhang W, Chen X, Yang J, Bhakat KK, Liu C. Kruppel-like factor 4 is acetylated by p300 andregulates gene transcription via modulation of histone acetylation. J Biol Chem 2007;282:33994e4002.[78] Matoba R, Niwa H, Masui S, Ohtsuka S, Carter MG, Sharov AA, et al. Dissect<strong>in</strong>g Oct3/4-regulated genenetworks <strong>in</strong> embryonic stem cells by expression profil<strong>in</strong>g. PLoS One 2006;1:e26.[79] Wadd<strong>in</strong>gton C. The strategy of the genes. london: Allen and Unw<strong>in</strong>; 1957.[80] Ieda M, Fu JD, Delgado-Olgu<strong>in</strong> P, Vedantham V, Hayashi Y, Bruneau BG, et al. Direct Reprogramm<strong>in</strong>g ofFibroblasts <strong>in</strong>to Functional Cardiomyocytes by Def<strong>in</strong>ed Factors. Cell 2010;142:375e86.[81] Yamanaka S. Elite and stochastic models for <strong>in</strong>duced pluripotent stem cell generation. Nature 2009;460:49e52.[82] Imamura M, Miura K, Iwabuchi K, Ichisaka T, Nakagawa M, Lee J, et al. Transcriptional repression and DNAhypermethylation of a small set of ES cell marker genes <strong>in</strong> male germl<strong>in</strong>e stem cells. BMC Dev Biol 2006;6:34.[83] Doi A, Park IH, Wen B, Murakami P, Aryee MJ, Irizarry R, et al. Differential methylation of tissue- and cancerspecificCpG island shores dist<strong>in</strong>guishes human <strong>in</strong>duced pluripotent stem cells, embryonic stem cells andfibroblasts. Nat Genet 2009;41:1350e3.[84] Hattori N, Nish<strong>in</strong>o K, Ko YG, Hattori N, Ohgane J, Tanaka S, et al. Epigenetic control of mouse Oct-4 geneexpression <strong>in</strong> embryonic stem cells and trophoblast stem cells. J Biol Chem 2004;279:17063e9.[85] Kimura H, Tada M, Nakatsuji N, Tada T. Histone code modifications on pluripotential nuclei of reprogrammedsomatic cells. Mol Cell Biol 2004;24:5710e20.[86] Knoepfler PS, Zhang XY, Cheng PF, Gafken PR, McMahon SB, Eisenman RN. Myc <strong>in</strong>fluences global chromat<strong>in</strong>structure. Embo J 2006;25:2723e34.[87] Shi Y, Do JT, Desponts C, Hahm HS, Scholer HR, D<strong>in</strong>g S. A comb<strong>in</strong>ed chemical and genetic approach for thegeneration of <strong>in</strong>duced pluripotent stem cells. Cell Stem Cell 2008;2:525e8.[88] Huangfu D, Maehr R, Guo W, Eijkelenboom A, Snitow M, Chen AE, et al. Induction of pluripotent stem cellsby def<strong>in</strong>ed factors is greatly improved by small-molecule compounds. Nat Biotechnol 2008;26:795e7.[89] Kubicek S, O’Sullivan RJ, August EM, Hickey ER, Zhang Q, Teodoro ML, et al. Reversal of H3K9me2 by a smallmolecule<strong>in</strong>hibitor for the G9a histone methyltransferase. Mol Cell 2007;25:473e81.


CHAPTER 24Stem Cell <strong>Epigenetics</strong> and <strong>Human</strong> <strong>Disease</strong>[90] Huangfu D, Osafune K, Maehr R, Guo W, Eijkelenboom A, Chen S, et al. Induction of pluripotent stem cellsfrom primary human fibroblasts with only Oct4 and Sox2. Nat Biotechnol 2008;26:1269e75.[91] Jones PA, Bayl<strong>in</strong> SB. The fundamental role of epigenetic events <strong>in</strong> cancer. Nat Rev Genet 2002;3:415e28.[92] Gaudet F, Hodgson JG, Eden A, Jackson-Grusby L, Dausman J, Gray JW, et al. Induction of tumors <strong>in</strong> mice bygenomic hypomethylation. <strong>Science</strong> 2003;300:489e92.[93] Fe<strong>in</strong>berg AP, Vogelste<strong>in</strong> B. Hypomethylation Dist<strong>in</strong>guishes Genes of Some <strong>Human</strong> Cancers from TheirNormal Counterparts. Nature 1983;301:89e92.[94] Greger V, Passarge E, Hopp<strong>in</strong>g W, Messmer E, Horsthemke B. Epigenetic changes may contribute to theformation and spontaneous regression of ret<strong>in</strong>oblastoma. Hum Genet 1989;83:155e8.[95] Jones PA, Bayl<strong>in</strong> SB. The epigenomics of cancer. Cell 2007;128:683e92.[96] Bayl<strong>in</strong> SB. DNA methylation and gene silenc<strong>in</strong>g <strong>in</strong> cancer. Nat Cl<strong>in</strong> Pract Oncol 2005;2(Suppl. 1):S4e11.[97] Widschwendter M, Fiegl H, Egle D, Mueller-Holzner E, Spizzo G, Marth C, et al. Epigenetic stem cell signature<strong>in</strong> cancer. Nat Genet 2007;39:157e8.[98] Ohm JE, McGarvey KM, Yu X, Cheng L, Schuebel KE, Cope L, et al. A stem cell-like chromat<strong>in</strong> pattern maypredispose tumor suppressor genes to DNA hypermethylation and heritable silenc<strong>in</strong>g. Nat Genet2007;39:237e42.[99] Schles<strong>in</strong>ger Y, Straussman R, Keshet I, Farkash S, Hecht M, Zimmerman J, et al. Polycomb-mediated methylationon Lys27 of histone H3 pre-marks genes for de novo methylation <strong>in</strong> cancer. Nat Genet 2007;39:232e6.[100] Shukla V, Vaissiere T, Herceg Z. Histone acetylation and chromat<strong>in</strong> signature <strong>in</strong> stem cell identity and cancer.Mutat Res 2008;637:1e15.[101] Moradei O, Vaisburg A, Martell RE. Histone deacetylase <strong>in</strong>hibitors <strong>in</strong> cancer therapy: new compounds andcl<strong>in</strong>ical update of benzamide-type agents. Curr Top Med Chem 2008;8:841e58.[102] Oki Y, Issa JP. Review: recent cl<strong>in</strong>ical trials <strong>in</strong> epigenetic therapy. Rev Recent Cl<strong>in</strong> Trials 2006;1:169e82.[103] Abdolmaleky HM, Smith CL, Faraone SV, Shafa R, Stone W, Glatt SJ, et al. Methylomics <strong>in</strong> psychiatry:Modulation of gene-environment <strong>in</strong>teractions may be through DNA methylation. Am J Med Genet BNeuropsychiatr Genet 2004;127B:51e9.[104] Veldic M, Guidotti A, Maloku E, Davis JM, Costa E. In psychosis, cortical <strong>in</strong>terneurons overexpress DNAmethyltransferase1. Proc Natl Acad Sci USA 2005;102:2152e7.[105] Chen Y, Sharma RP, Costa RH, Costa E, Grayson DR. On the epigenetic regulation of the human reel<strong>in</strong>promoter. Nucleic Acids Res 2002;30:2930e9.[106] Tremolizzo L, Doueiri MS, Dong E, Grayson DR, Davis J, P<strong>in</strong>na G, et al. Valproate corrects the schizophrenialikeepigenetic behavioral modifications <strong>in</strong>duced by methion<strong>in</strong>e <strong>in</strong> mice. Biol Psychiatry 2005;57:500e9.[107] Fuso A, Sem<strong>in</strong>ara L, Cavallaro RA, D’Anselmi F, Scarpa S. S-adenosylmethion<strong>in</strong>e/homocyste<strong>in</strong>e cycle alterationsmodify DNA methylation status with consequent deregulation of PS1 and BACE and beta-amyloidproduction. Mol Cell Neurosci 2005;28:195e204.[108] Mulder C, Schoonenboom NS, Jansen EE, Verhoeven NM, van Kamp GJ, Jakobs C, et al. The transmethylationcycle <strong>in</strong> the bra<strong>in</strong> of Alzheimer patients. Neurosci Lett 2005;386:69e71.[109] Jiang YH, Sahoo T, Michaelis RC, Bercovich D, Bressler J, Kashork CD, et al. A mixed epigenetic/genetic modelfor oligogenic <strong>in</strong>heritance of autism with a limited role for UBE3A. Am J Med Genet A 2004;131:1e10.[110] Unternaehrer JJ, Daley GQ. Induced pluripotent stem cells for modell<strong>in</strong>g human diseases. Philos Trans R SocLond B Biol Sci 2011;366:2274e85.[111] Hotta A, Cheung AY, Farra N, Vijayaragavan K, Segu<strong>in</strong> CA, Draper JS, et al. Isolation of human iPS cells us<strong>in</strong>gEOS lentiviral vectors to select for pluripotency. Nat Methods 2009;6:370e6.[112] Marchetto MC, Carromeu C, Acab A, Yu D, Yeo GW, Mu Y, et al. A model for neural development andtreatment of Rett syndrome us<strong>in</strong>g human <strong>in</strong>duced pluripotent stem cells. Cell 2010;143:527e39.[113] Cheung AY, Horvath LM, Grafodatskaya D, Pasceri P, Weksberg R, Hotta A, et al. Isolation of MECP2-null RettSyndrome patient hiPS cells and isogenic controls through X-chromosome <strong>in</strong>activation. Hum Mol Genet2011;20:2103e15.[114] Butler MG. Genomic impr<strong>in</strong>t<strong>in</strong>g disorders <strong>in</strong> humans: a m<strong>in</strong>i-review. J Assist Reprod Genet 2009;26:477e86.[115] Chamberla<strong>in</strong> SJ, Chen PF, Ng KY, Bourgois-Rocha F, Lemtiri-Chlieh F, Lev<strong>in</strong>e ES, et al. Induced pluripotentstem cell models of the genomic impr<strong>in</strong>t<strong>in</strong>g disorders Angelman and PradereWilli syndromes. Proc NatlAcad Sci USA 2010;107:17668e73.[116] Yang J, Cai J, Zhang Y, Wang X, Li W, Xu J, et al. Induced pluripotent stem cells can be used to model thegenomic impr<strong>in</strong>t<strong>in</strong>g disorder PradereWilli syndrome. J Biol Chem 2010;285:40303e11.[117] Rougeulle C, Glatt H, Lalande M. The Angelman syndrome candidate gene, UBE3A/E6-AP, is impr<strong>in</strong>ted <strong>in</strong>bra<strong>in</strong>. Nat Genet 1997;17:14e5.[118] Chamberla<strong>in</strong> SJ, Lalande M. Neurodevelopmental disorders <strong>in</strong>volv<strong>in</strong>g genomic impr<strong>in</strong>t<strong>in</strong>g at human chromosome15q11-q13. Neurobiol Dis 2010;39:13e20.499


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>500[119] Kipl<strong>in</strong>g D, Davis T, Ostler EL, Faragher RG. What can progeroid syndromes tell us about human ag<strong>in</strong>g?<strong>Science</strong> 2004;305:1426e31.[120] Eriksson M, Brown WT, Gordon LB, Glynn MW, S<strong>in</strong>ger J, Scott L, et al. Recurrent de novo po<strong>in</strong>t mutations <strong>in</strong>lam<strong>in</strong> A cause Hutch<strong>in</strong>soneGilford progeria syndrome. Nature 2003;423:293e8.[121] Dechat T, Pfleghaar K, Sengupta K, Shimi T, Shumaker DK, Solimando L, et al. Nuclear lam<strong>in</strong>s: majorfactors <strong>in</strong> the structural organization and function of the nucleus and chromat<strong>in</strong>. Genes Dev2008;22:832e53.[122] Scaffidi P, Misteli T. Reversal of the cellular phenotype <strong>in</strong> the premature ag<strong>in</strong>g disease Hutch<strong>in</strong>soneGilfordprogeria syndrome. Nat Med 2005;11:440e5.[123] Scaffidi P, Misteli T. Lam<strong>in</strong> A-dependent misregulation of adult stem cells associated with accelerated age<strong>in</strong>g.Nat Cell Biol 2008;10:452e9.[124] Chiba S. Notch signal<strong>in</strong>g <strong>in</strong> stem cell systems. Stem Cells 2006;24:2437e47.[125] Hennekam RC. Hutch<strong>in</strong>soneGilford progeria syndrome: review of the phenotype. Am J Med Genet A2006;140:2603e24.[126] Scaffidi P, Misteli T. Lam<strong>in</strong> A-dependent nuclear defects <strong>in</strong> human ag<strong>in</strong>g. <strong>Science</strong> 2006;312:1059e63.[127] Espada J, Varela I, Flores I, Ugalde AP, Cad<strong>in</strong>anos J, Pendas AM, et al. Nuclear envelope defects cause stem celldysfunction <strong>in</strong> premature-ag<strong>in</strong>g mice. J Cell Biol 2008;181:27e35.[128] Liu GH, Barkho BZ, Ruiz S, Diep D, Qu J, Yang SL, et al. Recapitulation of premature age<strong>in</strong>g with iPSCs fromHutch<strong>in</strong>soneGilford progeria syndrome. Nature 2011;472:221e5.[129] Zhang J, Lian Q, Zhu G, Zhou F, Sui L, Tan C, et al. A human iPSC model of Hutch<strong>in</strong>soneGilford Progeriareveals vascular smooth muscle and mesenchymal stem cell defects. Cell Stem Cell 2011;8:31e45.[130] Bar-Nur O, Russ HA, Efrat S, Benvenisty N. Epigenetic memory and preferential l<strong>in</strong>eage-specific differentiation<strong>in</strong> <strong>in</strong>duced pluripotent stem cells derived from human pancreatic islet Beta cells. Cell Stem Cell2011;9:17e23.[131] Kim K, Doi A, Wen B, Ng K, Zhao R, Cahan P, et al. Epigenetic memory <strong>in</strong> <strong>in</strong>duced pluripotent stem cells.Nature 2010;467:285e90.[132] Lunyak VV, Rosenfeld MG. Epigenetic regulation of stem cell fate. Hum Mol Genet 2008;17:R28e36.[133] Shafa M, Krawetz R, Rancourt DE. Return<strong>in</strong>g to the stem state: epigenetics of recapitulat<strong>in</strong>g pre-differentiationchromat<strong>in</strong> structure. Bioessays 2010;32:791e9.[134] Shi Y, Desponts C, Do JT, Hahm HS, Scholer HR, D<strong>in</strong>g S. Induction of pluripotent stem cells from mouseembryonic fibroblasts by Oct4 and Klf4 with small-molecule compounds. Cell Stem Cell 2008;3:568e74.[135] Huangfu D, Maehr R, Guo W, Eijkelenboom A, Snitow M, Chen AE, et al. Induction of pluripotent stem cellsby def<strong>in</strong>ed factors is greatly improved by small-molecule compounds. Nat Biotechnol 2008;26:795e7.[136] Mikkelsen TS, Hanna J, Zhang XL, Ku MC, Wernig M, Schorderet P, et al. Dissect<strong>in</strong>g direct reprogramm<strong>in</strong>gthrough <strong>in</strong>tegrative genomic analysis (vol 454, pg 49, 2008). Nature 2008;454. 794-794.[137] Veldic M, Guidotti A, Maloku E, Davis JM, Costa E. In psychosis, cortical <strong>in</strong>terneurons overexpress DNAmethyltransferase1. Proc Natl Acad Sci USA 2005;102:2152e7.[138] Chen Y, Sharma RP, Costa RH, Costa E, Grayson DR. On the epigenetic regulation of the human reel<strong>in</strong>promoter. Nucleic Acids Res 2002;30:2930e9.[139] Fuso A, Sem<strong>in</strong>ara L, Cavallaro RA, D’Anselmi F, Scarpa S. S-adenosylmethion<strong>in</strong>e/homocyste<strong>in</strong>e cycle alterationsmodify DNA methylation status with consequent deregulation of PS1 and BACE and beta-amyloidproduction. Mol Cell Neurosci 2005;28:195e204.[140] Weksberg R, Smith AC, Squire J, Sadowski P. BeckwitheWiedemann syndrome demonstrates a role forepigenetic control of normal development. Hum Mol Genet 2003;12. Spec No 1:R61e8.[141] Ehrlich M. The ICF syndrome, a DNA methyltransferase 3B deficiency and immunodeficiency disease. Cl<strong>in</strong>Immunol 2003;109:17e28.[142] Van den Veyver IB, Zoghbi HY. Mutations <strong>in</strong> the gene encod<strong>in</strong>g methyl-CpG-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> 2 cause Rettsyndrome. Bra<strong>in</strong> Dev 2001;23(Suppl. 1):S147e51.[143] Gibbons RJ, McDowell TL, Raman S, O’Rourke DM, Garrick D, Ayyub H, et al. Mutations <strong>in</strong> ATRX, encod<strong>in</strong>ga SWI/SNF-like prote<strong>in</strong>, cause diverse changes <strong>in</strong> the pattern of DNA methylation. Nat Genet 2000;24:368e71.[144] Nicholls RD, Knepper JL. Genome organization, function, and impr<strong>in</strong>t<strong>in</strong>g <strong>in</strong> PradereWilli and Angelmansyndromes. Annu Rev Genomics Hum Genet 2001;2:153e75.[145] Tassone F, Hagerman PJ. Expression of the FMR1 gene. Cytogenet Genome Res 2003;100:124e8.[146] Bello MJ, Alonso ME, Am<strong>in</strong>oso C, Anselmo NP, Arjona D, Gonzalez-Gomez P, et al. Hypermethylation of theDNA repair gene MGMT: association with TP53 G: C to A: T transitions <strong>in</strong> a series of 469 nervous systemtumors. Mutat Res 2004;554:23e32.[147] Esteller M, Corn PG, Bayl<strong>in</strong> SB, Herman JG. A gene hypermethylation profile of human cancer. Cancer Res2001;61:3225e9.


CHAPTER 24Stem Cell <strong>Epigenetics</strong> and <strong>Human</strong> <strong>Disease</strong>[148] Widschwendter M, Jiang G, Woods C, Muller HM, Fiegl H, Goebel G, et al. DNA hypomethylation andovarian cancer biology. Cancer Res 2004;64:4472e80.[149] Li LC, Ok<strong>in</strong>o ST, Dahiya R. DNA methylation <strong>in</strong> prostate cancer. Biochim Biophys Acta 2004;1704:87e102.[150] Richardson B. Impact of ag<strong>in</strong>g on DNA methylation. Age<strong>in</strong>g Res Rev 2003;2:245e61.[151] Sekigawa I, Okada M, Ogasawara H, Kaneko H, Hishikawa T, Hashimoto H. DNA methylation <strong>in</strong> systemiclupus erythematosus. Lupus 2003;12:79e85.501


This page <strong>in</strong>tentionally left blank


CHAPTER 25Non-Cod<strong>in</strong>g RNARegulatory Networks,<strong>Epigenetics</strong>, andProgramm<strong>in</strong>g Stem CellRenewal andDifferentiation:Implications for Stem CellTherapy503Rajesh C. MirandaTexas A&M Health <strong>Science</strong> Center, Bryan, TX, USACHAPTER OUTLINE25.1 Major Types of Stem Cells 50425.1.1 Stem Cell Renewal and thePluripotency TranscriptionFactor Network: Lessons fromInduced Pluripotent Stem Cell(IPSC) Biology 50525.1.2 Regulatory Networks andControl of Stem CellDifferentiation 50625.2 A Brief Overview of<strong>Epigenetics</strong> 50825.2.1 DNA Methylation, HistoneModifications, and GeneImpr<strong>in</strong>t<strong>in</strong>g 50825.2.2 <strong>Epigenetics</strong> of Non-Cod<strong>in</strong>gRNAs 50925.3 Epigenetic Programm<strong>in</strong>g of StemCells 51025.3.1 Epigenetic Programm<strong>in</strong>g andthe Core PluripotencyTranscription FactorNetwork 51025.3.2 Epigenetic Programm<strong>in</strong>g, Non-Cod<strong>in</strong>g RNAs, and Early StemCell DifferentiationPrograms 51225.3.3 Epigenetic Mutations andSomatic Cell Mosaicism 51325.4 F<strong>in</strong>al Comments 514References 514T. Tollefsbol (Ed): <strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>. DOI: 10.1016/B978-0-12-388415-2.00025-1Copyright Ó 2012 Elsevier Inc. All rights reserved.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>25.1 MAJOR TYPES OF STEM CELLSStem cells have a near <strong>in</strong>f<strong>in</strong>ite capacity for self-renewal and can be guided by specific signal<strong>in</strong>gmechanisms to differentiate <strong>in</strong>to selected cell l<strong>in</strong>eages. The capacity for self-renewal, i.e. forparent stem cells to produce daughter cells that are functionally and phenotypically identicalto the parent cell, means that at least theoretically, every tissue and organ with a residentpopulation of stem cells has a nearly <strong>in</strong>def<strong>in</strong>ite capacity for repair and regeneration. Moreover,the capacity of restricted-potential adult stem cells to replenish lost and damaged cells and torepair tissues, with programm<strong>in</strong>g by growth factors, makes these cells useful to harness fortherapeutic purposes.An extensive review of the types of stem cells and their properties is beyond the scope of thischapter. (The reader is referred to several well-annotated public resources (e.g. at the NationalInstitutes of Health http://stemcells.nih.gov/<strong>in</strong>fo/basics/; The Howard Hughes Medical Institutehttp://www.hhmi.org/bio<strong>in</strong>teractive/stemcells/lectures.html).)504Briefly, major classes of <strong>in</strong>digenous stem cells <strong>in</strong>clude embryonic (ES), fetal, and adult (ortissue) stem cells. These cells range <strong>in</strong> their capacity for differentiation. “Totipotent” stem cells,which reta<strong>in</strong> a capacity to generate an entire organism, can be derived from nuclear transfer<strong>in</strong>to an oocyte, or follow<strong>in</strong>g fertilization, from early divisions of the zygote and result<strong>in</strong>gmorula (Figure 25.1). (For an <strong>in</strong>terest<strong>in</strong>g essay <strong>in</strong>to the history of nuclear transfer and clon<strong>in</strong>g,see [1].) The cells of the morula subsequently form a blastocyst, where they differentiate <strong>in</strong>totwo cell layers, an outer trophoectoderm and an <strong>in</strong>ner cell mass. “Pluripotent” stem cells canbe isolated from the <strong>in</strong>ner cell mass, and exhibit the possibility of differentiation <strong>in</strong>to ectoderm,mesoderm, and endoderm cell l<strong>in</strong>eages, but are not, by themselves, capable of form<strong>in</strong>gan entire organism. F<strong>in</strong>ally, with<strong>in</strong> each fetal and adult tissue, resident stem cells cont<strong>in</strong>ue togenerate, with variable efficiency, replacement cells through the life of the tissue and organism.These cells normally exhibit restricted potential, rang<strong>in</strong>g from multipotent, to bi- or evenunipotency, from tissue-specific cell types. For example, hematopoietic stem cells are multipotentbecause they can generate a wide variety of lymphoid, erythroid, and myeloid cellFIGURE 25.1Schematic for the general maturation ofstem cells and loss of differentiationpotential. This figure is reproduced <strong>in</strong> thecolor plate section.


CHAPTER 25Implications for Stem Cell Therapyl<strong>in</strong>eages. Mesenchymal stem cells also exhibit multipotency as they can generate osteoblasts,chondroblasts, adipocytes, and fibroblasts among a wide variety of cells. In contrast, neuralstem cells exhibit a more restricted capacity to differentiate <strong>in</strong>to neurons and glia, andepidermal stem cells exhibit unipotential capacity to differentiate <strong>in</strong>to kerat<strong>in</strong>ocytes.F<strong>in</strong>ally, <strong>in</strong>creas<strong>in</strong>g evidence suggests that adult multipotential stem cells, even with<strong>in</strong> a s<strong>in</strong>gletissue are heterogeneous and exhibit a hierarchy of “stemness” that may make them more orless suited as therapeutic targets. For example, the G-prote<strong>in</strong>-coupled receptor Lgr5 (Gpr49)marks a population of rapidly cycl<strong>in</strong>g cells with<strong>in</strong> <strong>in</strong>test<strong>in</strong>al crypts that have the capacity togenerate every other cell of the <strong>in</strong>test<strong>in</strong>al epithelium [2]. However, when crypt Lgr5þ cells weredepleted, a rare population of slowly proliferat<strong>in</strong>g cells that are positive for Bmi1 (a member ofthe polycomb transcription regulatory complex), exhibit a parallel capacity to regenerate the<strong>in</strong>test<strong>in</strong>al epithelium <strong>in</strong>clud<strong>in</strong>g the Lgr5þ population [3]. Bmi1þ and Lgr5þ cells mayrepresent long- vs. short-term enteric stem cell populations that may be selectively targeted topromote long-range tissue re-eng<strong>in</strong>eer<strong>in</strong>g vs. shorter-term repair.25.1.1 Stem Cell Renewal and the Pluripotency Transcription FactorNetwork: Lessons from Induced Pluripotent Stem Cell (IPSC) BiologyNon-prote<strong>in</strong>-cod<strong>in</strong>g RNA networks and epigenetics have particular relevance for stem cellbiology <strong>in</strong> the context of recent successful efforts to transform somatic cells <strong>in</strong>to stem cells, i.e.<strong>in</strong>duced pluripotency. In 2007, two <strong>in</strong>dependent research groups published evidence for theexistence of a m<strong>in</strong>imal network of transcription factors that could be coord<strong>in</strong>ately activated, to<strong>in</strong>duce differentiated human cells to revert to a pluripotent state [4,5]. The research group ledby Sh<strong>in</strong>ya Yamanaka used a comb<strong>in</strong>ation of four transcription factors, Oct3/4 (POU5F1),Sox2, Klf4, and c-Myc to reprogram human dermal fibroblasts to express properties ofembryonic stem cells (Figure 25.2). The research group lead by James Thompson <strong>in</strong> contrast,used an overlapp<strong>in</strong>g comb<strong>in</strong>ation of transcription factors, OCT3/4, SOX2, NANOG, andLIN28, to accomplish the same purpose. Both research groups showed that the resultant IPSCshad atta<strong>in</strong>ed the capacity to differentiate <strong>in</strong>to cells belong<strong>in</strong>g to all three germ layers,suggest<strong>in</strong>g that these cells were pluripotent. A large amount of subsequent research <strong>in</strong>dicatesthat Sox2, c-Myc, Oct3/4, Klf4, and NANOG constitute a core network of transcription factorsthat ma<strong>in</strong>ta<strong>in</strong>s pluripotency of embryonic stem cells and confers pluripotency characteristics505FIGURE 25.2Schematic for the <strong>in</strong>duction of pluripotency <strong>in</strong>somatic cells by the pluripotency network oftranscription factors. This figure is reproduced <strong>in</strong>the color plate section.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>to a wide variety of differentiated somatic cell types derived from diverse tissue sources(reviewed <strong>in</strong> [6,7]).The pluripotency factor network has evolved functionally through vertebrate evolution, totarget new downstream genes, and presumably new tissue- and species-specific stem cellrenewal and differentiation programs. For example, the Fibroblast Growth Factor-4 (FGF4)gene is present <strong>in</strong> vertebrates, but only emerged <strong>in</strong> recent eutherian mammal evolution, asa positively regulated target of Oct3/4 [8]. FGF4 <strong>in</strong> turn mediates the transition between stemcell renewal and cell differentiation [9], imply<strong>in</strong>g that Oct3/4-<strong>in</strong>duced, FGF4 autocr<strong>in</strong>e/paracr<strong>in</strong>e signal<strong>in</strong>g may have evolved to specifically limit the renewal capacity of the pluripotencytranscription factor network <strong>in</strong> mammals. Interest<strong>in</strong>gly, the FGF family itself hasexpanded <strong>in</strong> numbers significantly dur<strong>in</strong>g vertebrate and even recent mammalian development[10], and has come to play a critical role <strong>in</strong> mediat<strong>in</strong>g l<strong>in</strong>eage specification of stem cells[11]. Though the core pluripotency network has rema<strong>in</strong>ed relatively evolutionarily conserved(stem cells <strong>in</strong> amphibian [12] and mammalian species [4,5] make use of an overlapp<strong>in</strong>gcomplement of pluripotency factors), the <strong>in</strong>tegration of the pluripotency network with l<strong>in</strong>eagespecification networks exhibits a great deal of evolutionarily diversity. As we will discuss below,some of this evolutionary recent regulatory complexity is due to the emergence of novelepigenetic programs.50625.1.2 Regulatory Networks and Control of Stem Cell DifferentiationGrowth factors constitute an effective means for guid<strong>in</strong>g multipotent stem cells down selecteddifferentiation pathways because of their capacity to activate specific transcription factornetworks (e.g. Figure 25.3A). FGF4 is an important example <strong>in</strong> this regard. FGF4 directs mouseembryonic stem cells towards a neural (ectoderm) l<strong>in</strong>eage by activat<strong>in</strong>g ERK1/2 [13]. Theconcurrent overexpression of Sprouty1 (Spry1) on the other hand, prevents neural l<strong>in</strong>eageprogression, and promotes mesoderm l<strong>in</strong>eage differentiation <strong>in</strong>stead [13]. Further, with<strong>in</strong> themesoderm l<strong>in</strong>eage, other growth factors, the Bone Morphogenetic Prote<strong>in</strong>s (BMP) 4 and 7 forexample, promote osteoblast differentiation by <strong>in</strong>duc<strong>in</strong>g the expression of the transcriptionfactor DLX5 [14,15]. Similarly, Wnt family members, Wnt1 and Wnt10b, direct differentiationof mesenchymal stem cells towards an osteoblast l<strong>in</strong>eage, while <strong>in</strong>hibit<strong>in</strong>g adipocyte l<strong>in</strong>eagedifferentiation [16], <strong>in</strong> part, by promot<strong>in</strong>g expression of osteoblast transcription factorslike Runx2 and Dlx5 while <strong>in</strong>hibit<strong>in</strong>g adipocyte-associated transcription factors like PPARg,C/EBPa, and C/EBPb. In contrast, BMP2 <strong>in</strong> cooperation with other signal<strong>in</strong>g molecules likeTGFb, drives mesenchymal stem cells <strong>in</strong>to a chrondrocyte l<strong>in</strong>eage [17]. Interest<strong>in</strong>gly, <strong>in</strong>portions of the cranial ectomesenchyme, Dlx5/6 <strong>in</strong>duces expression of the transcription factorHand2, which acts as a negative regulator of Dlx5/6. The expression of Hand2 results <strong>in</strong> thetransformation of primitive stem cells <strong>in</strong>to tissues of the tongue, whereas its absence results <strong>in</strong>a Dlx5/6-driven transformation to bone [18]. Specific l<strong>in</strong>eage commitment is, therefore,dependent on the contextual cues provided by compet<strong>in</strong>g signal<strong>in</strong>g molecules. Moreover,a limited set of <strong>in</strong>teract<strong>in</strong>g secreted signal<strong>in</strong>g factors and a related network of <strong>in</strong>tracellularsignal<strong>in</strong>g cascades and transcription factors can clearly drive divergent differentiation froma common cohort of stem cells.The core pluripotency network arguably emerged early <strong>in</strong> vertebrate evolution to support twoma<strong>in</strong> functions, to promote stem cell renewal while simultaneously suppress<strong>in</strong>g differentiation,and appears to be largely similar from one stem cell compartment to the next. Differentiation,on the other hand, is likely to <strong>in</strong>volve a diversity of maturation genes that adaptdifferentiat<strong>in</strong>g cells to specific tissues and organs. It is likely therefore that there is a layer ofcellular regulation that adapts the common pluripotency network to cell-, tissue-, and evenorganism-specific differentiation. Several pieces of evidence <strong>in</strong>directly po<strong>in</strong>t to an <strong>in</strong>terven<strong>in</strong>gregulatory layer between stem cell renewal and differentiation. Firstly, though stem cells reside<strong>in</strong> every tissue and organ, not all stem cells have an equivalent capacity to regenerate or repair


CHAPTER 25Implications for Stem Cell Therapy507FIGURE 25.3(A) Schematic of the compet<strong>in</strong>g network of transcription factors and signal<strong>in</strong>g molecules that direct stem cells down specific mesoderm or ectoderm l<strong>in</strong>eages.Green boxes <strong>in</strong>dicate core transcription factors necessary for osteoblast and adipocyte l<strong>in</strong>eage specific differentiation. (B) Overlay of the epigenetic/ncRNAregulatory network. This schematic depicts examples of ncRNAs (blue boxes) and chromat<strong>in</strong> modification factors (p<strong>in</strong>k boxes) that control the balance betweensignal<strong>in</strong>g molecules and transcription factors, to direct stem cell differentiation. This figure is reproduced <strong>in</strong> the color plate section.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>tissues and organs. Even cells that share a common developmental l<strong>in</strong>eage, i.e. mesenchymalstem cells, can exhibit vary<strong>in</strong>g differentiation potential related to their tissues of orig<strong>in</strong> [19,20],suggest<strong>in</strong>g the emergence of non-genetically programmed regulatory mechanisms. Secondly,the biology of stem cells is <strong>in</strong>timately associated with evolution and speciation. Similar typesof stem cells <strong>in</strong> different organisms produce diverse body plans and exhibit divergent regenerativecapacities. For example, <strong>in</strong> the python but not humans, an episode of feed<strong>in</strong>g producesa large and rapid <strong>in</strong>crease <strong>in</strong> enterocytes <strong>in</strong> the small <strong>in</strong>test<strong>in</strong>e [21], suggest<strong>in</strong>g that stem cellrenewal and maturation is remarkably attuned to the environment, but <strong>in</strong> a species-specificmanner. Stem cells <strong>in</strong> amphibian [12] and mammalian species [4,5] make use of an overlapp<strong>in</strong>gcomplement of pluripotency factors, yet amphibian, but not mammalian stem cellshave the capacity to regenerate complex tissues like limbs. F<strong>in</strong>ally, not only do stem cells <strong>in</strong>older organisms exhibit dim<strong>in</strong>ished regenerative capacities [22], but stem cells can exhibitaltered patterns of l<strong>in</strong>eage commitment with age; i.e. ag<strong>in</strong>g mesenchymal stem cells favoradipocyte rather than osteoblast differentiation [23]. The question is why is there suchdiversity <strong>in</strong> stem cell differentiation potential from tissue type, speciation, and age? Oneanswer to this question might lie <strong>in</strong> the existence of a new and relatively poorly understoodnetwork of regulatory mechanisms collectively termed, “epigenetics”.50825.2 A BRIEF OVERVIEW OF EPIGENETICS<strong>Epigenetics</strong>, literally “over the genome”, represents a newly discovered and dynamic regulatorynetwork that is superimposed over, and therefore <strong>in</strong>fluences, the <strong>in</strong>terpretation of genetic<strong>in</strong>formation conta<strong>in</strong>ed with<strong>in</strong> a cell. At the level of the organism, epigenetics serves to promoteadaptation and is <strong>in</strong>creas<strong>in</strong>gly thought to be a major mechanism for speciation, and at themolecular level, a mechanism to control cellular differentiation and homeostasis. Epigeneticregulatory networks are <strong>in</strong>creas<strong>in</strong>gly be<strong>in</strong>g found to be critical facilitators of the successfultransformation of stem cells <strong>in</strong>to tissues and organs, but may also serve the aberrant transformationof stem cells <strong>in</strong> cancer. A detailed and comprehensive overview of the field ofepigenetics is well beyond the scope of this chapter. Excellent recent reviews have outl<strong>in</strong>ed thehistory and basic mechanisms underly<strong>in</strong>g epigenetics [24], and detailed their relevance totissue and organism development [25,26] and to cancer mechanisms [27]. Briefly, the term“epigentics” was co<strong>in</strong>ed by the developmental biologist, Conrad Wadd<strong>in</strong>gton, based on earlyobservations that an environmental stimulus (heat) adm<strong>in</strong>istered to a cohort of fruitflies,produced multigenerational alterations <strong>in</strong> w<strong>in</strong>g structure without any apparent change <strong>in</strong> thefly genome [28]. A variety of cellular mechanisms that regulate nuclear chromat<strong>in</strong> structureand control gene transcription and translation are collectively classified as “epigeneticmechanisms”, if these mechanisms result <strong>in</strong> relatively irreversible changes <strong>in</strong> the function ofcells and tissues.25.2.1 DNA Methylation, Histone Modifications, and Gene Impr<strong>in</strong>t<strong>in</strong>gActivity of DNA methylases (DNMTs) results <strong>in</strong> methylation of genes at chromosomal loci thatare GC-rich (extensive regions of GC enrichment are termed CpG islands), and can result <strong>in</strong>the gene <strong>in</strong>activation by the recruitment of methyl-CpG-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s and the segregationof chromosome regions conta<strong>in</strong><strong>in</strong>g those genes <strong>in</strong>to the nuclear heterochromat<strong>in</strong>. Similarly,post-translational histone modifications can also alter the compactness of nucleosomes toregulate gene expression. The methylation of histones, such as di- or trimethylation of histoneH3 on lys<strong>in</strong>e-4 (H3K4me2 and H3K4me3), result <strong>in</strong> <strong>in</strong>creased activation, whereas di- andtrimethylation on H3K9 and histone acetylation are associated with repression [29]. Themethylation and demethylation of chromat<strong>in</strong> is an important component of the stem celldifferentiation process. For example, adipose-derived mesenchymal stem cells exhibit demethylationat Dlx5 and other osteoblast-specific transcription factors dur<strong>in</strong>g the process oftransformation <strong>in</strong>to osteoblasts [30]. The process of demethylation results <strong>in</strong> permissive


CHAPTER 25Implications for Stem Cell Therapyactivation of the Dlx5 locus and presumably plays an important and contextual role <strong>in</strong>enabl<strong>in</strong>g growth-factor-mediated transformation of stem cells.Dur<strong>in</strong>g vertebrate evolution the emergence of mammals, particularly eutherian mammals, wasaccompanied by an important variation <strong>in</strong> the epigenetic program, the phenomenon of geneimpr<strong>in</strong>t<strong>in</strong>g, where one allele (either the paternal or the maternal allele) of a gene undergoesDNA methylation, and is silenced. The dom<strong>in</strong>ant model for transcription at these loci is that itproceeds from the rema<strong>in</strong><strong>in</strong>g active allele. Frequently, the non-silenced allele exhibits posttranslationalhistone modifications like trimethylation of lys<strong>in</strong>e 4 (H3K4me3) that are knownto facilitate transcription activation [31]. The human genome is predicted to conta<strong>in</strong> as manyas 156 impr<strong>in</strong>ted genes [32], and many of these do not overlap with the cohort of impr<strong>in</strong>tedgenes <strong>in</strong> the mouse [33], suggest<strong>in</strong>g the likelihood of shifts <strong>in</strong> impr<strong>in</strong>t<strong>in</strong>g with mammalianspeciation. The net effect is to decrease the gene dosage <strong>in</strong> tissues and the emergence of thisphenomenon with mammalian evolution is thought to be a mechanism for the control of fetalsize. Paternal alleles are thought to promote, while maternal alleles are thought to constra<strong>in</strong>,fetal growth (reviewed <strong>in</strong> [34]). The implication of impr<strong>in</strong>t<strong>in</strong>g as an epigenetic phenomenonthat regulates stem cells is enormous. Because of their capacity to control tissue growth [35],itis likely that impr<strong>in</strong>ted genes play an important role <strong>in</strong> stem cell maturation [36]. The speciesvariation <strong>in</strong> gene impr<strong>in</strong>t<strong>in</strong>g suggests that the epigenetic controls over stem cell renewal andmaturation are likely to be species-specific. Moreover, gene impr<strong>in</strong>t<strong>in</strong>g may vary as a functionof the state of cellular differentiation. For example, neurons of the adult bra<strong>in</strong> have recentlybeen shown to exhibit monoallelic expression of the Angelman’s syndrome locus, Ube3a,whereas Glial Fibrillary Acidic Prote<strong>in</strong> (GFAP)-positive cells of the ventricular zone have beenfound to exhibit biallelic expression from this locus [37]. S<strong>in</strong>ce GFAP-positive cells adjacent tothe lateral ventricles of the bra<strong>in</strong> are presumptive neural stem cells [38] these data suggest thatthe gene dosage of Ube3a is decreased dur<strong>in</strong>g the differentiation of neural stem cells byepigenetic mechanisms. These data suggest that the epigenetic programm<strong>in</strong>g of stem cells mayvary as a function of both species and tissue of orig<strong>in</strong>, and that the replication of tissue- andspecies-specific epigenetic programs will be critical for the successful therapeutic manipulationof stem cells.50925.2.2 <strong>Epigenetics</strong> of Non-Cod<strong>in</strong>g RNAsIn the abstract to a 2007 article [39], John Mattick proposed that, “the epigenetic trajectories ofdifferentiation and development are primarily programmed by feed-forward (non-cod<strong>in</strong>g) RNAregulatory networks and that most of the <strong>in</strong>formation required for multicellular development isembedded <strong>in</strong> these networks” (author <strong>in</strong>serted text <strong>in</strong> parenthses). S<strong>in</strong>ce “differentiation” and“development” programs implicitly <strong>in</strong>volve the maturation of stem cells, the hypothesisadvanced by Mattick implies that non-cod<strong>in</strong>g RNAs (ncRNAs) are a significant locus ofepigenetic control of stem cell maturation programs.Sequenc<strong>in</strong>g the human genome has shown unexpectedly that the human genome conta<strong>in</strong>sa surpris<strong>in</strong>gly small number of prote<strong>in</strong>-cod<strong>in</strong>g genes [40]. The Ensembl database (http://www.ensembl.org/, Genebuild 2011) counts 20 599 prote<strong>in</strong>-cod<strong>in</strong>g genes, compared to the mouseat 21 873, the zebrafish at 18 572, and the <strong>in</strong>vertebrate, Caenorhabditis elegans at 20 389 knownprote<strong>in</strong>-cod<strong>in</strong>g genes. Clearly the prote<strong>in</strong> cod<strong>in</strong>g gene content of animal chromosomes doesnot change dramatically with vertebrate and mammalian evolution. The approximately 2000additional prote<strong>in</strong>-cod<strong>in</strong>g genes that separate mammals, <strong>in</strong>clud<strong>in</strong>g humans, from zebrafish,do not account for the evolutionary complexity of humans, and consequently, cannot readilyexpla<strong>in</strong> the complexity of stem cell programs that generate the human body plan. However,recent evidence <strong>in</strong>dicates that other products of genome transcription, namely non-cod<strong>in</strong>gRNAs, do exhibit <strong>in</strong>creased representation with<strong>in</strong> genomes with <strong>in</strong>creas<strong>in</strong>g evolutionarycomplexity [39]. A 2007 report of the ENCODE (Encyclopedia of DNA Elements) project,suggested that the human genome is “pervasively transcribed, such that the majority of its bases are


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>associated with at least one primary transcript” [41]. This paper showed that the human genome isfilled with large numbers of transcription start sites that code for ncRNAs and that regions ofthe genome that had hitherto been thought of as “silent”, were <strong>in</strong> fact, transcriptionally active.It is likely that, <strong>in</strong> terms of sheer numbers, ncRNAs will dwarf the numbers of prote<strong>in</strong>-cod<strong>in</strong>ggenes. RNA molecules can participate <strong>in</strong> a variety of cell functions because they exhibit twoimportant functions. Firstly, their primary nucleotide sequence complementarity can give RNAmolecules the capacity to b<strong>in</strong>d other RNA and DNA molecules with exquisite specificity.Secondly, RNAs can fold <strong>in</strong>to a variety of functional shapes <strong>in</strong> the presence of helicasechaperones to participate <strong>in</strong> RNAeprote<strong>in</strong> complexes [42]. Therefore, ncRNAs can effectivelyserve as receptors or adapter molecules to target regulatory prote<strong>in</strong> complexes to DNA or otherRNAs, with important consequences for stem cell renewal.The family of ncRNAs ranges <strong>in</strong> size from small, miRNAs and siRNAs (17e24 nucleotides),and piRNAs (25e30 nucleotides), to extremely long ncRNAs like the 19.3-kb X-chromosome<strong>in</strong>activation factor, XIST [43]. Both long ncRNAs (reviewed <strong>in</strong> [44]) and short ncRNAs likepiRNAs [45] have roles <strong>in</strong> chromat<strong>in</strong> reorganization, and consequently, epigenetic programm<strong>in</strong>g.NcRNAs like miR9/miR9* and miR124 can control chromat<strong>in</strong> remodel<strong>in</strong>g complexes[46] to promote the renewal or maturation of neural stem cells. However, recent deepsequenc<strong>in</strong>gdata suggest an additional layer of regulation, <strong>in</strong> that substantial numbers ofprocessed miRNAs traffic back <strong>in</strong>to the nucleus [47] where they may also directly act asepigenetic factors.25.3 EPIGENETIC PROGRAMMING OF STEM CELLSWe will now look at the epigenetic programm<strong>in</strong>g of stem cells.51025.3.1 Epigenetic Programm<strong>in</strong>g and the Core PluripotencyTranscription Factor NetworkRecent evidence po<strong>in</strong>ts to a complex <strong>in</strong>terplay between core pluripotency factors, epigenetics,and non-cod<strong>in</strong>g RNAs. Several miRNA families can serve as substitutes for or enhance theactivity of core pluripotency factors to transform somatic cells <strong>in</strong>to stem cells [48,49].The miR17-92 miRNA cluster, among others, for example, has been recently found to be highlyexpressed dur<strong>in</strong>g early stages of <strong>in</strong>duced stem cell programm<strong>in</strong>g [49] and therefore, it is<strong>in</strong>structive to exam<strong>in</strong>e the <strong>in</strong>teractions of members of this cluster with the core pluripotencyfactor network. The pluripotency factor c-Myc, for example, is thought to promote cell proliferation<strong>in</strong> part, by b<strong>in</strong>d<strong>in</strong>g to the common promoter for miR17-5p and miR20a, <strong>in</strong> the oncogenicmiR17-92 microRNA cluster (Figure 25.4) to promote the expression of miR20a andtherefore repress translation of the ret<strong>in</strong>oblastoma-like prote<strong>in</strong>-2 (Rbl2) [50]). Rbl2 and othermembers of the ret<strong>in</strong>oblastoma family promote and stabilize nuclear heterochromat<strong>in</strong> byrecruit<strong>in</strong>g histone deacetylases and <strong>in</strong>creas<strong>in</strong>g DNA methylation [51,52],suggest<strong>in</strong>gthatmiR20amight <strong>in</strong>directly <strong>in</strong>terfere with DNA methylation. However, miR20a and miR17-5p (anothermember of the miR17-92 cluster), also translocate to the nucleus, and recruit Argonautcomplexes to promoters of target genes (<strong>in</strong> this case, a cycl<strong>in</strong>-dependent k<strong>in</strong>ase <strong>in</strong>hibitorCDKN2B/INK4B), result<strong>in</strong>g <strong>in</strong> <strong>in</strong>creased trimethylation of histone H3 at Lys<strong>in</strong>e 9 (H3K9me3,a repressive epigenetic marks), and, like Rbl2, promote the formation of heterochromat<strong>in</strong> [53].These apparently contradictory data suggest that Myc-mediated epigenetic programm<strong>in</strong>g iscomplex, but taken as a whole, prevents cell cycle arrest. Recent observations [54] of genomewideDNA methylation patterns have reported a diversification <strong>in</strong> the patterns of moderatelymethylated genes dur<strong>in</strong>g stem cell maturation. Some genes that are moderately methylateddur<strong>in</strong>g stem cell renewal, become hypomethylated, while others exhibit <strong>in</strong>creased methylation.The association between Myc and the miR17-92 cluster provides a mechanistic hypothesis thatexpla<strong>in</strong>s the observed diversification <strong>in</strong> DNA methylation patterns that reshape the nuclearheterochromat<strong>in</strong> landscape dur<strong>in</strong>g transitions between stem cell renewal and differentiation.


CHAPTER 25Implications for Stem Cell TherapyFIGURE 25.4Example of <strong>in</strong>teractions between a pluripotency factor, Myc, andthe ncRNA/epigenetics network. Myc promotes the expressionof the miR17-92 miRNA cluster, which is processed,transported to the cytoplasm and directs translational repressionof cell cycle arrest genes by the RNA-<strong>in</strong>duced silenc<strong>in</strong>g complex(RISC). However, miRNAs are also localized to the nucleus,where they can b<strong>in</strong>d to nascent mRNA transcripts and recruitArgonaut prote<strong>in</strong>s (Ago2) and promote histone methylationmarks (e.g. H3K9me3) that result <strong>in</strong> transcription suppression ofcell cycle arrest prote<strong>in</strong>s. Collectively, these factors contribute toMyc-mediated epigenetic control over stem cell renewal andma<strong>in</strong>tenance of pluripotency. This figure is reproduced <strong>in</strong> thecolor plate section.Follow<strong>in</strong>g expression of the pluripotency factor network, Myc-mediated transcription at themiR17-92 cluster may favor direct miRNA-mediated chromat<strong>in</strong> remodel<strong>in</strong>g and <strong>in</strong>directly, bytranslation repression, prevent <strong>in</strong>activation of cell cycle genes.Myc’s role <strong>in</strong> pluripotency is complex. Myc normally promotes or represses gene transcription,but this function does not appear to be an important factor <strong>in</strong> the phase of DNA replication topromote cell cycle. Dur<strong>in</strong>g this phase, Orc1, a member of the DNA “orig<strong>in</strong> of replication(ORC)” complex, sequesters and <strong>in</strong>activates Myc control of transcription [55], and Myc itselflocalizes to early DNA replication complexes to promote DNA replication [56]. Interest<strong>in</strong>gly,the Myc gene itself serves as a site for ORC assembly and <strong>in</strong>itiation of DNA replication. Anetwork of small ncRNAs termed “yRNAs” [57,58] promote DNA replication by stabiliz<strong>in</strong>gORCs [59] to chromat<strong>in</strong> dur<strong>in</strong>g periods of DNA replication. However, ORCs also recognizerepressive epigenetic heterochromat<strong>in</strong> marks and localize to heterochromatic regions of thegenome dur<strong>in</strong>g other periods of the cell cycle [60]. The oscillation between Myc control oftranscription and Myc-ORC control of DNA replication recruits layers of epigenetic mechanismsand is critical for the processes of stem cell renewal and maturation.511A further analysis of Myc function is also <strong>in</strong>structive <strong>in</strong> terms of the assessment of the contributionof long ncRNAs to stem cell programm<strong>in</strong>g. Myc also directly b<strong>in</strong>ds to, and stronglyrepresses, the transcription of Gata6, a transcription factor that promotes endoderm differentiationof stem cells. However, while the expression of the Gata6 mRNA transcript is represseddur<strong>in</strong>g stem cell renewal, not all transcription at the Gata6 gene locus is suppressed. Recentevidence shows that the Gata6 gene locus is also the source of a long ncRNA [61], termedGata6bt (for Gata6 bidirectional transcript) that is transcribed <strong>in</strong> the antisense orientationrelative to Gata6. The expression of Gata6bt is <strong>in</strong>versely correlated with the mRNA transcript forGata6, be<strong>in</strong>g highly expressed <strong>in</strong> stem cells and suppressed dur<strong>in</strong>g differentiation. Gata6bt, likeMyc, may serve as a negative regulator of Gata6 transcription [61], to ma<strong>in</strong>ta<strong>in</strong> stem cellpluripotency, and like other large antisense ncRNAs (HOTAIR for example) [62]), may regulatechromat<strong>in</strong> state by serv<strong>in</strong>g as scaffolds for the assembly of histone modification complexes.Other members of the pluripotency network are also subject to epigenetic regulatoryprograms. Both Oct4 and NANOG are subject to extensive regulation by DNA methylation andhistone modifications [63]. Moreover, <strong>in</strong> silico screens have found that mammalian genomes


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>are specifically enriched <strong>in</strong> pseudogenes for Oct4 and NANOG. The human genome conta<strong>in</strong>ssix pseudogenes for Oct3/4 and ten pseudogenes for Nanog, compared to a relative paucity ofpsuedogenes for other non-pluripotency-related transcription factors [64]. The Oct4 pseudogenefamily has been recently found to exert complex and mutually <strong>in</strong>terdependent epigeneticregulation of the Oct4 promoter. Oct4-pseudogene-5 for example, generates an antisensetranscript (asOct4-pg5), that serves as a negative regulator of Oct4, <strong>in</strong> part by recruit<strong>in</strong>g thehistone methyl transferase, Ezh2 to the Oct3/4 promoter and <strong>in</strong>creas<strong>in</strong>g H3K9me2 andH3K9me3 histone methylation marks (Figure 25.3B) [65]). The more extensive NANOGpseudogene family may serve a similar epigenetic regulatory purpose. Interest<strong>in</strong>gly, theevolutionarily recent appearance of NANOG pseudogene-8 (NANOGP8), follow<strong>in</strong>g thedivergence between humans and chimpanzees [66], suggests that human stem cells may haveacquired novel epigenetic regulatory mechanisms that are not present <strong>in</strong> stem cells of othermammals or vertebrates. An analysis of the EST database for expressed sequences ascribed totranscription from the human NANOGP8 genomic locus (http://genome.ucsc.edu;chr15:35,371,776-35,450,927), <strong>in</strong>dicates that this locus is the orig<strong>in</strong> of multiple non-cod<strong>in</strong>gRNA transcripts. NANOGP8 has been found to promote cell proliferation and is also the orig<strong>in</strong>of a NANOG-like prote<strong>in</strong> that is expressed <strong>in</strong> cancers [67], open<strong>in</strong>g up the possibility that thisevolutionarily recent pseudogene may have the capacity to promote the oncogenic transformationstem cells.512F<strong>in</strong>ally, a recent study by Guttman et al., showed that a large network of long <strong>in</strong>tergenicncRNAs (termed l<strong>in</strong>cRNA), ma<strong>in</strong>ta<strong>in</strong> the pluripotency of stem cells [68]. Guttman et al. wereable to identify at least 26 l<strong>in</strong>cRNAs that positively regulated the activity of Oct4 and NANOG.Us<strong>in</strong>g Oct4 and Nanog promoter l<strong>in</strong>ked to a Luciferase reporter, the authors were able to showthat a suppression of any one of these l<strong>in</strong>cRNAs resulted <strong>in</strong> significant suppression of promoteractivity for these pluripotency factors. Interest<strong>in</strong>gly, these data imply that each of the 26l<strong>in</strong>cRNAs exhibits <strong>in</strong>dependent, non-redundant control over the Oct4 and Nanog promotersand suggests that all of the l<strong>in</strong>cRNAs are required to ma<strong>in</strong>ta<strong>in</strong> at least these components of them<strong>in</strong>imal pluripotency transcription factor network. These data lend further support toMattick’s hypothesis that extensive ncRNA regulatory networks [39] are likely to control stemcell renewal.25.3.2 Epigenetic Programm<strong>in</strong>g, Non-Cod<strong>in</strong>g RNAs, and Early Stem CellDifferentiation ProgramsWe previously presented evidence for FGF4 as an example of an early differentiation gene thatdirected stem cells towards a neural ectoderm l<strong>in</strong>eage <strong>in</strong> preference to mesoderm or endoderml<strong>in</strong>eages. Recent evidence <strong>in</strong>dicates that the FGF4 locus is <strong>in</strong>activated epigenetically, bydimethyation of histone H3 (H3K9me2 and H3K27me2), and that a histone demethylase,JHDM1D/KIAA1718, results <strong>in</strong> the elim<strong>in</strong>ation of these repressive methylation marks,<strong>in</strong>duction of FGF4 expression and consequently, neural l<strong>in</strong>eage commitment [69]. Spry1, thenegative regulator of FGF4 signal<strong>in</strong>g, is a target of the miRNA, miR21 [70], which is highlyexpressed <strong>in</strong> neural l<strong>in</strong>eage-committed stem cells [71]. Therefore, <strong>in</strong> the presence of miR21,FGF4-programmed stem cells will preferentially commit to neural l<strong>in</strong>eages (Figure 25.3B).Impr<strong>in</strong>ted gene loci play an important role <strong>in</strong> tissue growth <strong>in</strong> mammals and therefore ananalysis of how they control stem cell differentiation is particularly important for the therapeuticuse of stem cells. The Mest/Peg (Paternally-Expressed Gene)-1 locus is a good exampleof the role of epigenetics <strong>in</strong> stem cell maturation. An analysis of the Peg-1 locus (humanchromosome 7:130,125,573-130,146,478) shows the presence of two CpG islands, co<strong>in</strong>cid<strong>in</strong>gwith transcription factor b<strong>in</strong>d<strong>in</strong>g sites and constitut<strong>in</strong>g two presumptive promoters. Interest<strong>in</strong>gly,these regions, particularly at the second CpG island also co<strong>in</strong>cide with a high densityof activation acetylation (H3K27Ac) and methylation (H3K4me3 and H3K4Me1) marks onhistones, suggest<strong>in</strong>g differential activation of maternal and paternal alleles. While the exon


CHAPTER 25Implications for Stem Cell Therapystructure of MEST appears to be conserved through vertebrate evolution, two ncRNA transcripts,a miRNA (miR335) and a long antisense transcript overlapp<strong>in</strong>g the presumptivepromoter region (MESTIT1) late <strong>in</strong> evolution, <strong>in</strong> eutherian mammals and primates specifically.Like MEST, MESTIT1 also exhibits monoallelic expression from the paternal allele and likeother long ncRNAs [62] may recruit heterochromat<strong>in</strong> marks to the parental gene. We previouslyobserved that the MEST <strong>in</strong>tronic miRNA, miR335 is expressed at moderate levels <strong>in</strong>neural stem cells and that miR335 knockdown results <strong>in</strong> <strong>in</strong>creased neural stem cell proliferation[71]. The MEST/miR335 locus appears to cooperatively specific differentiation ofmesenchymal stem cells, by <strong>in</strong>teract<strong>in</strong>g with core differentiation signal<strong>in</strong>g programs. Wepreviously discussed evidence, for example, that Wnt signal<strong>in</strong>g directs mesenchymal stem cellstowards osteoblast-specific differentiation and <strong>in</strong>hibits adipocyte differentiation. MEST hasrecently been reported to block Wnt signal<strong>in</strong>g by <strong>in</strong>hibit<strong>in</strong>g post-translational process<strong>in</strong>g ofthe Wnt co-receptor LRP6, and knockdown of MEST prevents adipogenesis and results <strong>in</strong>reduced expression of the adipogenic factors C/EBPa and PPARg (Figure 25.3B) [72]. TheMEST <strong>in</strong>tronic miRNA on the other hand, is highly expressed <strong>in</strong> bone-marrow-derivedmesenchymal stem cells compared to differentiated cells belong<strong>in</strong>g to the mesoderm l<strong>in</strong>eage,and its expression decreases dur<strong>in</strong>g osteoblast and adipocyte differentiation [73]. MiR335promotes Wnt signal<strong>in</strong>g by suppress<strong>in</strong>g the expression of the Wnt antagonist DKK1 [74].However, miR335 also acts as a direct negative regulator of Runx2, a factor required forosteogenic differentiation [73]. Collectively, these data suggest that osteoblast differentiationfrom mesenchymal stem cells is facilitated by careful temporal choreography of gene expressionfrom the impr<strong>in</strong>ted MEST/miR335 locus. Runx2 and other important components of theosteoblastic transformation of mesenchymal stem cells, like DLX5 [14,15] are also loci for the<strong>in</strong>teractions between epigenetics and ncRNA networks. The human Runx2 (humanchr6:45,296,054-45,518,818) and DLX5 (human chr7:96,649,667-96,654,230) gene loci forexample, conta<strong>in</strong> CpG islands, and demethylation leads to <strong>in</strong>creased expression of thesefactors and supports osteoblast differentiation [75]. DLX2, 5, and 6 are coexpressed strongly <strong>in</strong>immature osteoblasts [76], but this cluster is also important for neural stem cell differentiation[77], and may provide redundant control of early stem cell maturation. The DLX5/6 gene lociare the site for transcription of two long ncRNAs, generated by alternate splic<strong>in</strong>g, the 2.7-kband 3.8-kb Evf1 and Evf2 RNAs. These ncRNAs expand activation of the DLX network byserv<strong>in</strong>g as transcription coactivators for DLX2, further <strong>in</strong>creas<strong>in</strong>g transcription of DLX5/6 [78].The Evf ncRNAs have evolved to coord<strong>in</strong>ate activity of the DLX gene family, and <strong>in</strong> coord<strong>in</strong>ationwith other epigenetic mechanisms that regulate the methylation state of DLX5, canpromote specific patterns of stem cell differentiation. The emerg<strong>in</strong>g story is that pluripotencyand differentiation-related transcription factors, and many of their associated signal<strong>in</strong>gpathways are subject to a layer of additional regulation from a network of prote<strong>in</strong>s andncRNAs that guide the renewal and differentiation potential of stem cells. F<strong>in</strong>ally, ncRNAslike miR-9/9* and miR-124 have reportedly been used to transform cells from one l<strong>in</strong>eage,mesoderm, to another, i.e. neural [79], without the requirement for pass<strong>in</strong>g through an<strong>in</strong>termediate “stem cell” stage. This suggests that the pluripotency factor network maybe dispensable, and that the direct ncRNA-mediated transformation of somatic cells may bea viable strategy for therapeutic purposes.51325.3.3 Epigenetic Mutations and Somatic Cell MosaicismA major barrier to the adoption of stem cell therapy is that stem cells develop epigeneticmutations, and that these may persist and <strong>in</strong>fluence differentiation patterns, cancer transformation,and premature stem cell senescence. For example, researchers have reported the lossof X-chromosome <strong>in</strong>activation <strong>in</strong> well-established human embryonic stem cell l<strong>in</strong>es [80]suggest<strong>in</strong>g that stem cells can experience epigenetic drift. Loss of expression of the ncRNA,XIST, is related to the loss of X-chromosome <strong>in</strong>activation and to the resistance of cancer tochemotherapy [81] suggest<strong>in</strong>g that disruptions <strong>in</strong> epigenetic programs may promote the


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>emergence of cancer stem cells. We [71] and others [54] have shown that ncRNAs coded with<strong>in</strong>impr<strong>in</strong>ted genomic regions, and methylation patterns <strong>in</strong> stem cells are both sensitive toteratogens like ethanol. This suggests that the environment can reprogram epigenetic controlsover stem cell renewal and maturation.Most epigenetic changes do not lead to alterations <strong>in</strong> the primary sequence of genes and arepotentially reversible. However, some epigenetic mutations do lead to genetic mosaicism <strong>in</strong>somatic stem cells, potentially lead<strong>in</strong>g to permanent alterations <strong>in</strong> differentiation. The retrotransposongenes, which constitute approximately 45% of the sequence of the human genome[82] are a good example of how mutations <strong>in</strong> the epigenome may produce genetic drift amongsomatic cells, and perhaps even among stem cells. Long <strong>in</strong>terspersed element 1 (LINE-1) retrotransposonscode for RNA molecules that can be reverse-transcribed and re<strong>in</strong>serted <strong>in</strong>to genes toalter their function either subtly, by alter<strong>in</strong>g promoter regions for example, or fundamentally, byshuffl<strong>in</strong>g exon structure of prote<strong>in</strong>-cod<strong>in</strong>g genes [82]. Retrotransposons are normally silenced <strong>in</strong>stem cell genome by piRNAs [45] and by recruitment of DNA methylation mechanisms [83].Recent research has shown that LINE-1 retrotransposons are activated dur<strong>in</strong>g neural stem celldifferentiation [84] and IPSCs obta<strong>in</strong>ed from patients with diagnosed mutations <strong>in</strong> Methyl CpGb<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>-2 (MeCP2) exhibited <strong>in</strong>crease LINE-1 activity dur<strong>in</strong>g neural differentiation [85].514Collectively these data show that not only are epigenetic mechanisms and RNA networks neededto ma<strong>in</strong>ta<strong>in</strong> chromosome stability <strong>in</strong> stem cells, but that evolution may have equippedmammalian and primate stem cells to actually utilize chromat<strong>in</strong> destabilization mechanisms todrive genetic diversification of somatic cells dur<strong>in</strong>g the process of stem cell differentiation.However, epigenetic mutations may contribute to senescence of adult tissue stem cells,compromis<strong>in</strong>g their regenerative capacity [22]. Moreover, epigenetically driven genetic diversificationof somatic cells means that these cells may not be equipped to recapitulate nativepluripotency states of embryonic stem cells derived from the blastocyst. For example, a recentstudy showed that IPSC cells derived from muscle-derived pericytes reta<strong>in</strong>ed their epigeneticprogramm<strong>in</strong>g, and preferentially differentiated <strong>in</strong>to muscle tissue [86]. Similar fate preferencewas shown for IPSCs derived from pancreatic beta cells [87]. Therefore IPSC-type stem cells mayhave limited functional therapeutic applicability to replacement of their tissues of orig<strong>in</strong>.25.4 FINAL COMMENTSThis real potential for stem differentiation for tissue repair and eng<strong>in</strong>eer<strong>in</strong>g <strong>in</strong> humanpopulations was emphasized by an early cl<strong>in</strong>ical report [88], outl<strong>in</strong><strong>in</strong>g the steps towardseng<strong>in</strong>eer<strong>in</strong>g a functional mandible <strong>in</strong> a human patient. In this study, the authors impregnatedan artificial mandible-shaped, titanium-encased bone m<strong>in</strong>eral matrix, with patient-derivedbone marrow (rich <strong>in</strong> adult mesenchymal stem cells) treated with bone morphogeneticprote<strong>in</strong>-7 (BMP7). The authors were able to show vascularization, osteogenesis, and successfulfunctional engraftment of the eng<strong>in</strong>eered mandible <strong>in</strong>to a patient. BMPs are now used cl<strong>in</strong>icallyto <strong>in</strong>duce bone formation and agents, by programm<strong>in</strong>g adult mesenchymal stem cells[89], show<strong>in</strong>g that the guided differentiation of restricted-potential adult stem cells haspractical value. The generation of embryonic stem cells and patient-specific stem cells by IPSCtechnology also bears significant promise, at least <strong>in</strong> animal studies of a variety of diseasemodel systems (for example [90e93]). A clearer understand<strong>in</strong>g of the epigenetic landscape ofthe stem cell dur<strong>in</strong>g renewal and through successive stages of maturation will be a criticalrequirement for the development of effective stem cell therapy.References[1] McLaren A. Clon<strong>in</strong>g: pathways to a pluripotent future. <strong>Science</strong> 2000;288(5472):1775e80.[2] Barker N, van Es JH, Kuipers J, et al. Identification of stem cells <strong>in</strong> small <strong>in</strong>test<strong>in</strong>e and colon by marker geneLgr5. Nature 2007;449(7165):1003e7.


CHAPTER 25Implications for Stem Cell Therapy[3] Tian H, Biehs B, Warm<strong>in</strong>g S, et al. A reserve stem cell population <strong>in</strong> small <strong>in</strong>test<strong>in</strong>e renders Lgr5-positive cellsdispensable. Nature 2011;478(7368):255e9.[4] Takahashi K, Tanabe K, Ohnuki M, et al. Induction of pluripotent stem cells from adult human fibroblasts bydef<strong>in</strong>ed factors. Cell 2007;131(5):861e72.[5] Yu J, Vodyanik MA, Smuga-Otto K, et al. Induced pluripotent stem cell l<strong>in</strong>es derived from human somatic cells.<strong>Science</strong> 2007;318(5858):1917e20.[6] Masip M, Veiga A, Izpisua Belmonte JC, Simon C. Reprogramm<strong>in</strong>g with def<strong>in</strong>ed factors: from <strong>in</strong>ducedpluripotency to <strong>in</strong>duced transdifferentiation. Mol Hum Reprod 2010;16(11):856e68.[7] Pan G, Thomson JA. Nanog and transcriptional networks <strong>in</strong> embryonic stem cell pluripotency. Cell Res2007;17(1):42e9.[8] Fernandez-Tresguerres B, Canon S, Rayon T, et al. Evolution of the mammalian embryonic pluripotency generegulatory network. Proc Natl Acad Sci USA 2010;107(46):19955e60.[9] Kunath T, Saba-El-Leil MK, Almousailleakh M, et al. FGF stimulation of the Erk1/2 signall<strong>in</strong>g cascade triggerstransition of pluripotent embryonic stem cells from self-renewal to l<strong>in</strong>eage commitment. Development2007;134(16):2895e902.[10] Itoh N, Ornitz DM. Functional evolutionary history of the mouse Fgf gene family. Dev Dyn 2008;237(1):18e27.[11] Villegas SN, Canham M, Brickman JM. FGF signall<strong>in</strong>g as a mediator of l<strong>in</strong>eage transitionseevidence fromembryonic stem cell differentiation. J Cell Biochem 2010;110(1):10e20.[12] Maki N, Suetsugu-Maki R, Tarui H, et al. Expression of stem cell pluripotency factors dur<strong>in</strong>g regeneration <strong>in</strong>newts. Dev Dyn 2009;238(6):1613e6.[13] Jung JE, Moon SH, Kim DK, et al. Sprouty1 Regulates Neural and Endothelial Differentiation of MouseEmbryonic Stem Cells. Stem Cells Dev 2011;21(4):554e61.[14] Holleville N, Quilhac A, Bontoux M, Monsoro-Burq AH. BMP signals regulate Dlx5 dur<strong>in</strong>g early avian skulldevelopment. Dev Biol 2003;257(1):177e89.[15] Miyama K, Yamada G, Yamamoto TS, et al. A BMP-<strong>in</strong>ducible gene, dlx5, regulates osteoblast differentiationand mesoderm <strong>in</strong>duction. Dev Biol 1999;208(1):123e33.[16] Bennett CN, Longo KA, Wright WS, et al. Regulation of osteoblastogenesis and bone mass by Wnt10b. ProcNatl Acad Sci USA 2005;102(9):3324e9.[17] zur Nieden NI, Kempka G, Rancourt DE, Ahr HJ. Induction of chondro-, osteo- and adipogenesis <strong>in</strong>embryonic stem cells by bone morphogenetic prote<strong>in</strong>-2: effect of cofactors on differentiat<strong>in</strong>g l<strong>in</strong>eages. BMCDev Biol 2005;5:1.[18] Barron F, Woods C, Kuhn K, et al. Downregulation of Dlx5 and Dlx6 expression by Hand2 is essential for<strong>in</strong>itiation of tongue morphogenesis. Development 2011;138(11):2249e59.[19] Sakaguchi Y, Sekiya I, Yagishita K, Muneta T. Comparison of human stem cells derived from variousmesenchymal tissues: superiority of synovium as a cell source. Arthritis Rheum 2005;52(8):2521e9.[20] Yoshimura H, Muneta T, Nimura A, et al. Comparison of rat mesenchymal stem cells derived from bonemarrow, synovium, periosteum, adipose tissue, and muscle. Cell Tissue Res 2007;327(3):449e62.[21] Secor SM, Whang EE, Lane JS, Ashley SW, Diamond J. Lum<strong>in</strong>al and systemic signals trigger <strong>in</strong>test<strong>in</strong>al adaptation<strong>in</strong> the juvenile python. Am J Physiol Gastro<strong>in</strong>test Liver Physiol 2000;279(6):G1177e87.[22] Rando TA. Stem cells, age<strong>in</strong>g and the quest for immortality. Nature 2006;441(7097):1080e6.[23] Moerman EJ, Teng K, Lipschitz DA, Lecka-Czernik B. Ag<strong>in</strong>g activates adipogenic and suppresses osteogenicprograms <strong>in</strong> mesenchymal marrow stroma/stem cells: the role of PPAR-gamma2 transcription factor and TGFbeta/BMPsignal<strong>in</strong>g pathways. Ag<strong>in</strong>g Cell 2004;3(6):379e89.[24] Holliday R. <strong>Epigenetics</strong>: a historical overview. <strong>Epigenetics</strong> 2006;1(2):76e80.[25] Bernal AJ, Jirtle RL. Epigenomic disruption: the effects of early developmental exposures. Birth Defects Res ACl<strong>in</strong> Mol Teratol 2010;88(10):938e44.[26] Sk<strong>in</strong>ner MK. Role of epigenetics <strong>in</strong> developmental biology and transgenerational <strong>in</strong>heritance. Birth Defects ResC Embryo Today 2011;93(1):51e5.[27] Sharma S, Kelly TK, Jones PA. <strong>Epigenetics</strong> <strong>in</strong> cancer. Carc<strong>in</strong>ogenesis 2010;31(1):27e36.[28] Wadd<strong>in</strong>gton CH. Genetic Assimilation of an acquired character. Evolution 1953;7:118e26.[29] Wang Z, Zang C, Rosenfeld JA, et al. Comb<strong>in</strong>atorial patterns of histone acetylations and methylations <strong>in</strong> thehuman genome. Nat Genet 2008;40(7):897e903.[30] Zhang RP, Shao JZ, Xiang LX. Gadd45a plays an essential role <strong>in</strong> active DNA demethylation dur<strong>in</strong>g term<strong>in</strong>alosteogenic differentiation of adipose-derived mesenchymal stem cells. J Biol Chem 2011;286(47):41083e94.[31] D<strong>in</strong>dot SV, Person R, Strivens M, Garcia R, Beaudet AL. Epigenetic profil<strong>in</strong>g at mouse impr<strong>in</strong>ted gene clustersreveals novel epigenetic and genetic features at differentially methylated regions. Genome Res2009;19(8):1374e83.515


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>516[32] Luedi PP, Dietrich FS, Weidman JR, et al. Computational and experimental identification of novel humanimpr<strong>in</strong>ted genes. Genome Res 2007;17(12):1723e30.[33] Morison IM, Ramsay JP, Spencer HG. A census of mammalian impr<strong>in</strong>t<strong>in</strong>g. Trends Genet 2005;21(8):457e65.[34] Abu-Amero S, Monk D, Apostolidou S, Stanier P, Moore G. Impr<strong>in</strong>ted genes and their role <strong>in</strong> human fetalgrowth. Cytogenet Genome Res 2006;113(1-4):262e70.[35] Constancia M, Hemberger M, Hughes J, et al. Placental-specific IGF-II is a major modulator of placental andfetal growth. Nature 2002;417(6892):945e8.[36] Hamidouche Z, Fromigue O, R<strong>in</strong>ge J, Haupl T, Marie PJ. Crosstalks between <strong>in</strong>tegr<strong>in</strong> alpha 5 and IGF2/IGFBP2signall<strong>in</strong>g trigger human bone marrow-derived mesenchymal stromal osteogenic differentiation. BMC CellBiol 2010;11:44.[37] D<strong>in</strong>dot SV, Antalffy BA, Bhattacharjee MB, Beaudet AL. The Angelman syndrome ubiquit<strong>in</strong> ligase localizes tothe synapse and nucleus, and maternal deficiency results <strong>in</strong> abnormal dendritic sp<strong>in</strong>e morphology. Hum MolGenet 2008;17(1):111e8.[38] van den Berge SA, Middeldorp J, Zhang CE, et al. Longterm quiescent cells <strong>in</strong> the aged human subventricularneurogenic system specifically express GFAP-delta. Ag<strong>in</strong>g Cell 2010;9(3):313e26.[39] Mattick JS. A new paradigm for developmental biology. J Exp Biol 2007;210(Pt 9):1526e47.[40] International <strong>Human</strong> Genome Sequenc<strong>in</strong>g Consortium. F<strong>in</strong>ish<strong>in</strong>g the euchromatic sequence of the humangenome. Nature 2004;431(7011):931e45.[41] Birney E, Stamatoyannopoulos JA, Dutta A, et al. Identification and analysis of functional elements <strong>in</strong> 1% ofthe human genome by the ENCODE pilot project. Nature 2007;447(7146):799e816.[42] Pan C, Russell R. Roles of DEAD-box prote<strong>in</strong>s <strong>in</strong> RNA and RNP Fold<strong>in</strong>g. RNA Biol 2010;7(6):667e76.[43] Hong YK, Ontiveros SD, Strauss WM. A revision of the human XIST gene organization and structuralcomparison with mouse Xist. Mamm Genome 2000;11(3):220e4.[44] Spitale RC, Tsai MC, Chang HY. RNA templat<strong>in</strong>g the epigenome: long noncod<strong>in</strong>g RNAs as molecular scaffolds.<strong>Epigenetics</strong> 2011;6(5):539e43.[45] Khurana JS, Theurkauf W. piRNAs, transposon silenc<strong>in</strong>g, and Drosophila germl<strong>in</strong>e development. J Cell Biol2010;191(5):905e13.[46] Yoo AS, Staahl BT, Chen L, Crabtree GR. MicroRNA-mediated switch<strong>in</strong>g of chromat<strong>in</strong>-remodell<strong>in</strong>g complexes<strong>in</strong> neural development. Nature 2009;460(7255):642e6.[47] Liao JY, Ma LM, Guo YH, et al. Deep sequenc<strong>in</strong>g of human nuclear and cytoplasmic small RNAs reveals anunexpectedly complex subcellular distribution of miRNAs and tRNA 3’ trailers. PLoS One 2010;5(5):e10563.[48] Anokye-Danso F, Trivedi CM, Juhr D, et al. Highly efficient miRNA-mediated reprogramm<strong>in</strong>g of mouse andhuman somatic cells to pluripotency. Cell Stem Cell 2011;8(4):376e88.[49] Li Z, Yang CS, Nakashima K, Rana TM. Small RNA-mediated regulation of iPS cell generation. EMBO J2011;30(5):823e34.[50] Smith KN, S<strong>in</strong>gh AM, Dalton S. Myc represses primitive endoderm differentiation <strong>in</strong> pluripotent stem cells.Cell Stem Cell 2010;7(3):343e54.[51] Gonzalo S, Garcia-Cao M, Fraga MF, et al. Role of the RB1 family <strong>in</strong> stabiliz<strong>in</strong>g histone methylation atconstitutive heterochromat<strong>in</strong>. Nat Cell Biol 2005;7(4):420e8.[52] Montoya-Durango DE, Liu Y, Teneng I, et al. Epigenetic control of mammalian LINE-1 retrotransposon byret<strong>in</strong>oblastoma prote<strong>in</strong>s. Mutat Res 2009;665(1e2):20e8.[53] Gonzalez S, Pisano DG, Serrano M. Mechanistic pr<strong>in</strong>ciples of chromat<strong>in</strong> remodel<strong>in</strong>g guided by siRNAs andmiRNAs. Cell Cycle 2008;7(16):2601e8.[54] Zhou FC, Balaraman Y, Teng M, et al. Alcohol Alters DNA Methylation Patterns and Inhibits Neural Stem CellDifferentiation. Alcohol Cl<strong>in</strong> Exp Res 2011;35(4):1e12.[55] Takayama MA, Taira T, Tamai K, Iguchi-Ariga SM, Ariga H. ORC1 <strong>in</strong>teracts with c-Myc to <strong>in</strong>hibit E-boxdependenttranscription by abrogat<strong>in</strong>g c-Myc-SNF5/INI1 <strong>in</strong>teraction. Genes Cells 2000;5(6):481e90.[56] Dom<strong>in</strong>guez-Sola D, Y<strong>in</strong>g CY, Grandori C, et al. Non-transcriptional control of DNA replication by c-Myc.Nature 2007;448(7152):445e51.[57] Christov CP, Gard<strong>in</strong>er TJ, Szuts D, Krude T. Functional requirement of noncod<strong>in</strong>g Y RNAs for human chromosomalDNA replication. Mol Cell Biol 2006;26(18):6993e7004.[58] Gard<strong>in</strong>er TJ, Christov CP, Langley AR, Krude T. A conserved motif of vertebrate Y RNAs essential for chromosomalDNA replication. RNA 2009;15(7):1375e85.[59] Norseen J, Thomae A, Sridharan V, et al. RNA-dependent recruitment of the orig<strong>in</strong> recognition complex.EMBO J 2008;27(22):3024e35.[60] Chakraborty A, Shen Z, Prasanth SG. “ORCanization” on heterochromat<strong>in</strong>: l<strong>in</strong>k<strong>in</strong>g DNA replication <strong>in</strong>itiationto chromat<strong>in</strong> organization. <strong>Epigenetics</strong> 2011;6(6):665e70.


CHAPTER 25Implications for Stem Cell Therapy[61] D<strong>in</strong>ger ME, Amaral PP, Mercer TR, et al. Long noncod<strong>in</strong>g RNAs <strong>in</strong> mouse embryonic stem cell pluripotencyand differentiation. Genome Res 2008;18(9):1433e45.[62] Tsai MC, Manor O, Wan Y, et al. Long noncod<strong>in</strong>g RNA as modular scaffold of histone modification complexes.<strong>Science</strong> 2010;329(5992):689e93.[63] Freberg CT, Dahl JA, Timoska<strong>in</strong>en S, Collas P. Epigenetic reprogramm<strong>in</strong>g of OCT4 and NANOG regulatoryregions by embryonal carc<strong>in</strong>oma cell extract. Mol Biol Cell 2007;18(5):1543e53.[64] Pa<strong>in</strong> D, Chirn GW, Strassel C, Kemp DM. Multiple retropseudogenes from pluripotent cell-specific geneexpression <strong>in</strong>dicates a potential signature for novel gene identification. J Biol Chem 2005;280(8):6265e8.[65] Hawk<strong>in</strong>s PG, Morris KV. Transcriptional regulation of Oct4 by a long non-cod<strong>in</strong>g RNA antisense to Oct4-pseudogene 5. Transcription 2010;1(3):165e75.[66] Fairbanks DJ, Maughan PJ. Evolution of the NANOG pseudogene family <strong>in</strong> the human and chimpanzeegenomes. BMC Evol Biol 2006;6:12.[67] Jeter CR, Badeaux M, Choy G, et al. Functional evidence that the self-renewal gene NANOG regulates humantumor development. Stem Cells 2009;27(5):993e1005.[68] Guttman M, Donaghey J, Carey BW, et al. l<strong>in</strong>cRNAs act <strong>in</strong> the circuitry controll<strong>in</strong>g pluripotency and differentiation.Nature 2011;477(7364):295e300.[69] Huang C, Xiang Y, Wang Y, et al. Dual-specificity histone demethylase KIAA1718 (KDM7A) regulates neuraldifferentiation through FGF4. Cell Res 2010;20(2):154e65.[70] Darimipoura<strong>in</strong> M, Wang S, Ittmann M, Kwabi-Addo B. Transcriptional and post-transcriptional regulation ofSprouty1, a receptor tyros<strong>in</strong>e k<strong>in</strong>ase <strong>in</strong>hibitor <strong>in</strong> prostate cancer. Prostate Cancer Prostatic Dis2011;14(4):279e85.[71] Sathyan P, Golden HB, Miranda RC. Compet<strong>in</strong>g <strong>in</strong>teractions between micro-RNAs determ<strong>in</strong>e neuralprogenitor survival and proliferation after ethanol exposure: evidence from an ex vivo model of the fetalcerebral cortical neuroepithelium. J Neurosci 2007;27(32):8546e57.[72] Jung H, Lee SK, Jho EH. Mest/Peg1 <strong>in</strong>hibits Wnt signall<strong>in</strong>g through regulation of LRP6 glycosylation. BiochemJ 2011;436(2):263e9.[73] Tome M, Lopez-Romero P, Albo C, et al. miR-335 orchestrates cell proliferation, migration and differentiation<strong>in</strong> human mesenchymal stem cells. Cell Death Differ 2011;18(6):985e95.[74] Zhang J, Tu Q, Bonewald LF, et al. Effects of miR-335-5p <strong>in</strong> modulat<strong>in</strong>g osteogenic differentiation byspecifically downregulat<strong>in</strong>g Wnt antagonist DKK1. J Bone M<strong>in</strong>er Res 2011;26(8):1953e63.[75] Zhou GS, Zhang XL, Wu JP, et al. 5-Azacytid<strong>in</strong>e facilitates osteogenic gene expression and differentiation ofmesenchymal stem cells by alteration <strong>in</strong> DNA methylation. Cytotechnology 2009;60:11e22.[76] Li H, Marijanovic I, Kronenberg MS, et al. Expression and function of Dlx genes <strong>in</strong> the osteoblast l<strong>in</strong>eage. DevBiol 2008;316(2):458e70.[77] Kohwi M, Petryniak MA, Long JE, et al. A subpopulation of olfactory bulb GABAergic <strong>in</strong>terneurons is derivedfrom Emx1- and Dlx5/6-express<strong>in</strong>g progenitors. J Neurosci 2007;27(26):6878e91.[78] Feng J, Bi C, Clark BS, et al. The Evf-2 noncod<strong>in</strong>g RNA is transcribed from the Dlx-5/6 ultraconserved regionand functions as a Dlx-2 transcriptional coactivator. Genes Dev 2006;20(11):1470e84.[79] Yoo AS, Sun AX, Li L, et al. MicroRNA-mediated conversion of human fibroblasts to neurons. Nature2011;476(7359):228e31.[80] Tanasijevic B, Dai B, Ezashi T, et al. Progressive accumulation of epigenetic heterogeneity dur<strong>in</strong>g human ES cellculture. <strong>Epigenetics</strong> 2009;4(5):330e8.[81] Huang KC, Rao PH, Lau CC, et al. Relationship of XIST expression and responses of ovarian cancer tochemotherapy. Mol Cancer Ther 2002;1(10):769e76.[82] Call<strong>in</strong>an PA, Batzer MA. Retrotransposable elements and human disease. Genome Dyn 2006;1:104e15.[83] Yu F, Z<strong>in</strong>gler N, Schumann G, Stratl<strong>in</strong>g WH. Methyl-CpG-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> 2 represses LINE-1 expression andretrotransposition but not Alu transcription. Nucleic Acids Res 2001;29(21):4493e501.[84] Muotri AR, Chu VT, Marchetto MC, et al. Somatic mosaicism <strong>in</strong> neuronal precursor cells mediated by L1retrotransposition. Nature 2005;435(7044):903e10.[85] Muotri AR, Marchetto MC, Coufal NG, et al. L1 retrotransposition <strong>in</strong> neurons is modulated by MeCP2. Nature2010;468(7322):443e6.[86] Quattrocelli M, Palazzolo G, Floris G, et al. Intr<strong>in</strong>sic cell memory re<strong>in</strong>forces myogenic commitment ofpericyte-derived iPSCs. J Pathol 2011;223(5):593e603.[87] Bar-Nur O, Russ HA, Efrat S, Benvenisty N. Epigenetic memory and preferential l<strong>in</strong>eage-specific differentiation <strong>in</strong><strong>in</strong>duced pluripotent stem cells derived from human pancreatic islet Beta cells. Cell Stem Cell 2011;9(1):17e23.[88] Warnke PH, Spr<strong>in</strong>ger IN, Wiltfang J, et al. Growth and transplantation of a custom vascularised bone graft <strong>in</strong>a man. Lancet 2004;364(9436):766e70.517


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>[89] Lavery K, Swa<strong>in</strong> P, Falb D, Alaoui-Ismaili MH. BMP-2/4 and BMP-6/7 differentially utilize cell surface receptorsto <strong>in</strong>duce osteoblastic differentiation of human bone marrow-derived mesenchymal stem cells. J Biol Chem2008;283(30):20948e58.[90] Hanna J, Wernig M, Markoulaki S, et al. Treatment of sickle cell anemia mouse model with iPS cells generatedfrom autologous sk<strong>in</strong>. <strong>Science</strong> 2007;318(5858):1920e3.[91] Lamba DA, McUsic A, Hirata RK, et al. Generation, purification and transplantation of photoreceptors derivedfrom human <strong>in</strong>duced pluripotent stem cells. PLoS One 2010;5(1):e8763.[92] Lu SJ, Li F, Y<strong>in</strong> H, et al. Platelets generated from human embryonic stem cells are functional <strong>in</strong> vitro and <strong>in</strong> themicrocirculation of liv<strong>in</strong>g mice. Cell Res 2011;21(3):530e45.[93] Mauritz C, Martens A, Rojas SV, et al. Induced pluripotent stem cell (iPSC)-derived Flk-1 progenitor cells engraft,differentiate, and improve heart function <strong>in</strong> a mouse model of acute myocardial <strong>in</strong>farction. Eur Heart J2011;32:2634e41.518


CHAPTER 26Ag<strong>in</strong>g and <strong>Disease</strong>: TheEpigenetic BridgeAndrea FusoSapienza University of Rome, Rome, ItalyCHAPTER OUTLINE26.1 Introduction 51926.2 Genes and Ag<strong>in</strong>g 52026.3 The Dynamic Methylome 52626.4 Epigenetic Dynamics<strong>in</strong> the Ag<strong>in</strong>g Bra<strong>in</strong> 52626.5 The Complexity of the Age-Associated EpigeneticChanges 52826.6 Healthy and PathologicalAg<strong>in</strong>g 52926.7 Environment, <strong>Epigenetics</strong>,and Ag<strong>in</strong>g 53026.8 <strong>Epigenetics</strong> and Age-Associated<strong>Disease</strong>s 53226.9 One Carbon Metabolism 53326.10 One-carbon Metabolism<strong>in</strong> Ag<strong>in</strong>g andNeurodegeneration 53526.11 <strong>Epigenetics</strong> andNeurodegeneration: TheAlzheimer’s <strong>Disease</strong>Paradigm 53626.12 Aged AD Mice and<strong>Epigenetics</strong> 53626.13 Conclusion 537References 53851926.1 INTRODUCTIONConrad Hal Wadd<strong>in</strong>gton, <strong>in</strong> 1942, was the first to def<strong>in</strong>e epigenetics as “the branch of biologywhich studies the causal <strong>in</strong>teractions between genes and their products, which br<strong>in</strong>g thephenotype <strong>in</strong>to be<strong>in</strong>g”. At that time, the biochemical nature of genes was unknown as well astheir role as repositories and transmitters of the genetic <strong>in</strong>formation. Wadd<strong>in</strong>gton imag<strong>in</strong>edthe epigenetics as a conceptual model to expla<strong>in</strong> his theory susta<strong>in</strong><strong>in</strong>g that different <strong>in</strong>teractionsbetween the genes and their surround<strong>in</strong>gs (or, we could say their “environment”) couldresult <strong>in</strong> different phenotypes, start<strong>in</strong>g from the same genetic material. He used the metaphorof the “epigenetic landscape” to expla<strong>in</strong> the biological development. Wadd<strong>in</strong>gton stated thatcell fates were established dur<strong>in</strong>g the development similarly to a stone (a marble) that rollsdown from high places to the po<strong>in</strong>t of lowest local elevation; the <strong>in</strong>creas<strong>in</strong>g irreversibilityassociated with cell-type differentiation was imag<strong>in</strong>ed as due to ridges, ris<strong>in</strong>g along the slopewhere the stone is roll<strong>in</strong>g down, direct<strong>in</strong>g the marble <strong>in</strong>to different valleys [1].More recently, Holliday def<strong>in</strong>ed epigenetics <strong>in</strong> a more formal way as “the study of the mechanismsof temporal and spatial control of gene activity dur<strong>in</strong>g the development of complexorganisms” [2]. Accord<strong>in</strong>g to this def<strong>in</strong>ition, the term “epigenetic” could be used to describeanyth<strong>in</strong>g other than a DNA sequence itself that is able to <strong>in</strong>fluence differentiation, physiology,T. Tollefsbol (Ed): <strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>. DOI: 10.1016/B978-0-12-388415-2.00026-3Copyright Ó 2012 Elsevier Inc. All rights reserved.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>and the fate of a cell or of a whole organism. Actually, the present use of the term is morenarrow, referr<strong>in</strong>g to the comb<strong>in</strong>ed epigenetic modifications of a given doma<strong>in</strong> of DNAsequence, characterized by heritability (over rounds of cell division and sometimes transgenerationally),that do not <strong>in</strong>volve changes to the underly<strong>in</strong>g DNA sequence. On this basis,epigenetics was recently def<strong>in</strong>ed as “the study of any potentially stable and, ideally, heritablechange <strong>in</strong> gene expression or cellular phenotype that occurs without changes <strong>in</strong> WatsoneCrickbase pair<strong>in</strong>g of DNA” [3]. Epigenetic modifications <strong>in</strong>clude (among others) methylation ofcytos<strong>in</strong>e residues of DNA and post-translational modifications of the histone prote<strong>in</strong>s.Specific comb<strong>in</strong>ations of epigenetic modifications determ<strong>in</strong>e the conformation of the chromat<strong>in</strong>fiber, thereby hav<strong>in</strong>g the possibility to regulate the transcriptional potential of theassociated genes.520Despite the advances <strong>in</strong> our knowledge about cell differentiation and epigenetic phenomena,and with the unavoidable adjustments and corrections, Wadd<strong>in</strong>gton’s model still representsa nice visualization of the epigenetics. As a matter of fact, it appears really useful to suggest thatag<strong>in</strong>g processes are particularly prone to epigenetic mechanisms. The notion Wadd<strong>in</strong>gtoncould not know, <strong>in</strong>deed, was that once differentiation has been completed (i.e. the “lowestlocal elevation” has been reached) cell fate could be not def<strong>in</strong>itive; on the contrary, thephenotype of differentiated cells still evolves either with the normal ag<strong>in</strong>g processes or asa consequence of external (pathogenic) stimuli. To resume and apply Wadd<strong>in</strong>gton’s model tothe ag<strong>in</strong>g, we can imag<strong>in</strong>e that erosive processes can change the shape of the slope and of thesurround<strong>in</strong>gs of the stone, caus<strong>in</strong>g the reprise of its roll<strong>in</strong>g down through new ridges andvalleys. Accord<strong>in</strong>g to this view, the term<strong>in</strong>ally differentiated cell is subjected to “environmental”stimuli (orig<strong>in</strong>ated either from the organism itself or from the external environment)able to <strong>in</strong>duce changes <strong>in</strong> gene expression through epigenetic mechanisms. The higher themounta<strong>in</strong>, the longer the slope; consequently, the stone encounters many more possibilities tobe subjected to changes of directions and shape. This view recalls the idea that a longer life (ofa cell or organism) is associated with a more frequent probability that epigenetic changes arise,possibly caus<strong>in</strong>g ag<strong>in</strong>g-associated dysregulation. On the basis of this metaphoric view, ag<strong>in</strong>g(and ag<strong>in</strong>g-associated diseases) represents the <strong>in</strong>evitable companion of a long life. Fortunately,epigenetic modifications are, by def<strong>in</strong>ition, reversible; consequently, they are potential targetsfor pharmacological <strong>in</strong>terventions aimed at re-establish<strong>in</strong>g the correct epigenome. Interest<strong>in</strong>gly,recent evidences <strong>in</strong>dicate that the epigenome can be also modulated by nonpharmacological<strong>in</strong>terventions like diet, physical exercise, lifestyle, and behavioral stresses.In the present chapter, evidences related to the connection between epigenetics and ag<strong>in</strong>g arepresented and discussed <strong>in</strong> the light of the most recent advances <strong>in</strong> this field of biomedicalresearch. Particular attention is devoted to the ag<strong>in</strong>g bra<strong>in</strong>, which appears to be the organ most<strong>in</strong>terest<strong>in</strong>g <strong>in</strong> normal and pathological ag<strong>in</strong>g processes, due to the relevance of neurodegenerationamong the age-associated diseases and to the recent scientific evidences <strong>in</strong>dicat<strong>in</strong>gsubstantial <strong>in</strong>volvement of epigenetic phenomena <strong>in</strong> bra<strong>in</strong> ag<strong>in</strong>g.26.2 GENES AND AGINGA common say<strong>in</strong>g among geriatricians sounds like “Life is a chronic disease with unpropitiousoutcome”. Probably, be<strong>in</strong>g faced daily with aged and diseased patients negatively <strong>in</strong>fluencesthe humor and mood of cl<strong>in</strong>icians and researchers work<strong>in</strong>g on ag<strong>in</strong>g, but the sentence isundeniably correct. Indeed, humans cannot escape (as far as we know) ag<strong>in</strong>g and, <strong>in</strong> that case,ag<strong>in</strong>g-related diseases [4]. Ag<strong>in</strong>g, although not considered a disease, is itself associated withprogressive, cognitive, physical, and physiological impairment; moreover, many diseases areage-related. The ma<strong>in</strong> risk factor for late-onset Alzheimer’s disease (LOAD), for <strong>in</strong>stance, isag<strong>in</strong>g, s<strong>in</strong>ce the <strong>in</strong>cidence of the disease is constantly <strong>in</strong>creas<strong>in</strong>g with age [5].As a matter of fact, the evolution of the human species has allowed the possibility to constantlyimprove life quality, particularly <strong>in</strong> terms of better hygiene, nutrition, protection, and medical


CHAPTER 26Ag<strong>in</strong>g and <strong>Disease</strong>: The Epigenetic Bridgecare. These improvements contributed not just to the <strong>in</strong>crease <strong>in</strong> average life expectancy butalso, <strong>in</strong> many cases, to reach and spend the oldest age <strong>in</strong> better physical and cognitivecondition than <strong>in</strong> the past. Despite this progress <strong>in</strong> life expectancy, it is <strong>in</strong>terest<strong>in</strong>g to note thatalmost no progress was observed for the oldest age that it is possible to reach (the “maximumlifespan potential”); moreover, <strong>in</strong> association with the <strong>in</strong>creased life expectancy, many (andsometimes new) diseases show an <strong>in</strong>creased morbidity dependent on ag<strong>in</strong>g [6].The existence of a genetic determ<strong>in</strong>ant of life duration is supported by the apparent impossibilityof go<strong>in</strong>g beyond a certa<strong>in</strong> maximum lifespan potential and also by the observations<strong>in</strong>dicat<strong>in</strong>g that this potential seems to be determ<strong>in</strong>ed and characteristic for each species. This<strong>in</strong>formation <strong>in</strong>duced the theory that even if we could cure or prevent the diseases mostresponsible for human death, we will be able to just further extend life expectancy, but won’tbe able to significantly overcome the maximum lifespan potential determ<strong>in</strong>ed by the advent offatal age-associated physiological impairment [7]. This hypothesis depicts the age-relateddiseases, ma<strong>in</strong>ly the neurodegenerative diseases, as the price to pay to survive cancer; <strong>in</strong> otherwords, the organism has two possible fates dur<strong>in</strong>g ag<strong>in</strong>g: fac<strong>in</strong>g cells prone to errors that wouldshift toward a relatively longer (cancerous) life, or cop<strong>in</strong>g with the progressive cell impairmentand death due to physiological cellular degeneration.Molecular and cellular degeneration, result<strong>in</strong>g <strong>in</strong> the ag<strong>in</strong>g of the whole organism, are oftenevident and clearly observable, although the <strong>in</strong>itial and causal molecular mechanisms aredifficult to study because of their extreme complexity, <strong>in</strong>terconnections, presence of sideeffects, and concurrent external and environmental factors [8]. The study of the picturerepresent<strong>in</strong>g the age-associated diseases is complicated by the possible early start of thepathological mechanisms, possibly <strong>in</strong>itiat<strong>in</strong>g <strong>in</strong> early age, and also by the above-cited difference<strong>in</strong> the regulation of ag<strong>in</strong>g mechanisms <strong>in</strong> different organisms, which makes it difficult touse surrogated animal models to study human ag<strong>in</strong>g.In this context, it is not easy to hypothesize a unify<strong>in</strong>g and comprehensive theory of ag<strong>in</strong>g.Initially, it was postulated that biological ag<strong>in</strong>g was dependent on changes resid<strong>in</strong>g <strong>in</strong> the<strong>in</strong>formation-conta<strong>in</strong><strong>in</strong>g molecules, lead<strong>in</strong>g to formulate the “error theory”, the “redundantmessage theory”, the “codon restriction theory”, and the “transcriptional event theory” [9].Ina second moment, the attention of the researchers has been shifted to the hypothesis that possible“gerontogenes”, ma<strong>in</strong>ly <strong>in</strong>volved <strong>in</strong> DNA repair, could be responsible for cellular senescence [10].To summarize, it is possible to differentiate two major groups of theories: stochastic and genetic;however, it becomes evident that these groups could be, <strong>in</strong> some cases, not mutually exclusive.A list of the pr<strong>in</strong>cipal theories expla<strong>in</strong><strong>in</strong>g causes and possible mechanisms of ag<strong>in</strong>g is reportedhere [8,11]:5211. Evolutionary: evolution presses the organisms to reach the reproductive age, procreate, andcare for the offspr<strong>in</strong>g. Accord<strong>in</strong>g to this po<strong>in</strong>t of view, the physiology of an organism afterthe end of the reproductive period could be the manifestation of the epigenetic eventsoccurr<strong>in</strong>g on the basis of the genetic development dur<strong>in</strong>g the previous stage of the life. Theconclusion is that cellular senescence could be the price to pay <strong>in</strong> order to avoid otherdamage, like tumorigenesis, potentially caused by the prolonged expression of the genes<strong>in</strong>volved <strong>in</strong> the reach<strong>in</strong>g of reproductive fitness [12].2. Stochastic: the stochastic theories are generally based on the idea that ag<strong>in</strong>g is caused bycasual damage to cell molecules, particularly to DNA.2.1. Mutation and repair: DNA is subjected to cont<strong>in</strong>uous damage that <strong>in</strong>ducesmutations. The effect is cumulative and becomes <strong>in</strong>creas<strong>in</strong>gly evident because of theloss of the DNA repair ability [9].2.2. Error-catastrophe: casual errors <strong>in</strong> the synthesized prote<strong>in</strong>s are normally bypassed bythe normal molecular turnover, but errors <strong>in</strong> the molecules <strong>in</strong>volved <strong>in</strong> prote<strong>in</strong> orDNA synthesis or <strong>in</strong> postsynthetic modification could represent the start of a process


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>522of error amplification, result<strong>in</strong>g <strong>in</strong> the accumulation of the wrong molecules able to<strong>in</strong>terfere with normal cell physiology [13].2.3. Prote<strong>in</strong> modification: the worsen<strong>in</strong>g of the enzymatic activities <strong>in</strong> ag<strong>in</strong>g could bea consequence of the altered postsynthetic modifications, altered turnover andprote<strong>in</strong>s cross-l<strong>in</strong>k<strong>in</strong>g [13].2.4. Oxidative stress: this is one of the most <strong>in</strong>vestigated areas of cellular senescence; the<strong>in</strong>volvement of free radicals and the alteration of the oxidative status <strong>in</strong> ag<strong>in</strong>g hasbeen characterized <strong>in</strong> several models and organisms and <strong>in</strong> different pathologiesassociated with older age, like Alzheimer’s disease and Park<strong>in</strong>son’s disease. Thebalance between pro- and antioxidants <strong>in</strong> the cell is f<strong>in</strong>ely and complexly regulatedand the impairment of this regulation is critical to mitochondrial, cellular, and tissuephysiology dur<strong>in</strong>g ag<strong>in</strong>g [14].3. Genetic: <strong>in</strong> the genetic (or developmental) theories, ag<strong>in</strong>g is considered as a programmedand genetically controlled process of maturation, successive to the development of theorganism or cell. These theories are supported by the elevated species-specificity of themaximum lifespan but are <strong>in</strong> contrast with the variable control and manifestation of ag<strong>in</strong>g<strong>in</strong> different <strong>in</strong>dividuals of the same species.3.1. Longevity genes: there are several evidences about the existence of genetic elementsable to regulate senescence, <strong>in</strong> particular responsible for the regulation of themaximum lifespan. Studies regard<strong>in</strong>g the role of genes <strong>in</strong>volved <strong>in</strong> the <strong>in</strong>crement oflifespan were primarily performed on “simple” eukaryotes like yeast and C. elegans,but significant data also arise from studies on D. melanogaster and rodents [15,16].3.2. Ag<strong>in</strong>g syndromes: the existence of a number of human genetic diseases (Hutch<strong>in</strong>son’ssyndrome, Werner’s syndrome, Down’s syndrome) display<strong>in</strong>g some characteristicsdist<strong>in</strong>ctive of accelerated ag<strong>in</strong>g, lead to hypothesize that ag<strong>in</strong>g could be a k<strong>in</strong>d ofdisease itself, regulated by specific genes. Recently, different transgenic mouse modelsshow<strong>in</strong>g ag<strong>in</strong>g phenotypes similar to those observed <strong>in</strong> humans were also settled [17].3.3. Neuroendocr<strong>in</strong>e theory: this is based on the importance of the hormones secreted <strong>in</strong>the bra<strong>in</strong> (hypothalamic, pituitary, and adrenal hormones) <strong>in</strong> the regulation oforganismic ag<strong>in</strong>g and on the decrement <strong>in</strong> bra<strong>in</strong> neurons [18].3.4. Immunologic theory: this is based on the decreased T-cell response and <strong>in</strong>creasedautoimmune reactions dur<strong>in</strong>g ag<strong>in</strong>g [19]. As for the neuroendocr<strong>in</strong>e theory, the weakpo<strong>in</strong>t is that complex immune and neuronal systems are not present <strong>in</strong> simpleeukaryotes although they show characteristics of ag<strong>in</strong>g comparable to higher organisms.3.5. Cellular senescence: cellular cultures were used as a model for the comprehension ofsenescence processes due to their usefulness <strong>in</strong> study<strong>in</strong>g the basic molecularmechanisms, unlike the whole organisms. Data on the genetic effectors responsiblefor the regulation of cell senescence susta<strong>in</strong> the hypothesis that organismic ag<strong>in</strong>greflects the senescence of s<strong>in</strong>gle cell l<strong>in</strong>es or tissues. Cellular senescence is often<strong>in</strong>dicated as “replicative senescence”, s<strong>in</strong>ce the genes <strong>in</strong>volved <strong>in</strong> this phenomenonare ma<strong>in</strong>ly genes related to the replication mach<strong>in</strong>ery and s<strong>in</strong>ce the cellularsenescence becomes evident through decl<strong>in</strong>e <strong>in</strong> growth rate and proliferative activityand alterations <strong>in</strong> the signal transduction and adaptive response pathways. All thesealterations characterize a senescent cell growth status, which is quite different fromthe young cells [20]. The first event characterized as a potential cellular clock was themechanism of telomere shorten<strong>in</strong>g [21]. The function of telomeres (sequences ofnon-cod<strong>in</strong>g DNA at the end of chromosomes) is to avoid the degradation of cod<strong>in</strong>gregions and fusion with other DNA sequences; the reduction of the telomeric lengthwith cell ag<strong>in</strong>g is the ma<strong>in</strong> evidence <strong>in</strong> cellular senescence. Another two genes of thereplicative mach<strong>in</strong>ery, ret<strong>in</strong>oblastoma and p53, are well known to be <strong>in</strong>volved <strong>in</strong> cellsenescence; their activity is generally <strong>in</strong>creased <strong>in</strong> senescent cells [22]. The complexityof the mechanisms of cell cycle progression and the balance between positive andnegative regulators is due to the importance of this regulation <strong>in</strong> order to avoid the


CHAPTER 26Ag<strong>in</strong>g and <strong>Disease</strong>: The Epigenetic Bridgeonset of tumorigenic events <strong>in</strong> the cell. These data are consistent with the hypothesisthat cellular senescence has evolved as a mechanism of tumor suppression [8].3.6. Cell death: strictly l<strong>in</strong>ked to the mechanisms of cellular replication and senescence,the mechanism of apoptosis is considered as a cause of ag<strong>in</strong>g s<strong>in</strong>ce it consists ofa process of active, gene-dependent and <strong>in</strong>jury-<strong>in</strong>dependent cell death [23].More recently, evidence that epigenetic mechanisms could have a role <strong>in</strong> cellular degenerationand ag<strong>in</strong>g has been supported by technical advances allow<strong>in</strong>g a detailed study of the epigenomeand of the epigenetic mechanisms and by the discovery of a complex, non-Mendelian,nature of many age-associated disorders.In yeast and mice, significant changes <strong>in</strong> gene expression dur<strong>in</strong>g cellular degeneration arerelated to significant and net loss of heterochromat<strong>in</strong>, with consequent overexpression ofheterochromat<strong>in</strong>-associated silenced genes [24]. Changes <strong>in</strong> chromat<strong>in</strong> structure are associatedwith epigenetic modifications that consist of DNA methylation, DNA hydroxymethylation,histone post-transcriptional modifications (methylation, acetylation, andphosphorylation), and ATP-mediated chromat<strong>in</strong> modifications. DNA is associated witheuchromat<strong>in</strong> <strong>in</strong> actively transcripted loci <strong>in</strong> proliferat<strong>in</strong>g cells; these loci ma<strong>in</strong>ly conta<strong>in</strong> genes<strong>in</strong>volved <strong>in</strong> cell growth regulation and <strong>in</strong> basal cell metabolism [25]. For this reason, it hasbeen proposed that loss of repressive chromat<strong>in</strong> doma<strong>in</strong>s (heterochromat<strong>in</strong>) may contributeto cellular degeneration and ag<strong>in</strong>g processes.The observed heterochromat<strong>in</strong> loss is <strong>in</strong> agreement with, and apparently l<strong>in</strong>ked to, thegenerally observed loss of DNA methylation <strong>in</strong> the elderly [26,27]. Other experimentalevidence also po<strong>in</strong>ts out the spread<strong>in</strong>g of DNA methylation and the presence of hypermethylatedDNA <strong>in</strong> senescence and cancer [28,29]; on the one hand, these contrast<strong>in</strong>g datastress the complexity of the epigenetic phenomena and contribute to confound the picture but,on the other, the contrast can be clarified when DNA methylation is analyzed at a sequencespecificlevel <strong>in</strong> regions and sequences undergo<strong>in</strong>g changes of methylation dur<strong>in</strong>g ag<strong>in</strong>g.Observed global changes <strong>in</strong> DNA methylation, at genomic level, could <strong>in</strong>deed be due tochanges <strong>in</strong> methylation of bulk or non-regulatory DNA regions. The role of these changes isnot yet ascerta<strong>in</strong>ed and we cannot conclude it is directly associated with changes <strong>in</strong> geneexpression, unlike methylation changes <strong>in</strong> promoters and other regulatory DNA sequences.However, the evidence about the general DNA methylation decreas<strong>in</strong>g <strong>in</strong> proliferat<strong>in</strong>g cellcultures and <strong>in</strong> normal organismic ag<strong>in</strong>g, but not <strong>in</strong> immortal cell l<strong>in</strong>es, and the concurrentimpairment of the DNA methyltransferase activity, stress the hypothesis that DNA methylationis susceptible to ag<strong>in</strong>g-l<strong>in</strong>ked variations and that these variations have a specific role <strong>in</strong> theag<strong>in</strong>g processes [30]. One of the ma<strong>in</strong> questions to be ascerta<strong>in</strong>ed <strong>in</strong> this sense <strong>in</strong>volves the“cause or consequence” role of DNA methylation <strong>in</strong> ag<strong>in</strong>g.DNA methylation is strictly related to histone modification and “pack<strong>in</strong>g”; <strong>in</strong> turn, histonemodifications can <strong>in</strong>fluence gene transcription by modulat<strong>in</strong>g chromat<strong>in</strong> assembly. Histoneacetylation is the most studied modification; it is regulated by the classical histone deacetylases(HDAC) family and by the SIR2 (silent <strong>in</strong>formation regulator) family. It is worthwhile tounderl<strong>in</strong>e that the SIR2 family was also identified as be<strong>in</strong>g <strong>in</strong>volved <strong>in</strong> the regulation of lifespan<strong>in</strong> other eukaryotes [31,32].Other epigenetic modifications, as histone methylation and ATP-mediated chromat<strong>in</strong>remodel<strong>in</strong>g, are not yet well characterized for their role <strong>in</strong> cellular degeneration and ag<strong>in</strong>g, but,as a matter of fact, it was shown that euchromat<strong>in</strong>ization of specific doma<strong>in</strong>s or, vice versa,heterochromat<strong>in</strong>ization of specific growth genes, due to alterations <strong>in</strong> balance of chromat<strong>in</strong>modify<strong>in</strong>g molecules, is modulated dur<strong>in</strong>g ag<strong>in</strong>g [33].Besides their role <strong>in</strong> normal cellular senescence, epigenetic mechanisms are also to beconsidered <strong>in</strong> the light of their association (DNA methylation <strong>in</strong> particular) to cancer onsetand development, s<strong>in</strong>ce it is well known that cancer onset <strong>in</strong>creases with ag<strong>in</strong>g.523


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>524Methylation, ag<strong>in</strong>g, and cancer are representatively connected <strong>in</strong> the observed CpG islandmethylation <strong>in</strong>creas<strong>in</strong>g associated to the estrogen receptor (ER) gene, which resulted ageassociatedly<strong>in</strong> the normal human colonic mucosa. ER gene hypermethylation was also found<strong>in</strong> all colonic tumors exam<strong>in</strong>ed <strong>in</strong>clud<strong>in</strong>g small adenomas, suggest<strong>in</strong>g that methylationdependent<strong>in</strong>activation of the ER gene <strong>in</strong> the ag<strong>in</strong>g colorectal mucosa is one early event <strong>in</strong>colorectal tumorigenesis. Other CpG islands <strong>in</strong> promoter regions of several genes exhibit agerelatedhypermethylation <strong>in</strong> colon mucosa [29,34]. Most of the CpG islands found hypermethylated<strong>in</strong> primary colon tumors were hypermethylated to a lesser extent <strong>in</strong> the ag<strong>in</strong>gcolon, but a m<strong>in</strong>or number of islands were hypermethylated only <strong>in</strong> subsets of colon cancers.These f<strong>in</strong>d<strong>in</strong>gs stress the hypothesis that two k<strong>in</strong>ds of methylation exist: (1) one age-relatedmethylation, presents <strong>in</strong> the normal mucosa as a function of the age and (2) a cancer-relatedmethylation, not observed <strong>in</strong> normal colon. The mechanisms responsible for the concurrent<strong>in</strong>duction of global DNA hypomethylation and sequence-specific <strong>in</strong>crease <strong>in</strong> de novo methylationwith ag<strong>in</strong>g are still an open field of research. It is well known that different DNAmethyltransferase(DNMT) enzymes with peculiar functions exist. Ma<strong>in</strong>tenance methylationof hemimethylated DNA dur<strong>in</strong>g cellular replication is guarantee by DNMT1, which is the mostabundant. Other DNMTs (namely, DNMT3a and DNMT3b) have a high aff<strong>in</strong>ity for unmethylatedDNA and were, for this reason, <strong>in</strong>dicated as de novo methyltransferases, able tomethylate DNA without pre-exist<strong>in</strong>g hemimethylation. These methylation mechanisms areschematized <strong>in</strong> Figure 26.1. S<strong>in</strong>ce the levels of DNMT1 decrease with ag<strong>in</strong>g, it can behypothesized that the overall decrease <strong>in</strong> DNA methylation can be referred to ma<strong>in</strong>tenancemethylation, whereas the sequence-specific <strong>in</strong>crease <strong>in</strong> de novo methylation depends on the<strong>in</strong>creased activity of the other DNMTs [35]. The observed general decrease <strong>in</strong> total genomicmethylation with ag<strong>in</strong>g <strong>in</strong> different organisms and the apparently f<strong>in</strong>ite number of cell divisionscharacteristic of most somatic cells stress the view of overall DNA methylation (or evenmethyl-cytos<strong>in</strong>e residues) loss as a cellular countdown mechanism to trigger normal cellularsenescence or degeneration.More recently, thanks to the power of the genome-wide studies compar<strong>in</strong>g younger to oldersubjects, it was possible to confirm on a large-scale basis that methylation changes (both <strong>in</strong> thedirection of hyper- and hypo-methylation) are associated with ag<strong>in</strong>g, both <strong>in</strong> humans [36e38]and <strong>in</strong> animal models [39]. Even after these recent results, the idea that the methylation statusof a larger part of the exam<strong>in</strong>ed genes and sequences seem unchanged dur<strong>in</strong>g ag<strong>in</strong>g [35] is, sofar, still preserved. This is not at all, of course, a “negative” or controversial result for thedisciples of the epigenetic theory of ag<strong>in</strong>g, but it just po<strong>in</strong>ts out the idea that the age-associatedepigenetic drift targets specific genes <strong>in</strong>volved <strong>in</strong> ag<strong>in</strong>g processes. The alterations <strong>in</strong> CpG islandmethylation are crucial to modulate the b<strong>in</strong>d<strong>in</strong>g of transcription factors and methyl-DNAb<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s. Aberrant methylation of CpG islands <strong>in</strong> the promoter region may contributeto the progressive <strong>in</strong>activation of growth-<strong>in</strong>hibitory genes dur<strong>in</strong>g ag<strong>in</strong>g, result<strong>in</strong>g <strong>in</strong> the clonalselection of cells with growth advantage towards cancer development.DNA methylation also presents a l<strong>in</strong>k with the regulation of telomerase activity, another keyfactor <strong>in</strong> the regulation of senescence. <strong>Human</strong> telomerase consists of an RNA component(hTR) and a catalytic subunit with reverse transcriptase activity (hTERT). Most differentiatedsomatic cells lack detectable telomerase activity, <strong>in</strong> contrast to the high activities found <strong>in</strong>immortalized cancer cells, germl<strong>in</strong>e cells, and stem cells and the high GC content of the hTERTpromoter <strong>in</strong>dicates that DNA methylation may be important <strong>in</strong> controll<strong>in</strong>g hTERT expression.One emerg<strong>in</strong>g theory <strong>in</strong>dicates that telomere control and DNA methylation may be strictly<strong>in</strong>volved <strong>in</strong> the ag<strong>in</strong>g processes; <strong>in</strong> fact DNMT1 expression results decreased <strong>in</strong> ag<strong>in</strong>g humanfibroblasts, with consequent decreas<strong>in</strong>g of methylation activity <strong>in</strong> the cells, but hTERT was ableto activate DNMT1 expression avoid<strong>in</strong>g the loss of methylation [40].All the above-reported data evidence that the methylation pattern established dur<strong>in</strong>g thedevelopment is not stable or def<strong>in</strong>itive <strong>in</strong> adult life and, <strong>in</strong> particular, dur<strong>in</strong>g ag<strong>in</strong>g. Therefore,


CHAPTER 26Ag<strong>in</strong>g and <strong>Disease</strong>: The Epigenetic Bridge525FIGURE 26.1Schematic representation of the DNA methylation reactions: ma<strong>in</strong>tenance methylation, passive demethylation, de novo methylation, and active demethylation.(A) After rounds of cell divisions, methylated sequences result <strong>in</strong> demethylated neosynthesized DNA molecules (passive methylation) unless the ma<strong>in</strong>tenancemethylase activity guarantees the transmission of the methylation pattern reta<strong>in</strong>ed by the parental DNA strand. (B) De novo methylase activity has the ability tomethylate DNA sequences <strong>in</strong>dependently on the presence of methyl groups <strong>in</strong> one of the two strands, whereas active demethylase activity has the ability toremove methyl groups; both these activities can be exerted <strong>in</strong> absence of cellular replication. This figure is reproduced <strong>in</strong> the color plate section.the epigenetic approach seems promis<strong>in</strong>g for the understand<strong>in</strong>g of the mechanisms thatregulate ag<strong>in</strong>g and age-related diseases, whereas the consideration that the preservation ofepigenetic patterns may help the ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g of a healthy status could provide the basis fora therapeutic and preventive action.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>52626.3 THE DYNAMIC METHYLOMEThe idea that epigenetic changes are responsible for ag<strong>in</strong>g and age-related diseases directlyimplies that the epigenome is a dynamic entity, undergo<strong>in</strong>g modulation after particularstimuli. DNA methylation was long considered as the repository of the epigenetic “message”s<strong>in</strong>ce the methylation pattern could be transmitted to the offspr<strong>in</strong>g and, once established atthe end of the developmental processes, could be ma<strong>in</strong>ta<strong>in</strong>ed dur<strong>in</strong>g cell replication <strong>in</strong>a semiconservative manner. This feature is permitted by the ability of the ma<strong>in</strong>tenanceDNMT(s) to replicate <strong>in</strong> the newly synthesized DNA strand the methylation pattern of theparent strand. For the last two decades it was supposed that the only mechanism lead<strong>in</strong>g toDNA demethylation could be the passive, replication-dependent, loss of methyl moietiesoccurr<strong>in</strong>g when the ma<strong>in</strong>tenance DNMT activity was defective dur<strong>in</strong>g cell replication. Thishypothesis stressed the impossibility that DNA demethylation could occur <strong>in</strong> adult organisms,at least <strong>in</strong> non-proliferat<strong>in</strong>g tissues, depict<strong>in</strong>g a DNA methylation pattern that is, onceestablished, relatively fixed and stable [41]. These assumptions were <strong>in</strong>itially contrasted by theidentification of putative DNA demethylase activity (Figure 26.1) <strong>in</strong> different candidateprote<strong>in</strong>s like 5-methylcytos<strong>in</strong>e-DNA glycosilase, Gadd45a, MBD2, MBD2b, MBD3, MBD4[42e48]. It was even evidenced that DNMTs could be associated with demethylase activity[45,49,50]. Among these putative demethylases, MBD2 is the best characterized thanks to thepioneer<strong>in</strong>g work by the group of M. Szyf: they hypothesized that MBD2 can directly b<strong>in</strong>d tomethylated CpGs remov<strong>in</strong>g only the methyl group without the cytos<strong>in</strong>e excision postulated byothers [46]. Data are still not def<strong>in</strong>itive and the possibility that MBD2 could <strong>in</strong>teract with other(effectors) prote<strong>in</strong>s could not be excluded; if the real nature of the active DNA demethylase isnot yet def<strong>in</strong>itely revealed, the existence of such enzymatic activity cannot be ignored anylonger [51]. In our laboratory, we obta<strong>in</strong>ed earlier <strong>in</strong>direct <strong>in</strong>dications that rapid demethylation,not compatible with the time necessary for cellular replication, occurred at a specific CpGsite of myogen<strong>in</strong> gene promoter dur<strong>in</strong>g myogenic differentiation <strong>in</strong> vitro [52]. More recently,we obta<strong>in</strong>ed evidence, <strong>in</strong> the same experimental system, that DNA demethylation pattern ofmyogen<strong>in</strong> gene promoter is regulated by a dynamic balance between DNA methylase anddemethylase activities and that <strong>in</strong>hibition of DNMTs results <strong>in</strong> improved active demethylation,non necessarily dependent on DNA replication [53]. In a different experimental model, morestrictly connected to age-related disease processes, we were able to demonstrate that DNAmethylase and demethylase activities could be modulated <strong>in</strong> the adult bra<strong>in</strong>. Us<strong>in</strong>g a transgenicmouse model of Alzheimer’s disease we showed that the <strong>in</strong>hibition of the metabolic pathwaythat generates the methyl donor S-adenosylmethion<strong>in</strong>e resulted <strong>in</strong> the impairment of themethylase activities and the improvement of the demethylase activity <strong>in</strong> mice bra<strong>in</strong> d a tissueknown for its scarce cellular proliferation. On the contrary, the supplementation of the methyldonor resulted <strong>in</strong> <strong>in</strong>creased DNA methylation and decreased DNA demethylation activities.These modulations had a direct effect on the methylation pattern of Presenil<strong>in</strong>1 (PSEN1) gene,which is <strong>in</strong>volved <strong>in</strong> amyloid process<strong>in</strong>g <strong>in</strong> Alzheimer’s disease [54,55]. We can thereforeconclude that the methylation pattern of specific genes is not fixed <strong>in</strong> adults and nonproliferat<strong>in</strong>gtissues but undergoes dynamic regulation under appropriate stimuli.26.4 EPIGENETIC DYNAMICS IN THE AGING BRAINThe bra<strong>in</strong> is certa<strong>in</strong>ly the human organ most hit by ag<strong>in</strong>g: it shows sensitive impairment <strong>in</strong>normal ag<strong>in</strong>g, and appears more prone than other organs to the occurrence of ag<strong>in</strong>g-relateddiseases. Neurodegenerative disorders represent the ma<strong>in</strong> class of age-associated diseases and,among these, Alzheimer’s disease represents the most prevalent form of neurodegenerativedisease. Moreover, many of these disorders have been recently associated with epigenetic events.For this reasons, it seems of particular relevance <strong>in</strong> the discussion of the epigenetic changesoccurr<strong>in</strong>g <strong>in</strong> the bra<strong>in</strong> and observed <strong>in</strong> adulthood and ag<strong>in</strong>g; an excellent and comprehensivereview of these mechanisms was recently published by J. Rogers and colleagues [56].


CHAPTER 26Ag<strong>in</strong>g and <strong>Disease</strong>: The Epigenetic BridgeAs previously discussed, the knowledge that postmitotic cells could present functional alterationsof the methylation patterns is a quite recent acquisition. In recent years, it wasdemonstrated that dynamic and rapid (active) DNA methylation and demethylation arepresent <strong>in</strong> postmitotic cells and, <strong>in</strong> particular, <strong>in</strong> neurons [45,50,57] stress<strong>in</strong>g the <strong>in</strong>volvementof epigenetic processes <strong>in</strong> neuronal plasticity. The paper by Murgatroyd and colleagues [50] hasto be considered as a milestone <strong>in</strong> this area. In their work, they demonstrated that early lifestress <strong>in</strong> mice was associated with behavioral changes <strong>in</strong> adult life via a mechanism <strong>in</strong>volv<strong>in</strong>gepigenetic modifications of hypothalamic neurons. Early exposure of mice to environmentalstress dur<strong>in</strong>g the first 10 days of life resulted <strong>in</strong> impaired avoidance learn<strong>in</strong>g, susta<strong>in</strong>edhyperactivity of the hypothalamicepituitaryeadrenal axis, and corticosterone and pituitaryadrenocorticotrop<strong>in</strong> prohormone hypersecretion. These effects were l<strong>in</strong>ked to arg<strong>in</strong><strong>in</strong>evasopress<strong>in</strong>(AVP) overexpression by parvocellular neurons of the hypothalamic paraventricularnucleus, and to hypomethylation of specific CpG sites with<strong>in</strong> the AVP gene. Theauthors also showed the age-dependent hypomethylation of AVP gene at different CpGsites <strong>in</strong> control mice, although hypomethylation of CpGs with<strong>in</strong> a CpG island <strong>in</strong>cluded <strong>in</strong> theAVP enhancer region were responsible for AVP overexpression <strong>in</strong> mice that underwent theearly-life stress. Hypomethylation of this CpG island was specific to the paraventricularnucleus, suggest<strong>in</strong>g a possible mechanism for the observed epigenetic change that <strong>in</strong>volvesthe b<strong>in</strong>d<strong>in</strong>g of MeCP2 prote<strong>in</strong> to the CpG sites <strong>in</strong> this island. The authors f<strong>in</strong>allydemonstrated that phosphorylation of MeCP2 by calmodul<strong>in</strong>-dependent prote<strong>in</strong> k<strong>in</strong>ase IIdecreased MeCP2 occupancy of CpGs <strong>in</strong> the CpG island, enhanc<strong>in</strong>g gene expression. Experimentalmodels tak<strong>in</strong>g advantage of early-life stresses represent promis<strong>in</strong>g approaches to studythe modification of adult stress response, cognition and behavior, <strong>in</strong>duced by epigeneticmodifications [58,59].A different approach was used to study the age-dependent decrease of caspase-3 <strong>in</strong> rat bra<strong>in</strong>,associated with alterations of the methylation pattern of specific CpG sites <strong>in</strong> the promoter ofthe gene [60]. The promoter sequence <strong>in</strong>terested by methylation alteration lies <strong>in</strong> a region ofthe promoter necessary for its activity. This region is predicted to b<strong>in</strong>d the transcription factorsETS-1 and -2, which are important, besides <strong>in</strong> controll<strong>in</strong>g caspase-3 transcription, <strong>in</strong> regulat<strong>in</strong>gneuronal differentiation and death. S<strong>in</strong>ce these two factors seem not to be altered dur<strong>in</strong>gag<strong>in</strong>g, it is possible to appreciate the relevance of methylation status, mediat<strong>in</strong>g transcriptionfactors b<strong>in</strong>d<strong>in</strong>g and activity.527Dynamic changes of methylation patterns even showed cyclical regulation associated withcyclical activation/<strong>in</strong>activation of transcription [45,57]. In this model, DNMT3a and 3b areresponsible for deam<strong>in</strong>ation of methylated CpG sites, lead<strong>in</strong>g to a process of active demethylationmediated by an excision/repair mechanism; DNA demethylation <strong>in</strong>duces genetranscription, followed by MeCP2 and DNMTs new recruitment responsible for new DNAmethylation and gene silenc<strong>in</strong>g.While much is known about DNA methylation mechanisms and the possible role of DNAmethylation dysregulation has become an <strong>in</strong>tense field of research <strong>in</strong> respect to ag<strong>in</strong>g and agerelatedneurodegeneration [61], little is known about the relevance to the ag<strong>in</strong>g processes ofDNA hydroxymethylation, a relatively new epigenetic modification recently identified [62].5-Hydroxymethylcytos<strong>in</strong>e (5-hmC) is generated by the action of the ten-eleven translocationenzymes (TET), which are able to hydroxylate methylated cytos<strong>in</strong>es [63,64]. 5-hmCs is thenformed after a previous cytos<strong>in</strong>e methylation and is not efficiently recognized by methylb<strong>in</strong>d<strong>in</strong>gprote<strong>in</strong>s [65,66]. Unlike DNA methylation, hydroxymethylation is generallyassociated with euchromat<strong>in</strong>ization and gene expression [67,68]. A role for this epigeneticmodification <strong>in</strong> ag<strong>in</strong>g could be hypothesized on the basis of the work by Chouliaras andcolleagues that recently showed an <strong>in</strong>crease <strong>in</strong> hydroxymethylation <strong>in</strong> ag<strong>in</strong>g mouse hippocampusassociated with <strong>in</strong>creases <strong>in</strong> Dnmt3a levels [69]; <strong>in</strong>terest<strong>in</strong>gly, this <strong>in</strong>crease wasprevented by caloric restriction (CR) [70].


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>As for DNA methylation and demethylation, a recent study suggested that also histone acetylationand deacetylation are subjected to dynamic and rapid changes that can <strong>in</strong>fluence genetranscription [71]. Histone modifications are <strong>in</strong>volved <strong>in</strong> several neurobiological processes <strong>in</strong>the differentiated bra<strong>in</strong>, operat<strong>in</strong>g dynamic regulatory mechanisms <strong>in</strong> postmitotic neurons:post-traumatic stress disorders [72], addiction [73], chol<strong>in</strong>e acetyltransferase activity regulation[74], GDNF and BDNF transcription [75], and microglial apoptosis [76]. Moreover,histone modifications are known to be associated with different neurological and neurodegenerativedisorders, <strong>in</strong>clud<strong>in</strong>g Park<strong>in</strong>son’s disease [75], motor neuron disease [77], multiplesclerosis [78], and X-l<strong>in</strong>ked mental retardation [79].Another class of molecules with transcriptional regulatory functions, today <strong>in</strong>cluded <strong>in</strong> the listof the epigenetic mechanisms, is represented by the microRNAs (miRNAs). The study of theseregulatory molecules is fac<strong>in</strong>g an <strong>in</strong>creas<strong>in</strong>g boost <strong>in</strong> the very recent times s<strong>in</strong>ce miRNAs werefound to be associated with different physiological and pathological processes, also <strong>in</strong>volv<strong>in</strong>gthe bra<strong>in</strong>, and appear modulated <strong>in</strong> postmitotic neurons [80]. For example, miRNA-329,miRNA-134, and miRNA-381, which are <strong>in</strong>duced by neuronal activity, seem to be essential forthe dendritic outgrowth of hippocampal neurons [81] and were also <strong>in</strong>vestigated <strong>in</strong> variousneurological disorders [82].52826.5 THE COMPLEXITY OF THE AGE-ASSOCIATED EPIGENETICCHANGESBoth normal and pathological ag<strong>in</strong>g processes <strong>in</strong> the bra<strong>in</strong> are characterized by <strong>in</strong>creased DNAdamage, synaptic dysfunction, bra<strong>in</strong> shr<strong>in</strong>kage, structural bra<strong>in</strong> changes, and cognitive decl<strong>in</strong>e[83e85]. As a matter of fact, specific bra<strong>in</strong> regions such as cortex and hippocampus appearmore prone to be <strong>in</strong>terested by these ag<strong>in</strong>g-related changes [86e88]. This region-specificitycould be also observed when alterations of gene transcription are analyzed. These alterationscan be summarized, on the one hand, <strong>in</strong> down-regulation of genes devoted to ma<strong>in</strong>ta<strong>in</strong>synaptic plasticity, neurotrophic support to neurons, neurotransmitter synthesis, and DNArepair mach<strong>in</strong>ery and, on the other, <strong>in</strong> up-regulation of immune-related genes [61,89,90].A role for epigenetic mechanisms, which are responsible for regulat<strong>in</strong>g gene expression, is nowclearly claimed <strong>in</strong> these ag<strong>in</strong>g-related changes [91e93].As previously stated for Alzheimer’s disease, ag<strong>in</strong>g is the most evident risk factor associatedwith ag<strong>in</strong>g-related diseases [94]. Unfortunately, the causal or consequential nature of thisassociation is not at all yet clear, although a number of possible concurrent alterations wereidentified both <strong>in</strong> normal and pathological ag<strong>in</strong>g; these <strong>in</strong>clude oxidative stress, sexualhormone effects, calcium dyshomeostasis, neurotransmitter and glucocorticoid deregulation,neuro<strong>in</strong>flammation, diabetes-associated alterations, neurovascular deficits, toxic prote<strong>in</strong>deposition, and transcriptional alterations of many genes [91,95e103]. Epigenetic modifications,be<strong>in</strong>g <strong>in</strong>volved <strong>in</strong> ag<strong>in</strong>g-associated changes <strong>in</strong> different experimental models andorganisms, could represent a l<strong>in</strong>k between normal and pathological ag<strong>in</strong>g [104].As <strong>in</strong> other organisms [105], progressive, age-related and genome-wide DNA hypomethylationwas observed <strong>in</strong> humans both <strong>in</strong> vivo and <strong>in</strong> <strong>in</strong> vitro models [106,107] and was associated withthe concurrent DNMT1 impairment [35,108]. Age-associated loss of DNA methylation wasalso associated with the parallel <strong>in</strong>crease <strong>in</strong> S-adenosylhomocyste<strong>in</strong>e [109] caus<strong>in</strong>g the <strong>in</strong>hibitionof methyltransferase reactions (as discussed <strong>in</strong> depth <strong>in</strong> the prosecution of this chapter).Besides this general loss of methylation, specific hypomethylation of both cod<strong>in</strong>g and noncod<strong>in</strong>gregions was observed dur<strong>in</strong>g ag<strong>in</strong>g.As for non-cod<strong>in</strong>g regions, s<strong>in</strong>ce these are normally repressed by methylation, it seems possiblethat hypomethylation processes activat<strong>in</strong>g repetitive sequences, retrotransposons, andendogenous retroviruses occurr<strong>in</strong>g with ag<strong>in</strong>g, could <strong>in</strong>duce chromosome <strong>in</strong>stability andretroviruses activation [110]. Moreover, a very recent paper evidenced that hypomethylation of


CHAPTER 26Ag<strong>in</strong>g and <strong>Disease</strong>: The Epigenetic Bridgenon-cod<strong>in</strong>g sequences like LINE-1 is possibly associated with neurodegeneration and, <strong>in</strong>particular, with LOAD [111].The effect of the age-related hypomethylation on cod<strong>in</strong>g regions and specific genes <strong>in</strong> humanswas also particularly studied <strong>in</strong> LOAD, probably because it is the most frequent pathology <strong>in</strong>older people. The work by Tohgi and his group, for example, showed hypomethylation ofAmyloid Precursor Prote<strong>in</strong> (APP) gene promoter <strong>in</strong> the bra<strong>in</strong> of older people and also complexvariations <strong>in</strong> methylation patterns of Tau and RAGE (Receptor for Advanced Glycation Endproducts)gene promoters [112e114]. Other researchers evidenced the age-dependent hypomethylationof regulatory regions of the immune/<strong>in</strong>flammatory antigen CD11a [115].Examples of gene-specific hypermethylation related to ag<strong>in</strong>g were also evidenced, togetherwith the silenc<strong>in</strong>g of the associated genes. This was particularly observed for genes associatedwith CpG islands like tumor-suppressor genes, ER genes, and <strong>in</strong>sul<strong>in</strong>-like growth factor 2[116,117].The very complex draft of the age-dependent changes <strong>in</strong> DNA methylation patterns, represent<strong>in</strong>gboth hypo- and hypermethylation events at specific DNA sequences, is furthercomplicated by the presence of tissue-specific patterns. Once aga<strong>in</strong>, it is the bra<strong>in</strong> that offersa clear example of this complexity, s<strong>in</strong>ce it was demonstrated that methylation patterns areregion-specific [118].F<strong>in</strong>ally, a further degree of complexity is added by the possibility that many of the abovediscussedchanges <strong>in</strong> the epigenome of the ag<strong>in</strong>g bra<strong>in</strong> could have an unsuspected early (evendevelopmental) orig<strong>in</strong>. The theory that early-life events could <strong>in</strong>duce epigenetic modificationsthat are phenotypically silent until middle or old age, accumulat<strong>in</strong>g potential toxic features(amyloid deposition, for example) dur<strong>in</strong>g many years of asymptomatic state, is wellsummarized by the LEARn (latent early-life associated regulation) model of age-relatedneurological disorders [119]. Experimental evidence of this model was given by the abovementionedeffect of early-life stress that results <strong>in</strong> epigenetic modifications and behavioraldeficits <strong>in</strong> adult age [50,59] and by the f<strong>in</strong>d<strong>in</strong>g that early exposure of monkeys to Pb decreasedDNMT activity and had effects on amyloid-beta (Ab) deposition <strong>in</strong> late life [87]. Also, the workperformed <strong>in</strong> our laboratory so far, seems to support the LEARn theory s<strong>in</strong>ce we observed thatearly <strong>in</strong>terventions on methylation mach<strong>in</strong>ery <strong>in</strong> AD transgenic mice are able to modulatePSEN1 expression and Ab deposition [54,55,120].52926.6 HEALTHY AND PATHOLOGICAL AGINGThe variability of age-associated cognitive change is related to many factors <strong>in</strong>fluenced bydemographic, social, educational, medical, nutritional, and biological stimuli. Biologicalfactors <strong>in</strong>clude both genetic and epigenetic mechanisms. Epigenetic modifications, such asDNA methylation and histone acetylation regulate replication and transcription and areresponsible for chromat<strong>in</strong> (re)model<strong>in</strong>g, chromosome stability, and DNA impr<strong>in</strong>t<strong>in</strong>g. Amongthe different epigenetic modifications, DNA methylation plays a pivotal role; methylationhomeostasis is fundamental for normal bra<strong>in</strong> physiology, whereas the impairment of methylationreactions and particularly DNA methylation are associated with markers related toneurodegeneration. Moreover, epigenetic mechanisms are greatly <strong>in</strong>volved <strong>in</strong> bra<strong>in</strong> developmentdur<strong>in</strong>g the early life and the adult neurogenesis and are also <strong>in</strong>volved <strong>in</strong> the onset anddevelopment of AD and other neurodegenerative disorders [121]. The hypothesis that genetranscription <strong>in</strong> bra<strong>in</strong> is epigenetically regulated by DNA methylation much more than tissuespecificexpression <strong>in</strong> other organs is now largely accepted. This hypothesis is supported by thestrong association between age-related low methylation status and cognitive deficits orneurological and neurodegenerative pathologies [4,26,27]. However, it is still not completelyclear whether epigenetic changes actually represent a cause or a consequence of the disease.This gap is due to the high complexity of the epigenetic mechanisms and of their regulation


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>dur<strong>in</strong>g ag<strong>in</strong>g; moreover it is possible that epigenetic studies on pathological ag<strong>in</strong>g could bebiased by the <strong>in</strong>volvement of subjects <strong>in</strong> an advanced stage of disease or, <strong>in</strong> any case, subjects <strong>in</strong>which the epigenetic changes started even many years before the comparison of the firstsymptoms. An active role for epigenetics <strong>in</strong> normal and pathological ag<strong>in</strong>g must meet twoconditions: specific epigenetic changes must occur dur<strong>in</strong>g ag<strong>in</strong>g and they must be functionallyassociated with the aged and/or the diseased phenotype. Assum<strong>in</strong>g that specific epigeneticmodifications can have a direct functional outcome <strong>in</strong> ag<strong>in</strong>g or age-related diseases, it is alsoessential to establish whether they depend on genetic, environmental, or stochastic factors[61]. Few objections could be moved to the statement that the two cited conditions (thespecificity of the epigenetic changes and the functional association to a phenotype) aredemonstrated <strong>in</strong> the relationship between ag<strong>in</strong>g and cancer. As a matter of fact, epigeneticmodifications play a major role <strong>in</strong> cancer, <strong>in</strong>fluenc<strong>in</strong>g tumor outcome by <strong>in</strong>terfer<strong>in</strong>g with keysenescence pathways [122]. As for other physiopathological mechanisms, a direct and causalrole can be, so far, just supposed; the complexity of the immune system, for example, opens thepossibility to predict that a higher-order, supragenetic regulation is <strong>in</strong>dispensable for generationof its constituents and control of its functions dur<strong>in</strong>g ag<strong>in</strong>g [123].530In human bra<strong>in</strong>, a recent study attempted to quantify the extent and the identity of epigeneticchanges <strong>in</strong> the ag<strong>in</strong>g process. DNA methylation at >27 000-CpG sites throughout the humangenome, was exam<strong>in</strong>ed <strong>in</strong> frontal cortex, temporal cortex, pons, and cerebellum from 387subjects between 1 and 102 years of age. The authors were able to demonstrate the presence ofCpG loci, ma<strong>in</strong>ly associated to CpG islands, which showed highly significant correlationbetween DNA methylation and age; they also confirmed the positive correlation between ageand DNA methylation level. Moreover, the loci show<strong>in</strong>g the most significant association withage were physically close to genes and <strong>in</strong>volved <strong>in</strong> DNA b<strong>in</strong>d<strong>in</strong>g and regulation of transcription,suggest<strong>in</strong>g that specific age-dependent DNA methylation changes could be responsiblefor regulat<strong>in</strong>g gene expression <strong>in</strong> the human bra<strong>in</strong> [124].A very <strong>in</strong>trigu<strong>in</strong>g and useful model to study normal and pathological ag<strong>in</strong>g is represented bythe monozygotic tw<strong>in</strong>s discordant for the occurrence of age-related diseases. Monozygotictw<strong>in</strong> sibl<strong>in</strong>gs share the same genotype because they are derived from the same zygote. Despitethe appearance, they frequently present phenotypic differences, such as their susceptibility todisease. Recent studies suggest that phenotypic discordance between monozygotic tw<strong>in</strong>s couldbe at least <strong>in</strong> part due to epigenetic differences and factors chang<strong>in</strong>g over their lifetime. Theepigenetic drift occurr<strong>in</strong>g dur<strong>in</strong>g the development is probably result<strong>in</strong>g by a comb<strong>in</strong>ation ofstochastic and environmental factors [125]. One example is represented by a study on tw<strong>in</strong>sdiscordant for Lewy body dementia that allowed postulat<strong>in</strong>g that epigenetic factors could playa role <strong>in</strong> Lewy body pathology [126]. In another elegant work tak<strong>in</strong>g advantage of a rare set ofmonozygotic tw<strong>in</strong>s discordant for AD, it was possible to observe significantly reduced levels ofDNA methylation <strong>in</strong> temporal neocortex neuronal nuclei of the AD tw<strong>in</strong>. This result stressedthe hypothesis that the effects of life events on AD risk could be mediated by epigeneticmechanisms, provid<strong>in</strong>g a more general potential explanation for AD discordance despitegenetic similarities [127].We can conclude that ag<strong>in</strong>g is a process characterized by genetic and epigenetic <strong>in</strong>teractions,where epigenetics has an important function <strong>in</strong> determ<strong>in</strong><strong>in</strong>g phenotypic differences.<strong>Epigenetics</strong> also plays a key role <strong>in</strong> the development of diseases associated with ag<strong>in</strong>g andexpla<strong>in</strong>s the relationship between <strong>in</strong>dividual genetic background, environment, ag<strong>in</strong>g,and disease [128].26.7 ENVIRONMENT, EPIGENETICS, AND AGINGThe discussion about the studies on monozygotic tw<strong>in</strong>s also helps to <strong>in</strong>troduce anotherfundamental concept regard<strong>in</strong>g epigenetic changes occurr<strong>in</strong>g with ag<strong>in</strong>g: it is the idea that


CHAPTER 26Ag<strong>in</strong>g and <strong>Disease</strong>: The Epigenetic Bridgeepigenetic mechanisms could be triggered by environmental factors or, <strong>in</strong> a slightly differentpo<strong>in</strong>t of view, that epigenetics exerts the role of mediator of environmental stimuli. The term“environmental” encompasses, <strong>in</strong> this case, many different processes and conditions occurr<strong>in</strong>goutside but enter<strong>in</strong>g <strong>in</strong> contact with the organism. These environmental factors <strong>in</strong>clude,among others: stresses (physical and behavioral), nutritional factors, pollutants and pesticides,chemicals, metals, physical exercise, and lifestyle. All these factors are able to cause, <strong>in</strong> theorganism, biological effects that could determ<strong>in</strong>e (or contribute to) the onset and progressionof the disease; many of these factors were found associated with the <strong>in</strong>duction of epigeneticchanges, as depicted <strong>in</strong> Figure 26.2. Obviously, these factors become <strong>in</strong>creas<strong>in</strong>gly relevant withag<strong>in</strong>g to the healthy or pathological status of an <strong>in</strong>dividual, due to the <strong>in</strong>creased possibility toencounter different environmental “hits” or to cumulate the reiterated effects of one of thesefactors.S<strong>in</strong>ce monozygotic tw<strong>in</strong>s are genetically identical, they are considered as ideal “experimentalmodels” to study the role of environmental factors as determ<strong>in</strong>ants of complex diseases andphenotypes. As <strong>in</strong> the case of the above-mentioned study on tw<strong>in</strong>s discordant for AD, forexample, the AD tw<strong>in</strong> presented a history of contact with chemical species [127]. Anotherexample of association between acute environmental stimuli and epigenetic-dependentdisease phenotypes is given by the observed <strong>in</strong>crease of CpG-island promoter hypermethylation<strong>in</strong> tumor-suppressor genes <strong>in</strong> the oral mucosa of smokers (reviewed <strong>in</strong> [125]).Environmental factors are <strong>in</strong>creas<strong>in</strong>gly claimed as responsible for neurodegeneration-relatedmodifications; a l<strong>in</strong>k between environmental-<strong>in</strong>duced epigenetic modification, oxidation, andrepair of AD-related genes, was discussed <strong>in</strong> depth <strong>in</strong> a recent review [129].One of the most <strong>in</strong>trigu<strong>in</strong>g <strong>in</strong>teractions between environment and epigenome is representedby the discussed results evidenc<strong>in</strong>g that even an apparently “mild” environmental factor as the531FIGURE 26.2Many environmental factors can <strong>in</strong>terferewith the organism, <strong>in</strong>duc<strong>in</strong>g epigeneticmodifications. DNA methylation patternsestablished after developmentalprocesses, for example, can be modified<strong>in</strong> sense of local and sequence-specifichyper- or (more frequently)hypomethylation. These alterations canbe responsible for deviations from thenormal ag<strong>in</strong>g processes, result<strong>in</strong>g <strong>in</strong>higher susceptibility to age-associateddisease. This figure is reproduced <strong>in</strong> thecolor plate section.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>behavioral stress (s<strong>in</strong>ce it does not <strong>in</strong>volve a physical contact of the <strong>in</strong>dividual with anychemical species), could result <strong>in</strong> a long-last<strong>in</strong>g alteration of epigenetic markers, lead<strong>in</strong>g tofunctional alterations. This surpris<strong>in</strong>g effect was very well demonstrated by the abovediscussedstudy show<strong>in</strong>g that per<strong>in</strong>atal behavioral stress <strong>in</strong> rats was responsible for altered AVPor BDNF expression, mediated by DNA methylation [50,59]. Another “mild” environmentalfactor that seems <strong>in</strong>volved <strong>in</strong> the development of healthy or pathological ag<strong>in</strong>g is representedby the physical exercise. Physical exercise improves the efficiency of the capillary system and<strong>in</strong>creases the oxygen supply to the bra<strong>in</strong>, thus enhanc<strong>in</strong>g metabolic activity and oxygen <strong>in</strong>take<strong>in</strong> neurons, and <strong>in</strong>creases neurotroph<strong>in</strong> levels and resistance to stress. Regular exercise andactive lifestyle dur<strong>in</strong>g adulthood have been associated with reduced risk and protective effectsfor mild cognitive impairment and AD. Similarly, studies <strong>in</strong> animal models show that physicalactivity has positive physiological and cognitive effects that correlate with changes <strong>in</strong> transcriptionalprofiles possibly mediated by epigenetic modifications [130].532Nutrition and diet represent another environmental factor that can exert its <strong>in</strong>fluence on ag<strong>in</strong>g.Dietary exposures can have consequences even many years later and this observation raisesquestions about the mechanisms through which such exposures are “remembered” and howthey can result <strong>in</strong> altered disease risk. There is grow<strong>in</strong>g evidence that epigenetic mechanismsmay mediate the effects of nutrients, micronutrients, and even non-nutrient dietary factorsmay be causal for the development of complex diseases [131]. Alterations <strong>in</strong> DNA methylationdur<strong>in</strong>g ag<strong>in</strong>g can depend on alterations <strong>in</strong> dietary status and the great <strong>in</strong>fluence of nutritionalcomponents on health and lifespan it is largely accepted. Among the various mechanisms bywhich nutritional elements could affect the progress of ag<strong>in</strong>g, two pathways <strong>in</strong>volve DNAmethylation: the first <strong>in</strong>volves the supply of metabolites of the S-adenosylmethion<strong>in</strong>e cycle(and will be extensively discussed <strong>in</strong> the next paragraph), whereas the second is referred toelements able to directly modify the DNMT activity (selenium, cadmium, and nickel).However, other nutritional factors seem able to determ<strong>in</strong>e epigenetic modifications withoutdirectly perturb<strong>in</strong>g the core of the methylation reactions. One example is given by the l<strong>in</strong>kbetween under- and overnutrition dur<strong>in</strong>g pregnancy and the consequent (later <strong>in</strong> life)development of diseases such as diabetes and obesity. Epigenetic modifications may be onemechanism by which exposure to an altered <strong>in</strong>trauter<strong>in</strong>e milieu may <strong>in</strong>fluence the onset ofthese disturbances much later <strong>in</strong> life. As a matter of fact, it was demonstrated that epigeneticmodifications affect<strong>in</strong>g processes important to glucose regulation and <strong>in</strong>sul<strong>in</strong> secretion arepresent <strong>in</strong> the pancreatic b-cells and muscle of the <strong>in</strong>trauter<strong>in</strong>e growth-retarded offspr<strong>in</strong>g,characteristics essential to the pathophysiology of type 2 diabetes. Moreover, epigeneticregulation of gene expression contributes to both adipocyte determ<strong>in</strong>ation and differentiation<strong>in</strong> <strong>in</strong> vitro models [132]. The epigenetic connection between nutrition and age-related diseaseswas well presented by Tollefsbol and colleagues <strong>in</strong> relation to cancer onset [35], whereas thework performed <strong>in</strong> my laboratory <strong>in</strong> relation to the connection between nutrition andepigenetics <strong>in</strong> LOAD and the role of nutrition <strong>in</strong> the modulation of methylation reaction weresummarized <strong>in</strong> a recent commentary [133] and will be here exposed more <strong>in</strong> detail.26.8 EPIGENETICS AND AGE-ASSOCIATED DISEASESEpigenetic modifications appear to be causative of, or at least <strong>in</strong>volved <strong>in</strong>, an <strong>in</strong>creas<strong>in</strong>g numberof human diseases. As previously discussed, a modern and develop<strong>in</strong>g concept po<strong>in</strong>ts out thefetal or per<strong>in</strong>atal orig<strong>in</strong> of adult diseases and the adaptation response to environmental stimulilead<strong>in</strong>g to <strong>in</strong>creased susceptibility to age-associated diseases [134]. Although the mechanismsmediat<strong>in</strong>g and express<strong>in</strong>g this “memory” of the early life throughout ag<strong>in</strong>g are not clearlyunraveled, it is clear that an epigenetic basis exists. Apparently, the consequent <strong>in</strong>creasedsusceptibility to the disease recapitulates as well the mechanisms typical of the decl<strong>in</strong>e observed<strong>in</strong> normal ag<strong>in</strong>g. The <strong>in</strong>volvement of multiple organ systems <strong>in</strong> the pathological ag<strong>in</strong>gphenotype can be assimilated to the “frail syndrome”. Frailty is def<strong>in</strong>ed as a non-specific state of


CHAPTER 26Ag<strong>in</strong>g and <strong>Disease</strong>: The Epigenetic Bridgeglobal vulnerability reflect<strong>in</strong>g multisystem pathology due to decreased adaptation to stressorsand reduced functional reserve that becomes not sufficient to ma<strong>in</strong>ta<strong>in</strong> and repair the ag<strong>in</strong>gbody [135,136]. Cl<strong>in</strong>ically, frailty is characterized by low physical activity, global weakness, lowmuscle strength, fatigability, general slowness, and loss of weight [137]. As a matter of fact, frailtyis strictly related to epigenetic mechanisms. It was demonstrated that global DNA methylationlevels were correlated to the frailty status and that the worsen<strong>in</strong>g of frailty was significantlyassociated with DNA demethylation [138]. Identification of the role of epigenetic drift <strong>in</strong> theonset of frail status also represents the opportunity to underl<strong>in</strong>e the connection betweenepigenetics and other age-associated diseases. Part of the frail phenotype is, <strong>in</strong> fact, connected toother diseases typical of old age and characterized by evident epigenetic bases. As a matter of fact,significant high levels of <strong>in</strong>flammatory markers and activation of the clott<strong>in</strong>g cascade were found<strong>in</strong> frail patients; these markers are, as is well known, also risk factors for cardiovascular diseases(CVD) [139]. An emerg<strong>in</strong>g theory identifies an epigenetic basis also for the chronic low-grade<strong>in</strong>flammation typical of ag<strong>in</strong>g, generated by the <strong>in</strong>crease <strong>in</strong> the production of pro<strong>in</strong>flammatorycytok<strong>in</strong>es and other markers that lead to the def<strong>in</strong>ition of “<strong>in</strong>flamm-ag<strong>in</strong>g” status. This status is,<strong>in</strong> turn, comparable to frailty due to the multiorgan (bra<strong>in</strong>, liver) and different tissue (adipose,muscle) contribution [140]. F<strong>in</strong>ally, this complex picture <strong>in</strong>volv<strong>in</strong>g <strong>in</strong>flammation and multiorgancontribution to the aged phenotype, f<strong>in</strong>ds a further piece of the jigsaw <strong>in</strong> the epigeneticbasis of another complex disease like diabetes. Diabetes also <strong>in</strong>volves alterations of the<strong>in</strong>flammatory markers and undergoes environmental <strong>in</strong>fluences mediated ma<strong>in</strong>ly by thenutrient <strong>in</strong>take; thus ag<strong>in</strong>g, frailty, nutrition, early life-events, and epigenetics are, once aga<strong>in</strong>,connected [134,141e143]. A very important concept emerg<strong>in</strong>g from these studies is thatmalnutrition is often associated with ag<strong>in</strong>g but that this deficit should be seen <strong>in</strong> terms of qualityand variety of foods rather than <strong>in</strong> terms of quantity [141].<strong>Epigenetics</strong>, disease, and ag<strong>in</strong>g are connected also <strong>in</strong> another complex relationship representedby the telomere attrition and the onset of cancer. Telomere attrition and mutations accumulatedue to the deficit of the DNA repair mach<strong>in</strong>ery occurr<strong>in</strong>g with age is one of the ma<strong>in</strong> causes ofage-associated genome <strong>in</strong>stability. Telomeres are composed by G-rich repetitive DNAsequences (TTAGGG); these sequences are bound to complexes and specialized prote<strong>in</strong>s,localized at the ends of l<strong>in</strong>ear chromosomes. Telomere length shortens with age as a consequenceof decreased DNA methylation, until the m<strong>in</strong>imal required length necessary toma<strong>in</strong>ta<strong>in</strong> telomere structure and function is reached [144e146]. The activation of the mechanismresponsible for the ma<strong>in</strong>tenance of telomeric length (telomerase) is considered a hallmarkof cancer while attrition is associated with the ag<strong>in</strong>g phenotype [147] depend<strong>in</strong>g on theresponse of the DNA damage checkpo<strong>in</strong>t [148]; as previously discussed, the human telomerasereverse transcriptase gene (hTERT) is regulated by epigenetic mechanisms [149]. Recently,different models of transgenic mice deleted for the shelter<strong>in</strong> prote<strong>in</strong>s (the major complexbound to telomeres) have been generated and could help the future study on the role oftelomeric attrition and <strong>in</strong>stability <strong>in</strong> ag<strong>in</strong>g and cancer [146]. Besides the epigenetic regulationof the telomeric length, cancer onset is clearly associated with epigenetics via the transcriptionalrepression of tumor-suppressor genes by CpG island promoter hypermethylation(reviewed <strong>in</strong> [150]); <strong>in</strong> addition to classical tumor-suppressor and DNA repair genes, cancerassociatedhypermethylation also affects genes <strong>in</strong>volved <strong>in</strong> premature ag<strong>in</strong>g and miRNAs withgrowth-<strong>in</strong>hibitory functions.533Epigenetic changes associated with ag<strong>in</strong>g and very often <strong>in</strong>duced by environmental stimuli,seem therefore responsible for the possible onset of different, although strictly <strong>in</strong>terconnected,pathologies typical of the elderly.26.9 ONE CARBON METABOLISMStudies on epigenetics exponentially <strong>in</strong>creased, as results evident by the raise, from a scarcedozen <strong>in</strong> 1980 to over 5000 papers published <strong>in</strong> 2010, of PubMed citations <strong>in</strong>volv<strong>in</strong>g


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>epigenetics. Despite this great <strong>in</strong>crease, one could suspect that a real knowledge of thebiochemical basis regulat<strong>in</strong>g epigenetic mechanisms (DNA methylation, for <strong>in</strong>stance) is oftenmiss<strong>in</strong>g <strong>in</strong> these works; as a matter of fact, we can see that PubMed citations <strong>in</strong> which“epigenetics” and “homocyste<strong>in</strong>e” are both present are only 96 <strong>in</strong> the period 1980e2010. Thisobservation can be considered as a clue that the biochemical pathway underneath the transferof methyl groups (the most relevant epigenetic modification) is quite unknown, even thoughthis pathway (known as “one-carbon metabolism”, be<strong>in</strong>g characterized by the transfer ofmonocarbonic units like the methyl group eCH 3 ) has a number of biochemical reactionscomparable to ATP pathway.534One carbon metabolism, also known as the homocyste<strong>in</strong>e (HCY) cycle, is a complexbiochemical pathway regulated by the presence of folate, vitam<strong>in</strong> B12, and vitam<strong>in</strong> B6 (amongother metabolites) and lead<strong>in</strong>g to the production of methyl donor molecule S-adenosylmethion<strong>in</strong>e(SAM). A schematic and partial (focused on SAM and B vitam<strong>in</strong>s) representation ofthis metabolic cycle is reported <strong>in</strong> Figure 26.3. SAM can donate the methyl group to differentsubstrates (lipids, prote<strong>in</strong>s, and DNA) be<strong>in</strong>g converted <strong>in</strong> S-adenosylhomocyste<strong>in</strong>e (SAH),a strong competitive <strong>in</strong>hibitor of methyltransferases. SAH is then hydrolyzed to adenos<strong>in</strong>e andHCY, a thiol-conta<strong>in</strong><strong>in</strong>g am<strong>in</strong>o acid produced dur<strong>in</strong>g the methion<strong>in</strong>e metabolism via theadenosylated compound SAM, once formed is either converted to cyste<strong>in</strong>e by transsulfuration(further lead<strong>in</strong>g to glutathione synthesis), remethylated to form methion<strong>in</strong>e, or exportedto blood [151e155]. In the remethylation pathway HCY is remethylated by the vitam<strong>in</strong>B12-dependent enzyme methion<strong>in</strong>e synthase (MS) us<strong>in</strong>g 5-methyltetrahydrofolate as cosubstrate.Alternatively, ma<strong>in</strong>ly <strong>in</strong> the liver, beta<strong>in</strong>e can donate a methyl group <strong>in</strong> a vitam<strong>in</strong>B12-<strong>in</strong>dependent reaction, catalyzed by beta<strong>in</strong>e-homocyste<strong>in</strong>e methyltransferase (BHMT). Inthe transsulfuration pathway, HCY can condense with ser<strong>in</strong>e to form cystathion<strong>in</strong>e <strong>in</strong> a reactioncatalyzed by the cystathion<strong>in</strong>e beta synthase (CBS), a vitam<strong>in</strong> B6-dependent enzyme, andthe cystathion<strong>in</strong>e is hydrolyzed to cyste<strong>in</strong>e (Cys); it is not known for sure whether this pathwayFIGURE 26.3Schematic representation of the most relevantreactions comprises of the “one-carbonmetabolism”, emphasiz<strong>in</strong>g a vitam<strong>in</strong> cofactorscycle (remethylation) and SAM-dependent DNAmethylation (transmethylation). Met, methion<strong>in</strong>e;SAM, S-adenosylmethion<strong>in</strong>e; SAH,S-adenosylhomocyste<strong>in</strong>e; HCY, homocyste<strong>in</strong>e;CYS, cystathion<strong>in</strong>e, GSH, glutathione; THF,tetrahydrofolate; MTHF, methyltetrahydrofolate;B12, vitam<strong>in</strong> B12; B6, vitam<strong>in</strong> B6; 1, methion<strong>in</strong>eadenosyltransferase (MAT); 2,methyltransferase(s); 3, SAH hydrolase;4, cystathion<strong>in</strong>eebesynthase (CBS); 5,methion<strong>in</strong>e synthase; 6,methylenetetrahydrofolate reductase (MTHFR).This figure is reproduced <strong>in</strong> the color platesection.


CHAPTER 26Ag<strong>in</strong>g and <strong>Disease</strong>: The Epigenetic Bridgeis normally active <strong>in</strong> the bra<strong>in</strong>, due to the lack of the enzyme g-cystathionase, but a recentstudy demonstrated the presence of a functional (maybe alternative) transsulfuration pathwayalso <strong>in</strong> this tissue [156]. Cys is used for prote<strong>in</strong> synthesis, metabolized to sulfate or used forglutathione (GSH) synthesis. The tripeptide GSH is the most abundant <strong>in</strong>tracellular nonprote<strong>in</strong>thiol, and it is a versatile reductant, serv<strong>in</strong>g multiple biological functions, act<strong>in</strong>g,among others, as a quencher of free radicals and a cosubstrate <strong>in</strong> the enzymatic reduction ofperoxides [157].HCY accumulation causes the accumulation of SAH because of the reversibility of the reactionconvert<strong>in</strong>g SAH to HCY and adenos<strong>in</strong>e (Ado); the equilibrium dynamics favors SAH synthesis.The reaction proceeds <strong>in</strong> the hydrolytic direction only if HCY and Ado are efficiently removed.At least <strong>in</strong> bra<strong>in</strong> and other tissues, SAH is a DNA methyltransferase <strong>in</strong>hibitor, which re<strong>in</strong>forcesDNA hypomethylation [158]. Therefore, an alteration of the metabolism through eitherremethylation or transsulfuration pathways can lead to hyperhomocyste<strong>in</strong>emia, decreas<strong>in</strong>g ofSAM/SAH ratio (often <strong>in</strong>dicated as the “methylation potential”, MP) and alteration of GSHlevels, suggest<strong>in</strong>g that hypomethylation is a mechanism through which HCY is <strong>in</strong>volved <strong>in</strong>vascular disease and AD, together with the oxidative damage [4,159,160].26.10 ONE-CARBON METABOLISM IN AGING ANDNEURODEGENERATIONMethylation of CpG d<strong>in</strong>ucleotides plays an important role <strong>in</strong> regulation of gene expression <strong>in</strong>the bra<strong>in</strong>. It was discovered that bra<strong>in</strong>-specific promoter-related sequences are surpris<strong>in</strong>glyenriched <strong>in</strong> CpG sites. This leads to the conclusion that it is likely that bra<strong>in</strong>-specific transcriptionis regulated by methylation at an epigenetic level much more frequently than tissuespecificexpression <strong>in</strong> other organs. Low methylation status is strongly associated withneurological and cognitive deficits. Many epidemiological studies have shown that factorsconnected to low methylation status such as elevated total homocyste<strong>in</strong>e, low folate or lowvitam<strong>in</strong> B12 levels are associated with <strong>in</strong>creased risk of cognitive decl<strong>in</strong>e, dementia, and bra<strong>in</strong>atrophy. In recent years, hyperhomocyste<strong>in</strong>emia has begun to be widely considered a riskfactor <strong>in</strong> AD and this may be ascribed to alteration of the S-adenosylmethion<strong>in</strong>e/homocyste<strong>in</strong>e(SAM/HCY) metabolism [161]. Indeed, a frequently observed condition <strong>in</strong> ADaffectedpeople is the <strong>in</strong>crease <strong>in</strong> plasmatic HCY, sometimes along with a decrease <strong>in</strong> vitam<strong>in</strong>B12, B6, and folate uptake [162]. In AD, the loss of precise control through gene methylationmay alter a delicate equilibrium among the three enzymes (a-, b-, and g-secretases) knownto be <strong>in</strong>volved <strong>in</strong> the production of either amyloid-beta (Ab) or other non-dangerous catabolites[163].535It is well established that a-secretase cleavage of APP does not produce the amyloidogenicpeptides and that, on the contrary, they are produced by the activity of b-secretases thatgenerate an N-term<strong>in</strong>al soluble fragment and a C-term<strong>in</strong>al fragment that is sequentiallycleaved by g-secretase to produce Ab peptides [164]. The alteration of SAM/SAH ratio is tightlyrelated to the altered expression of two genes <strong>in</strong>volved <strong>in</strong> APP metabolism, f<strong>in</strong>ally produc<strong>in</strong>gthe accumulation of Ab peptide <strong>in</strong> the senile plaque. In the last 50 years, SAM has been shownto be perhaps the most frequently used substrate, after ATP, and therefore occupies a centralposition <strong>in</strong> human health, disease, and ag<strong>in</strong>g. SAM is known to be the primary methyl-donorpresent <strong>in</strong> eukaryotes and it is <strong>in</strong>volved <strong>in</strong> methylation of target molecules as DNA, RNA,prote<strong>in</strong>s, lipids, and polyam<strong>in</strong>es synthesis [165]. SAM appears to be altered <strong>in</strong> some neurologicaldisorders, <strong>in</strong>clud<strong>in</strong>g LOAD [26]. About 95% of SAM is engaged <strong>in</strong> methylation reactions.These metabolic alterations may be responsible for the generalized reduction of DNAmethylation observed <strong>in</strong> ag<strong>in</strong>g, which can lead to overactivation of methylation-controlledgenes. DNA methylation represents an important mechanism for epigenetic control of geneexpression and the ma<strong>in</strong>tenance of genome <strong>in</strong>tegrity. Methyl deficiency results <strong>in</strong> global


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>hypomethylation of the genome. Hence, methyl deficiency leads to disturbances <strong>in</strong> geneexpression. Interest<strong>in</strong>gly, the ag<strong>in</strong>g process leads to similar changes <strong>in</strong> the methylation pattern.53626.11 EPIGENETICS AND NEURODEGENERATION: THEALZHEIMER’S DISEASE PARADIGMThe sporadic form of Alzheimer’s disease (LOAD) is considered a multifactorial disease.Genetic, nutritional, metabolic, environmental, and social factors are associated with onsetand progression of the disease [75,120,166] but, despite the extent of researches <strong>in</strong> this area,most causes of LOAD rema<strong>in</strong> obscure. The complex, <strong>in</strong>terconnected, non-Mendelian, etiologyof LOAD suggests that epigenetic components could be <strong>in</strong>volved and epigenetic modificationscould represent candidate effector mechanisms mediat<strong>in</strong>g the above-mentioned risk factors <strong>in</strong>AD onset and progression. Despite the fact that it is already well known that epigeneticchanges could act <strong>in</strong> several physiological and pathological processes, few papers po<strong>in</strong>ted theattention on epigenetic regulation of ag<strong>in</strong>g and neurodegeneration. Nevertheless, it is evidentthat whereas genetic factors are clearly associated with the early-onset form of AD, epigeneticfactors could be more easily l<strong>in</strong>ked to LOAD, s<strong>in</strong>ce the epigenome is prone to changes dur<strong>in</strong>gdevelopment and also ag<strong>in</strong>g [167e169]. As a matter of fact, the epigenetic mechanisms can beconsidered as a l<strong>in</strong>k between environmental stimuli and their effect on the genome and on thepathologies. For these reasons, LOAD research is fac<strong>in</strong>g an <strong>in</strong>creas<strong>in</strong>g <strong>in</strong>terest <strong>in</strong> the study ofepigenetic mechanisms possibly <strong>in</strong>volved <strong>in</strong> the pathology [56,133,170]. Due to the largedevelopment of wide-scale analysis techniques and their application to the study of epigenome,it has been possible to demonstrate that DNA methylation <strong>in</strong> <strong>in</strong>dividuals changes overtime [171]. Moreover, the DNA methylation pattern <strong>in</strong> human bra<strong>in</strong> is different <strong>in</strong> differentbra<strong>in</strong> areas [120] and can dynamically change dur<strong>in</strong>g the lifespan [172]. F<strong>in</strong>ally, it has beendemonstrated that the existence of epigenetic changes <strong>in</strong> LOAD patients can easily contributeto the onset and development of disease [173]. In particular, Wang and colleagues [173]evidenced that, among others, PS1 gene was differentially methylated and that there wasa correlation between gene demethylation and LOAD. In agreement with this result, we alsodemonstrated that PSEN1 promoter demethylation is responsible for the overexpression of thegene [54,55,120].As previously described, methylation metabolism is dependent on the “one-carbon” metabolism(Figure 26.3). High HCY and low B vitam<strong>in</strong> levels are positively associated with LOAD[174e176], even if the causeeeffect relationship is still not completely demonstrated. In thisscenario, epigenome modulation could represent the molecular l<strong>in</strong>k between hyperhomocyste<strong>in</strong>emiaand LOAD course and onset [177e179]. Moreover, one-carbon metabolismalteration and consequent methylation reactions unbalance (i.e. loss of methyl groups) dur<strong>in</strong>gag<strong>in</strong>g represents one of the mechanisms by which environmental and dietary factors canpromote LOAD [75,158,180e182]. F<strong>in</strong>ally, the f<strong>in</strong>d<strong>in</strong>g that one-carbon metabolism alterationscan affect also tau process<strong>in</strong>g [183] stress the hypothesis that this metabolism, andmethylation reactions <strong>in</strong> particular, could reta<strong>in</strong> a central role <strong>in</strong> LOAD, connect<strong>in</strong>g bothamyloidogenic and neurofibrillary pathways.Two genes directly <strong>in</strong>volved <strong>in</strong> AD showed changes <strong>in</strong> methylation pattern. It was demonstratedthat the APP gene promoter was differentially modulated [184] and it was foundhypomethylated <strong>in</strong> postmortem bra<strong>in</strong>s of AD patients [112,185]. Besides APP, also PSEN1 hasbeen found hypomethylated both <strong>in</strong> AD patients [173] and <strong>in</strong> our experimental modelscharacterized by unbalanced methylation pathway [54,55,120].26.12 AGED AD MICE AND EPIGENETICSThese studies, spac<strong>in</strong>g from specific gene to large-scale studies, both <strong>in</strong> LOAD patients and <strong>in</strong>experimental models, are very well summarized <strong>in</strong> two recent reviews [56,186]. Studies on


CHAPTER 26Ag<strong>in</strong>g and <strong>Disease</strong>: The Epigenetic BridgeLOAD subjects evidenced that DNA methylation, the ma<strong>in</strong> epigenetic modification, is associatedwith Alzheimer’s mechanisms. Studies on experimental models, although notsupported by a f<strong>in</strong>al proof <strong>in</strong> AD patients, had the fundamental value of <strong>in</strong>vestigat<strong>in</strong>g themolecular mechanisms l<strong>in</strong>k<strong>in</strong>g DNA methylation to the onset and the progression of the ADlikefeatures observed <strong>in</strong> these models. Several groups <strong>in</strong>vestigated the role of epigenetics <strong>in</strong> AD<strong>in</strong> different areas of cell biology as Ab process<strong>in</strong>g and scaveng<strong>in</strong>g, tau phosphorylation,<strong>in</strong>flammation, apoptosis, cell cycle dysregulation, and ApoE expression (summarized <strong>in</strong> [56]).In our laboratory, dur<strong>in</strong>g the last 10 years, we addressed our studies to the area of DNAmethylation <strong>in</strong> human neuroblastoma cells [187,188] and then <strong>in</strong> a mur<strong>in</strong>e model ofAlzheimer’s disease [120], develop<strong>in</strong>g an experimental model of nutritionally <strong>in</strong>ducedhyperhomocyste<strong>in</strong>emia. Tak<strong>in</strong>g advantage of the massive and rapid amyloid deposition of theTgCRND8 mice, carry<strong>in</strong>g a double-mutated human APP gene, we were able to demonstratethat hyperhomocyste<strong>in</strong>emia <strong>in</strong>duced by B vitam<strong>in</strong>s deficiency (folate, B12, and B6) caused analteration <strong>in</strong> SAM and SAH levels, <strong>in</strong>hibit<strong>in</strong>g SAM-dependent methyltransferases. This alterationwas responsible for dysequilibrium <strong>in</strong> the DNA methylation mach<strong>in</strong>ery consist<strong>in</strong>g of thedecrease of the methyltransferase and <strong>in</strong>crease of the demethylase activities [54]. The DNAmethylation impairment was correlated to the site-specific demethylation of PSEN1 promoter,with consequent gene overexpression [55]. Other studies found similar results <strong>in</strong> a differentcellular model [189], also suggest<strong>in</strong>g the role of DNA hypomethylation <strong>in</strong> the <strong>in</strong>crease ofPSEN1 expression and amyloid production.If the <strong>in</strong>volvement of DNA methylation <strong>in</strong> LOAD is now largely accepted, its causal orconsequential role still rema<strong>in</strong>s to be ascerta<strong>in</strong>ed [121,190]. One hypothesis is that alterationof methylation reactions could be dependent on other major pathways <strong>in</strong>volved <strong>in</strong> LOADonset, represent<strong>in</strong>g just a marker of the disease. Experimental evidences <strong>in</strong> this sense are givenby the f<strong>in</strong>d<strong>in</strong>g that exogenous Ab 1-40 seems to <strong>in</strong>duce the hypermethylation of the Neprilys<strong>in</strong>gene [191]. To prove that PSEN1 hypomethylation is a cause and not a consequence of the ADlikephenotype <strong>in</strong> our mouse model, we analyzed the methylation pattern <strong>in</strong> old mice(present<strong>in</strong>g <strong>in</strong>creased Ab pathology) versus young mice and <strong>in</strong> transgenic versus wild-typemice (which are not affected at all by Ab pathology); our results (submitted at the moment thischapter was written) clearly <strong>in</strong>dicate that PSEN1 methylation is not dependent on Ab levels,s<strong>in</strong>ce similar methylation patterns were found <strong>in</strong> young and old mice, both transgenic andwild-type. This result further stresses the idea that epigenetics plays a pivotal and fundamentalrole <strong>in</strong> the onset and progression of age-related diseases like LOAD.53726.13 CONCLUSIONThe great technological advance <strong>in</strong> biomedical research makes possible the constant <strong>in</strong>crease<strong>in</strong> our ability to study the extremely complex phenomena encompass<strong>in</strong>g the different, buthighly <strong>in</strong>terconnected, epigenetic modifications. This ability opened the w<strong>in</strong>dow, <strong>in</strong> recentyears, on a previously hidden scenario where epigenetics reta<strong>in</strong>s a causal role <strong>in</strong> mediat<strong>in</strong>g theeffects that environmental stimuli exert <strong>in</strong> the organism. This grow<strong>in</strong>g area of the science isparticularly relevant to the study of ag<strong>in</strong>g-associated processes, because the ag<strong>in</strong>g organismis <strong>in</strong>creas<strong>in</strong>gly exposed to cont<strong>in</strong>uous and different external stimuli. Now that the w<strong>in</strong>dow isopen on the mechanisms possibly responsible for the shift<strong>in</strong>g from healthy to pathologicalag<strong>in</strong>g, new questions rise together with the first results. How many, and how long, stimuli arenecessary to <strong>in</strong>duce changes <strong>in</strong> the normal ag<strong>in</strong>g processes? In which manner does theorganism translate these stimuli <strong>in</strong> processes that alter epigenetic modifications? Why doenvironmental-<strong>in</strong>duced epigenetic changes have different tissue, cell population, genomicsequence, or even DNA site specificity? Alternatively, is this different susceptibility anendogenous characteristic of these tissue, cells, or sequences? Are these “toxic” epigeneticmodifications really reversible? And, <strong>in</strong> this case, are these modifications “druggable”, <strong>in</strong> orderto hypothesize epigenetic <strong>in</strong>tervention and therapies? Decipher<strong>in</strong>g the epigenome and its


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>relevance to the ag<strong>in</strong>g processes is probably one of the most promis<strong>in</strong>g challenges for theresearchers <strong>in</strong> the com<strong>in</strong>g years and, due to its extreme complexity, it surely requires the<strong>in</strong>teraction of different skills and knowledge <strong>in</strong> an <strong>in</strong>terdiscipl<strong>in</strong>ary effort.538References[1] Wadd<strong>in</strong>gton CH. Endeavour 1942;1:18e20.[2] Holliday R. Mechanisms for the control of gene activity dur<strong>in</strong>g development. Biol Rev Cambr Philos Soc1990;65:431e71.[3] Goldberg AD, Allis CD, Bernste<strong>in</strong> E. <strong>Epigenetics</strong>: a landscape takes shape. Cell 2007;128:635e8.[4] Scarpa S, Cavallaro RA, D’Anselmi F, Fuso A. Gene silenc<strong>in</strong>g through methylation: an epigenetic <strong>in</strong>terventionon Alzheimer disease. J. Alzheimer’s Dis 2006;9:407e14.[5] Reitz C, Brayne C, Mayeux R. Epidemiology of Alzheimer disease. Nat Rev Neurol 2011;7:137e52.[6] Cutler RG. Evolutionary perspective of human longevity. In: Hazzard WR, Andres R, Bierman EL, Blass JP,editors. Pr<strong>in</strong>ciples of geriatric medic<strong>in</strong>e and gerontology. New York: McGraw-Hill; 1990. p. 15e21.[7] Roush W. Live long and prosper? <strong>Science</strong> 1996;273:42e6.[8] Troen BR. The biology of ag<strong>in</strong>g. Mt S<strong>in</strong>ai J Med 2003;70:3e22.[9] Hayflick L. Current theories of biological age<strong>in</strong>g. Federation Proceed<strong>in</strong>gs 1975;34:9e13.[10] Rattan SI. Cellular and molecular determ<strong>in</strong>ants of age<strong>in</strong>g. Indian J Exp Biol 1996;34:1e6.[11] Hamet P, Tremblay J. Genes of Ag<strong>in</strong>g. Metab 2003;52:5e9.[12] Kirkwood TB. <strong>Human</strong> senescence. Bioassays 1996;18:1009e16.[13] Orgel LE. The ma<strong>in</strong>tenance of the accuracy of prote<strong>in</strong> synthesis and its relevance to ag<strong>in</strong>g. Proc Natl Acad SciUSA 1963;49:517e21.[14] Suzuki YJ, Forman HJ, Sevanian A. Oxidants as stimulators of signal transduction. Free Radic Biol Med1997;22:269e85.[15] F<strong>in</strong>ch CE, Tanzi RE. Genetics of ag<strong>in</strong>g. <strong>Science</strong> 1997;278:407e11.[16] Perls T, Levenson R, Regan M, Puca A. What does it take to live to 100? Mech Age<strong>in</strong>g Dev2002;123:231e42.[17] Kuro-o M. <strong>Disease</strong> model: human ag<strong>in</strong>g. Trends Mol Med 2001;7:179e81.[18] Denckla WD. A time to die. Life Sci 1975;16:31e44.[19] Walford RL. Immunologic theory of ag<strong>in</strong>g: current status. Federation Proc 1974;33:2020e7.[20] Rub<strong>in</strong> H. Cell ag<strong>in</strong>g <strong>in</strong> vivo and <strong>in</strong> vitro. Mech Age<strong>in</strong>g Dev 1997;98:1e35.[21] Harley CB. Telomere loss: mitotic clock or genetic time bomb? Mutation Res 1991;256:271e82.[22] Campisi J. Ag<strong>in</strong>g and cancer: the double-edged sword of replicative senescence. J Am Geriatric Soc1997;45:482e8.[23] Jacobson MD, Weil M, Raff MC. Programmed cell death <strong>in</strong> animal development. Cell 1997;88:347e54.[24] Villeponteau B. The heterochromat<strong>in</strong> loss model of ag<strong>in</strong>g. Exp Gerontol 1997;32:383e94.[25] Howard BH. Replicative senescence: considerations relat<strong>in</strong>g to the stability of heterochromat<strong>in</strong> doma<strong>in</strong>s. ExpGerontol 1996;31:281e93.[26] Bottiglieri T, Hyland K. S-adenosylmethion<strong>in</strong>e levels <strong>in</strong> psychiatric and neurological disorders: a review. ActaNeurologica Scand<strong>in</strong>ava: Supplements 1994;154:19e26.[27] Morrison LD, Smith DD, Kish SJ. Bra<strong>in</strong> S-adenosylmethion<strong>in</strong>e levels are severely decreased <strong>in</strong> Alzheimer’sdisease. J Neurochem 1996;67:1328e31.[28] Tollefsbol TO, Andrews LG. Mechanisms for methylation-mediated gene silenc<strong>in</strong>g and ag<strong>in</strong>g. Med Hypotheses1993;41:83e92.[29] Ahuja N, Issa JP. Ag<strong>in</strong>g, methylation and cancer. Histol Histopathol 2000;15:835e42.[30] Young JI, Smith JR. DNA methyltransferase <strong>in</strong>hibition <strong>in</strong> normal human fibroblasts <strong>in</strong>duces a p21-dependentcell cycle withdrawal. J Biol Chem 2001;276:19610e6.[31] Raz<strong>in</strong> A. CpG methylation, chromat<strong>in</strong> structure and gene silenc<strong>in</strong>g-a three-way connection. EMBO J1998;17:4905e8.[32] Guarente L. Sir2 l<strong>in</strong>ks chromat<strong>in</strong> silenc<strong>in</strong>g, metabolism, and ag<strong>in</strong>g. Gene Dev 2000;14:1021e6.[33] Bandyopadhyay D, Medrano EE. The emerg<strong>in</strong>g role of epigenetics <strong>in</strong> cellular and organismal ag<strong>in</strong>g. ExpGerontol 2003;38:1299e307.


CHAPTER 26Ag<strong>in</strong>g and <strong>Disease</strong>: The Epigenetic Bridge[34] Yuasa Y. DNA methylation <strong>in</strong> cancer and age<strong>in</strong>g. Mech Age<strong>in</strong>g Dev 2002;123:1649e54.[35] Liu L, Wylie RC, Andrews LG, Tollefsbol TO. Ag<strong>in</strong>g, cancer and nutrition: the DNA methylation connection.Mech Age<strong>in</strong>g Dev 2003;124:989e98.[36] Teschendorff AE, Menon U, Gentry-Maharaj A, Ramus SJ, Weisenberger DJ, Shen H, et al. Age- dependentDNA methylation of genes that are suppressed <strong>in</strong> stem cells is a hallmark of cancer. Genome Res2010;20:440e6.[37] Rakyan VK, Down TA, Maslau S, Andrew T, Yang TP, Beyan H, et al. <strong>Human</strong> ag<strong>in</strong>g-associated DNA hypermethylationoccurs preferentially at bivalent chromat<strong>in</strong> doma<strong>in</strong>s. Genome Res 2010;20:434e9.[38] Christensen BC, Houseman EA, Marsit CJ, Zheng S, Wrensch MR, Wiemels JL, et al. Ag<strong>in</strong>g and environmentalexposures alter tissue-specific DNA methylation dependent upon CpG island context. PLoS Genet2009;5e1000602:[39] Maegawa S, H<strong>in</strong>kal G, Kim HS, Shen L, Zhang L, Zhang J, et al. Widespread and tissue specific age-related DNAmethylation changes <strong>in</strong> mice. Genome Res 2010;20:332e40.[40] Young JI, Sedivy JM, Smith JR. Telomerase expression <strong>in</strong> normal human fibroblasts stabilizes DNA5-methylcytos<strong>in</strong>e transferase I. J Biol Chem 2003;278:19904e8.[41] Szyf M. A dynamic methylome; implications of non-CpG methylation/demethylation. Cell Cycle 2010;9:3846e7.[42] Zhu B, Zheng Y, Hess D, Angliker H, Schwarz S, Siegmann M, et al. 5-methylcytos<strong>in</strong>e-DNA glycosylase activityis present <strong>in</strong> a cloned G/T mismatch DNA glycosylase associated with the chicken embryo DNA demethylationcomplex. Proc Natl Acad Sci USA 2000;97:5135e9.[43] Barreto G, Schäfer A, Marhold J, Stach D, Swam<strong>in</strong>athan SK, Handa V, et al. Gadd45a promotes epigenetic geneactivation by repair-mediated DNA demethylation. Nature 2007;445:671e5.[44] Brown SE, Suderman MJ, Hallett M, Szyf M. DNA demethylation <strong>in</strong>duced by the methyleCpGeb<strong>in</strong>d<strong>in</strong>gdoma<strong>in</strong> prote<strong>in</strong> MBD3. Gene 2008;420:99e106.[45] Métivier R, Gallais R, Tiffoche C, Le Péron C, Jurkowska RZ, Carmouche RP, et al. Cyclical DNA methylation ofa transcriptionally active promoter. Nature 2008;452:45e50.[46] Bhattacharya SK, Ramchandani S, Cervoni N, Szyf M. A mammalian prote<strong>in</strong> with specific demethylase activityfor mCpG DNA. Nature 1999;397:579e83.[47] Cervoni N, Bhattacharya S, Szyf M. DNA demethylase is a processive enzyme. J Biol Chem 1999;274:8363e6.[48] Detich N, Hamm S, Just G, Knox JD, Szyf M. The methyl donor S-Adenosylmethion<strong>in</strong>e <strong>in</strong>hibits activedemethylation of DNA: a candidate novel mechanism for the pharmacological effects of S-Adenosylmethion<strong>in</strong>e.J Biol Chem 2003;278:20812e20.[49] Levenson JM, Roth TL, Lub<strong>in</strong> FD, Miller CA, Huang IC, Desai P, et al. Evidence that DNA (cytos<strong>in</strong>e-5)methyltransferase regulates synaptic plasticity <strong>in</strong> the hippocampus. J Biol Chem 2006;281:15763e73.[50] Murgatroyd C, Patchev AV, Wu Y, Micale V, Bockmühl Y, Fischer D, et al. Dynamic DNA methylation programspersistent adverse effects of early-life stress. Nat Neurosci 2009;12:1559e66.[51] Szyf M. <strong>Epigenetics</strong>, DNA methylation, and chromat<strong>in</strong> modify<strong>in</strong>g drugs. Annu Rev Pharmacol Toxicol2009;49:243e63.[52] Lucarelli M, Fuso A, Strom R, Scarpa S. The dynamics of myogen<strong>in</strong> site-specific demethylation is stronglycorrelated with its expression and with muscle differentiation. J Biol Chem 2001;276:7500e6.[53] Fuso A, Ferraguti G, Grandoni F, Ruggeri R, Scarpa S, Strom R, et al. Early demethylation of non-CpG, CpCrich,elements <strong>in</strong> the myogen<strong>in</strong> 5 0 -flank<strong>in</strong>g region: a prim<strong>in</strong>g effect on the spread<strong>in</strong>g of active demethylation.Cell Cycle 2010;9:3965e76.[54] Fuso A, Nicolia V, Cavallaro RA, Scarpa S. DNA methylase and demethylase activities are modulated by onecarbonmetabolism <strong>in</strong> Alzheimer’s disease models. J Nutr Biochem 2011;122:242e51.[55] Fuso A, Nicolia V, Pasqualato A, Fiorenza MT, Cavallaro RA, Scarpa S. Changes <strong>in</strong> Presenil<strong>in</strong> 1 gene methylationpattern <strong>in</strong> diet-<strong>in</strong>duced B vitam<strong>in</strong> deficiency. Neurobiol Ag<strong>in</strong>g 2011;32:187e99.[56] Mastroeni D, Grover A, Delvaux E, Whiteside C, Coleman PD, Rogers J. Epigenetic mechanisms <strong>in</strong> Alzheimer’sdisease. Neurobiol Ag<strong>in</strong>g 2011;7:1161e80.[57] Kangaspeska S, Stride B, Métivier R, Polycarpou-Schwarz M, Ibberson D, Carmouche RP, et al. Transientcyclical methylation of promoter DNA. Nature 2008;452:12e5.[58] Weaver IC, Champagne FA, Brown SE, Dymov S, Sharma S, Meaney MJ, et al. Reversal of maternalprogramm<strong>in</strong>g of stress responses <strong>in</strong> adult offspr<strong>in</strong>g through methyl supplementation: alter<strong>in</strong>g epigeneticmark<strong>in</strong>g later <strong>in</strong> life. J Neurosci 2005;25:11045e54.[59] Roth TL, Sweatt JD. Epigenetic mark<strong>in</strong>g of the BDNF gene by early-life adverse experiences. Horm Behav2011;59:315e20.[60] Yakovlev A, Khafizova M, Abdullaev Z, Louk<strong>in</strong>ov D, Kondratyev A. Epigenetic regulation of caspase-3 geneexpression <strong>in</strong> rat bra<strong>in</strong> development. Gene 2010;450:103e8.539


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>540[61] Calvanese V, Lara E, Kahn A, Fraga MF. The role of epigenetics <strong>in</strong> ag<strong>in</strong>g and age-related diseases. Age<strong>in</strong>g Res Rev2009;8:268e76.[62] Kriaucionis S, He<strong>in</strong>tz N. The nuclear DNA base 5-hydroxymethylcytos<strong>in</strong>e is present <strong>in</strong> Purk<strong>in</strong>je neurons andthe bra<strong>in</strong>. <strong>Science</strong> 2009;324:929e30.[63] Tahiliani M, Koh KP, Shen Y, Pastor WA, Bandukwala H, Brudno Y, et al. Conversion of 5-methylcytos<strong>in</strong>e to5- hydroxymethylcytos<strong>in</strong>e <strong>in</strong> mammalian DNA by MLL partner TET1. <strong>Science</strong> 2009;324:930e5.[64] Ito S, D’Alessio AC, Taranova OV, Hong K, Sowers LC, Zhang Y. Role of Tet prote<strong>in</strong>s <strong>in</strong> 5mC to 5hmCconversion, ES-cell self-renewal and <strong>in</strong>ner cell mass specification. Nature 2010;466:1129e33.[65] Williams K, Christensen J, Pedersen MT, Johansen JV, Cloos PA, Rappsilber J, et al. TET1 and hydroxymethylcytos<strong>in</strong>e<strong>in</strong> transcription and DNA methylation fidelity. Nature 2011;473:343e8.[66] Val<strong>in</strong>luck V, Tsai HH, Rogstad DK, Burdzy A, Bird A, Sowers LC. Oxidative damage to methyl-CpG sequences<strong>in</strong>hibits the b<strong>in</strong>d<strong>in</strong>g of the methyl-CpG b<strong>in</strong>d<strong>in</strong>g doma<strong>in</strong> (MBD) of methyl-CpG b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> 2 (MeCP2).Nucleic Acids Res 2004;32:4100e8.[67] Ficz G, Branco MR, Seisenberger S, Santos F, Krueger F, Hore TA, et al. Dynamic regulation of5-hydroxymethylcytos<strong>in</strong>e <strong>in</strong> mouse ES cells and dur<strong>in</strong>g differentiation. Nature 2011;473:398e402.[68] Song CX, Szulwach KE, Fu Y, Dai Q, Yi C, Li X, et al. Selective chemical label<strong>in</strong>g reveals the genome-widedistribution of 5-hydroxymethylcytos<strong>in</strong>e. Nat Biotechnol 2011;29:68e72.[69] Chouliaras L, van den Hove DL, Kenis G, Dela Cruz J, Lemmens MA, van Os J, et al. Caloric restrictionattenuates age-related changes of DNA methyltransferase 3a <strong>in</strong> mouse hippocampus. Bra<strong>in</strong> Behav Immun2011;25:616e23.[70] Chouliaras L, van den Hove DL, Kenis G, Keitel S, Hof PR, van Os J, et al. Prevention of age-related changes <strong>in</strong>hippocampal levels of 5-methylcytid<strong>in</strong>e by caloric restriction. Neurobiol Ag<strong>in</strong>g 2011. DOI: http://dx.doi.org/10.1016/j.neurobiolag<strong>in</strong>g.2011.06.003.[71] Clayton AL, Hazzal<strong>in</strong> CA, Mahadevan LC. Enhanced histone acetylation and transcription: a dynamicperspective. Mol Cell 2006;23:289e96.[72] Sokolova NE, Shiryaeva NV, Dyuzhikova NA, Savenko YN, Vaido AI. Effect of long-term mental and pa<strong>in</strong> stresson the dynamics of H4 histone acetylation <strong>in</strong> hippocampal neurons of rats with different levels of nervoussystem excitability. Bull Exp Biol Med 2006;142:341e3.[73] Impey S. A histone deacetylase regulates addiction. Neuron 2007;56:415e7.[74] Aizawa S, Yamamuro Y. Involvement of histone acetylation <strong>in</strong> the regulation of chol<strong>in</strong>e acetyltransferase gene<strong>in</strong> NG108-15 neuronal cells. Neurochem Int 2010;56:627e33.[75] Wu J, Basha MR, Zawia NH. The environment, epigenetics and amyloidogenesis. J Mol Neurosci 2008;34:1e7.[76] Chen PS, Wang CC, Bortner CD, Peng GS, Wu X, Pang H, et al. Valproic acid and other histone deacetylase<strong>in</strong>hibitors <strong>in</strong>duce microglial apoptosis and attenuate lipopolysaccharide-<strong>in</strong>duced dopam<strong>in</strong>ergic neurotoxicity.Neuroscience 2007;149:203e12.[77] Echaniz-Laguna A, Bousiges O, Loeffler JP, Boutillier AL. Histone deacetylase <strong>in</strong>hibitors: therapeutic agents andresearch tools for decipher<strong>in</strong>g motor neuron diseases. Curr Med Chem 2008;15:1263e73.[78] Gray SG, Dangond F. Rationale for the use of histone deacetylase <strong>in</strong>hibitors as a dual therapeutic modality <strong>in</strong>multiple sclerosis. <strong>Epigenetics</strong> 2006;1:67e75.[79] Tahiliani M, Mei P, Fang R, Leonor T, Rutenberg M, Shimizu F, et al. The histone H3K4 demethylase SMCXl<strong>in</strong>ks REST target genes to X-l<strong>in</strong>ked mental retardation. Nature 2007;447:601e5.[80] Yoo AS, Staahl BT, Chen L, Crabtree GR. MicroRNA-mediated switch<strong>in</strong>g of chromat<strong>in</strong>-remodell<strong>in</strong>g complexes<strong>in</strong> neural development. Nature 2009;460:642e6.[81] Khudayberdiev S, Fiore R, Schratt G. MicroRNA as modulators of neuronal responses. Commun Integr Biol2009;2:411e3.[82] Maes OC, Chertkow HM, Wang E, Schipper HM. MicroRNA: Implications for Alzheimer <strong>Disease</strong> and other<strong>Human</strong> CNS. Disorders. Curr Genomics 2009;10:154e68.[83] Morrison JH, Hof PR. Life and death of neurons <strong>in</strong> the ag<strong>in</strong>g bra<strong>in</strong>. <strong>Science</strong> 1997;278:412e9.[84] Hof PR, Morrison JH. The ag<strong>in</strong>g bra<strong>in</strong>: morphomolecular senescence of cortical circuits. Trends Neurosci2004;27:607e13.[85] Dickste<strong>in</strong> DL, Kabaso D, Rocher AB, Luebke JI, Wearne SL, Hof PR. Changes <strong>in</strong> the structural complexity of theaged bra<strong>in</strong>. Ag<strong>in</strong>g Cell 2007;6:275e84.[86] Rutten BP, Schmitz C, Gerlach OH, Oyen HM, de Mesquita EB, et al. The ag<strong>in</strong>g bra<strong>in</strong>: accumulation of DNAdamage or neuron loss? Neurobiol Ag<strong>in</strong>g 2007;28:91e8.[87] Wu W, Brickman AM, Luchs<strong>in</strong>ger J, Ferrazzano P, Pichiule P, Yoshita M, et al. The bra<strong>in</strong> <strong>in</strong> the age of old:the hippocampal formation is targeted differentially by diseases of late life. Ann Neurol2008;64:698e706.


CHAPTER 26Ag<strong>in</strong>g and <strong>Disease</strong>: The Epigenetic Bridge[88] Luebke JI, Weaver CM, Rocher AB, Rodriguez A, Crim<strong>in</strong>s JL, Dickste<strong>in</strong> DL, et al. Dendritic vulnerability <strong>in</strong>neurodegenerative disease: <strong>in</strong>sights from analyses of cortical pyramidal neurons <strong>in</strong> transgenic mouse models.Bra<strong>in</strong> Struct Funct 2010;214:181e99.[89] Lee CK, We<strong>in</strong>druch R, Prolla TA. Gene-expression profile of the age<strong>in</strong>g bra<strong>in</strong> <strong>in</strong> mice. Nat Genet2000;25:294e7.[90] Lu T, Pan Y, Kao SY, Li C, Kohane I, Chan J, et al, B. A. Gene regulation and DNA damage <strong>in</strong> the age<strong>in</strong>g humanbra<strong>in</strong>. Nature 2004;429:883e91.[91] Berchtold NC, Cribbs DH, Coleman PD, Rogers J, Head E, Kim R, et al. Gene expression changes <strong>in</strong> the courseof normal bra<strong>in</strong> ag<strong>in</strong>g are sexually dimorphic. Proc Natl Acad Sci USA 2008;105:15605e10.[92] Penner MR, Roth TL, Barnes CA, Sweatt JD. An epigenetic hypothesis of ag<strong>in</strong>g-related cognitive dysfunction.Front Ag<strong>in</strong>g Neurosci 2010;2:9.[93] Peleg S, Sananbenesi F, Zovoilis A, Burkhardt S, Bahari-Javan S, Agis-Balboa RC, et al. Altered histone acetylationis associated with age-dependent memory impairment <strong>in</strong> mice. <strong>Science</strong> 2010;328:753e6.[94] Kukull WA, Higdon R, Bowen JD, McCormick WC, Teri L, Schellenberg GD, et al. Dementia and Alzheimerdisease <strong>in</strong>cidence: a prospective cohort study. Arch Neurol 2002;59:1737e46.[95] Crouch PJ, Cimd<strong>in</strong>s K, Duce JA, Bush AI, Trounce IA. Mitochondria <strong>in</strong> ag<strong>in</strong>g and Alzheimer’s disease.Rejuvenation Res 2007;10:349e57.[96] Fuller SJ, Tan RS, Mart<strong>in</strong>s RN. Androgens <strong>in</strong> the etiology of Alzheimer’s disease <strong>in</strong> ag<strong>in</strong>g men and possibletherapeutic <strong>in</strong>terventions. J Alzheimer’s Dis 2007;12:129e42.[97] Thibault O, Gant JC, Landfield PW. Expansion of the calcium hypothesis of bra<strong>in</strong> ag<strong>in</strong>g and Alzheimer’sdisease: m<strong>in</strong>d<strong>in</strong>g the store. Ag<strong>in</strong>g Cell 2007;6:307e17.[98] Landfield PW, Blalock EM, Chen KC, Porter NM. A new glucocorticoid hypothesis of bra<strong>in</strong> ag<strong>in</strong>g: implicationsfor Alzheimer’s disease. Curr Alzheimer Res 2007;4:205e12.[99] Duenas-Gonzalez A, Candelaria M, Perez-Plascencia C, Perez-Cardenas E, de la Cruz-Hernandez E,Herrera LA. Valproic acid as epigenetic cancer drug: precl<strong>in</strong>ical, cl<strong>in</strong>ical and transcriptional effects on solidtumors. Cancer Treat Rev 2008;34:206e22.[100] Craft S. Insul<strong>in</strong> resistance syndrome and Alzheimer’s disease: age- and obesity-related effects on memory,amyloid, and <strong>in</strong>flammation. Neurobiol Ag<strong>in</strong>g 2005;26:65e9.[101] Bailey TL, Rivara CB, Rocher AB, Hof PR. The nature and effects of cortical microvascular pathology <strong>in</strong> ag<strong>in</strong>gand Alzheimer’s disease. Neurol Res 2004;26:57357e8.[102] Selkoe DJ. Ag<strong>in</strong>g, amyloid, and Alzheimer’s disease: a perspective <strong>in</strong> honor of Carl Cotman. Neurochem Res2003;28:1705e13.[103] Maeda S, Sahara N, Saito Y, Murayama S, Ikai A, Takashima A. Increased levels of granular tau oligomers: anearly sign of bra<strong>in</strong> ag<strong>in</strong>g and Alzheimer’s disease. Neurosci Res 2006;54:197e201.[104] Fraga MF, Esteller M. <strong>Epigenetics</strong> and ag<strong>in</strong>g: the targets and the marks. Trends Genet 2007;23:413e8.[105] Mays-Hoopes LL. DNA methylation <strong>in</strong> ag<strong>in</strong>g and cancer. J Gerontol 1989;44:35e6.[106] Yu MK. <strong>Epigenetics</strong> and chronic lymphocytic leukemia. Am J Hematol 2006;81:864e9.[107] Wilson VL, Jones PA. DNA methylation decreases <strong>in</strong> ag<strong>in</strong>g but not <strong>in</strong> immortal cells. <strong>Science</strong>1983;220:1055e10057.[108] Lopat<strong>in</strong>a N, Haskell JF, Andrews LG, Poole JC, Saldanha S, et al. Differential ma<strong>in</strong>tenance and de novomethylat<strong>in</strong>g activity by three DNA methyltransferases <strong>in</strong> ag<strong>in</strong>g and immortalized fibroblasts. J Cell Biochem2002;84:324e34.[109] Varela-Moreiras G, Pérez-Olleros L, García-Cuevas M, Ruiz-Roso B. Effects of age<strong>in</strong>g on folate metabolism <strong>in</strong>rats fed a long-term folate deficient diet. Int J Vitam Nutr Res 1994;64:294e9.[110] Richardson BC. Role of DNA methylation <strong>in</strong> the regulation of cell function: autoimmunity, ag<strong>in</strong>g and cancer.J Nutr 2002;132:2401Se5S.[111] Bollati V, Galimberti D, Pergoli L, Dalla Valle E, Barretta F, Cort<strong>in</strong>i F, et al. DNA methylation <strong>in</strong> repetitiveelements and Alzheimer disease. Bra<strong>in</strong> Behav Immun 2011;25:1078e83.[112] Tohgi H, Utsugisawa K, Nagane Y, Yoshimura M, Genda Y, Ukitsu M. Reduction with age <strong>in</strong> methylcytos<strong>in</strong>e <strong>in</strong>the promoter region -224 approximately -101 of the amyloid precursor prote<strong>in</strong> gene <strong>in</strong> autopsy human cortex.Mol. Bra<strong>in</strong> Res 1999;70:288e92.[113] Tohgi H, Utsugisawa K, Nagane Y, Yoshimura M, Ukitsu M, Genda Y. The methylation status of cytos<strong>in</strong>es <strong>in</strong>a tau gene promoter region alters with age to downregulate transcriptional activity <strong>in</strong> human cerebral cortex.Neurosci Lett 1999;275:89e92.[114] Tohgi H, Utsugisawa K, Nagane Y, Yoshimura M, Ukitsu M, Genda Y. Decrease with age <strong>in</strong> methylcytos<strong>in</strong>es <strong>in</strong>the promoter region of receptor for advanced glycated end products (RAGE) gene <strong>in</strong> autopsy human cortex.Bra<strong>in</strong> Res Mol Bra<strong>in</strong> Res 1999;65:124e8.541


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>542[115] Zhang Z, Deng C, Lu Q, Richardson B. Age-dependent DNA methylation changes <strong>in</strong> the ITGAL (CD11a)promoter. Mech Age<strong>in</strong>g Dev 2002;123:1257e68.[116] Issa JP, Ottaviano YL, Celano P, Hamilton SR, Davidson NE, Bayl<strong>in</strong> SB. Methylation of the oestrogen receptorCpG island l<strong>in</strong>ks age<strong>in</strong>g and neoplasia <strong>in</strong> human colon. Nat Genet 1994;7:536e40.[117] Issa JP, Vert<strong>in</strong>o PM, Boehm CD, Newsham IF, Bayl<strong>in</strong> SB. Switch from monoallelic to biallelic human IGF2promoter methylation dur<strong>in</strong>g ag<strong>in</strong>g and carc<strong>in</strong>ogenesis. Proc Natl Acad Sci USA 1996;93:11757e62.[118] Ladd-Acosta C, Pevsner J, Sabunciyan S, Yolken RH, Webster MJ, D<strong>in</strong>k<strong>in</strong>s T, et al. DNA methylation signatureswith<strong>in</strong> the human bra<strong>in</strong>. Am J Hum Genet 2007;81:1304e15.[119] Lahiri DK, Maloney B, Basha MR, Ge YW, Zawia NH. How and when environmental agents and dietaryfactors affect the course of Alzheimer’s disease: the “LEARn” model (latent early-life associated regulation)may expla<strong>in</strong> the trigger<strong>in</strong>g of AD. Curr Alzheimer Res 2007;4:219e28.[120] Fuso A, Nicolia V, Cavallaro RA, Ricceri L, D’Anselmi F, Coluccia P, et al. B-Vitam<strong>in</strong> deprivation <strong>in</strong>duceshyperhomocyste<strong>in</strong>emia and bra<strong>in</strong> S-adenosylhomocsyte<strong>in</strong>e, depletes bra<strong>in</strong> S-adenosylmethion<strong>in</strong>e, andenhances PSEN1 and BACE expression and Amyloid-b deposition <strong>in</strong> mice. Mol Cell Neurosci2008;37:731e46.[121] Chouliaras L, Rutten BP, Kenis G, Peerbooms O, Visser PJ, Verhey F, et al. Epigenetic regulation <strong>in</strong> thepathophysiology of Alzheimer’s disease. Prog Neurobiol 2010;90:498e510.[122] Carnero A, Lleonart ME. Epigenetic mechanisms <strong>in</strong> senescence, immortalisation and cancer. Biol Rev CambPhilos Soc 2011;86:443e55.[123] Fernández-Morera JL, Calvanese V, Rodríguez-Rodero S, Menández-Torre E, Fraga MF. Epigenetic regulationof the immune system <strong>in</strong> health and disease. Tissue Antigens 2010;76:431e9.[124] Hernandez DG, Nalls MA, Gibbs JR, Arepalli S, van der Brug M, Chong S, et al. Dist<strong>in</strong>ct DNA methylationchanges highly correlated with chronological age <strong>in</strong> the human bra<strong>in</strong>. Hum Mol Genet2011;20:1164e72.[125] Poulsen P, Esteller M, Vaag A, Fraga MF. The epigenetic basis of tw<strong>in</strong> discordance <strong>in</strong> age-related diseases.Pediatr Res 2007;61:38Re42R.[126] Wang CS, Burke JR, Steffens DC, Hulette CM, Breitner JC, Plassman BL. Tw<strong>in</strong> pairs discordant for neuropathologicallyconfirmed Lewy body dementia. J Neurol Neurosurg Psychiatry 2009;80:562e5.[127] Mastroeni D, McKee A, Grover A, Rogers J, Coleman PD. Epigenetic differences <strong>in</strong> cortical neurons froma pair of monozygotic tw<strong>in</strong>s discordant for Alzheimer’s disease. PLoS One 2009;4e6617:[128] Rodríguez-Rodero S, Fernández-Morera JL, Fernandez AF, Menéndez-Torre E, Fraga MF. Epigenetic regulationof ag<strong>in</strong>g. Discov Med 2010;10:225e33.[129] Coppedè F, Migliore L. Evidence l<strong>in</strong>k<strong>in</strong>g genetics, environment, and epigenetics to impaired DNA repair <strong>in</strong>Alzheimer’s disease. J Alzheimer’s Dis 2010;20:953e66.[130] Kaliman P, Párrizas M, Lalanza JF, Cam<strong>in</strong>s A, Escorihuela RM, Pallàs M. Neurophysiological and epigeneticeffects of physical exercise on the ag<strong>in</strong>g process. Age<strong>in</strong>g Res Rev 2011;10(4):475e86.[131] McKay JA, Mathers JC. Diet <strong>in</strong>duced epigenetic changes and their implications for health. Acta Physiol2011;202:103e18.[132] Simmons R. <strong>Epigenetics</strong> and maternal nutrition: nature v. nurture. Proc Nutr Soc 2011;70:73e81.[133] Fuso A, Scarpa S. One-carbon metabolism and Alzheimer’s disease: is it all a methylation matter? NeurobiolAg<strong>in</strong>g 2011;32:1192e5.[134] Thompson RF, E<strong>in</strong>ste<strong>in</strong> FH. Epigenetic basis for fetal orig<strong>in</strong>s of age-related disease. J Womens Health(Larchmt) 2010;19:581e7.[135] Fulop T, Larbi A, Witkowski JM, McElhaney J, Loeb M, Mitnitski A, et al. Ag<strong>in</strong>g, frailty and age-relateddiseases. Biogerontology 2010;11:547e63.[136] Lang PO, Michel JP, Zekry D. Frailty syndrome: a transitional state <strong>in</strong> a dynamic process. Gerontology2009;55:539e49.[137] Top<strong>in</strong>ková E. Ag<strong>in</strong>g, disability and frailty. Ann Nutr Metab 2008;52:6e11.[138] Bellizzi D, D’Aquila P, Montesanto A, Corsonello A, Mari V, Mazzei B, et al. Global DNA methylation <strong>in</strong> oldsubjects is correlated with frailty. Age 2012;34:169e79.[139] Phan HM, Alpert JS, Fa<strong>in</strong> M. Frailty, <strong>in</strong>flammation, and cardiovascular disease: evidence of a connection. AmJ Geriatr Cardiol 2008;17:101e7.[140] Ceven<strong>in</strong>i E, Caruso C, Candore G, Capri M, Nuzzo D, Duro G, et al. Age-related <strong>in</strong>flammation: thecontribution of different organs, tissues and systems. How to face it for therapeutic approaches. Curr PharmDes 2010;16:609e18.[141] Kaiser M, Band<strong>in</strong>elli S, Lunenfeld B. Frailty and the role of nutrition <strong>in</strong> older people. A review of the currentliterature. Acta Biomed 2010;81:37e45.[142] Choi SW, Friso S. <strong>Epigenetics</strong>: A New Bridge between Nutrition and Health. Adv Nutr 2010;1:8e16.


CHAPTER 26Ag<strong>in</strong>g and <strong>Disease</strong>: The Epigenetic Bridge[143] Ozanne SE, Sandovici I, Constância M. Maternal diet, ag<strong>in</strong>g and diabetes meet at a chromat<strong>in</strong> loop. Ag<strong>in</strong>g2011;3:548e54.[144] Gonzalo S. Epigenetic alterations <strong>in</strong> ag<strong>in</strong>g. J Appl Physiol 2010;109:586e97.[145] Kaszubowska L. Telomere shorten<strong>in</strong>g and age<strong>in</strong>g of the immune system. J Physiol Pharmacol 2008;59:169e86.[146] Donate LE, Blasco MA. Telomeres <strong>in</strong> cancer and age<strong>in</strong>g. Philos Trans R Soc Lond B Biol Sci 2011;366:76e84.[147] Gonzalez-Suarez I, Gonzalo S. Crosstalk between chromat<strong>in</strong> structure, nuclear compartmentalization, andtelomere biology. Cytogenet Genome Res 2008;122:202e10.[148] Artandi SE, De P<strong>in</strong>ho RA. Telomeres and telomerase <strong>in</strong> cancer. Carc<strong>in</strong>ogenesis 2010;31:9e18.[149] Zhu J, Zhao Y, Wang S. Chromat<strong>in</strong> and epigenetic regulation of the telomerase reverse transcriptase gene.Prote<strong>in</strong> Cell 2010;1:22e32.[150] Esteller M. Epigenetic gene silenc<strong>in</strong>g <strong>in</strong> cancer: the DNA hypermethylome. Hum Mol Genet 2007;16:R50e9.[151] Chiang PK. Biological effects of <strong>in</strong>hibitors of S-adenosylhomocyste<strong>in</strong>e hydrolase. Pharmacol Ther1998;77:115e34.[152] Chiang PK, Gordon RK, Tal J, Zeng GC, Doctor BP, Pardhasaradhi K, et al. S-Adenosylmethion<strong>in</strong>e andmethylation. FASEB J 1996;10:471e80.[153] Med<strong>in</strong>a MA, Urdiales JL, Amores-Sanchez MI. Roles of homocyste<strong>in</strong>e <strong>in</strong> cell metabolism: old and newfunctions. Eur J Biochem 2001;268:3871e82.[154] F<strong>in</strong>kelste<strong>in</strong> JD. Homocyste<strong>in</strong>e: a History <strong>in</strong> progress. Nutr Rev 2000;58:193e204.[155] Williams KT, Schal<strong>in</strong>ke KL. New <strong>in</strong>sight <strong>in</strong>to the regulation of methyl group and homocyste<strong>in</strong>e metabolism.J Nutr 2007;137:311e4.[156] Vitvitsky V, Thomas M, Ghorpade A, Gendelman HE, Banerjee R. A functional transsulfuration pathway <strong>in</strong>the bra<strong>in</strong> l<strong>in</strong>ks to glutathione homeostasis. J Biol Chem 2006;281:35785e93.[157] Liu H, Wang H, Shenvi S, Hagen TM, Liu RM. Glutathione metabolism dur<strong>in</strong>g ag<strong>in</strong>g and <strong>in</strong> Alzheimerdisease. Ann N Y Acad Sci 2004;1019:346e9.[158] Caudill MA, Wang JC, Melnyk S, Pogribny IP, Jernigan S, Coll<strong>in</strong>s MD, et al. Intracellular S-adenosylhomocyste<strong>in</strong>econcentrations predict global DNA hypomethylation <strong>in</strong> tissues of methyl-deficient cystathion<strong>in</strong>ebeta-synthase heterozygous mice. J Nutr 2001;131:2811e8.[159] Ho PI, Coll<strong>in</strong>s SC, Dhitavat S, Ortiz D, Ashl<strong>in</strong>e D, Rogers E, et al. Homocyste<strong>in</strong>e potentiates beta-amyloidneurotoxicity: role of oxidative stress. J Neurochem 2001;78:249e53.[160] Ulrey CL, Liu L, Andrews LG, Tollefsbol TO. The impact of metabolism on DNA methylation. Hum. Mol.Genet 2005;14:R139e47.[161] Seshadri S, Beiser A, Selhub J, Jacques PF, Rosenberg IH, D’Agost<strong>in</strong>o RB, et al. Plasma homocyste<strong>in</strong>e as a riskfactor for dementia and Alzheimer’s disease. N Engl J Med 2002;346:476e83.[162] Quadri P, Fragiacomo C, Pezzati R, Zanda E, Forloni G, Tettamanti M, et al. Homocyste<strong>in</strong>e, folate, andvitam<strong>in</strong> B-12 <strong>in</strong> mild cognitive impairment, Alzheimer disease, and vascular dementia. Am J Cl<strong>in</strong> Nutr2004;80:114e22.[163] De Strooper B. Alzheimer’s disease. Clos<strong>in</strong>g <strong>in</strong> on gamma-secretase. Nature 2000;405:627e9.[164] Vassar R, Bennett BD, Babu-Khan S, Kahn S, Mendiaz EA, Denis P, et al. Beta-secretase cleavage of Alzheimer’samyloid precursor prote<strong>in</strong> by the transmembrane aspartic protease BACE. <strong>Science</strong> 1999;286:735e41.[165] Fontecave M, Atta M, Mulliez E. S-adenosylmethion<strong>in</strong>e: noth<strong>in</strong>g goes to waste. Trends Biochem Sci2004;29:243e9.[166] Cacabelos R. Influence of pharmacogenetic factors on Alzheimer’s disease therapeutics. Neurodegener Dis2008;5:176e8.[167] Liddell MB, Lovestone S, Owen MJ. Genetic risk of Alzheimer’s disease: advis<strong>in</strong>g relatives. Br J Psychiatry2001;178:7e11.[168] Kam<strong>in</strong>sky Z, Wang SC, Petronis A. Complex disease, gender and epigenetics. Ann Med 2006;38:530e44.[169] Dol<strong>in</strong>oy DC, Das R, Weidman JR, Jirtle RL. Metastable epialleles, impr<strong>in</strong>t<strong>in</strong>g, and the fetal orig<strong>in</strong>s of adultdiseases. Pediatr Res 2007;61:30Re7R.[170] Liu L, van Groen T, Kadish I, Tollefsbol TO. DNA methylation impacts on learn<strong>in</strong>g and memory <strong>in</strong> ag<strong>in</strong>g.Neurobiol Ag<strong>in</strong>g 2009;30:549e60.[171] Bjornsson HT, Sigurdsson MI, Fall<strong>in</strong> MD, Irizarry RA, Aspelund T, Cui H, et al. Intra-<strong>in</strong>dividual change overtime <strong>in</strong> DNA methylation with familial cluster<strong>in</strong>g. J Am Med Ass 2008;299:2877e83.[172] Siegmund KD, Connor CM, Campan M, Long TI, Weisenberger DJ, B<strong>in</strong>iszkiewicz D, et al. DNA methylation<strong>in</strong> the human cerebral cortex is dynamically regulated throughout the life span and <strong>in</strong>volves differentiatedneurons. PLoS ONE 2007;2e895:[173] Wang SC, Oelze B, Schumacher A. Age-specific epigenetic drift <strong>in</strong> late-onset Alzheimer’s disease. PLoS ONE2008;3e2698:543


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>544[174] Gillette Guyonnet S, Abellan Van Kan G, Andrieu S, Barberger Gateau P, Berr C, Bonnefoy M, et al. IANA taskforce on nutrition and cognitive decl<strong>in</strong>e with ag<strong>in</strong>g. J Nutr Health Ag<strong>in</strong>g 2007;11:132e52.[175] Haan MN, Miller JW, Aiello AE, Whitmer RA, Jagust WJ, Mungas DM, et al. Homocyste<strong>in</strong>e, B vitam<strong>in</strong>s, andthe <strong>in</strong>cidence of dementia and cognitive impairment: results from the Sacramento Area Lat<strong>in</strong>o Study onAg<strong>in</strong>g. Am J Cl<strong>in</strong> Nutr 2007;85:511e7.[176] Luchs<strong>in</strong>ger JA, Tang MX, Miller J, Green R, Mehta PD, Mayeux R. Relation of plasma homocyste<strong>in</strong>e to plasmaamyloid beta levels. Neurochem Res 2007;32:775e81.[177] Lee ME, Wang H. Homocyste<strong>in</strong>e and hypomethylation. A novel l<strong>in</strong>k to vascular disease. Trends Cardiovasc.Med 1999;9:49e54.[178] Miller AL. The methion<strong>in</strong>e-homocyste<strong>in</strong>e cycle and its effects on cognitive diseases. Altern Med Rev2003;8:7e19.[179] Kennedy BP, Bottiglieri T, Arn<strong>in</strong>g E, Ziegler MG, Hansen LA, Masliah EJ. Elevated S-adenosylhomocyste<strong>in</strong>e <strong>in</strong>Alzheimer bra<strong>in</strong>: <strong>in</strong>fluence on methyltransferases and cognitive function. Neural Transm 2004;111:547e67.[180] Grant WB, Campbell A, Itzhaki RF, Savory J. The significance of environmental factors <strong>in</strong> the etiology ofAlzheimer’s disease. J Alz Dis 2002;4:179e89.[181] Dosunmu R, Wu J, Basha MR, Zawia NH. Environmental and dietary risk factors <strong>in</strong> Alzheimer’s disease.Expert Rev Neurother 2007;7:887e900.[182] Liu L, Li Y, Tollefsbol TO. Gene-environment <strong>in</strong>teractions and epigenetic basis of human diseases. Curr IssuesMol Biol 2008;10:25e36.[183] Sontag E, Nunbhakdi-Craig V, Sontag JM, Diaz-Arrastia R, Ogris E, Dayal S, et al. Prote<strong>in</strong> phosphatase 2Amethyltransferase l<strong>in</strong>ks homocyste<strong>in</strong>e metabolism with tau and amyloid precursor prote<strong>in</strong> regulation.J Neurosci 2007;27:2751e9.[184] Rogaev EI, Lukiw WJ, Lavrush<strong>in</strong>a O, Rogaeva EA, St George-Hyslop PH. The upstream promoter of the betaamyloidprecursor prote<strong>in</strong> gene (APP) shows differential patterns of methylation <strong>in</strong> human bra<strong>in</strong>. Genomics1994;22:340e7.[185] West RL, Lee JM, Maroun LE. Hypomethylation of the amyloid precursor prote<strong>in</strong> gene <strong>in</strong> the bra<strong>in</strong> of anAlzheimer’s disease patient. J Mol Neurosci 1995;6:141e6.[186] Flem<strong>in</strong>g JL, Phiel CJ, Toland AE. The Role for Oxidative Stress <strong>in</strong> Aberrant DNA Methylation <strong>in</strong> Alzheimer’s<strong>Disease</strong>. Curr Alzheimer Res 2011. [In Press].[187] Fuso A, Sem<strong>in</strong>ara L, Cavallaro RA, D’Anselmi F, Scarpa. S. S-adenosylmethion<strong>in</strong>e/homocyste<strong>in</strong>e cycle alterationsmodify DNA methylation status with consequent deregulation of PS1 and BACE and beta-amyloidproduction. Mol Cell Neurosci 2005;28:195e204.[188] Fuso A, Cavallaro RA, Zampelli A, D’Anselmi F, Piscopo P, Confaloni A, et al. Gamma-Secretase is differentiallymodulated by alterations of homocyste<strong>in</strong>e cycle <strong>in</strong> neuroblastoma and glioblastoma cells.J Alzheimer’s Dis 2007;11:275e90.[189] L<strong>in</strong> HC, Hsieh HM, Chen YH, Hu ML. S-Adenosylhomocyste<strong>in</strong>e <strong>in</strong>creases beta-amyloid formation <strong>in</strong> BV-2microglial cells by <strong>in</strong>creased expressions of beta-amyloid precursor prote<strong>in</strong> and presenil<strong>in</strong> 1 and by hypomethylationof these gene promoters. Neurotoxicology 2009;30:622e7.[190] Lee J, Ryu H. Epigenetic modification is l<strong>in</strong>ked to Alzheimer’s disease: is it a maker or a marker? BMB Rep2010;43:649e55.[191] Chen KL, Wang SS, Yang YY, Yuan RY, Chen RM, Hu CJ. The epigenetic effects of amyloid-beta(1-40) onglobal DNA and neprilys<strong>in</strong> genes <strong>in</strong> mur<strong>in</strong>e cerebral endothelial cells. Biochem Biophys Res Commun2009;378:57e61.


CHAPTER 27Early-Life EpigeneticProgramm<strong>in</strong>g of <strong>Human</strong><strong>Disease</strong> and Ag<strong>in</strong>gAlexander M. VaisermanInstitute of Gerontology, Kiev, Ukra<strong>in</strong>eCHAPTER OUTLINE27.1 Introduction 54527.2 Intrauter<strong>in</strong>e GrowthRestriction 54727.3 Fetal Macrosomia 54827.4 Endocr<strong>in</strong>e Programm<strong>in</strong>g Dur<strong>in</strong>gIntrauter<strong>in</strong>e Development 54927.5 Intrauter<strong>in</strong>e Growth Restrictionand Reprogramm<strong>in</strong>g of theHypothalamicePituitaryeAdrenalAxis 55027.6 Early-life Programm<strong>in</strong>g of theGrowth Hormone/Insul<strong>in</strong>-LikeGrowth Factors Axis 55027.7 Early Interventions to Prevent andTreat Endocr<strong>in</strong>eeMetabolicDisturbances 55127.8 Early-Life Nutritional Programm<strong>in</strong>gof Adult Health and Ag<strong>in</strong>g 55227.9 The Thrifty Phenotype and ThriftyEpigenotype Concepts 55227.10 Prenatal Fam<strong>in</strong>e and Adult HealthOutcomes 55427.11 Effect of Prenatal Exposure toMethyl Donors on DevelopmentalProgramm<strong>in</strong>g 55427.12 Long-Term Programm<strong>in</strong>gEffects of PrenatalStress 55627.13 Long-Term Impacts of MaternalSubstance Use Dur<strong>in</strong>gPregnancy 55727.14 Programm<strong>in</strong>g Effect of Early-LifeExposure to EnvironmentalToxicants 55827.15 Epigenetic Risks of AssistedReproductive Technologies 55927.16 Conclusions and FutureDirections 560References 56154527.1 INTRODUCTIONAg<strong>in</strong>g is a complex process result<strong>in</strong>g from the progressive reduction of an <strong>in</strong>dividual’s ability toma<strong>in</strong>ta<strong>in</strong> homeostasis and <strong>in</strong>volv<strong>in</strong>g numerous factors centered on transcriptional changeswith advanced age [1]. The genetic component of ag<strong>in</strong>g received <strong>in</strong>itially all of the attention.However, epigenetic mechanisms <strong>in</strong>clud<strong>in</strong>g DNA methylation, histone modifications, andnon-cod<strong>in</strong>g RNAs have now emerged as key contributors to the alterations of genome structureand function that accompany ag<strong>in</strong>g [2]. Ag<strong>in</strong>g is associated with <strong>in</strong>creased stochastic deregulationof gene expression caused by errors <strong>in</strong> ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g the established epigenetic patterns.Such stochastic changes <strong>in</strong> the epigenome were called “epimutations” by Rob<strong>in</strong> Holliday [3].T. Tollefsbol (Ed): <strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>. DOI: 10.1016/B978-0-12-388415-2.00027-5Copyright Ó 2012 Elsevier Inc. All rights reserved.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>Epimutations have been found to be crucially important as causal factors <strong>in</strong> the age-related<strong>in</strong>crease <strong>in</strong> <strong>in</strong>cidence of cancer [4], but can also play a pivotal role <strong>in</strong> driv<strong>in</strong>g other ag<strong>in</strong>gassociateddiseases. It has been suggested that accumulation of epimutations over a lifetime isa major contributor to age-related decl<strong>in</strong>e of gene function [5]. However, the available data<strong>in</strong>dicate that most of the detectable epigenetic modifications are systematic, and DNA respondsto the environmental stimuli by modify<strong>in</strong>g its epigenetic status <strong>in</strong> an adaptive manner, <strong>in</strong> orderto ma<strong>in</strong>ta<strong>in</strong> a proper functionality [6,7]. Grav<strong>in</strong>a and Vijg [7] suggested that ag<strong>in</strong>g <strong>in</strong> part isdriven by an epigenetic-mediated loss of phenotypic plasticity. Particularly, age-associated DNAhypomethylation can <strong>in</strong>itiate chromosome <strong>in</strong>stability while DNA hypermethylation <strong>in</strong>promoter regions suppresses expression of normal genes (e.g. tumor-suppressor genes).Thus, theoretically, age-related hypo- or hypermethylation can impair or enhance normal generesponsiveness to environmental signals, <strong>in</strong> turn contribut<strong>in</strong>g to generalized functionaldecl<strong>in</strong>e and failure of homeostasis [7]. Such changes may be a result of an “epigenetic drift”caused by <strong>in</strong>sufficient ma<strong>in</strong>tenance of epigenetic marks, but can also be <strong>in</strong>duced by environmentalfactors. Fraga et al. [8] found that patterns of DNA methylation and histone acetylationdiverge with age <strong>in</strong> all sets of monozygotic tw<strong>in</strong>s studied, but those tw<strong>in</strong>s who haddifferent lifestyles, and had spent less of their lives together, were found to be the mostepigenetically dissimilar. These results suggest that epigenetic divergence that occurs overa lifetime is not solely due to <strong>in</strong>tr<strong>in</strong>sic epigenetic drift, but can be at least partially l<strong>in</strong>ked toenvironmental factors. Thus, age-associated changes <strong>in</strong> the epigenome could be seen asa process of lay<strong>in</strong>g down memories of the environments encountered throughout life. Thereare, however, specific stages of development dur<strong>in</strong>g which the epigenetic landscape is morelabile than it is dur<strong>in</strong>g adulthood, therefore the environment with the potential for the mostprofound <strong>in</strong>fluence on epigenetic states is that encountered <strong>in</strong> prenatal life [9,10].546Recently, several new hypotheses have been proposed postulat<strong>in</strong>g the important role of earlylifeevents <strong>in</strong> determ<strong>in</strong><strong>in</strong>g late-life diseases <strong>in</strong>clud<strong>in</strong>g cardiovascular disease, type 2 diabetes,osteoporosis, depression, cognitive impairments, and cancer [11e16]. Accord<strong>in</strong>g to the“developmental programm<strong>in</strong>g” concept proposed by Alan Lucas 20 years ago, events dur<strong>in</strong>gcritical or sensitive periods of development may program long-term or life-time structure orfunction of the organism [17]. The developmental orig<strong>in</strong>s of adult health and disease(DOHaD) hypothesis states that adverse <strong>in</strong>fluences early <strong>in</strong> development, and particularlydur<strong>in</strong>g <strong>in</strong>trauter<strong>in</strong>e life, can program the risks for adverse health outcomes <strong>in</strong> adult life [18,19].This hypothesis has s<strong>in</strong>ce been confirmed <strong>in</strong> a number of animal and human studies [20e23].Increas<strong>in</strong>g evidence has been accumulated <strong>in</strong>dicat<strong>in</strong>g the important role of epigenetic regulation<strong>in</strong> developmental programm<strong>in</strong>g. The genome undergoes major epigenetic alterationsdur<strong>in</strong>g early development, when genome-wide changes <strong>in</strong> epigenetic marks orchestratechromat<strong>in</strong> <strong>in</strong> a way dest<strong>in</strong>ed to form different organs and tissues <strong>in</strong> the body. Once established,the epigenetic marks are stably ma<strong>in</strong>ta<strong>in</strong>ed through somatic cell divisions and createunique, l<strong>in</strong>eage-specific patterns of gene expression. In mammalian development, there aretwo ma<strong>in</strong> periods of epigenetic modification: gametogenesis and early embryogenesis [24].Dur<strong>in</strong>g germ cell development, genome-wide DNA demethylation occurs followed by remethylationbefore fertilization. Early embryogenesis is then characterized by a second genomewidedemethylation wave, and patterns of methylation are re-established after implantation.The postfertilization demethylation and remethylation phases are likely to play a role <strong>in</strong> theremoval of acquired epigenetic modifications, which can be <strong>in</strong>fluenced by <strong>in</strong>dividual geneticand environmental factors [25]. The epigenome is therefore likely to be particularly vulnerableto the adverse <strong>in</strong>fluences dur<strong>in</strong>g gametogenesis and early embryogenesis [24]. Nutritional andendocr<strong>in</strong>e factors have been repeatedly shown to be able to reprogram the epigenotype of theembryo [26,27]. In human be<strong>in</strong>gs, the w<strong>in</strong>dow of epigenetic developmental plasticity extendsfrom preconception to early childhood and <strong>in</strong>volves epigenetic responses to environmentalchanges, which exert their effects dur<strong>in</strong>g life-history phase transitions [28].


CHAPTER 27Early-Life Epigenetic Programm<strong>in</strong>g of <strong>Human</strong> <strong>Disease</strong> and Ag<strong>in</strong>gPresently, alteration <strong>in</strong> epigenetic pathways is considered to be a key mechanism connect<strong>in</strong>gthe early-life events with age-associated diseases <strong>in</strong>clud<strong>in</strong>g cancer, neurodegenerative diseases,and type 2 diabetes [29e32]. The accelerated adult atherogenesis associated with maternalhyperlipidemia is another example of the long-term epigenetic programm<strong>in</strong>g [33,34]. Dysregulation<strong>in</strong> epigenetic pathways can contribute to ag<strong>in</strong>g <strong>in</strong> general as well [2,28,35]. The role ofearly-life epigenetic events <strong>in</strong> developmental programm<strong>in</strong>g of adult disease and ag<strong>in</strong>g has beenrepeatedly reported <strong>in</strong> animal models. However, human data are comparably scarce. Thepurpose of this chapter is to provide a summary of theoretical models and recent researchf<strong>in</strong>d<strong>in</strong>gs which <strong>in</strong>dicate that early-life conditions can program human adult health and ag<strong>in</strong>gvia epigenetic mechanisms.27.2 INTRAUTERINE GROWTH RESTRICTIONIt is well-established that <strong>in</strong>trauter<strong>in</strong>e growth restriction (IUGR) <strong>in</strong>duced by dietary manipulation,hypoxia, prenatal glucocorticoid adm<strong>in</strong>istration, placental <strong>in</strong>sufficiency, maternalstress, smok<strong>in</strong>g, alcohol consumption, etc., can lead to postnatal abnormalities <strong>in</strong> cardiovascular,metabolic, and endocr<strong>in</strong>e function <strong>in</strong> rats, gu<strong>in</strong>ea pigs, sheep, pigs, horses, andprimates [27,36]. In humans, IUGR result<strong>in</strong>g <strong>in</strong> low birth weight and subsequent rapid catchupgrowth has been repeatedly implicated as an important risk factor for the development ofa variety of disorders <strong>in</strong>clud<strong>in</strong>g postnatal hypertension, glucose <strong>in</strong>tolerance, obesity, type 2diabetes, and cardiovascular disease, manifest<strong>in</strong>g as late as decades later [27,37]. Multiplecohort studies have provided evidence that be<strong>in</strong>g born small for gestational age (SGA)<strong>in</strong>creases the risk for adult diseases through various pathways of metabolic dysregulation.SGA <strong>in</strong>fants can exhibit rapid postnatal weight ga<strong>in</strong> (catch-up growth), altered bodycomposition, <strong>in</strong>creased visceral adiposity and low adiponect<strong>in</strong> levels which predispose tometabolic syndrome (a prediabetic condition associated with abdom<strong>in</strong>al obesity, arterialhypertension, and <strong>in</strong>sul<strong>in</strong> resistance), as well as to cardiovascular disease and type 2 diabetesmellitus <strong>in</strong> adulthood [38]. The precise mechanisms of IUGR-related metabolic programm<strong>in</strong>gare poorly understood. Recently, epigenetics has been proposed as an important mechanismfor IUGR-associated programmed changes through environmentally <strong>in</strong>duced changes <strong>in</strong> geneexpression [39e42].547The DOHaD hypothesis has been <strong>in</strong>vestigated <strong>in</strong> a wide range of epidemiological and animalstudies; these <strong>in</strong>vestigations highlight adaptations made by the nutritionally manipulatedfetus that aim to ma<strong>in</strong>ta<strong>in</strong> energy homeostasis to ensure survival. One consequence of suchdevelopmental adaptation may be a long-term resett<strong>in</strong>g of cellular energy homeostasis viaepigenetic modification of genes <strong>in</strong>volved <strong>in</strong> a number of key regulatory pathways. Forexample, reduced maternal-fetal nutrition dur<strong>in</strong>g early and mid gestation affects adipose tissuedevelopment and adiposity of the fetus by sett<strong>in</strong>g an <strong>in</strong>creased number of adipocyte precursorcells [43]. The changes <strong>in</strong> the development of key endocr<strong>in</strong>e axes <strong>in</strong>clud<strong>in</strong>g the reprogramm<strong>in</strong>gof the hypothalamic-pituitary-adrenal (HPA) axis and <strong>in</strong>sul<strong>in</strong>-signal<strong>in</strong>g pathways could be<strong>in</strong>volved [44].In adults born with IUGR, abnormalities <strong>in</strong> the circulat<strong>in</strong>g concentrations of <strong>in</strong>sul<strong>in</strong>, <strong>in</strong>sul<strong>in</strong>likegrowth factors (IGFs), catecholam<strong>in</strong>es, cortisol, and growth hormone (GH) wererepeatedly observed [44,45]. Most of the present data regard<strong>in</strong>g IUGR-related epigeneticmodifications have been generated <strong>in</strong> animal experiments [40,41], while only few humanstudies have been carried out to date. Recently, Lee et al. [46] identified differentially expressedgenes related to the glycolytic pathway between uncomplicated pregnancies and pregnancieswith IUGR. They also compared the concentrations of <strong>in</strong>sul<strong>in</strong> and IGFs <strong>in</strong> cord blood betweenthe two groups. Microarray experiments identified <strong>in</strong>creased expression of glycolytic enzymerelatedgenes, <strong>in</strong>clud<strong>in</strong>g lactate dehydrogenase C, dihydrolipoamide S-acetyltransferase,6 phosphofructo-2-k<strong>in</strong>ase/fructose-2, 6-biphosphatase 2, oxoglutarate dehydrogenase, phosphorylase,and IGF-2 and decreased expression of IGF-1 <strong>in</strong> placentas from pregnancies with


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>IUGR. There were significantly lower concentrations of glucose, <strong>in</strong>sul<strong>in</strong>, IGF-1, and IGF-2 <strong>in</strong>the fetal cord blood of pregnancies with IUGR. Microarray analysis revealed <strong>in</strong>creasedexpression of enzyme genes related to the tricarboxylic acid cycle pathway <strong>in</strong> placentas frompregnancies with IUGR. In the McCarthy et al. [47] study, microarray technology was used toidentify genes, which may impair placentation result<strong>in</strong>g <strong>in</strong> IUGR. The RNA was isolated fromboth IUGR term placentas and normal term placentas. Microarray experiments identified<strong>in</strong>creased expression of certa<strong>in</strong> genes <strong>in</strong>clud<strong>in</strong>g lept<strong>in</strong>, soluble vascular endothelial growthfactor receptor, human chorionic gonadotrop<strong>in</strong>, follistat<strong>in</strong>-like 3, and hypoxia-<strong>in</strong>duciblefactor 2alpha <strong>in</strong> the IUGR. These results were confirmed by real-time PCR. The up-regulationof soluble vascular endothelial growth factor receptor and hypoxia-<strong>in</strong>ducible factor 2alpha atthis period <strong>in</strong> pregnancy <strong>in</strong>dicate that placental angiogenesis is altered <strong>in</strong> IUGR and thathypoxia is a major contributor to maldevelopment of the placental vasculature. IUGR isknown to be associated with <strong>in</strong>creased expression of genes regulat<strong>in</strong>g both cell proliferationand the <strong>in</strong>tr<strong>in</strong>sic pathway of apoptosis. Specifically, IUGR is associated with altered cellturnover <strong>in</strong> the villous trophoblast, an essential functional cell type of the human placenta.The <strong>in</strong>tr<strong>in</strong>sic pathway of apoptosis, particularly p53, is important <strong>in</strong> regulat<strong>in</strong>g placental cellturnover <strong>in</strong> response to damage. In the recent Heazell et al. [48] study, p53 mRNA and prote<strong>in</strong>expression were <strong>in</strong>creased <strong>in</strong> IUGR, which localized to the syncytiotrophoblast.548Currently, a genome-wide epigenetic profil<strong>in</strong>g has become feasible, and a recent study byE<strong>in</strong>ste<strong>in</strong> et al. [49] employ<strong>in</strong>g such a strategy found that the umbilical cord blood cells of<strong>in</strong>fants with IUGR demonstrate subtle but widespread DNA methylation changes across thegenome. E<strong>in</strong>ste<strong>in</strong> et al. [49] hypothesized that IUGR-related programm<strong>in</strong>g is mediated bypermanent epigenetic alterations <strong>in</strong> stem cell populations, and focused their study specificallyon DNA methylation <strong>in</strong> CD34 þ hematopoietic stem and progenitor cells from cord bloodfrom neonates with IUGR and control subjects. They found that changes <strong>in</strong> cytos<strong>in</strong>e methylationoccur <strong>in</strong> response to IUGR of moderate degree and <strong>in</strong>volv<strong>in</strong>g a restricted number of loci.They also identify specific loci that are targeted for dysregulation of DNA methylation, <strong>in</strong>particular the hepatocyte nuclear factor 4alpha (HNF4A) gene, a well-known diabetes candidategene, and other loci encod<strong>in</strong>g HNF4A-<strong>in</strong>teract<strong>in</strong>g prote<strong>in</strong>s. This genome-wide studysuggests that many genes are epigenetically susceptible to alterations <strong>in</strong> maternal nutrition,and that comprehensive effects on the epigenome can be <strong>in</strong>duced by mild as well as severe<strong>in</strong>trauter<strong>in</strong>e <strong>in</strong>sults. It gives the possibility that the epigenetic alterations underly<strong>in</strong>g developmentalprogramm<strong>in</strong>g are not restricted to a few specific genes. It is also possible that smallbut widespread epigenetic alterations <strong>in</strong>duced by a poor <strong>in</strong>trauter<strong>in</strong>e environment canpersisted over a lifetime and hence can lead to the acceleration of an age-associated epigeneticdecl<strong>in</strong>e [10]. A schematic diagram of hypothetical mechanisms potentially underly<strong>in</strong>g the l<strong>in</strong>kbetween IUGR and its long-term adverse health outcomes is presented <strong>in</strong> Figure 27.127.3 FETAL MACROSOMIAWhile the DOHaD hypothesis has focused on the adverse effects of maternal undernutrition,exposure to overnutrition <strong>in</strong> fetal life also results <strong>in</strong> a series of central and peripheral neuroendocr<strong>in</strong>eresponses that <strong>in</strong> turn program development of the fat cell and central appetiteregulatory system [19]. Epidemiologic studies have found that higher maternal gestationalweight ga<strong>in</strong> is associated with fetal macrosomia (arbitrarily def<strong>in</strong>ed as a birth weight of morethan 4000 g) and consequent risk for obesity and its cardiometabolic complications amongoffspr<strong>in</strong>g. There is also some evidence that epigenetic changes might occur <strong>in</strong> response tomaternal overnutrition [50,51]. Altered epigenetic regulation can be <strong>in</strong>duced by both maternalunder- and overnutrition with<strong>in</strong> genes that control lipid and carbohydrate metabolism andwith<strong>in</strong> genes <strong>in</strong>volved <strong>in</strong> the central appetiteeenergy balance neural network [51].In the context of the ’’small baby syndrome hypothesis,’’ it has usually been suggested that therelationship between birth weight and type 2 diabetes is <strong>in</strong>versely l<strong>in</strong>ear, imply<strong>in</strong>g that high


CHAPTER 27Early-Life Epigenetic Programm<strong>in</strong>g of <strong>Human</strong> <strong>Disease</strong> and Ag<strong>in</strong>gFIGURE 27.1Hypothetical model of causes and consequences of IUGR. Exposure to adverse environmental/lifestyle factors (maternal stress, undernutrition, smok<strong>in</strong>g,alcohol consumption, etc.) dur<strong>in</strong>g fetal life can result <strong>in</strong> IUGR and cause epigenetic reprogramm<strong>in</strong>g of gene expression. IUGR conditions are associated withdecreased anabolic hormone levels, <strong>in</strong>creased catabolic hormone concentrations, higher apoptosis rate, and reprogramm<strong>in</strong>g of the HPA axis and GHeIGFaxis <strong>in</strong> the fetus. IUGR leads to low birth weight (


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>FIGURE 27.2Hypothetical model of causes and consequences of fetal macrosomia. Prenatal overnutrition and maternal antibiotics, as well as low physical activitydur<strong>in</strong>g pregnancy can result <strong>in</strong> fetal macrosomia (larger than normal fetal size and weight). Fetal macrosomia-related epigenetic reprogramm<strong>in</strong>g causes<strong>in</strong>creased anabolic hormone levels, decreased catabolic hormone concentrations, and up-regulation of adipogenic genes. It results <strong>in</strong> high birth weight(>4.0 kg) of the baby and high serum IGF-1 levels <strong>in</strong> adulthood. Fetal macrosomia is associated with long-term health problems <strong>in</strong>clud<strong>in</strong>g some cancersand probably type 2 diabetes.by alter<strong>in</strong>g the delivery, uptake, and metabolic fate of nutrients <strong>in</strong> the fetoplacental tissues. Inutero manipulation of glucocorticoid, androgen, and thyroid hormone levels alters fetaldevelopment and has long-term consequences for cardiovascular, reproductive, and metabolicfunction [27].55027.5 INTRAUTERINE GROWTH RESTRICTIONAND REPROGRAMMING OF THEHYPOTHALAMICePITUITARYeADRENAL AXISSeveral experimental and human studies suggest that IUGR permanently resets the HPA axis thatplays an essential role <strong>in</strong> the body’s response to stressful events. Programm<strong>in</strong>g of the HPA axis<strong>in</strong>volves epigenetic remodel<strong>in</strong>g of chromat<strong>in</strong>, lead<strong>in</strong>g to alterations <strong>in</strong> the expression of genes <strong>in</strong>many organs and tissues <strong>in</strong>volved <strong>in</strong> HPA activation and response, <strong>in</strong>clud<strong>in</strong>g the hippocampusand peripheral tissues [62]. HPA axis reprogramm<strong>in</strong>g may <strong>in</strong>volve persistently altered expressionof the hippocampal glucocorticoid receptor (GR), an important regulator of HPA axis reactivity,and postnatal changes <strong>in</strong> hippocampal GR variant and total mRNA expression may underlieIUGR-associated HPA axis reprogramm<strong>in</strong>g [63]. Crucial to proper <strong>in</strong>fant growth and developmentis the placenta, and alterations to placental gene function may reflect differences <strong>in</strong> the<strong>in</strong>trauter<strong>in</strong>e environment which functionally contribute to <strong>in</strong>fant growth and may affect theconsequent health outcomes. To exam<strong>in</strong>e whether epigenetic alterations of the GR gene arel<strong>in</strong>ked to <strong>in</strong>fant growth, Filiberto et al. [64] analyzed 480 human placentas, and exam<strong>in</strong>ed howdifferential methylation of the GR gene exon 1F is associated with fetal growth. A significantassociation between differential methylation of the GR gene and large-for-gestational-age (LGA)status was revealed. This work is one of the first to l<strong>in</strong>k <strong>in</strong>fant growth as a measure of the<strong>in</strong>trauter<strong>in</strong>e environment and epigenetic alterations of the GR gene and suggests that DNAmethylation may be a critical determ<strong>in</strong>ant of placental function.27.6 EARLY-LIFE PROGRAMMING OF THE GROWTH HORMONE/INSULIN-LIKE GROWTH FACTORS AXISThe GH/IGF axis plays a fundamental role <strong>in</strong> somatic growth and cellular differentiation, aswell as <strong>in</strong> metabolism and survival. The processes l<strong>in</strong>k<strong>in</strong>g nutrition, metabolism, and growthare thought to <strong>in</strong>volve a complex <strong>in</strong>terrelationship among <strong>in</strong>sul<strong>in</strong>, GH, IGFs, and <strong>in</strong>sul<strong>in</strong>-likegrowth factor b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s (IGFBPs). Fetal <strong>in</strong>sul<strong>in</strong> and IGFs are thought to have a centralrole <strong>in</strong> the regulation of fetal growth, and the IGFs and IGFBPs are shown to be nutritionallyregulated <strong>in</strong> the fetus [65]. Fetal growth retardation leads to long-term abnormalities <strong>in</strong> theGH/IGF axis [27,36,65], e.g., subjects born SGA had significantly lower mean serum IGF-I,


CHAPTER 27Early-Life Epigenetic Programm<strong>in</strong>g of <strong>Human</strong> <strong>Disease</strong> and Ag<strong>in</strong>gIGF-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>-3 (IGFBP-3) and IGF-I/IGFBP-3 ratio <strong>in</strong> adulthood than those bornappropriate for gestational age [66].Intrauter<strong>in</strong>e programm<strong>in</strong>g of the GHeIGF axis has been proposed as a potential candidatemechanism to expla<strong>in</strong> the l<strong>in</strong>k between low birth weight and adult disease [13,65,67].Accumulat<strong>in</strong>g data from experimental and molecular epidemiological studies <strong>in</strong>dicate thatgrowth factors may be important <strong>in</strong> the pathophysiological processes underly<strong>in</strong>g adult-onsetchronic disease, <strong>in</strong>clud<strong>in</strong>g type 2 diabetes, coronary heart disease, and cancer, and IGF-1signal<strong>in</strong>g is one of the key pathways implicated <strong>in</strong> ag<strong>in</strong>g and longevity [68]. Abnormalities <strong>in</strong>the <strong>in</strong>sul<strong>in</strong> signal<strong>in</strong>g pathway generate age-related diseases and <strong>in</strong>creased mortality, whereasthe GH/IGF-1 axis could potentially modulate human longevity [69].A number of experimental and observational studies suggest that higher levels of circulat<strong>in</strong>g IGF-1may <strong>in</strong>crease risk of several cancers dur<strong>in</strong>g adulthood <strong>in</strong>clud<strong>in</strong>g breast, colorectal, lung, andprostate cancers [70e73], and reduce risk of type 2 diabetes and coronary heart disease [67,74].Overexpression of IGF-1 occurs <strong>in</strong> tumors diagnosed <strong>in</strong> childhood (osteosarcoma, Wilms tumor,neuroblastoma, etc.) and <strong>in</strong> adults (breast, ovaries, colon, and prostate cancer), and congenitalIGF-1 deficiency acts as a protect<strong>in</strong>g factor for the development of cancer [75]. Low-birth-weight<strong>in</strong>fants, on the contrary, have low levels of IGF-1 [76] and high risk of <strong>in</strong>sul<strong>in</strong> resistance and type 2diabetes [77], ischemic heart disease [78], cognitive decl<strong>in</strong>e [79], and osteoporosis [80] <strong>in</strong>adulthood. Ben-Shlomo et al. [81] suggest that low birth weight followed by accelerated catch-upgrowth dur<strong>in</strong>g <strong>in</strong>fancy and childhood is associated with lower life course IGF-1 levels and this mayact as one of the biological pathways l<strong>in</strong>k<strong>in</strong>g early growth with adult <strong>in</strong>sul<strong>in</strong> resistance, hypertension,and cardiovascular disease. Similarly, this pattern should be associated with decreasedcancer risk and is consistent with the observational data on breast-feed<strong>in</strong>g, which has been shownto be protective for IGF-related premenopausal breast cancers [73].27.7 EARLY INTERVENTIONS TO PREVENT AND TREATENDOCRINEeMETABOLIC DISTURBANCESSeveral nutritional and hormonal <strong>in</strong>terventions have been proposed to prevent and treat theendocr<strong>in</strong>eemetabolic disturbances. Milk <strong>in</strong>take <strong>in</strong> childhood and <strong>in</strong> adulthood is positivelyassociated with higher levels of circulat<strong>in</strong>g IGF-1, and higher circulat<strong>in</strong>g IGF-1 promotes l<strong>in</strong>eargrowth. A recent study conducted by Mart<strong>in</strong> et al. [82] <strong>in</strong>dicated that milk consumption <strong>in</strong>childhood appears to have long-term, programm<strong>in</strong>g effects. Specifically, some studies suggestthat the long-term effect of higher levels of milk <strong>in</strong>take <strong>in</strong> early childhood is opposite to theexpected short-term effect, because milk <strong>in</strong>take <strong>in</strong> early-life is <strong>in</strong>versely associated with IGF-1levels throughout adult life. The authors hypothesized that this long-term programm<strong>in</strong>g effectis via a resett<strong>in</strong>g of pituitary control <strong>in</strong> response to raised levels of IGF-1 <strong>in</strong> childhood. Sucha programm<strong>in</strong>g effect of milk <strong>in</strong>take <strong>in</strong> early life could potentially have implications for cancerand ischemic heart disease risk many years later. Early-life diet has been found to affect the riskof childhood leukemia. In the study by Tower and Spector [83], maternal dietary DNAtopoisomerase II (DNAt2) <strong>in</strong>hibitor <strong>in</strong>take was associated with <strong>in</strong>fant acute myeloid leukemiawith the MLL gene translocation. Increased <strong>in</strong>take of fruits and vegetables has been associatedwith decreased leukemia risk, and lack of maternal folate supplementation has been associatedwith <strong>in</strong>creased childhood leukemia risk, possibly by caus<strong>in</strong>g DNA hypomethylation and<strong>in</strong>creased DNA strand breaks. Adm<strong>in</strong>istration of low-dose GH therapy, at a dose that m<strong>in</strong>imizesthe lipolytic effects of GH and has the ability to <strong>in</strong>crease IGF-1 levels, were shown toenhance <strong>in</strong>sul<strong>in</strong> sensitivity <strong>in</strong> young healthy adults and <strong>in</strong> GH-deficient adults and <strong>in</strong>creases<strong>in</strong>sul<strong>in</strong> secretion <strong>in</strong> <strong>in</strong>dividuals with impaired glucose tolerance [84]. IGF system is emerg<strong>in</strong>gas a promis<strong>in</strong>g new target <strong>in</strong> cancer therapy. Different strategies are be<strong>in</strong>g pursued to target thispathway. Several monoclonal antibodies and tyros<strong>in</strong>e k<strong>in</strong>ase <strong>in</strong>hibitors target<strong>in</strong>g the IGF-1receptor are <strong>in</strong> cl<strong>in</strong>ical development [85,86].551


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>27.8 EARLY-LIFE NUTRITIONAL PROGRAMMING OF ADULTHEALTH AND AGINGThere is <strong>in</strong>creas<strong>in</strong>g epidemiological evidence l<strong>in</strong>k<strong>in</strong>g prenatal and early-life nutritional statusto later adiposity and metabolic disease risk [87,88]. Different gestational dietary stressors(undernutrition, overnutrition, or a modified supply of key nutrients) can elicit similarmetabolic responses <strong>in</strong> offspr<strong>in</strong>g [9]. High carbohydrate/prote<strong>in</strong> ratio <strong>in</strong> the maternal diet wasshown to be l<strong>in</strong>ked to impaired glucose homeostasis and raised blood pressure <strong>in</strong> offspr<strong>in</strong>g[89,90]. In the longitud<strong>in</strong>al studies conducted <strong>in</strong> the United K<strong>in</strong>gdom and F<strong>in</strong>land, it has beenshown that subjects that were underweight at birth had high rates of coronary heart disease,high blood pressure, high cholesterol concentrations, and abnormal glucosee<strong>in</strong>sul<strong>in</strong>metabolism <strong>in</strong> adulthood [52,91,92]. The authors suggested that a high carbohydrate <strong>in</strong>take <strong>in</strong>early pregnancy suppresses placental growth, especially if comb<strong>in</strong>ed with a low dairy prote<strong>in</strong><strong>in</strong>take <strong>in</strong> late pregnancy, and such an effect could have long-term consequences for theoffspr<strong>in</strong>g’s risk of cardiovascular disease [91].While the molecular basis of prenatal nutritional programm<strong>in</strong>g is unknown, available animaland human data suggest that epigenetic changes <strong>in</strong> gene expression play a substantial role <strong>in</strong>the l<strong>in</strong>k between the maternal diet, and altered metabolism and body composition <strong>in</strong> the adultoffspr<strong>in</strong>g [93e95]. Accord<strong>in</strong>g to the “epigenetic programm<strong>in</strong>g” hypothesis, suboptimalmaternal diet <strong>in</strong>duces epimutations <strong>in</strong> offspr<strong>in</strong>g dur<strong>in</strong>g early embryonic development, andthat altered expression of affected genes is ma<strong>in</strong>ta<strong>in</strong>ed <strong>in</strong>to adulthood, eventually affect<strong>in</strong>ghealth [9]. Epigenetic mechanisms play a key role <strong>in</strong> mediat<strong>in</strong>g between the early-life nutrient<strong>in</strong>puts and the ensu<strong>in</strong>g phenotypic changes throughout the entire life and seem to beresponsible, <strong>in</strong> part, for the biological changes that occur dur<strong>in</strong>g ag<strong>in</strong>g [96].552In animal models, maternal diet alters offspr<strong>in</strong>g body composition, accompanied by epigeneticchanges <strong>in</strong> metabolic control genes. To study whether such processes operate <strong>in</strong> humans,Godfrey et al. [95] analyzed the associations of methylation status of 68 CpGs 5 0 from fivecandidate genes <strong>in</strong> umbilical cord tissue DNA from healthy neonates with maternal pregnancydiet and with child’s adiposity at age 9 years. Replication was sought <strong>in</strong> a second <strong>in</strong>dependentcohort. In cohort 1, ret<strong>in</strong>oid X receptor-a (RXRA) chr9:136355885þ and endothelial nitricoxide synthase (eNOS) chr7:150315553þ methylation had <strong>in</strong>dependent associations with sexadjustedchildhood fat mass and fat mass percentage. Regression analyses <strong>in</strong>clud<strong>in</strong>g sex andneonatal epigenetic marks expla<strong>in</strong>ed >25% of the variance <strong>in</strong> childhood adiposity. Highermethylation of RXRA chr9:136355885þ, but not of eNOS chr7:150315553þ, was associatedwith lower maternal carbohydrate <strong>in</strong>take <strong>in</strong> early pregnancy, previously l<strong>in</strong>ked with higherneonatal adiposity <strong>in</strong> this population. In cohort 2, cord eNOS chr7:150315553þ methylationshowed no association with adiposity, but RXRA chr9:136355885þ methylation showedsimilar associations with fat mass and fat mass percentage.27.9 THE THRIFTY PHENOTYPE AND THRIFTY EPIGENOTYPECONCEPTSIn 1962, James Neel proposed the “thrifty genotype” hypothesis, the idea that the same genesthat helped our ancestors survive occasional fam<strong>in</strong>es are now be<strong>in</strong>g challenged by modern lifeconditions <strong>in</strong> which food is plentiful [97]. Thirty years ago, <strong>in</strong> 1992, Hales and Barkersuggested the “thrifty phenotype” hypothesis, the concept that environmental factors act<strong>in</strong>g <strong>in</strong>early life, <strong>in</strong> particular undernutrition, might <strong>in</strong>fluence later risk of type 2 diabetes [98].Accord<strong>in</strong>g to this hypothesis, undernutrition dur<strong>in</strong>g <strong>in</strong> utero development results <strong>in</strong> long-termadaptive changes <strong>in</strong> glucosee<strong>in</strong>sul<strong>in</strong> metabolism (<strong>in</strong>clud<strong>in</strong>g reduced capacity for <strong>in</strong>sul<strong>in</strong>secretion and <strong>in</strong>sul<strong>in</strong> resistance) that, due to an enhanced ability to store fat, improves survivalunder postnatal conditions of nutritional deprivation. However, w<strong>in</strong>dows of plasticity closeearly <strong>in</strong> life, and postnatal environmental exposures may result <strong>in</strong> the selected trajectory


CHAPTER 27Early-Life Epigenetic Programm<strong>in</strong>g of <strong>Human</strong> <strong>Disease</strong> and Ag<strong>in</strong>gbecom<strong>in</strong>g <strong>in</strong>appropriate, result<strong>in</strong>g <strong>in</strong> adverse effects on adult health. If mismatch existsbetween the environment predicted <strong>in</strong> utero and the actual environment experienced <strong>in</strong>subsequent life (e.g. excess food consumption), diabetes and other features of the metabolicsyndrome will result. Fetal growth restriction followed by rapid weight ga<strong>in</strong> dur<strong>in</strong>g early<strong>in</strong>fancy (“catch-up growth”) has also been proposed to play an important role <strong>in</strong> promot<strong>in</strong>gcentral adiposity and <strong>in</strong>sul<strong>in</strong> resistance [99].Epigenetic regulation of gene expression is one mechanism by which genetic susceptibility andenvironmental <strong>in</strong>sults can lead to type 2 diabetes. Several studies clearly show that environmentaleffects can <strong>in</strong>duce epigenetic alterations, ultimately affect<strong>in</strong>g expression of key genes l<strong>in</strong>ked to thedevelopment of type 2 diabetes <strong>in</strong>clud<strong>in</strong>g genes critical for pancreatic development and beta-cellfunction, peripheral glucose uptake, and <strong>in</strong>sul<strong>in</strong> resistance, as well as atherosclerosis [100].Recently, Re<strong>in</strong>hard Stöger used elements of the thrifty phenotype and thrifty genotype concepts tosynthesize a “thrifty epigenotype” hypothesis [101]. Accord<strong>in</strong>g to this hypothesis, it has beenpostulated that: (1) metabolic thrift, the capacity for efficient acquisition, storage, and use ofenergy, is an ancient, complex trait; (2) the environmentally responsive gene network encod<strong>in</strong>gthis trait is subject to genetic canalization and thereby has become robust aga<strong>in</strong>st mutationalperturbations; (3) DNA sequence polymorphisms play a m<strong>in</strong>or role <strong>in</strong> the etiology of obesity andtype 2 diabetes d <strong>in</strong>stead, disease susceptibility is predom<strong>in</strong>antly determ<strong>in</strong>ed by epigeneticvariations; (4) correspond<strong>in</strong>g epigenotypes have the potential to be <strong>in</strong>herited across generations;and (5) lept<strong>in</strong> is a candidate gene for the acquisition of a thrifty epigenotype. Accord<strong>in</strong>g to Stöger’svisual metaphor, the efficiency of anabolic metabolism (“metabolic thrift”) is built upon largegene networks that form a rigid canal. In the absence of nutritional extremes (either over- orundernutrition), metabolism develops <strong>in</strong>to the healthy norm. However, under conditions of<strong>in</strong>trauter<strong>in</strong>e malnutrition, compensatory epigenetic changes can be <strong>in</strong>duced <strong>in</strong> adipogenic andenergy metabolism gene networks, and this can change the shape of the canal <strong>in</strong> such a way thatmetabolic phenotype is optimized for survival <strong>in</strong> these conditions. If the “thrifty epigenotype”hypothesis is correct, then the “thrifty epigenotype” is anticipated to be present at significantlyhigher frequencies <strong>in</strong> human populations experienc<strong>in</strong>g recurrent food shortages [101].Individualsexposed to these conditions will have a characteristic epigenetic profile, which could differmarkedly from those for residents of developed countries.553Lept<strong>in</strong> is thought to be one of the best thrifty gene candidates s<strong>in</strong>ce it encodes a hormoneregulat<strong>in</strong>g appetite and energy homeostasis [101]. Lept<strong>in</strong> is secreted by adipocytes, and serumlept<strong>in</strong> level is thought to signal nutritional status to the hypothalamus and thus help governappetite and energy expenditure. Lept<strong>in</strong> has been shown to be implicated <strong>in</strong> nutritionalprogramm<strong>in</strong>g dur<strong>in</strong>g fetal and neonatal growth with long-term effects on susceptibility toobesity, diabetes, and coronary heart disease [102]. The failure of elevated lept<strong>in</strong> levels tosuppress feed<strong>in</strong>g and mediate weight loss <strong>in</strong> common forms of obesity def<strong>in</strong>es a state of socalledlept<strong>in</strong> resistance. The mechanisms underly<strong>in</strong>g lept<strong>in</strong> resistance rema<strong>in</strong> a matter ofdebate, but there is <strong>in</strong>creas<strong>in</strong>g evidence that it may be programmed dur<strong>in</strong>g the fetal andneonatal life [103]. The promoter region of the lept<strong>in</strong> gene is methylated <strong>in</strong> somatic tissues ofhuman and mouse and displays epigenetic variation [104], and it is the gene for whichproximal promoter demethylation has been shown to <strong>in</strong>duce its transcription <strong>in</strong> matureadipocytes [105]. Recently, new evidence of the key role of lept<strong>in</strong> <strong>in</strong> epigenetic programm<strong>in</strong>g ofhuman metabolic disorders was obta<strong>in</strong>ed. Bouchard et al. [106] exam<strong>in</strong>ed the possibility oflept<strong>in</strong> gene epigenetic adaptation to impaired glucose metabolism dur<strong>in</strong>g pregnancy bystudy<strong>in</strong>g whether the DNA methylation profile of lept<strong>in</strong> gene is altered <strong>in</strong> the offspr<strong>in</strong>g ofmothers with gestational impaired glucose tolerance. They have shown that placental lept<strong>in</strong>gene DNA methylation levels were correlated with glucose levels <strong>in</strong> women with impairedglucose tolerance, and with decreased lept<strong>in</strong> gene expression <strong>in</strong> the whole cohort studied. Theauthors hypothesized that <strong>in</strong>creas<strong>in</strong>g maternal glycemia results <strong>in</strong> fetal lept<strong>in</strong> gene DNAdemethylation, which leads to higher mRNA levels and subsequently higher lept<strong>in</strong> levels,possibly promot<strong>in</strong>g lept<strong>in</strong> resistance and obesity development.


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>55427.10 PRENATAL FAMINE AND ADULT HEALTH OUTCOMESThe opportunities to directly exam<strong>in</strong>e the relationship between early-life nutrition and late-lifedisease are limited because few cohort studies have <strong>in</strong>formation from birth until old age. Thestrong evidence l<strong>in</strong>k<strong>in</strong>g early-life conditions with adult disease risk has been accumulatedfrom natural experiments, i.e. naturally occurr<strong>in</strong>g circumstances <strong>in</strong> which subsets of thepopulation have different levels of exposure to a supposed causal factor [23]. The fam<strong>in</strong>e hasmultiple beneficial features for use as a natural experiment. A relationship between theprenatal exposure to the fam<strong>in</strong>e and various adverse metabolic and mental phenotypes later <strong>in</strong>life <strong>in</strong>clud<strong>in</strong>g a higher body mass <strong>in</strong>dex, elevated plasma lipids, <strong>in</strong>creased risks of obesity,cardiovascular disease, and psychiatric disorders was found <strong>in</strong> a large number of epidemiologicalstudies. The bulk of these data were obta<strong>in</strong>ed <strong>in</strong> observational cohort studies of thelong-term health consequences of the prenatal exposure to the Dutch fam<strong>in</strong>e of 1944e45 andto the Ch<strong>in</strong>ese fam<strong>in</strong>e of 1959e61 [107e109]. These associations were dependent on thetim<strong>in</strong>g of the exposure dur<strong>in</strong>g gestation and lactation periods. Early gestation seems to be anespecially vulnerable period [110]. The mechanisms contribut<strong>in</strong>g to associations between theprenatal exposure to fam<strong>in</strong>e and adult health outcomes are still unknown but may <strong>in</strong>volve thepersistent epigenetic alterations [108]. In the Heijmans et al. [110] study, the level of methylationof the IGF-2 gene, a key factor <strong>in</strong> human growth and development, was estimated toexam<strong>in</strong>e whether the prenatal exposure to fam<strong>in</strong>e can lead to persistent changes <strong>in</strong> theepigenome. It has been found that <strong>in</strong>dividuals who were exposed to fam<strong>in</strong>e <strong>in</strong> early gestationhad a much lower level of methylation of IGF-2 than controls 60 years after the exposure [110].More recently, this observation was extended by the study a set of 15 additional candidate lociimplicated <strong>in</strong> growth, metabolic, and cardiovascular disorders [111]. Methylation of six ofthese loci has been shown to be associated with <strong>in</strong> utero exposure to fam<strong>in</strong>e. IGF-2 washypomethylated <strong>in</strong> <strong>in</strong>dividuals whose mothers were exposed to fam<strong>in</strong>e periconceptually,whereas <strong>in</strong>terleuk<strong>in</strong>-10, guan<strong>in</strong>e nucleotide-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>, lept<strong>in</strong>, ATP-b<strong>in</strong>d<strong>in</strong>g cassette A1,and maternally expressed 3(meg3) were hypermethylated. Taken together, these data <strong>in</strong>dicatethat differences <strong>in</strong> DNA methylation <strong>in</strong>duced by exposure to prenatal fam<strong>in</strong>e may persistdur<strong>in</strong>g the human life course. Moreover, Tobi et al. [112] <strong>in</strong>vestigated whether prenatal growthrestriction early <strong>in</strong> pregnancy can be associated with changes <strong>in</strong> DNA methylation at loci thatwere previously shown by them to be sensitive to early gestational fam<strong>in</strong>e exposure. Theycompared 38 <strong>in</strong>dividuals born preterm (


CHAPTER 27Early-Life Epigenetic Programm<strong>in</strong>g of <strong>Human</strong> <strong>Disease</strong> and Ag<strong>in</strong>gprocess is accompanied by alterations <strong>in</strong> DNA methylation, the dim<strong>in</strong>ished activity of DNAmethyltransferases (DNMTs) can be a potential mechanism for the decreased genomic DNAmethylation dur<strong>in</strong>g ag<strong>in</strong>g, along with reduced folate <strong>in</strong>take and altered folate metabolism [114].Current research suggests that compounds that serve as metabolic methyl group donors andcofactors (chol<strong>in</strong>e, beta<strong>in</strong>e, methion<strong>in</strong>e, folic acid, and vitam<strong>in</strong> B 12 ) play a key role among thenutrients that alter the epigenetic status dur<strong>in</strong>g the fetal development [115e118]. For example,Yajnik et al. [119] studied the association between maternal vitam<strong>in</strong> B 12 , folate, and totalhomocyste<strong>in</strong>e (tHcy) status dur<strong>in</strong>g pregnancy, and offspr<strong>in</strong>g adiposity and <strong>in</strong>sul<strong>in</strong> resistance at6 years <strong>in</strong> the Pune (India). They measured maternal nutritional <strong>in</strong>take and circulat<strong>in</strong>gconcentrations of folate, vitam<strong>in</strong> B 12 , tHcy, and methylmalonic acid at 18 and 28 weeks ofgestation. These parameters were correlated with offspr<strong>in</strong>g anthropometry, body composition,and <strong>in</strong>sul<strong>in</strong> resistance at 6 years. Although short and th<strong>in</strong>, the 6-year-old children were relativelyadipose compared with the UK standards. Higher maternal erythrocyte folate concentrationsat 28 weeks predicted higher offspr<strong>in</strong>g adiposity. Low maternal vitam<strong>in</strong> B12 predictedhigher risk of <strong>in</strong>sul<strong>in</strong> resistance <strong>in</strong> children. The offspr<strong>in</strong>g of mothers with a comb<strong>in</strong>ation ofhigh folate and low vitam<strong>in</strong> B12 concentrations were the most <strong>in</strong>sul<strong>in</strong> resistant. Consistentdata were obta<strong>in</strong>ed <strong>in</strong> the Krishnaveni et al. [120] study, where low plasma vitam<strong>in</strong> B 12concentrations comb<strong>in</strong>ed with high folate concentrations <strong>in</strong> pregnancy were associated witha higher <strong>in</strong>cidence of gestational diabetes and later diabetes <strong>in</strong> Mysore (India).The early-life dietary manipulation of methyl group donors (either deficiency or supplementation)can have a profound impact on the gene expression profile and, consequently, onthe homeostatic mechanisms that ensure the normal course of physiological processes [117].Recently, Steegers-Theunissen et al. [121] exam<strong>in</strong>ed whether periconceptional maternal folicacid use and markers of global DNA methylation potential (S-adenosylmethion<strong>in</strong>e andS-adenosylhomocyste<strong>in</strong>e blood levels) <strong>in</strong> mothers and children affect methylation of the IGF-2gene differentially methylated region (DMR) <strong>in</strong> the child. Children of mothers who used folicacid had a 4.5% higher methylation of the IGF-2 DMR than children who were not exposed tofolic acid. An <strong>in</strong>verse association between IGF-2 DMR methylation and birth weight wasobserved. These results <strong>in</strong>dicate plasticity of IGF-2 methylation by periconceptional folic aciduse. The authors concluded that periconceptional folic acid adm<strong>in</strong>istration is associated withepigenetic changes <strong>in</strong> IGF-2 <strong>in</strong> the child that may affect <strong>in</strong>trauter<strong>in</strong>e programm<strong>in</strong>g of growthand development with consequences for adult health and disease. Hoyo et al. [122] evaluatedexposure to maternal folic acid supplementation before and dur<strong>in</strong>g pregnancy <strong>in</strong> relation toaberrant DNA methylation at two DMRs regulat<strong>in</strong>g IGF-2 expression <strong>in</strong> <strong>in</strong>fants. Aberrantmethylation at these regions has been associated with IGF-2 deregulation and <strong>in</strong>creasedsusceptibility to several chronic diseases. Compared to <strong>in</strong>fants born to women report<strong>in</strong>g nofolic acid <strong>in</strong>take before or dur<strong>in</strong>g pregnancy, methylation levels at the H19 DMR decreasedwith <strong>in</strong>creas<strong>in</strong>g folic acid <strong>in</strong>take. Ba et al. [123] <strong>in</strong>vestigated the relationship between folate,vitam<strong>in</strong> B 12 , and methylation of the IGF-2 gene <strong>in</strong> maternal and cord blood. The methylationpatterns of IGF-2 <strong>in</strong> promoter 2 (P2) and promoter 3 (P3) <strong>in</strong> cord blood were not associatedwith serum folate levels <strong>in</strong> either cord or maternal blood, whereas the P3 methylation patternswere associated with serum levels of vitam<strong>in</strong> B 12 <strong>in</strong> mother’s blood. Methylation patterns <strong>in</strong> P2of maternal blood were associated with serum levels of vitam<strong>in</strong> B 12 <strong>in</strong> mother’s blood,exposure to passive smok<strong>in</strong>g, and mother’s weight ga<strong>in</strong> dur<strong>in</strong>g pregnancy.555Chol<strong>in</strong>e <strong>in</strong>take exceed<strong>in</strong>g current dietary recommendations has also been shown to be able topreserve markers of cellular methylation and attenuate DNA damage <strong>in</strong> a genetic subgroup offolate-compromised men [124], <strong>in</strong>dicat<strong>in</strong>g the epigenetic contribution to the l<strong>in</strong>k betweenchol<strong>in</strong>e and folate pathways. Although the molecular mechanisms by which methyl donorsalter the epigenetic profile are still unclear, it was commonly considered that the primarymechanisms could be S-adenosylmethion<strong>in</strong>e (SAM) availability. However, recent animalstudies have demonstrated that SAM concentrations <strong>in</strong> rat liver are very stable, <strong>in</strong>dicat<strong>in</strong>g that


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>gene-specific epigenetic mechanisms are <strong>in</strong>volved rather than simply SAM availability formethylation reactions [125,126].55627.12 LONG-TERM PROGRAMMING EFFECTSOF PRENATAL STRESSA large number of animal and epidemiological studies have revealed that prenatal stress as wellas excess exogenous glucocorticoids or <strong>in</strong>hibition of 11b-hydroxysteroid dehydrogenase type 2(the placental barrier to maternal glucocorticoids) are l<strong>in</strong>ked with adverse health outcomes<strong>in</strong>clud<strong>in</strong>g low birth weight, risk of cardio-metabolic disorders, neuroendocr<strong>in</strong>e dysfunction,and psychiatric diseases <strong>in</strong> later life [127e130], and with an <strong>in</strong>creased risk of <strong>in</strong>fectiousdiseases [131] <strong>in</strong> the offspr<strong>in</strong>g. Molecular mechanisms underly<strong>in</strong>g the programm<strong>in</strong>g effects ofearly life stress and glucocorticoids have been suggested to <strong>in</strong>clude epigenetic changes <strong>in</strong> targetgene promoters, notably affect<strong>in</strong>g tissue-specific expression of the <strong>in</strong>tracellular GR [132]. Thelong-term effects of maternal behavior on the stress responsiveness and behavior of theoffspr<strong>in</strong>g dur<strong>in</strong>g adulthood are well documented <strong>in</strong> animal models, and these experimentalf<strong>in</strong>d<strong>in</strong>gs have been extended to humans by identify<strong>in</strong>g an association between early-lifeadversity and epigenetic marks <strong>in</strong> adult life [133,134]. In humans, childhood abuse was foundto alter HPA stress responses and to <strong>in</strong>crease the risk of suicide. To test the hypothesis thatepigenetic differences <strong>in</strong> critical loci <strong>in</strong> the bra<strong>in</strong> are <strong>in</strong>volved <strong>in</strong> the pathophysiology ofsuicide, McGowan et al. [133] <strong>in</strong>vestigated the extent of DNA methylation <strong>in</strong> the promoter ofgenes that encoded rRNA genes <strong>in</strong> the bra<strong>in</strong>s of suicide victims. Suicide subjects were selectedfor a history of early childhood neglect/abuse, which is associated with decreased hippocampalvolume and cognitive impairments. rRNA was significantly hypermethylatedthroughout the promoter and 5 0 regulatory region <strong>in</strong> the bra<strong>in</strong> of suicide subjects, consistentwith reduced rRNA expression <strong>in</strong> the hippocampus. More recently, McGowan et al. [134]exam<strong>in</strong>ed epigenetic differences <strong>in</strong> a neuron-specific glucocorticoid receptor (NR3C1)promoter between postmortem hippocampus obta<strong>in</strong>ed from suicide victims, who were orwere not abused as children. In this study, expression of total glucocorticoid receptor mRNAwas significantly reduced <strong>in</strong> suicide victims with a history of childhood abuse relative to nonabusedsuicide victims or controls; there was no difference between non-abused suicidevictims and controls. There was also a significant effect on the expression of transcriptsconta<strong>in</strong><strong>in</strong>g the exon 1 F NR3C1 promoter. The glucocorticoid receptor 1 F expression wassignificantly lower <strong>in</strong> samples from suicide victims with a history of childhood abusecompared with suicide victims without childhood abuse or controls. Similar to the f<strong>in</strong>d<strong>in</strong>gswith total glucocorticoid receptor mRNA expression, there was no difference between nonabusedsuicide victims and controls subjects. These f<strong>in</strong>d<strong>in</strong>gs suggest a common effect ofparental care on the epigenetic regulation of hippocampal GR expression.It is known that cesarean section can cause more severe stress <strong>in</strong> newborn <strong>in</strong>fants comparedwith that of those born by vag<strong>in</strong>al delivery, who adapt to the new conditions better. Cesareansection is associated with significant risks for a baby’s short- and long-term physical andemotional health compared to vag<strong>in</strong>al birth, <strong>in</strong>clud<strong>in</strong>g diabetes, leukemia, respiratory problems,and asthma later <strong>in</strong> life [135]. To study whether the mode of delivery affects epigeneticactivity <strong>in</strong> newborn <strong>in</strong>fants, Schl<strong>in</strong>zig et al. [135] analyzed the level of DNA methylation <strong>in</strong>leukocytes of 21 newborn <strong>in</strong>fants delivered by cesarean section and 16 <strong>in</strong>fants born by vag<strong>in</strong>aldelivery. In this study, <strong>in</strong>fants born by cesarean section exhibited significantly higher DNAmethylation level <strong>in</strong> leukocytes compared with that of those born by vag<strong>in</strong>al delivery.The secretion of glucocorticoids is a classic endocr<strong>in</strong>e response to stress. The exposure to excessglucocorticoids <strong>in</strong> early life can permanently alter tissue glucocorticoid signal<strong>in</strong>g, and theseeffects may have short-term adaptive benefits but <strong>in</strong>crease the risk of later disease [129].Currently, multiple courses of synthetic glucocorticoids are recommended for various conditions.Prenatal exposure to glucocorticoids is very efficient <strong>in</strong> reduc<strong>in</strong>g the <strong>in</strong>cidence of


CHAPTER 27Early-Life Epigenetic Programm<strong>in</strong>g of <strong>Human</strong> <strong>Disease</strong> and Ag<strong>in</strong>grespiratory distress syndrome <strong>in</strong> preterm babies. However, despite the beneficial therapeuticeffect of antenatally adm<strong>in</strong>istered glucocorticoids, their prenatal adm<strong>in</strong>istration can result <strong>in</strong>transgenerational effects with respect to the risk of develop<strong>in</strong>g several metabolic and cardiovasculardisorders <strong>in</strong> later life which implies that these epigenetic effects can persist acrossgenerations [132,136].27.13 LONG-TERM IMPACTS OF MATERNAL SUBSTANCE USEDURING PREGNANCYIt is well known that heavy prenatal alcohol exposure can severely affect human physical andneurobehavioral development. Epidemiological data offer some evidence that paternalalcohol consumption can affect birth weight, congenital heart defects, and mild cognitiveimpairments [137e139]. The detrimental effects follow<strong>in</strong>g <strong>in</strong> utero alcohol exposure (fetalalcohol spectrum disorders, FASD) have been recognized for centuries, but only recently newdata were obta<strong>in</strong>ed provid<strong>in</strong>g a molecular <strong>in</strong>sight <strong>in</strong>to the mechanisms <strong>in</strong>volved <strong>in</strong> FASD.A substantial amount of data have been accumulated to support the role of environmentally<strong>in</strong>duced epigenetic remodel<strong>in</strong>g dur<strong>in</strong>g gametogenesis and after conception as a key mechanismfor the deleterious effects of prenatal alcohol exposure that persist <strong>in</strong>to adulthood [139].For example, the Ouko et al. [140] study has demonstrated a l<strong>in</strong>k between chronic alcohol use<strong>in</strong> men and hypomethylation of paternally impr<strong>in</strong>ted loci <strong>in</strong> sperm DNA <strong>in</strong> genomic regionscritical for embryonic development (H19 and IG-DMR) thus provid<strong>in</strong>g a mechanism forpaternal effects <strong>in</strong> the etiology of FASDs. There was a pattern of <strong>in</strong>creased demethylation withalcohol consumption at the two impr<strong>in</strong>ted loci with a significant difference observed at the IG-DMR between the non-dr<strong>in</strong>k<strong>in</strong>g and heavy-alcohol-consum<strong>in</strong>g groups. Greater <strong>in</strong>ter<strong>in</strong>dividualvariation <strong>in</strong> average methylation was observed at the H19 DMR and <strong>in</strong>dividual cloneswere more extensively demethylated than those of the IG-DMR. CpG site #4 <strong>in</strong> the IG-DMRwas preferentially demethylated among all <strong>in</strong>dividuals and, along with the H19 DMR CpG site#7 located with<strong>in</strong> the CTCF b<strong>in</strong>d<strong>in</strong>g site 6, showed significant demethylation <strong>in</strong> the alcoholconsum<strong>in</strong>ggroups compared with the control group. The authors hypothesized that, shouldthese epigenetic changes <strong>in</strong> impr<strong>in</strong>ted genes be transmitted through fertilization, they wouldalter the critical gene expression dosages required for normal prenatal development result<strong>in</strong>g<strong>in</strong> offspr<strong>in</strong>g with features of FASD.557Three developmental periods are particularly vulnerable: preconception, preimplantation, andgastrulation. These periods of teratogenesis correlate with peak periods of epigenetic reprogramm<strong>in</strong>gwhich, together with the evidence that ethanol <strong>in</strong>terferes with one-carbon metabolism,DNA methylation, histone modifications, and non-cod<strong>in</strong>g RNA, suggests an importantrole for epigenetic mechanisms <strong>in</strong> the etiology of FASDs [141]. Moreover, alcohol is known tolead to a variety of nutritional disturbances <strong>in</strong>clud<strong>in</strong>g nutrient <strong>in</strong>take, absorption, utilization,and excretion. Whereas some nutrients can essentially affect gene expression, the alcohol<strong>in</strong>ducednutrient disbalance may be a major contributor to impaired gene expression <strong>in</strong> FASD.Several metabolites, namely, acetate, S-adenosylmethion<strong>in</strong>e, nicot<strong>in</strong>amide aden<strong>in</strong>e d<strong>in</strong>ucleotide,and z<strong>in</strong>c are supposed to be especially relevant to alcohol metabolism and ALD [142].A wide range of fetal abnormalities and birth defects have been repeatedly reported <strong>in</strong> animalsand humans after preconceptional alcohol exposure. The mechanisms, particularly <strong>in</strong> males,are likely to <strong>in</strong>volve alcohol-<strong>in</strong>duced epigenetic changes <strong>in</strong> the gametes or, alternatively,selection effects with<strong>in</strong> the germl<strong>in</strong>e, result<strong>in</strong>g <strong>in</strong> the ontogenesis of “FASD-like” phenotypes <strong>in</strong>unexposed generations [137].Maternal smok<strong>in</strong>g can also result <strong>in</strong> <strong>in</strong>trauter<strong>in</strong>e growth restriction. Children born to motherswho smoke are at an <strong>in</strong>creased risk of obesity, hypertension, and diabetes [143,144]. Maternalsmok<strong>in</strong>g may be <strong>in</strong>volved <strong>in</strong> fetal programm<strong>in</strong>g [145], and <strong>in</strong> utero tobacco exposure wasshown to be associated with epigenetic changes <strong>in</strong> the offspr<strong>in</strong>g [146]. To demonstrate thatdifferences <strong>in</strong> DNA methylation patterns occur <strong>in</strong> children exposed to prenatal tobacco


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>smok<strong>in</strong>g and that variation <strong>in</strong> detoxification genes may alter these associations Breton et al.[147] measured methylation of DNA repetitive elements LINE1 and AluYb8 <strong>in</strong> buccal cells of348 children. DNA methylation patterns were associated with <strong>in</strong> utero exposure to maternalsmok<strong>in</strong>g. Exposed children had significantly lower methylation of AluYb8. Differences <strong>in</strong>smok<strong>in</strong>g-related effects on LINE1 methylation were observed <strong>in</strong> children with the commonGSTM1-null genotype. Differential methylation of CpG loci <strong>in</strong> eight genes was identifiedthrough the screen. Two genes, AXL and PTPRO, showed significant <strong>in</strong>creases <strong>in</strong> methylationand <strong>in</strong> exposed children. The authors concluded that life-long effects of <strong>in</strong> utero exposures maybe mediated through alterations <strong>in</strong> DNA methylation, and variants <strong>in</strong> detoxification genes maymodulate the effects of <strong>in</strong> utero exposure through epigenetic mechanisms. In a more recentstudy, Breton et al. [148] have shown that <strong>in</strong> utero tobacco smoke exposure negatively impactsrespiratory health <strong>in</strong> later life and some of the detrimental effects of this exposure are likely dueto epigenetic modifications. In a genome-wide survey of site-specific CPG methylation <strong>in</strong>asthmatics, they have demonstrated evidence of widespread epigenetic consequences of <strong>in</strong>utero smoke exposure, with differential methylation of more than 25% of all genes tested,<strong>in</strong>clud<strong>in</strong>g numerous cancer- and asthma-related genes.558It is known that prenatal exposure to maternal cigarette smok<strong>in</strong>g (PEMCS) is associated withvariations <strong>in</strong> bra<strong>in</strong> and behavior <strong>in</strong> adolescence. Toledo-Rodriguez et al. [149] have found thatepigenetic mechanisms may mediate some of the consequences of PEMCS through methylationof DNA <strong>in</strong> genes important for bra<strong>in</strong> development, such as the bra<strong>in</strong>-derived neurotrophicfactor (BDNF). They used bisulfite sequenc<strong>in</strong>g to assess DNA methylation of the BDNFpromoter <strong>in</strong> the blood of adolescents whose mothers smoked dur<strong>in</strong>g pregnancy. PEMCS wasassociated with higher rates of DNA methylation <strong>in</strong> the BDNF-6 exon. These results suggestthat PEMCS may lead to long-term down-regulation of BDNF expression via the <strong>in</strong>crease <strong>in</strong>DNA methylation <strong>in</strong> its promoter region. Such mechanisms could, <strong>in</strong> turn, lead to modifications<strong>in</strong> both development and plasticity of the bra<strong>in</strong> exposed <strong>in</strong> utero to maternal cigarettesmok<strong>in</strong>g.27.14 PROGRAMMING EFFECT OF EARLY-LIFE EXPOSURETO ENVIRONMENTAL TOXICANTSPrenatal and early postnatal exposure to environmental toxicantswas repeatedly found to beassociated with aberrant DNA methylation of regulatory sequences <strong>in</strong> susceptible genes,lead<strong>in</strong>g to <strong>in</strong>appropriate gene expression and disease pathogenesis <strong>in</strong> later life [150,151]. Theendocr<strong>in</strong>e-disrupt<strong>in</strong>g chemicals (EDCs) are of specific concern among the detrimental environmentalfactors because they are widespread <strong>in</strong> the environment. <strong>Human</strong> epidemiologicalstudies have shown that women exposed to EDCs such as agricultural pesticides haveprolonged/irregular estrous cycles and difficulty <strong>in</strong> achiev<strong>in</strong>g pregnancy, failed assistedreproductive technology attempts, and loss of pregnancies [152,153]. An exposure to EDCsdur<strong>in</strong>g early development could alter the epigenetic programm<strong>in</strong>g of the genome and therebyresult <strong>in</strong> adult-onset disease [154].Per<strong>in</strong>atal diethylstilbesterol (DES) exposure, which is associated with several reproductive tractabnormalities and <strong>in</strong>creased vag<strong>in</strong>al and cervical cancer risk <strong>in</strong> women, provides a clearexample of how estrogenic xenobiotic exposure dur<strong>in</strong>g a critical period of development canabnormally demethylate DNA sequences dur<strong>in</strong>g organ development and possibly <strong>in</strong>creasecancer risk later <strong>in</strong> life [150,155]. Importantly, these effects can be epigenetically transmitted tothe next generation [155,156]. Increas<strong>in</strong>g epidemiologic evidence l<strong>in</strong>ks specific pesticides,polychlor<strong>in</strong>ated biphenyls (PCBs), and <strong>in</strong>organic arsenic exposures to elevated prostate cancerrisk <strong>in</strong> adulthood [157]. Importantly, the prostate seems to be particularly sensitive to theseendocr<strong>in</strong>e disruptors dur<strong>in</strong>g the critical developmental w<strong>in</strong>dows <strong>in</strong>clud<strong>in</strong>g <strong>in</strong> utero andneonatal time po<strong>in</strong>ts as well as dur<strong>in</strong>g puberty.


CHAPTER 27Early-Life Epigenetic Programm<strong>in</strong>g of <strong>Human</strong> <strong>Disease</strong> and Ag<strong>in</strong>gIn utero and early postnatal exposure to bisphenol A (BPA), an estrogenic environmentaltox<strong>in</strong> widely used <strong>in</strong> the production of plastics, was shown to be able to produce a broadrange of adverse health effects, <strong>in</strong>clud<strong>in</strong>g impaired bra<strong>in</strong> development, sexual differentiation,behavior, and immune function, which could extend to future generations [158].Molecular mechanisms that underlie the longlast<strong>in</strong>g effects of BPA likely <strong>in</strong>volve disruptionof epigenetic programm<strong>in</strong>g of gene expression dur<strong>in</strong>g development. Low-dose exposures toBPA were shown to be able to affect the prostate epigenome dur<strong>in</strong>g development and, asa consequence, promote prostate disease with ag<strong>in</strong>g [151]. The alterations <strong>in</strong> DNA methylationpatterns <strong>in</strong> multiple cell signal<strong>in</strong>g genes <strong>in</strong> BPA-exposed prostates were obta<strong>in</strong>edsuggest<strong>in</strong>g that exposure to the environmentally relevant doses of BPA can be impr<strong>in</strong>ted <strong>in</strong>the develop<strong>in</strong>g prostate through epigenetic alterations [151,157]. Substantial evidence<strong>in</strong>dicates that exposure to BPA dur<strong>in</strong>g early development may also <strong>in</strong>crease breast cancerrisk later <strong>in</strong> life. Recently, Weng et al. [159] exam<strong>in</strong>ed epigenetic changes <strong>in</strong> breast epithelialcells treated with low-dose BPA. They identified 170 genes with similar expression changes<strong>in</strong> response to BPA. Functional analysis confirms that gene suppression was mediated <strong>in</strong>part through an estrogen receptor a (ERa)-dependent pathway. As a result of exposure toBPA or other estrogen-like chemicals, the expression of lysosomal-associated membraneprote<strong>in</strong> 3 (LAMP3) became epigenetically silenced <strong>in</strong> breast epithelial cells. Furthermore,<strong>in</strong>creased DNA methylation <strong>in</strong> the promoter CpG island regions of LAMP3 loci wasobserved <strong>in</strong> ERa-positive breast tumors.There is also conv<strong>in</strong>c<strong>in</strong>g evidence that prenatal environmental exposures can <strong>in</strong>fluence the riskfor subsequent asthma. Mart<strong>in</strong>o and Prescott [160] exam<strong>in</strong>ed the epigenetic regulation ofimmune development and the early immune profiles that contribute to allergic risk. Theyreviewed new evidence that key environmental exposures, such as microbial exposure, dietarychanges, tobacco smoke, and pollutants, can <strong>in</strong>duce epigenetic changes <strong>in</strong> gene expression andalter disease risk. In particular, transplacental exposure to high levels of airborne traffic-relatedpolycyclic aromatic hydrocarbons (PAHs) can cause aberrant DNA methylation changeslead<strong>in</strong>g to dysregulation of gene expression and childhood asthma [161]. In the Perera et al.study [162] it has been reported that methylation of ACSL3, a gene expressed <strong>in</strong> lung andthymus tissue, may be a possible biomarker l<strong>in</strong>k<strong>in</strong>g prenatal exposure to PAHs to childhoodasthma. In this study, methylation of the ACSL3 5 0 -CGI was found to be significantly associatedwith maternal airborne PAH exposure and with a parental report of asthma symptoms <strong>in</strong>children prior to age 5.55927.15 EPIGENETIC RISKS OF ASSISTED REPRODUCTIVETECHNOLOGIESAssisted reproductive technologies (ARTs) are methods of treat<strong>in</strong>g <strong>in</strong>fertility <strong>in</strong> whichpregnancy is achieved by artificial or partially artificial means. They generally <strong>in</strong>cludea stage of embryo culture that precisely co<strong>in</strong>cides with zygotic epigenetic resett<strong>in</strong>g. In vitrostudies on the human embryos have suggested that impr<strong>in</strong>ted genes <strong>in</strong> humans may besusceptible to ART conditions. Genomic impr<strong>in</strong>t<strong>in</strong>g is an epigenetic phenomenon bywhich certa<strong>in</strong> genes are expressed <strong>in</strong> a parent-of-orig<strong>in</strong>-dependent manner, i.e. primarily orexclusively from either the maternal or paternal allele [163]. Impr<strong>in</strong>ted expression isa clear example of epigenetic <strong>in</strong>heritance, because genetically identical sequences aredifferentially transcribed depend<strong>in</strong>g on the sex of the parent from which the gene orig<strong>in</strong>ates[164]. Most impr<strong>in</strong>ted genes conta<strong>in</strong> differentially methylated regions, where themethylation state of the parental alleles differs [165]. This variation allows for differentialregulation of these alleles dependent on parental orig<strong>in</strong> of the allele and leads to preferentialexpression of a specific allele, depend<strong>in</strong>g on its parental orig<strong>in</strong> [25]. The underly<strong>in</strong>gmechanisms by which culture media <strong>in</strong>duce abnormal epigenetic modifications are stillnot clear but it has been suggested that embryonic developmental tim<strong>in</strong>g can be disturbed


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>by the synthetic media and that this <strong>in</strong>terferes with epigenetic reprogramm<strong>in</strong>g and geneexpression [166].Several reports have raised concerns that children conceived by ART are at <strong>in</strong>creased risk ofhav<strong>in</strong>g impr<strong>in</strong>t<strong>in</strong>g disorders. Among them, BeckwitheWiedemann syndrome and Angelmansyndrome are the most extensively studied [167]. It was found the association of <strong>in</strong> vitrofertilization with BeckwitheWiedemann syndrome, and with epigenetic alterations of LIT1and H19 impr<strong>in</strong>ted genes [168] and abnormal impr<strong>in</strong>t<strong>in</strong>g of the KCNQ1OT gene [169,170].Intracytoplasmic sperm <strong>in</strong>jection was shown to <strong>in</strong>crease the risk of Angelman syndrome andsome impr<strong>in</strong>t<strong>in</strong>g defects [171,172]. Although these studies exam<strong>in</strong>ed only few cases, almostevery case showed loss of methylation at impr<strong>in</strong>t<strong>in</strong>g control regions rather than the geneticdefects generally responsible for these syndromes. Later, the conclusion that <strong>in</strong>fertility andovulation <strong>in</strong>duction are risk factors for ART-related Angelman syndrome was supported by thef<strong>in</strong>d<strong>in</strong>gs from the surveys conducted <strong>in</strong> Great Brita<strong>in</strong> [173] and Holland [174]. More recentstudies on global methylation changes <strong>in</strong> ART-conceived patients with other impr<strong>in</strong>t<strong>in</strong>gdisorders (<strong>in</strong>clud<strong>in</strong>g PradereWilli syndrome, SilvereRussell syndrome, transient neonataldiabetes mellitus, and maternal hypomethylation syndrome) reveal conflict<strong>in</strong>g results. Tierl<strong>in</strong>get al. [175] reported no association with ART and impr<strong>in</strong>t<strong>in</strong>g <strong>in</strong> a study of ten loci known to beimpr<strong>in</strong>ted, whereas Katari et al. [176] observed altered expression <strong>in</strong> several genes implicated<strong>in</strong> metabolic disorders, such as obesity and diabetes. Evidence from cl<strong>in</strong>ical reports suggeststhat the association between impr<strong>in</strong>t<strong>in</strong>g syndromes and ART may be restricted to syndromeswhere the impr<strong>in</strong>t<strong>in</strong>g change takes the form of hypomethylation on the maternal allele [177].Further studies are required to establish the level of risk of impr<strong>in</strong>t<strong>in</strong>g disorders <strong>in</strong> patientsconceived by ART.56027.16 CONCLUSIONS AND FUTURE DIRECTIONSThe relationship between adverse events dur<strong>in</strong>g prenatal and/or early postnatal life anddevelopment of chronic diseases later <strong>in</strong> life has been reported <strong>in</strong> a number of recent experimentaland epidemiological studies. In these studies, it has been highlighted as the key role ofepigenetic mechanisms <strong>in</strong> mediat<strong>in</strong>g the l<strong>in</strong>k between nutritional, hormonal, and metabolicenvironment early <strong>in</strong> life and lifelong health outcomes. Over recent years, there have beenconducted numerous animal studies and limited human studies aimed at understand<strong>in</strong>g thespecific epigenetic mechanisms underly<strong>in</strong>g developmental programm<strong>in</strong>g of later lifepathology and ag<strong>in</strong>g.<strong>Epigenetics</strong> has substantial potential for develop<strong>in</strong>g biological markers to predict whichexposures would put exposed subjects at risk and which <strong>in</strong>dividuals will be more susceptible todevelop disease. In human studies, this will require the use of highly sensitive laboratorymethods, so that epigenetic alterations can be detected well ahead of disease diagnosis [178].Given the reversibility of epigenetic modifications, the understand<strong>in</strong>g of epigenetic mechanismsmay represent a promis<strong>in</strong>g novel therapeutic target for prevention or reversion of humanage-related disorders and healthy life extension. These therapeutic strategies may <strong>in</strong>cludechanges <strong>in</strong> nutrition and lifestyle as well as pharmacological treatments. Several epigeneticdrugs, such as DNA demethylat<strong>in</strong>g agents and histone deacetylase <strong>in</strong>hibitors have already beensuccessfully tested <strong>in</strong> cl<strong>in</strong>ical trials [179]. However, all these drug candidates are very unspecificand, therefore, can cause large-scale epigenetic deregulation. In the future, it will be essential todevelop therapies that target only specific elements of the epigenome. Such preventiveapproaches <strong>in</strong>itiated <strong>in</strong> pre- and early postnatal periods of human development seem to beparticularly promis<strong>in</strong>g. If one could modify the <strong>in</strong>correct or deleterious epigenetic patternsthrough specific nutritional or pharmacological <strong>in</strong>terventions dur<strong>in</strong>g early ontogenesis, then itwould be possible to correct the disrupted gene expression programs to treat age-relateddiseases and to achieve better health and longevity.


CHAPTER 27Early-Life Epigenetic Programm<strong>in</strong>g of <strong>Human</strong> <strong>Disease</strong> and Ag<strong>in</strong>gReferences[1] Br<strong>in</strong>k TC, Regenbrecht C, Demetrius L, Lehrach H, Adjaye J. Activation of the immune response is a key featureof ag<strong>in</strong>g <strong>in</strong> mice. Biogerontology 2009;10:721e34.[2] Gonzalo S. Epigenetic alterations <strong>in</strong> ag<strong>in</strong>g. J Appl Physiol 2010;109:586e97.[3] Holliday R. The <strong>in</strong>heritance of epigenetic defects. <strong>Science</strong> 1987;238:163e70.[4] Fe<strong>in</strong>berg AP, Ohlsson R, Henikoff S. The epigenetic progenitor orig<strong>in</strong> of human cancer. Nat Rev Genet2006;7:21e33.[5] Calvanese V, Lara E, Kahn A, Fraga MF. The role of epigenetics <strong>in</strong> age<strong>in</strong>g and <strong>in</strong> age-related diseases. Age<strong>in</strong>g ResRev 2009;8:268e76.[6] Tremblay J, Hamet P. Impact of genetic and epigenetic factors from early life to later disease. Metab2008;57:S27e31.[7] Grav<strong>in</strong>a S, Vijg J. Epigenetic factors <strong>in</strong> ag<strong>in</strong>g and longevity. Pflugers Arch 2010;459:247e58.[8] Fraga MF, Ballestar E, Paz MF, Ropero S, Setien F, Ballestar ML, et al. Epigenetic differences arise dur<strong>in</strong>g thelifetime of monozygotic tw<strong>in</strong>s. Proc Natl Acad Sci USA 2005;102:10604e9.[9] Li CC, Maloney CA, Cropley JE, Suter CM. Epigenetic programm<strong>in</strong>g by maternal nutrition: shap<strong>in</strong>g futuregenerations. Epigenomics 2010;2:539e49.[10] Thompson RF, E<strong>in</strong>ste<strong>in</strong> FH. Epigenetic basis for fetal orig<strong>in</strong>s of age-related disease. J Women’s Health2010;19:581e7.[11] Zwaan BJ. L<strong>in</strong>k<strong>in</strong>g development and ag<strong>in</strong>g. Sci Ag<strong>in</strong>g Knowledge Environ 2003;47:32.[12] Gavrilov LA, Gavrilova NS. Early-life programm<strong>in</strong>g of ag<strong>in</strong>g and longevity: the idea of high <strong>in</strong>itial damage load(the HIDL hypothesis). Ann N Y Acad Sci 2004;1019:496e501.[13] Kajantie E. Early-life events. Effects on ag<strong>in</strong>g. Hormones 2008;7:101e13.[14] Chen JH, Cottrell EC, Ozanne SE. Early growth and age<strong>in</strong>g. Nestle Nutr Workshop Ser Pediatr Program2010;65:41e50.[15] Fernandez-Capetillo O. Intrauter<strong>in</strong>e programm<strong>in</strong>g of age<strong>in</strong>g. EMBO Rep 2010;11:32e6.[16] Mentis AF, Kararizou E. Does age<strong>in</strong>g orig<strong>in</strong>ate <strong>in</strong> utero? Biogerontology 2010;11:725e9.[17] Lucas A. Programm<strong>in</strong>g by early nutrition <strong>in</strong> man. Ciba Found Symp 1991;156:38e50.[18] Barker DJ. The developmental orig<strong>in</strong>s of adult disease. J Am Coll Nutr 2004;23:588Se95S.[19] McMillen IC, MacLaughl<strong>in</strong> SM, Muhlhausler BS, Gentili S, Duffield JL, Morrison JL. Developmental orig<strong>in</strong>s ofadult health and disease: the role of periconceptional and foetal nutrition. Basic Cl<strong>in</strong> Pharmacol Toxicol2008;102:82e9.[20] Bateson P, Barker D, Clutton-Brock T, Deb D, D’Ud<strong>in</strong>e B, Foley RA, et al. Developmental plasticity and humanhealth. Nature 2004;430:419e21.[21] Gluckman PD, Hanson MA, Mitchell MD. Developmental orig<strong>in</strong>s of health and disease: reduc<strong>in</strong>g the burdenof chronic disease <strong>in</strong> the next generation. Genome Med 2010;2:14.[22] Hanson M, Godfrey KM, Lillycrop KA, Burdge GC, Gluckman PD. Developmental plasticity and developmentalorig<strong>in</strong>s of non-communicable disease: Theoretical considerations and epigenetic mechanisms. ProgBiophys Mol Biol 2011;106:272e80.[23] Vaiserman A. Early-life orig<strong>in</strong> of adult disease: evidence from natural experiments. Exp Gerontol2011;46:189e92.[24] Vickaryous N, Whitelaw E. The role of the early embryonic environment on epigenotype and phenotype.Reprod Fertil Dev 2005;17:335e40.[25] Reik W, Dean W, Walter J. Epigenetic reprogramm<strong>in</strong>g <strong>in</strong> mammalian development. <strong>Science</strong>2001;293:1089e93.[26] Waterland RA, Carza C. Potential mechanisms of metabolic impr<strong>in</strong>t<strong>in</strong>g that lead to chronic disease. Am J Cl<strong>in</strong>Nutr 1999;69:179e97.[27] Fowden AL, Giussani DA, Forhead AJ. Intrauter<strong>in</strong>e programm<strong>in</strong>g of physiological systems: causes andconsequences. Physiology (Bethesda) 2006;21:29e37.[28] Hochberg Z, Feil R, Constancia M, Fraga M, Junien C, Carel JC, et al. Child health, developmental plasticity,and epigenetic programm<strong>in</strong>g. Endocr Rev 2011;32:159e224.[29] Wolfson M, Tacutu R, Budovsky A, Aizenberg N, Fraifeld VE. MicroRNAs: relevance to ag<strong>in</strong>g and age-associateddiseases. Open Longevity <strong>Science</strong> 2008;2:66e75.[30] Szyf M. The early life environment and the epigenome. Biochim Biophys Acta 2009;1790:878e85.[31] Gluckman PD, Hanson MA, Buklijas T, Low FM, Beedle AS. Epigenetic mechanisms that underp<strong>in</strong> metabolicand cardiovascular diseases. Nat Rev Endocr<strong>in</strong>ol 2009;5:401e8.561


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>562[32] Gluckman PD, Hanson MA, Low FM. The role of developmental plasticity and epigenetics <strong>in</strong> human health.Birth Defects Res C Embryo Today 2011;93:12e8.[33] Mukherjee S. Adult onset atherosclerosis may have its orig<strong>in</strong> <strong>in</strong> the foetal state <strong>in</strong> utero. Curr Op<strong>in</strong> Lipidol2009;20:155e6.[34] Lund G, Za<strong>in</strong>a S. Atherosclerosis: an epigenetic balanc<strong>in</strong>g act that goes wrong. Curr Atheroscler Rep2011;13:208e14.[35] Rodríguez-Rodero S, Fernández-Morera JL, Fernandez AF, Menéndez-Torre E, Fraga MF. Epigenetic regulationof ag<strong>in</strong>g. Discov Med 2010;10:225e33.[36] Fowden AL, Giussani DA, Forhead AJ. Endocr<strong>in</strong>e and metabolic programm<strong>in</strong>g dur<strong>in</strong>g <strong>in</strong>trauter<strong>in</strong>e development.Early Hum Dev 2005;81:723e34.[37] Cota BM, Allen PJ. The developmental orig<strong>in</strong>s of health and disease hypothesis. Pediatr Nurs 2010;36:157e67.[38] Varvarigou AA. Intrauter<strong>in</strong>e growth restriction as a potential risk factor for disease onset <strong>in</strong> adulthood. J PediatrEndocr<strong>in</strong>ol Metab 2010;23:215e24.[39] Cutfield WS, Hofman PL, Mitchell M, Morison IM. Could epigenetics play a role <strong>in</strong> the developmental orig<strong>in</strong>sof health and disease? Pediatr Res 2007;61:68Re75R.[40] Hall JG. Review and hypothesis: syndromes with severe <strong>in</strong>trauter<strong>in</strong>e growth restriction and very short statured are they related to the epigenetic mechanism(s) of fetal survival <strong>in</strong>volved <strong>in</strong> the developmental orig<strong>in</strong>s ofadult health and disease? Am J Med Genet A 2010;152A:512e27.[41] Wadhwa PD, Buss C, Entr<strong>in</strong>ger S, Swanson JM. Developmental orig<strong>in</strong>s of health and disease: brief history ofthe approach and current focus on epigenetic mechanisms. Sem<strong>in</strong> Reprod Med 2009;27:358e68.[42] Joss-Moore LA, Metcalfe DB, Albert<strong>in</strong>e KH, McKnight RA, Lane RH. <strong>Epigenetics</strong> and fetal adaptation toper<strong>in</strong>atal events: Diversity through fidelity. J Anim Sci 2010;88:E216e222.[43] Symonds ME, Sebert SP, Hyatt MA, Budge H. Nutritional programm<strong>in</strong>g of the metabolic syndrome. Nat RevEndocr<strong>in</strong>ol 2009;5:604e10.[44] Fowden AL, Forhead AJ. Endocr<strong>in</strong>e mechanisms of <strong>in</strong>trauter<strong>in</strong>e programm<strong>in</strong>g. Reproduction 2004;127:515e26.[45] Phillips DI. Fetal growth and programm<strong>in</strong>g of the hypothalamicepituitaryeadrenal axis. Cl<strong>in</strong> Exp PharmacolPhysiol 2001;28:967e70.[46] Lee MH, Jeon YJ, Lee SM, Park MH, Jung SC, Kim YJ. Placental gene expression is related to glucose metabolismand fetal cord blood levels of <strong>in</strong>sul<strong>in</strong> and <strong>in</strong>sul<strong>in</strong>-like growth factors <strong>in</strong> <strong>in</strong>trauter<strong>in</strong>e growth restriction.Early Hum Dev 2010;86:45e50.[47] McCarthy C, Cotter FE, McElwa<strong>in</strong>e S, Twomey A, Mooney EE, Ryan F, et al. Altered gene expression patterns <strong>in</strong><strong>in</strong>trauter<strong>in</strong>e growth restriction: potential role of hypoxia. Am J Obstet Gynecol 2007;196:70:e1e6.[48] Heazell AE, Sharp AN, Baker PN, Crocker IP. Intra-uter<strong>in</strong>e growth restriction is associated with <strong>in</strong>creasedapoptosis and altered expression of prote<strong>in</strong>s <strong>in</strong> the p53 pathway <strong>in</strong> villous trophoblast. Apoptosis2011;16:135e44.[49] E<strong>in</strong>ste<strong>in</strong> F, Thompson RF, Bhagat TD, Fazzari MJ, Verma A, Barzilai N, et al. Cytos<strong>in</strong>e methylation dysregulation<strong>in</strong> neonates follow<strong>in</strong>g <strong>in</strong>trauter<strong>in</strong>e growth restriction. PLoS One 2010;5:e8887.[50] Gemma C, Sookoian S, Alvariñas J, García SI, Qu<strong>in</strong>tana L, Kanevsky D, et al. Maternal pregestational BMI isassociated with methylation of the PPARGC1A promoter <strong>in</strong> newborns. Obesity 2009;17:1032e9.[51] Lillycrop KA, Burdge GC. Epigenetic changes <strong>in</strong> early life and future risk of obesity. Int J Obes (Lond)2011;35:72e83.[52] Godfrey KM, Barker DJ. Fetal nutrition and adult disease. Am J Cl<strong>in</strong> Nutr 2000;71:1344Se52S.[53] Harder T, Rodekamp E, Schellong K, Dudenhausen JW, Plagemann A. Birth weight and subsequent risk of type2 diabetes: a meta-analysis. Am J Epidemiol 2007;165:849e57.[54] Neitzke U, Harder T, Plagemann A. Intrauter<strong>in</strong>e growth restriction and developmental programm<strong>in</strong>g of themetabolic syndrome: a critical appraisal. Microcirculation 2011;18:304e11.[55] Ekbom A, Hsieh CC, Lipworth L, Wolk A, Pontén J, Adami HO, et al. Per<strong>in</strong>atal characteristics <strong>in</strong> relation to<strong>in</strong>cidence of and mortality from prostate cancers. BMJ 1996;313:337e41.[56] Vatten L. Can prenatal factors <strong>in</strong>fluence future breast cancer risk? Lancet 1996;348:1531.[57] Park SK, Kang D, McGlynn KA, Garcia-Closas M, Kim Y, Yoo KY, et al. Intrauter<strong>in</strong>e environments and breastcancer risk: meta-analysis and systematic review. Breast Cancer Res 2008;10. R8.[58] Trichopoulos D. Hypothesis: Does breast cancer orig<strong>in</strong>ate <strong>in</strong> utero? Lancet 1990;335. 9390-9340.[59] Oken E. Maternal and child obesity: the causal l<strong>in</strong>k. Obstet Gynecol Cl<strong>in</strong> North Am 2009;36:361e77.[60] Ornoy A. Prenatal orig<strong>in</strong> of obesity and their complications: Gestational diabetes, maternal overweight and theparadoxical effects of fetal growth restriction and macrosomia. Reprod Toxicol 2011;32:205e12.[61] Heerwagen MJ, Miller MR, Barbour LA, Friedman JE. Maternal obesity and fetal metabolic programm<strong>in</strong>g:a fertile epigenetic soil. Am J Physiol Regul Integr Comp Physiol 2010;299:R711e722.


CHAPTER 27Early-Life Epigenetic Programm<strong>in</strong>g of <strong>Human</strong> <strong>Disease</strong> and Ag<strong>in</strong>g[62] Grace CE, Kim SJ, Rogers JM. Maternal <strong>in</strong>fluences on epigenetic programm<strong>in</strong>g of the develop<strong>in</strong>ghypothalamic-pituitary-adrenal axis. Birth Defects Res A Cl<strong>in</strong> Mol Teratol 2011;91:797e805.[63] Ke X, Schober ME, McKnight RA, O’Grady S, Caprau D, Yu X, et al. Intrauter<strong>in</strong>e growth retardation affectsexpression and epigenetic characteristics of the rat hippocampal glucocorticoid receptor gene. PhysiolGenomics 2010;42:177e89.[64] Filiberto AC, Maccani MA, Koestler DC, Wilhelm-Benartzi C, Avissar-Whit<strong>in</strong>g M, Banister CE, et al. Birthweightis associated with DNA promoter methylation of the glucocorticoid receptor <strong>in</strong> human placenta. <strong>Epigenetics</strong>2011;6:566e72.[65] Holt RI. Fetal programm<strong>in</strong>g of the growth hormone-<strong>in</strong>sul<strong>in</strong>-like growth factor axis. Trends Endocr<strong>in</strong>ol Metab2002;13:392e7.[66] Verkauskiene R, Jaquet D, Deghmoun S, Chevenne D, Czernichow P, Lévy-Marchal C. Smallness for gestationalage is associated with persistent change <strong>in</strong> <strong>in</strong>sul<strong>in</strong>-like growth factor I (IGF-I) and the ratio of IGF-I/IGFb<strong>in</strong>d<strong>in</strong>gprote<strong>in</strong>-3 <strong>in</strong> adulthood. J Cl<strong>in</strong> Endocr<strong>in</strong>ol Metab 2005;10:5672e6.[67] Sandhu M. Insul<strong>in</strong>-like growth factor-I and risk of type 2 diabetes and coronary heart disease: molecularepidemiology. Endocr Dev 2005;9:44e54.[68] R<strong>in</strong>con M, Muzumdar R, Atzmon G, Barzilai N. The paradox of the <strong>in</strong>sul<strong>in</strong>/IGF-1 signal<strong>in</strong>g pathway <strong>in</strong>longevity. Mech Age<strong>in</strong>g Dev 2004;125:397e403.[69] R<strong>in</strong>con M, Rud<strong>in</strong> E, Barzilai N. The <strong>in</strong>sul<strong>in</strong>/IGF-1 signal<strong>in</strong>g <strong>in</strong> mammals and its relevance to human longevity.Exp Gerontol 2005;40:873e7.[70] Holly JMP, Gunnell DJ, Davey, Smith G. Growth hormone, IGF-I and cancer. Less <strong>in</strong>tervention to avoid cancer?More <strong>in</strong>tervention to prevent cancer? J Endocr<strong>in</strong>ol 1999;162:321e30.[71] Jenk<strong>in</strong>s PJ, Bust<strong>in</strong> SA. Evidence for a l<strong>in</strong>k between IGF-I and cancer. Eur J Endocr<strong>in</strong>ol 2004;151(Suppl 1):S17e22.[72] Moschos SJ, Mantzoros CS. The role of the IGF system <strong>in</strong> cancer: from basic to cl<strong>in</strong>ical studies and cl<strong>in</strong>icalapplications. Oncology 2002;63:317e32.[73] Renehan AG, Zwahlen M, M<strong>in</strong>der C, O’Dwyer ST, Shalet SM, Egger M. Insul<strong>in</strong>-like growth factor (IGF)-1,IGF b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>-3, and cancer risk: systematic review and meta-regression analysis. Lancet2004;363:1346e53.[74] Mart<strong>in</strong> RM, Holly JM, Middleton N, Davey, Smith G, Gunnell D. Childhood diet and <strong>in</strong>sul<strong>in</strong>-like growthfactors <strong>in</strong> adulthood: 65-year follow-up of the Boyd Orr Cohort. Eur J Cl<strong>in</strong> Nutr 2007;61:1281e92.[75] Shevah O, Laron Z. Patients with congenital deficiency of IGF-I seem protected from the development ofmalignancies: a prelim<strong>in</strong>ary report. Growth Horm IGF Res 2007;17:54e7.[76] Ong K, Kratzsch J, Kiess W, Costello M, Scott C, Dunger D. Size at birth and cord blood levels of <strong>in</strong>sul<strong>in</strong>,<strong>in</strong>sul<strong>in</strong>-like growth factor I (IGF-I), IGF-II, IGF-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>-1 (IGFBP-1), IGFBP-3, and the soluble IGF-II/mannose-6-phosphate receptor <strong>in</strong> term human <strong>in</strong>fants. The ALSPAC Study Team. Avon Longitud<strong>in</strong>al Study ofPregnancy and Childhood. J Cl<strong>in</strong> Endocr<strong>in</strong>ol Metab 2000;85:4266e9.[77] Sandhu MS, Heald AH, Gibson JM, Cruickshank JK, Dunger DB, Wareham NJ. Circulat<strong>in</strong>g concentrations of<strong>in</strong>sul<strong>in</strong>-like growth factor-I and development of glucose <strong>in</strong>tolerance: a prospective observational study. Lancet2002;359:1740e5.[78] Juul A, Scheike T, Davidsen M, Gyllenborg J, Jørgensen T. Low serum <strong>in</strong>sul<strong>in</strong>-like growth factor I is associatedwith <strong>in</strong>creased risk of ischemic heart disease: a population-based case-control study. Circulation2002;106:939e44.[79] Kalmijn S, Janssen JA, Pols HA, Lamberts SW, Breteler MM. A prospective study on circulat<strong>in</strong>g <strong>in</strong>sul<strong>in</strong>-likegrowth factor I (IGF-I), IGF-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s, and cognitive function <strong>in</strong> the elderly. J Cl<strong>in</strong> Endocr<strong>in</strong>ol Metab2000;85:4551e5.[80] Liu JM, Zhao HY, N<strong>in</strong>g G, Chen Y, Zhang LZ, Sun LH, et al. IGF-1 as an early marker for low bonemass or osteoporosis <strong>in</strong> premenopausal and postmenopausal women. J Bone M<strong>in</strong>er Metab 2008;26:159e64.[81] Ben-Shlomo Y, Holly J, McCarthy A, Savage P, Davies D, Davey Smith G. Prenatal and postnatal milksupplementation and adult <strong>in</strong>sul<strong>in</strong>-like growth factor I: long-term follow-up of a randomized controlled trial.Cancer Epidemiol Biomarkers Prev 2005;14:1336e9.[82] Mart<strong>in</strong> RM, Holly JM, Gunnell D. Milk and l<strong>in</strong>ear growth: programm<strong>in</strong>g of the IGF-I axis and implication forhealth <strong>in</strong> adulthood. Nestle Nutr Workshop Ser Pediatr Program 2011;67:79e97.[83] Tower RL, Spector LG. The epidemiology of childhood leukemia with a focus on birth weight and diet. Crit RevCl<strong>in</strong> Lab Sci 2007;44:203e42.[84] Dunger D, Yuen K, Ong K. Insul<strong>in</strong>-like growth factor I and impaired glucose tolerance. Horm Res2004;62:101e7.[85] López-Calderero I, Sánchez Chávez E, García-Carbonero R. The <strong>in</strong>sul<strong>in</strong>-like growth factor pathway as a targetfor cancer therapy. Cl<strong>in</strong> Transl Oncol 2010;12:326e38.563


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>564[86] Rosenzweig SA, Atreya HS. Def<strong>in</strong><strong>in</strong>g the pathway to <strong>in</strong>sul<strong>in</strong>-like growth factor system target<strong>in</strong>g <strong>in</strong> cancer.Biochem Pharmacol 2010;80:1115e24.[87] Gluckman PD, Hanson MA, Cooper C, Thornburg KL. Effect of <strong>in</strong> utero and early-life conditions on adulthealth and disease. N Engl J Med 2008;359:61e73.[88] Hoffman DJ. Early nutrition and adult health: Perspectives for <strong>in</strong>ternational and community nutritionprograms and policies. Nutr Res Pract 2010;4:449e54.[89] Campbell DM, Hall MH, Barker DJ, Cross J, Shiell AW, Godfrey KM. Diet <strong>in</strong> pregnancy and the offspr<strong>in</strong>g’sblood pressure 40 years later. Br J Obstet Gynaecol 1996;103:273e80.[90] Siemel<strong>in</strong>k M, Verhoef A, Dormans JA, Span PN, Piersma AH. Dietary fatty acid composition dur<strong>in</strong>g pregnancyand lactation <strong>in</strong> the rat programs growth and glucose metabolism <strong>in</strong> the offspr<strong>in</strong>g. Diabetologia2002;45:1397e403.[91] Godfrey K, Rob<strong>in</strong>son S, Barker DJP, Osmond C, Cox V. Maternal nutrition <strong>in</strong> early and late pregnancy <strong>in</strong>relation to placental and fetal growth. BMJ 1996;312:410e4.[92] Forsen T, Osmond C, Eriksson JG, Barker DJ. Growth of girls who later develop coronary heart disease. Heart2004;90:20e4.[93] Burdge GC, Slater-Jefferies J, Torrens C, Phillips ES, Hanson MA, Lillycrop KA. Dietary prote<strong>in</strong> restriction ofpregnant rats <strong>in</strong> the F0 generation <strong>in</strong>duces altered methylation of hepatic gene promoters <strong>in</strong> the adult maleoffspr<strong>in</strong>g <strong>in</strong> the F1 and F2 generations. Br J Nutr 2007;97:435e9.[94] Lillycrop KA, Hanson MA, Burdge GC. <strong>Epigenetics</strong> and the <strong>in</strong>fluence of maternal diet. In: Newnham JP,Ross MG, editors. Early Life Orig<strong>in</strong>s of <strong>Human</strong> Health and <strong>Disease</strong>. Basel: Karger; 2009. p. 11e20.[95] Godfrey KM, Sheppard A, Gluckman PD, Lillycrop KA, Burdge GC, McLean C, et al. Epigenetic gene promotermethylation at birth is associated with child’s later adiposity. Diabetes 2011;60:1528e34.[96] Niculescu MD, Lupu DS. Nutritional <strong>in</strong>fluence on epigenetics and effects on longevity. Curr Op<strong>in</strong> Cl<strong>in</strong> NutrMetab Care 2011;14:35e40.[97] Neel JV. Diabetes mellitus: a ‘‘thrifty’’ genotype rendered detrimental by ‘‘progress’’? Am J Hum Genet1962;14:353e62.[98] Hales CN, Barker DJ. Type 2 (non-<strong>in</strong>sul<strong>in</strong>-dependent) diabetes mellitus: the thrifty phenotype hypothesis.Diabetologia 1992;35:595e601.[99] Dulloo AG. Thrifty energy metabolism <strong>in</strong> catch-up growth trajectories to <strong>in</strong>sul<strong>in</strong> and lept<strong>in</strong> resistance. BestPract Res Cl<strong>in</strong> Endocr<strong>in</strong>ol Metab 2008;22:155e71.[100] P<strong>in</strong>ney SE, Simmons RA. Epigenetic mechanisms <strong>in</strong> the development of type 2 diabetes. Trends EndocrMetab 2010;21:223e9.[101] Stöger R. The thrifty epigenotype: an acquired and heritable predisposition for obesity and diabetes?Bioessays 2008;30:156e66.[102] Houseknecht KL, Spurlock ME. Lept<strong>in</strong> regulation of lipid homeostasis: dietary and metabolic implications.Nutr Res Rev 2003;16:83e96.[103] Djiane J, Attig L. Role of lept<strong>in</strong> dur<strong>in</strong>g per<strong>in</strong>atal metabolic programm<strong>in</strong>g and obesity. J Physiol Pharmacol2008;59:55e63.[104] Stöger R. In vivo methylation patterns of the Lept<strong>in</strong> promoter <strong>in</strong> human and mouse. <strong>Epigenetics</strong>2006;1:155e62.[105] Melzner I, Scott V, Dorsch K, Fischer P, Wabitsch M, Brüderle<strong>in</strong> S, et al. Lept<strong>in</strong> gene expression <strong>in</strong> humanpreadipocytes is switched on by maturation-<strong>in</strong>duced demethylation of dist<strong>in</strong>ct CpGs <strong>in</strong> its proximalpromoter. J Biol Chem 2002;277:45420e7.[106] Bouchard L, Thibault S, Guay SP, Santure M, Monpetit A, St-Pierre J, et al. Lept<strong>in</strong> gene epigenetic adaptationto impaired glucose metabolism dur<strong>in</strong>g pregnancy. Diabetes Care 2010;33:2436e41.[107] Pa<strong>in</strong>ter RC, Roseboom TJ, Bleker OP. Prenatal exposure to the Dutch fam<strong>in</strong>e and disease <strong>in</strong> later life: anoverview. Reprod Toxicol 2005;20:345e52.[108] Heijmans BT, Tobi EW, Lumey LH, Slagboom PE. The epigenome: archive of the prenatal environment.<strong>Epigenetics</strong> 2009;4:526e31.[109] Lumey LH, Ste<strong>in</strong> AD, Susser E. Prenatal fam<strong>in</strong>e and adult health. Annu Rev Public Health 2011;32:237e62.[110] Heijmans BT, Tobi EW, Ste<strong>in</strong> AD, Putter H, Blauw GJ, Susser ES, et al. Persistent epigenetic differencesassociated with prenatal exposure to fam<strong>in</strong>e <strong>in</strong> humans. Proc Natl Acad Sci USA 2008;105:17046e9.[111] Tobi EW, Lumey LH, Talens RP, Kremer D, Putter H, Ste<strong>in</strong> AD, et al. DNA methylation differences afterexposure to prenatal fam<strong>in</strong>e are common and tim<strong>in</strong>g- and sex-specific. Hum Mol Genet 2009;18:4046e53.[112] Tobi EW, Heijmans BT, Kremer D, Putter H, Delemarre-van de Waal HA, F<strong>in</strong>ken MJ, et al. DNA methylation ofIGF2, GNASAS, INSIGF and LEP and be<strong>in</strong>g born small for gestational age. <strong>Epigenetics</strong> 2011;6:171e6.[113] Hughes LA, van den Brandt PA, de Bruïne AP, Wouters KA, Hulsmans S, Spiertz A, et al. Early life exposure tofam<strong>in</strong>e and colorectal cancer risk: a role for epigenetic mechanisms. PLoS One 2009;4:e7951.


CHAPTER 27Early-Life Epigenetic Programm<strong>in</strong>g of <strong>Human</strong> <strong>Disease</strong> and Ag<strong>in</strong>g[114] Kim KC, Friso S, Choi SW. DNA methylation, an epigenetic mechanism connect<strong>in</strong>g folate to healthyembryonic development and ag<strong>in</strong>g. J Nutr Biochem 2009;20:917e26.[115] Zeisel SH. Gene response elements, genetic polymorphisms and epigenetics <strong>in</strong>fluence the human dietaryrequirement for chol<strong>in</strong>e. IUBMB Life 2007;59:380e7.[116] Zeisel SH. Is maternal diet supplementation beneficial? Optimal development of <strong>in</strong>fant depends on mother’sdiet. Am J Cl<strong>in</strong> Nutr 2009;89:685Se7S.[117] Zeisel SH. Nutritional genomics: def<strong>in</strong><strong>in</strong>g the dietary requirement and effects of chol<strong>in</strong>e. J Nutr2011;141:531e4.[118] Zeisel SH, da Costa KA. Chol<strong>in</strong>e: an essential nutrient for public health. Nutr Rev 2009;67:615e23.[119] Yajnik CS, Deshpande SS, Jackson AA, Refsum H, Rao S, Fisher DJ, et al. Vitam<strong>in</strong> B 12 and folate concentrationsdur<strong>in</strong>g pregnancy and <strong>in</strong>sul<strong>in</strong> resistance <strong>in</strong> the offspr<strong>in</strong>g: the Pune Maternal Nutrition Study.Diabetologia 2008;51:29e38.[120] Krishnaveni GV, Hill JC, Veena SR, Bhat DS, Wills AK, Karat CL, et al. Low plasma vitam<strong>in</strong> B12 <strong>in</strong> pregnancyis associated with gestational ’diabesity’ and later diabetes. Diabetologia 2009;52:2350e8.[121] Steegers-Theunissen RP, Obermann-Borst SA, Kremer D, L<strong>in</strong>demans J, Siebel C, Steegers EA, et al. Periconceptionalmaternal folic acid use of 400 microg per day is related to <strong>in</strong>creased methylation of the IGF2gene <strong>in</strong> the very young child. PLoS One 2009;4:e7845.[122] Hoyo C, Murtha AP, Schildkraut JM, Jirtle RL, Demark-Wahnefried W, Forman MR, et al. Methylation variationat IGF2 differentially methylated regions and maternal folic acid use before and dur<strong>in</strong>g pregnancy.<strong>Epigenetics</strong> 2011;6:928e36.[123] Ba Y, Yu H, Liu F, Geng X, Zhu C, Zhu Q, et al. Relationship of folate, vitam<strong>in</strong> B12 and methylation of<strong>in</strong>sul<strong>in</strong>-like growth factor-II <strong>in</strong> maternal and cord blood. Eur J Cl<strong>in</strong> Nutr 2011;65:480e5.[124] Sh<strong>in</strong> W, Yan J, Abratte CM, Vermeylen F, Caudill MA. Chol<strong>in</strong>e <strong>in</strong>take exceed<strong>in</strong>g current dietary recommendationspreserves markers of cellular methylation <strong>in</strong> a genetic subgroup of folate-compromised men. J Nutr2010;140:975e80.[125] M<strong>in</strong> H. Effects of dietary supplementation of high-dose folic acid on biomarkers of methylat<strong>in</strong>g reaction <strong>in</strong>vitam<strong>in</strong> B(12)-deficient rats. Nutr Res Pract 2009;3:122e7.[126] Engeham SF, Haase A, Langley-Evans SC. Supplementation of a maternal low-prote<strong>in</strong> diet <strong>in</strong> rat pregnancywith folic acid ameliorates programm<strong>in</strong>g effects upon feed<strong>in</strong>g behaviour <strong>in</strong> the absence of disturbances to themethion<strong>in</strong>e-homocyste<strong>in</strong>e cycle. Br J Nutr 2010;103:996e1007.[127] Kajantie E. Fetal orig<strong>in</strong>s of stress-related adult disease. Ann NY Acad Sci 2006;1083:11e27.[128] Beydoun H, Saftlas AF. Physical and mental health outcomes of prenatal maternal stress <strong>in</strong> human andanimal studies: a review of recent evidence. Paediatr Per<strong>in</strong>at Epidemiol 2008;22:438e66.[129] Cottrell EC, Seckl JR. Prenatal stress, glucocorticoids and the programm<strong>in</strong>g of adult disease. Front BehavNeurosci 2009;3:19.[130] Virk J, Li J, Vestergaard M, Obel C, Lu M, Olsen J. Early life disease programm<strong>in</strong>g dur<strong>in</strong>g the preconceptionand prenatal period: mak<strong>in</strong>g the l<strong>in</strong>k between stressful life events and type-1 diabetes. PLoS One2010;5: e11523.[131] Nielsen NM, Hansen AV, Simonsen J, Hviid A. Prenatal stress and risk of <strong>in</strong>fectious diseases <strong>in</strong> offspr<strong>in</strong>g. Am JEpidemiol 2011;173:990e7.[132] Harris A, Seckl J. Glucocorticoids, prenatal stress and the programm<strong>in</strong>g of disease. Horm Behav2011;59:279e89.[133] McGowan PO, Sasaki A, Huang TC, Unterberger A, Suderman M, Ernst C, et al. Promoter-wide hypermethylationof the ribosomal RNA gene promoter <strong>in</strong> the suicide bra<strong>in</strong>. PLoS One 2008;3:e2085.[134] McGowan PO, Sasaki A, D’Alessio AC, Dymov S, Labonté B, Szyf M, et al. Epigenetic regulation of theglucocorticoid receptor <strong>in</strong> human bra<strong>in</strong> associates with childhood abuse. Nat Neurosci 2009;12:342e8.[135] Schl<strong>in</strong>zig T, Johansson S, Gunnar A, Ekström TJ, Norman M. Epigenetic modulation at birth d altered DNAmethylation<strong>in</strong> white blood cells after Caesarean section. Acta Paediatr 2009;98:1096e9.[136] Seckl JR, Holmes MC. Mechanisms of disease: glucocorticoids, their placental metabolism and fetal‘programm<strong>in</strong>g’ of adult pathophysiology. Nat Cl<strong>in</strong> Pract Endocr<strong>in</strong>ol Metab 2007;3:479e88.[137] Abel E. Paternal contribution to fetal alcohol syndrome. Addict Biol 2004;9:127e33.[138] Riley EP, McGee CL. Fetal alcohol spectrum disorders: an overview with emphasis on changes <strong>in</strong> bra<strong>in</strong> andbehavior. Exp Biol Med (Maywood) 2005;230:357e65.[139] Ramsay M. Genetic and epigenetic <strong>in</strong>sights <strong>in</strong>to fetal alcohol spectrum disorders. Genome Med2010;2:27e34.[140] Ouko LA, Shantikumar K, Knezovich J, Haycock P, Schnugh DJ, Ramsay M. Effect of alcohol consumption onCpG methylation <strong>in</strong> the differentially methylated regions of H19 and IG-DMR <strong>in</strong> male gametes: implicationsfor fetal alcohol spectrum disorders. Alcohol Cl<strong>in</strong> Exp Res 2009;33:1615e27.565


<strong>Epigenetics</strong> <strong>in</strong> <strong>Human</strong> <strong>Disease</strong>566[141] Haycock PC. Fetal alcohol spectrum disorders: the epigenetic perspective. Biol Reprod 2009;81:607e17.[142] Moghe A, Joshi-Barve S, Ghare S, Gobejishvili L, Kirpich I, McCla<strong>in</strong> CJ, et al. Histone modifications andalcohol-<strong>in</strong>duced liver disease: Are altered nutrients the miss<strong>in</strong>g l<strong>in</strong>k? World J Gastroenterol2011;17:2465e72.[143] Montgomery SM, Ekbom A. Smok<strong>in</strong>g dur<strong>in</strong>g pregnancy and diabetesmellitus <strong>in</strong> a British longitud<strong>in</strong>al birthcohort. BMJ 2002;324:26e7.[144] Somm E, Schwitzgebel VM, Vauthay DM, Aubert ML, Hüppi PS. Prenatal nicot<strong>in</strong>e exposure and theprogramm<strong>in</strong>g of metabolic and cardiovascular disorders. Mol Cell Endocr<strong>in</strong>ol 2009;304:69e77.[145] Rogers JM. Tobacco and pregnancy: overview of exposures and effects. Birth Defects Res C Embryo Today2008;84:1e15.[146] Suter M, Abramovici A, Aagaard-Tillery K. Genetic and epigenetic <strong>in</strong>fluences associated with <strong>in</strong>trauter<strong>in</strong>egrowth restriction due to <strong>in</strong> utero tobacco exposure. Pediatr Endocr<strong>in</strong>ol Rev 2010;8:94e102.[147] Breton CV, Byun HM, Wenten M, Pan F, Yang A, Gilliland FD. Prenatal tobacco smoke exposure affects globaland gene-specific DNA methylation. Am J Respir Crit Care Med 2009;180:462e7.[148] Breton CV, Siegmund KD, Kong H, Qiu W, Islam KTS, Salam MT, et al. The epigenetic impact of <strong>in</strong> uterosmoke exposure <strong>in</strong> later life: a genome-wide survey of site-specific CPG methylation <strong>in</strong> asthmatics. Am JRespir Crit Care Med 2011;183:A1005.[149] Toledo-Rodriguez M, Lotfipour S, Leonard G, Perron M, Richer L, Veillette S, et al. T. Maternal smok<strong>in</strong>gdur<strong>in</strong>g pregnancy is associated with epigenetic modifications of the bra<strong>in</strong>-derived neurotrophic factor-6 exon<strong>in</strong> adolescent offspr<strong>in</strong>g. Am J Med Genet B Neuropsychiatr Genet 2010;153B:1350e4.[150] Li S, Hurst<strong>in</strong>g SD, Davis BJ, McLachlan JA, Barrett JC. Environmental exposure, DNA methylation, and generegulation: lessons from diethylstilbesterol-<strong>in</strong>duced cancers. Ann NY Acad Sci 2003;983:161e9.[151] Ho SM, Tang WY, Belmonte DF, Pr<strong>in</strong>s GS. Developmental exposure to estradiol and bisphenol A <strong>in</strong>creasessusceptibility to prostate carc<strong>in</strong>ogenesis and epigenetically regulates phosphodiesterase type 4 variant 4.Cancer Res 2006;66:5624e32.[152] Fuortes L, Clark MK, Kirchner HL, Smith EM. Association between female <strong>in</strong>fertility and agricultural workhistory. Am J Ind Med 1997;31:445e51.[153] Younglai EV, Holloway AC, Foster WG. Environmental and occupational factors affect<strong>in</strong>g fertility and IVFsuccess. Hum Reprod Update 2005;11:43e57.[154] Zama AM, Uzumcu M. Fetal and neonatal exposure to the endocr<strong>in</strong>e disruptor methoxychlor causesepigenetic alterations <strong>in</strong> adult ovarian genes. Endocr<strong>in</strong>ology 2009;150:4681e91.[155] Newbold RR. Lessons learned from per<strong>in</strong>atal exposure to diethylstilbestrol. Toxicol Appl Pharmacol2004;199:142e50.[156] Sk<strong>in</strong>ner MK. Environmental epigenetic transgenerational <strong>in</strong>heritance and somatic epigenetic mitotic stability.<strong>Epigenetics</strong> 2011;6:838e42.[157] Pr<strong>in</strong>s GS. Endocr<strong>in</strong>e disruptors and prostate cancer risk. Endocr Relat Cancer 2008;15:649e56.[158] Kundakovic M, Champagne FA. Epigenetic perspective on the developmental effects of bisphenol A. Bra<strong>in</strong>Behav Immun 2011;25:1084e93.[159] Weng YI, Hsu PY, Liyanarachchi S, Liu J, Deatherage DE, Huang YW, et al. Epigenetic <strong>in</strong>fluences of low-dosebisphenol A <strong>in</strong> primary human breast epithelial cells. Toxicol Appl Pharmacol 2010;248:111e21.[160] Mart<strong>in</strong>o D, Prescott S. <strong>Epigenetics</strong> and prenatal <strong>in</strong>fluences on asthma and allergic airways disease. Chest2011;139:640e7.[161] Miller RL, Ho SM. Environmental epigenetics and asthma: current concepts and call for studies. Am J RespirCrit Care Med 2008;177:567e73.[162] Perera F, Tang WY, Herbstman J, Tang D, Lev<strong>in</strong> L, Miller R, et al. Relation of DNA methylation of 5 0 -CPGisland of ACSL3 to transplacental exposure to airborne polycyclic aromatic hydrocarbons and childhoodasthma. PLoS One 2009;4:e4488.[163] Koerner MV, Barlow DP. Genomic impr<strong>in</strong>t<strong>in</strong>g d an epigenetic gene-regulatory model. Curr Op<strong>in</strong> Genet Dev2010;20:164e70.[164] Uribe-Lewis S, Woodf<strong>in</strong>e K, Stojic L, Murrell A. Molecular mechanisms of genomic impr<strong>in</strong>t<strong>in</strong>g and cl<strong>in</strong>icalimplications for cancer. Expert Rev Mol Med 2011;13:e2.[165] Neumann B, Kubicka P, Barlow DP. Characteristics of impr<strong>in</strong>ted genes. Nat Genet 1995;9:12e3.[166] De Rycke M, Liebaers I, Van Steirteghem A. Epigenetic risks related to assisted reproductive technologies.Hum Reprod 2002;17:2487e94.[167] Odom LN, Segars J. Impr<strong>in</strong>t<strong>in</strong>g disorders and assisted reproductive technology. Curr Op<strong>in</strong> Endocr<strong>in</strong>olDiabetes Obes 2010;17:517e22.[168] De Baun MR, Niemitz EL, Fe<strong>in</strong>berg AP. Association of <strong>in</strong> vitro fertilisation with BeckwitheWiedemannsyndrome and epigenetic alterations of LIT1 and H19. Am J Hum Genet 2003;72:156e60.


CHAPTER 27Early-Life Epigenetic Programm<strong>in</strong>g of <strong>Human</strong> <strong>Disease</strong> and Ag<strong>in</strong>g[169] Gicquel C, Gaston V, Mandelbaum J, Siffroi JP, Flahault A, Le Bouc Y. In vitro fertilisation may <strong>in</strong>crease therisk of BeckitheWiedemann Syndrome related to the abnormal impr<strong>in</strong>t<strong>in</strong>g of the KCNQ1OT gene. Am JHum Genet 2003;72:1338e41.[170] Lim D, Bowd<strong>in</strong> SC, Tee L, Kirby GA, Blair E, Fryer A, et al. Cl<strong>in</strong>ical and molecular genetic features ofBeckwitheWiedemann syndrome associated with assisted reproductive technologies. Hum Reprod2009;24:741e7.[171] Cox GF, Bürger J, Lip V, Mau UA, Sperl<strong>in</strong>g K, Wu BL, et al. Intracytoplasmic sperm <strong>in</strong>jection may <strong>in</strong>crease therisk of impr<strong>in</strong>t<strong>in</strong>g defects. Am J Hum Genet 2002;71:162e4.[172] Orstavik KH, Eiklid K, van der Hagen CB, Spetalen S, Kierulf K, Skjeldal O, et al. Another case of impr<strong>in</strong>t<strong>in</strong>gdefect <strong>in</strong> a girl with Angelman Syndrome who was conceived by <strong>in</strong>tracytoplasmic sperm <strong>in</strong>jection. Am J HumGenet 2003;72:218e9.[173] Sutcliffe AG, Peters CJ, Bowd<strong>in</strong> S, Temple K, Reardon W, Wilson L, et al. Assisted reproductive therapies andimpr<strong>in</strong>t<strong>in</strong>g disorders: a prelim<strong>in</strong>ary British survey. Hum Reprod 2006;21:1009e11.[174] Doornbos ME, Maas SM, McDonnell J, Vermeiden JP, Hennekam RC. Infertility, assisted reproductiontechnologies and impr<strong>in</strong>t<strong>in</strong>g disturbances: a Dutch study. Hum Reprod 2007;22:2476e80.[175] Tierl<strong>in</strong>g S, Souren NY, Gries J, Loporto C, Groth M, Lutsik P, et al. Assisted reproductive technologies do notenhance the variability of DNA methylation impr<strong>in</strong>ts <strong>in</strong> human. J Med Genet 2010;47:371e6.[176] Katari S, Turan N, Bibikova M, Er<strong>in</strong>le O, Chalian R, Foster M, et al. DNA methylation and gene expressiondifferences <strong>in</strong> children conceived <strong>in</strong> vitro or <strong>in</strong> vivo. Hum Mol Genet 2009;18:3769e78.[177] Amor DJ, Halliday J. A review of known impr<strong>in</strong>t<strong>in</strong>g syndromes and their association with assisted reproductiontechnologies. Hum Reprod 2008;23:2826e34.[178] Baccarelli A, Bollati V. <strong>Epigenetics</strong> and environmental chemicals. Curr Op<strong>in</strong> Pediatr 2009;21:243e51.[179] Mani S, Herceg Z. DNA demethylat<strong>in</strong>g agents and epigenetic therapy of cancer. Adv Genet 2010;70:327e40.567


This page <strong>in</strong>tentionally left blank


INDEXPage references followed by t, f, and b denote tables, figures, and boxes, respectively.AAcetylationhistone. See Histone acetylationnon-histone prote<strong>in</strong>s, 118e122, 388e389Adipocyte differentiation, 339e340, 347Adipocyte metabolismdiabetes, 337, 341obesity, 278e279, 282, 285Adiponect<strong>in</strong> (Adpn), 337Adult stem cells. See Stem cellsAdult T-cell leukemia/lymphoma (ATLL),426Age-specific epigenetic drift, 182, 281, 546Aggresomes, 345Ag<strong>in</strong>g, 6, 519e538environmental factors, 530e532,531f, 546epigenetic mechanisms, 281, 523e525,525fanimal models, 536e537complexity of, 528e529disease, 532e533early-life programm<strong>in</strong>g, 529,532e533, 545e560neurodegeneration, 526e528one-carbon metabolism, 533e536,534fhealthy versus pathological, 529e530life expectancy, 520e521premature, 494e495theories of, 521e523Ag<strong>in</strong>g syndromes, 522Agouti gene, 279f, 279e280, 308Air pollutants, 378e379Airway disease, 387e392epigenetic modifications, 388f, 388e390epigenetic therapies, 389e392, 391fAlcohol exposure, prenatal, 557Allele-specific expression (ASE), 276Allergic diseases, 4e5, 369e380developmental issues, 371e374, 374fenvironmental factors, 370e371,374e379, 375t, 559epidemiology of, 369e370epigenetic modifications, 374e379, 375tpathophysiology of, 370e371Alpha thalassemia/mental retardationsyndrome X-l<strong>in</strong>ked homologprote<strong>in</strong> (ATRX), 161e162,164e165ALZGene database, 176Alzheimer’s disease (AD), 127, 175e187ag<strong>in</strong>g process and, 520, 526, 528e529,536environmental factors, 176e177epigenetic modifications, 176e177DNA methylation, 177e183, 179f,180t, 186e187, 536e537histone modifications, 183e185, 184fone-carbon metabolism, 177f,177e183, 187RNA-mediated mechanisms, 185e186,186t, 186fstem cells, 492epigenetic therapies, 184e185, 187pathophysiology of, 175e176American Psychiatric Association, 132Amyloid precursor prote<strong>in</strong> (APP), 176,529, 536Anaplasma phagocytophilium, 418e419Angelman syndrome (AS), 168e169assisted reproductive technologies and,560epigenetic mechanisms, 195, 268, 307obesity <strong>in</strong>, 276tphenotypes, 267t, 276tstem cells and, 493e494Ankylos<strong>in</strong>g spondylitis (AS), 207t, 217Antagomirs, 240e243Antioxidants, 391Arg<strong>in</strong><strong>in</strong>e deim<strong>in</strong>ation, 58Arg<strong>in</strong><strong>in</strong>e methyltransferases. See HistonemethyltransferasesArgonaute (Ago) prote<strong>in</strong>s, 91Aspir<strong>in</strong>, 347Assisted reproductive technologies (ARTs),307, 559e560Asthma. See Airway disease; AllergicdiseasesAtherosclerosis, 401e402, 404e405, 406t.See also Cardiovascular diseaseAutism spectrum disorder (ASD), 127diagnosis of, 133e134environmental factors, 194e196epigenetic therapies, 198e199epigenomic programm<strong>in</strong>g, 131genetics, 136e138genomic <strong>in</strong>stability, 142impr<strong>in</strong>t<strong>in</strong>g disorders, 195e196, 196fmaternal duplications, 169pathogenesis of, 194, 194f, 199, 199fstem cell epigenetics, 492e493Autoimmune disorders, 3, 205e217.See also specific disorderepidemiology of, 225e226epigenetic biomarkers, 243, 246epigenetic deregulation <strong>in</strong>, 206e217,207tepigenetic therapies, 225e246antagomirs, 240e243conventional therapy balanced with,244e245DNA methylation, 236e237drug development, 226, 227f, 227bdrug targets, 229e230, 231b, 232ffuture research, 245e246HDAC <strong>in</strong>hibitors, 212e213,228e236, 348microRNA, 237e240, 241t, 245e246pathophysiologic basis, 226e229,229fside effects, 243e244, 244borgan-specific, 206, 206tstem cell epigenetics, 492tAutoimmune thyroid diseases (AITDs),207t, 215e216Azacytid<strong>in</strong>e, 208, 237, 392, 459, 490tBBAC array-based methylated CpG islandamplification (BAMCA), 38e39Bacterial <strong>in</strong>fection. See also Infectiousdiseases; specific diseaseepigenetic modifications, 417e421, 422timmune development and, 374e376Beckwith-Wiedemann syndrome (BWS),264e265assisted reproductive technologies and,560epigenetic mechanisms, 268, 278, 307phenotypes, 266tstem cells and, 494Bekhterev syndrome, 217Biological modulators, 230Biomarkers, 560autoimmune disorders, 243, 246cardiovascular disease, 408e409DNA methylation as, 40e41, 42fendometrial cancer, 477f, 477e478endometriosis, 42obesity, 312e314Bipolar disease (BD), 127diagnosis of, 132e133environmental factors, 138e139epigenetic modifications, 136e138,142e143, 157treatment of, 130e131Bisphenol A (BPA), 559Bisulfite and restriction analysis(COBRA), 10Bisulfite conversion, of unmethylatedcytos<strong>in</strong>es, 9Bisulfite treatment and PCR-based SSCP(BiPS), 9BIX-01294, 490tBlack carbon particles, 378e379B lymphocyte-<strong>in</strong>duced maturationprote<strong>in</strong>-1 (BLIMP-1), 346569


INDEX570Bone homeostasis, HDAC <strong>in</strong>hibitors <strong>in</strong>,233e234Bone morphogenetic prot<strong>in</strong>-7 (BMP7),514Bra<strong>in</strong>ag<strong>in</strong>g process <strong>in</strong>, 526e529, 535e536disorders of See Neurobehavioraldiseases; specific diseaseBroccoli, 376e377Butyrates, 231, 234e235, 347e348CCampylobacter rectus, 417e418Cancer, 2e3ag<strong>in</strong>g process and, 523e524, 533, 546developmental orig<strong>in</strong>s of, 551DNA methylation alterations <strong>in</strong>, 31e47,97, 111e112diagnosis, 40e41, 42fDNMTs, 33e35, 117e118endometrial cancer, 472e474, 474fhistone modifications, 36e37precancerous conditions, 31e33prognostic <strong>in</strong>dicators, 44e47, 45te46trisk estimation, 42e44signal pathways, 35e36subclassification profil<strong>in</strong>g, 37e40,39f, 40ftherapy, 47, 117e118DNA replication errors <strong>in</strong>, 15, 16fendometrial. See Endometrial cancerepigenetic therapies, 44e47acetylation of non-histone prote<strong>in</strong>s,118e122DNMT <strong>in</strong>hibitors, 117e118, 119te121tendometrial cancer, 476e478HDAC <strong>in</strong>hibitors, 113, 113t, 229histone acetylation, 112e113, 112f,114fhistone methylation, 114e117, 115tmicroRNA, 102histone modifications <strong>in</strong>, 66e69, 97,111e112acetylation, 70e71, 70f, 112e113, 112f,114fH3, 68e69H4, 66e68methylation, 70f, 71e73, 114e117,115tubiquit<strong>in</strong>ation, 70f, 73e74<strong>in</strong>fectious diseases and, 419e420,424e425, 428e429, 431, 432microRNA <strong>in</strong>, 92e93, 420cl<strong>in</strong>ical applications, 102e103deregulation mechanisms, 95e99oncomirs, 31, 93e95treatment resistance, 99e102stem cell epigenetics, 491, 492tCarc<strong>in</strong>ogenetic risk estimation, 42e44Cardiac hypertrophy, 396, 405e408, 406tCardiovascular disease (CVD), 5,395e410biomarkers, 408e409diabetes and, 401e402epigenetic modifications, 396e398, 406tatherosclerosis, 401e402, 404e405endothelial homeostasis, 398e401,399fheart failure, 405e408, 406tsmooth muscle homeostasis,401e404, 403f<strong>in</strong>ternet resources, 409tmicroRNA <strong>in</strong>, 408e409obesity and, 298Celiac disease, 207t, 217Cell death theory of ag<strong>in</strong>g, 523Cellular reprogramm<strong>in</strong>g, 487e491Cellular senescence, 522e523Chaperones, 343e346, 389e390Charcot-Marie-Tooth disease, 193Chemotherapyprognostic <strong>in</strong>dicators, 44e47treatment resistance, 99e100Chlamydophila spp., 421Chol<strong>in</strong>e, 555e556CHOP (C/EBP Homologous Prote<strong>in</strong>), 345ChromaSig, 21Chromat<strong>in</strong>, 254, 508alterationsag<strong>in</strong>g, 523autoimmune disorders, 228diabetes, 334e339<strong>in</strong>fectious disease, 427e428biological role of, 53e54epigenomic modulation of, 30, 129e132,129forganization of, 54e55, 65, 155e156,157fstem cells <strong>in</strong>, 481e482, 485e487remodel<strong>in</strong>g, dur<strong>in</strong>g cellularreprogramm<strong>in</strong>g, 487e491structure, 183, 254t, 254e255Chromat<strong>in</strong> immunoprecipitationcomb<strong>in</strong>ed with Mpss (ChIPsequenc<strong>in</strong>g), 19e21, 243Chromat<strong>in</strong> loop<strong>in</strong>g, 262Chromat<strong>in</strong>ome, 288Chromat<strong>in</strong> signatures, 20e21Chromosomal aberrationsimpr<strong>in</strong>t<strong>in</strong>g disorders, 265e268microRNA <strong>in</strong>, 95e96Chromosomal band structures, 15e19,18t, 18fChronic obstructive pulmonary disease(COPD). See Airway diseaseCis mechanism, 63e64, 258Clon<strong>in</strong>g of microRNA, 13Clozap<strong>in</strong>e, 145, 198Cluster<strong>in</strong>, 184e185Coca<strong>in</strong>e, 145, 196Co-factors, 74, 555Coff<strong>in</strong>-Lowry syndrome, 165e166Cohes<strong>in</strong>, 164e165Colony Stimulat<strong>in</strong>g Factor 1 (CSF-1),233e234Common fragile sites (CFSs), 17Comparative genomic hybridization(CGH), 9, 39, 40fComputational tools, 11, 21Conflict theory of impr<strong>in</strong>t<strong>in</strong>g, 258,276e277Congenital neurobiological disorders,195e196Copy number abnormalities, 264Cornelia de Lange syndrome (CdLS),162e164Corticosteroids, 389e391, 391fCortisone, 226CpG methylation patterns. See MethylCpG-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>CREB b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> (CBP), 165, 388,396, 404Crohn’s disease (CD)epigenetic deregulation <strong>in</strong>, 207t,216e217treatment of, 226, 234e235Cross-talkbetween epigenetic systems, 257histone, 61, 65, 75Curcum<strong>in</strong>, 351, 391Cytok<strong>in</strong>e profile, 230Cytomegalovirus, 425e426Cytos<strong>in</strong>es, unmethylated, bisulfiteconversion of, 9DDacogen, 117Darw<strong>in</strong>ian <strong>in</strong>heritance, 197Decitab<strong>in</strong>e, 459DeCODE Genetics, 287Demethylation agents (DMAs). See alsoEpigenetic therapies; specificagentendometrial cancer, 476e478endometriosis, 448, 458e459Dermatomyositis, 240Deubiquit<strong>in</strong>at<strong>in</strong>g enzyme (Dubs), 62t, 63<strong>in</strong> cancer, 70f, 73e74Development. See also Maternal factorsag<strong>in</strong>g process and, 529, 532e533,545e560allergic diseases and, 371diabetes and, 324e326, 324f, 340e341,347e348DNA methylation dur<strong>in</strong>g, 255e257,256f, 281e282, 554e556endocr<strong>in</strong>e programm<strong>in</strong>g dur<strong>in</strong>g,549e550environmental toxicants dur<strong>in</strong>g,558e559GH/IGF axis dur<strong>in</strong>g, 550e551HPA axis dur<strong>in</strong>g, 550methyl donors dur<strong>in</strong>g, 554e556neuronal. See Neurodevelopmentaldisorders; specific disorderobesity and, 281e283, 298e307prenatal stress, 556e557stem cells <strong>in</strong>. See Stem cellsDevelopmental Orig<strong>in</strong>s of Health and<strong>Disease</strong> (DOHaD) hypothesis,371, 546e548“Developmental programm<strong>in</strong>g” concept,546Diabetes, 4, 321e326cardiovascular complications, 401e402classification of, 321diagnosis of, 321


INDEXenvironmental factors, 326, 553epidemiology of, 321, 332epigenetic modifications, 322,332e334, 333fchromat<strong>in</strong> regulation, 334e339DNA methylation, 332e333, 334<strong>in</strong>sul<strong>in</strong> resistance, 325e326, 342<strong>in</strong>sul<strong>in</strong> secretion, 322e324microRNA, 339e341prote<strong>in</strong> function, 334epigenetic therapies, 346e353microRNA-based, 352e353nutrition-based, 350e352gestational, 284e285juvenile (type 1), 207t, 217,321pathogenesis of, 342e346, 345bDiabetogenes, 334Diagnosis. See also Biomarkersautoimmune disorders, 243, 246cancer, 40e41, 42f, 102diabetes, 321endometrial cancer, 475e476endometriosis, 444, 460neurobehavioral diseases, 132e136, 142Diagnostic and Statistical Manual of MentalDisorders (DSM), 132Diesel exhaust particles (DEP), 378Diet. See NutritionDiethylstilbestrol (DES), 447, 454, 558Differentially methylated regions (DMRs),258, 261e263, 261f<strong>Disease</strong>-modify<strong>in</strong>g antirheumatic drugs(DMARDs), 226, 233, 242tDNA demethylation, 30, 130, 255e256DNA methylation, 508e509alterationsag<strong>in</strong>g, 281, 523e529, 525f,536e537, 546Alzheimer’s disease, 177e183, 179f,180t, 186e187, 536e537cancer. See Cancercardiovascular disease, 396, 398,400e405diabetes, 322e324, 332e333, 334endometriosis, 446e449, 450t<strong>in</strong>fectious disease, 31e33, 428e430rheumatoid arthritis, 210e212, 211fsmok<strong>in</strong>g, 377systemic lupus erythematosus,208e210, 209f, 228, 236e237analysis methods, 8e11, 17tbiological roles of, 29e30, 129e130, 255cross-talk with other systems, 257dur<strong>in</strong>g development, 255e257,281e282, 554e556histone modifications and, 36e37impr<strong>in</strong>ted genes, 154, 195, 260obesity, 275, 279e280, 306processes, 154e155, 155f, 156fstem cells and, 483transcriptional silenc<strong>in</strong>g by, 255DNA methyltransferases (DNMTs), 30,111e112, 129, 154, 508ag<strong>in</strong>g, 524, 526, 555Alzheimer’s disease, 179e180, 179f,186e187autoimmune disorders, 211e212,236e237cancer, 33e35, 117e118endometrial, 472e473cardiovascular disease, 396, 398, 401dur<strong>in</strong>g development, 255e257, 256fdiabetes, 339e340endometriosis, 448neurodevelopmental disorders, 157e158obesity, 306stem cells and, 483, 485DNA methyltransferase <strong>in</strong>hibitors(DNMTi), 227b, 234bautoimmune disorders, 236e237cancer, 117e118, 119te121tdrug <strong>in</strong>teractions, 244e245side effects, 243e244, 244bDNA replication tim<strong>in</strong>g, 14e15, 16f, 17tearly/late-switch regions, 17e18, 18fDNA tissue samples, 8Dopam<strong>in</strong>e metabolism, 145Drug-<strong>in</strong>duced lupus, 236e237Drugs. See also specific drugautoimmune disorders, 226epigenetic. See Epigenetic therapiesmaternal use of, 557e558neurobehavioral diseases, 139, 145e146neurodevelopmental disorders, 196Dutch Hunger W<strong>in</strong>ter, 282e283, 298,302, 326, 554EEarly-life programm<strong>in</strong>g. See DevelopmentE-cadher<strong>in</strong>, 419, 448E2 conjugat<strong>in</strong>g enzymes, 62t, 63Embryogenesis. See DevelopmentEmbryonic stem cells (ESCs). See Stem cellsENCODE (Encyclopedia of DNAElements), 509e510Endocr<strong>in</strong>e-disrupt<strong>in</strong>g chemicals (EDCs),558Endocr<strong>in</strong>e programm<strong>in</strong>g, 549e551Endometrial cancer, 5, 471e478carc<strong>in</strong>ogenesis <strong>in</strong>, 471, 471fdiagnosis, 475e476epidemiology of, 471e472epigenetic modificationsDNA methylation, 472e474, 474fmicroRNA methylation, 474e475epigenetic therapy, 476e478Endometriosis, 5, 443e461def<strong>in</strong>ition of, 443diagnosis and classification of, 444, 460epidemiology of, 444epigenetic modifications, 446, 450tcause versus consequence, 454DNA methylation, 446e449histone modifications, 446, 449e453future research, 460e461review methods, 445e446treatment of, 444e445, 455e459Endophenotypes, 133, 134bEndoplasmic reticulum (ER) stress,343e346Endothelial cells (ECs), 396homeostasis, 398e401, 399f, 406tEnsembl database, 509Environmental factors, 520. See also specificfactorag<strong>in</strong>g process, 530e532, 531f, 546allergic diseases, 370e371, 374e379,375t, 559Alzheimer’s disease, 176e177diabetes, 326, 553endometriosis, 454<strong>in</strong>trauter<strong>in</strong>e. See Development; Maternalnutritionneurobehavioral diseases, 138e139neurobiological disorders, 194e196,197f, 199e200obesity, 273e274, 310Epialleles, 286e287Epigenetic code, 75Epigenetic drift, 182, 281, 546Epigenetic drugs. See Epigenetic therapies;specific drugEpigenetic field for cancerization, 476Epigenetic <strong>in</strong>heritance, transgenerational,197e198, 309e310Epigenetic landscape model(Wadd<strong>in</strong>gton), 128, 128f,488e489, 519e520Epigenetic modificationschromat<strong>in</strong> structure, 254e255, 254tDNA methylation, 508e509. See alsoDNA methylationhistone modifications, 508e509. See alsoHistone modificationsmicroRNA, 97e98. See also MicroRNAreversibility of, 47, 75, 199e200, 200f,228, 514, 560types of, 254e257, 520“Epigenetic programm<strong>in</strong>g” hypothesis,552<strong>Epigenetics</strong>overview of, 1e6, 508e510def<strong>in</strong>ition of, 128, 193e194, 206, 254,482, 508, 519e520variation methods, 2, 7e22. See alsospecific methodEpigenetic therapies, 75, 560airway disease, 389e392, 391fAlzheimer’s disease, 184e185, 187autoimmune disorders. See Autoimmunedisordersbalanc<strong>in</strong>g conventional therapy with,244e245bipolar disease, 130e131cancer. See Cancercellular reprogramm<strong>in</strong>g, 490, 490tdiabetes, 346e353drug categories, 226, 227b. See also specificdrugendometriosis, 455e459future research, 245neurobehavioral diseases, 145e146neurobiological disorders, 198e199obesity. See Obesityside effects, 243e244, 244b,458e459Epigenome, 128e129reference, 47571


INDEX572Epigenome-wide association studies(EWASs), 19obesity, 284, 288e289, 310Epigenomic programm<strong>in</strong>g, 131e132Epigenomicschromat<strong>in</strong> modulation, 129e132, 129fdef<strong>in</strong>ition of, 128e129gender differences, 131, 195Epi-microRNAs, 334, 339e341, 475Epimutations, 170, 264, 513e514,545e546Epistasis, 137EpiTYPER, 10Epste<strong>in</strong>-Barr virus (EBV), 422e424, 425tERb promoter, 448Error-catastrophe theory of ag<strong>in</strong>g, 421e422ES cell models, 158Escherichia coli, 421Estrogen receptor (ER) gene, 524Euchromat<strong>in</strong>, 183, 254Eukaryotic l<strong>in</strong>ear motifs (ELMs), 12Evolutionag<strong>in</strong>g theory, 521genomic impr<strong>in</strong>t<strong>in</strong>g, 258parental conflict theory, 258, 276e277Exercise, ag<strong>in</strong>g and, 532FFam<strong>in</strong>es, 282e283, 298, 302, 326, 554Fat, dietarydiabetes and, 336maternal, 304e305Fetal alcohol spectrum disorders (FASD),557Fetal growth retardation. See Intrauter<strong>in</strong>egrowth restrictionFetal macrosomia, 548e549, 550fFetal plasticity, 282e283, 305e306, 546Fetal stem cells. See Stem cellsFibroblast Growth Factor-4 (FGF4), 506,512Field effect, 34, 43e44Fluorescence correlation spectroscopy,243Fluorescence <strong>in</strong> situ hybridization (FISH),14Fluorescent recovery after photobleach<strong>in</strong>g(FRAP), 486Fluoxet<strong>in</strong>e, 145FMR1 prote<strong>in</strong> (FMRP), 167, 170Folic acidallergic diseases and, 376diabetes and, 326metabolic pathwayag<strong>in</strong>g process, 533e536, 534f,554e555Alzheimer’s disease, 177e183, 177f,187FMTD hub, 143e145, 144ftherapeutic use of, 198e199, 311Fragile sites, 141Fragile X-associated primary ovarian<strong>in</strong>sufficiency (FXPOI), 166e167Fragile X-associated tremor/ataxiasyndrome (FXTAS), 166e167Fragile X (FRAX) syndrome, 127, 141,166e167Frailty, 532e533GGammaherpesvirus, 422e425, 425tGarlic, 376e377GATA-4, 406e407Gene mutations. See MutationsGene silenc<strong>in</strong>g, 90f, 91e92autoimmune disorders, 237by DNA methylation, 255endometrial cancer, 473<strong>in</strong>fectious disease, 420, 426e428, 431neurodevelopmental disorders, 167obesity, 306Genetic theories of ag<strong>in</strong>g, 522Genome-wide analysis methods, 15,17t, 19e21Genome-wide association studies(GWASs), 19ag<strong>in</strong>g, 524, 528, 548diabetes, 323e324, 334endometrial cancer, 476neurological diseases, 136, 176, 199obesity, 275, 287e288, 303, 310Genome-wide demethylation, 255e256Genomic impr<strong>in</strong>t<strong>in</strong>g. See Impr<strong>in</strong>ted genesGenomic <strong>in</strong>stability, 14e15, 16f, 18neurobehavioral diseases, 139e142obesity, 311Gestational diabetes, 284e285G<strong>in</strong>gival epithelial cells (GECs), 417Global chromat<strong>in</strong> dynamism, 485Glucocorticoid receptor (GR)airway disease, 389f, 389e390allergic disease, 373autoimmune disorders, 235e236, 244diabetes, 346<strong>in</strong>trauter<strong>in</strong>e development, 550, 556e557Glucocorticoid response elements (GREs),235Glucose-dependent <strong>in</strong>sul<strong>in</strong>otropicpolypeptide receptor (GIPR),338Glucose Regulated Prote<strong>in</strong> 78 (GRP78 orBiP), 346Glut4, 338e340, 339fGluten-sensitive enteropathy(celiac disease), 207t, 217Glyc<strong>in</strong>e, 311Grave’s disease (GD), 215e216Growth factors, 506e508Growth hormone/<strong>in</strong>sul<strong>in</strong>-like growthfactor (GH/IGF) axis, 550e551Growth-regulatory region (GRR)11p15,260e261, 261f, 263, 278HHaloperidol, 145Hashimoto’s thyroiditis (HT), 215e216H19 DMR, 261f, 261e262Heart failure, 405e408, 406t. See alsoCardiovascular diseaseHeat shock prote<strong>in</strong>s (HSP), 343e344, 389Helicobacter pylori, 419e421Hepatitis B virus (HBV), 428e429, 430tHepatitis C virus (HCV), 429e430, 430tHepatocyte nuclear factor 4 alpha(HNF4a), 338Hereditary sensory neuropathy withdementia and hear<strong>in</strong>g loss(HSAN1), 158e159Herpesviruses, 422e425, 425tHeterochromat<strong>in</strong>, 183, 255, 482Hidden Markov model (HMM), 21Highly active antiretroviral therapy(HAART), 426e427Highly methylated doma<strong>in</strong>s (HMDs), 131Histones, 54, 111, 130, 256cross-talk, 61, 65, 75mark<strong>in</strong>g, 60, 450, 487Histone acetylation, 55e57, 56tag<strong>in</strong>g, 523, 528airway disease, 388f, 388e390Alzheimer’s disease, 183e185, 184fcancer, 70f, 70e71, 112f, 112e113, 114fcardiovascular disease, 396e405, 399fdur<strong>in</strong>g development, 256e257diabetes, 336e337Histone acetyltransferases (HATs), 55e57,56t, 155airway disease, 388, 388fallergic diseases, 372, 377Alzheimer’s disease, 183autoimmune disorders, 228cancer, 70f, 70e71, 112f, 112e113cardiovascular disease, 396, 405e407diabetes, 333e336, 335t, 342e345endometriosis, 450e451“Histone bivalent” model, 486e487Histone code, 333, 387, 482Histone deacetylases (HDACs), 55e57,56t, 155ag<strong>in</strong>g, 528airway disease, 388f, 388e390, 391fallergic diseases, 372, 377Alzheimer’s disease, 183e185autoimmune disorders, 210, 212e213,228e229cancer, 70f, 70e71, 112f, 113cardiovascular disease, 396e408, 397tdiabetes, 333e336, 335t, 340, 342e346,345bendometriosis, 451<strong>in</strong>fectious disease, 425e426obesity, 306Histone deacetylases (HDACs) <strong>in</strong>hibitors,227b. See also specific drug orcompoundairway disease, 391Alzheimer’s disease, 184e185, 187autoimmune disorders, 212e213,228e236anti-<strong>in</strong>flammatory effects, 235e236bone homeostasis, 233e234diabetes, 348<strong>in</strong>flammatory bowel disease, 234e235multiple sclerosis, 235rheumatoid arthritis, 233, 234bcancer, 113, 113t, 229


INDEXcardiovascular disease, 397e400,399f, 405cellular reprogramm<strong>in</strong>g, 490, 490tdiabetes, 336e339, 339f, 341e343,343f, 346e353dietary, 350e352, 376e377, 391drug <strong>in</strong>teractions, 244e245endometriosis, 455e459, 458f<strong>in</strong>fectious disease, 427mechanism of action, 457t, 457e458side effects, 243e244, 244b, 458e459specificity, 350structural classes of, 398tHistone demethylases (HDMs), 58e60, 60fcancer, 70f, 71e73, 116e117diabetes, 333, 336endometriosis, 452e453<strong>in</strong>fectious disease, 431KDM families, 453tstem cells and, 487e488Histone demethylases (HDMs) <strong>in</strong>hibitors,116e117Histone epigenomic modifiers, 285e286Histone methylation, 57e61, 59t, 60fag<strong>in</strong>g, 523Alzheimer’s disease, 183e185, 184fcancer, 70f, 71e73, 114e117, 115tdur<strong>in</strong>g development, 256e257<strong>in</strong>fectious disease, 428e431stem cells and, 483, 490Histone methyltransferases (HMTs),58e60, 59t, 60f, 155cancer, 70f, 71e73cardiovascular disease, 396endometriosis, 451e452<strong>in</strong>fectious disease, 421, 431obesity, 306site-specific list, 452tHistone modifications, 54e55, 55f,155e156, 508e509acetylation. See Histone acetylationag<strong>in</strong>g, 523e525, 528airway disease, 388f, 388e390Alzheimer’s disease, 183e185, 184fanalysis methods, 11e12, 17t, 20e21autoimmune disorders, 210, 228cancer. See Cancercardiovascular disease, 396e405, 399fcellular reprogramm<strong>in</strong>g and, 490cross-talk with other systems, 257dur<strong>in</strong>g development, 256e257diabetes, 322e324, 333DNA methylation and, 36e37endometriosis, 446, 449e453, 450t<strong>in</strong>fectious diseases, 418e419, 421e431methylation. See Histone methylationmode of action, 63e65phosphorylation, 61e62, 62t“reader/effector” modules, 453rheumatoid arthritis, 212e213ubiquit<strong>in</strong>ation, 62t, 62e63“writers” and “erasers” of, 450e453, 460Histone modification signature, 75Histone posttranslational modifications(PTMs), 11, 20, 53e54, 333Histone variants, 485e486Holliday, Rob<strong>in</strong>, 519, 545Homocyste<strong>in</strong>e cycle. See One-carbonmetabolismHormones. See also specific hormone<strong>in</strong> diabetes, 336e337dur<strong>in</strong>g <strong>in</strong>trauter<strong>in</strong>e development,549e550HOXA10 gene, 447, 454<strong>Human</strong> cytomegalovirus (HCMV),425e426<strong>Human</strong> immunodeficiency virus (HIV),426e428, 428t<strong>Human</strong> islet-derived precursor cells(hIPCs), 348e349<strong>Human</strong> mesenchymal stem cells (hMSCs),494<strong>Human</strong> papillomavirus (HPV), 431<strong>Human</strong> T-cell lymphotropic virus (HTLV),426, 428tHunt<strong>in</strong>gt<strong>in</strong> gene, 136Hunt<strong>in</strong>gton’s disease (HD), 127, 136,141e142Hutch<strong>in</strong>son-Gilford progeria syndrome(HGPS), 494e495Hybridizationcomparative genomic, 9, 39, 40ffluorescence <strong>in</strong> situ (FISH), 14of microRNA, 13Hydatidiform mole, 265, 276Hygiene hypothesis, 374e375Hyperdynamic (‘breath<strong>in</strong>g’) theory ofchromat<strong>in</strong> architecture, 486Hyperglycemic memory, 336Hypernutrition, 284e285, 302e305Hypothalamicepituitaryeadrenal (HPA)axis, 379, 550IICD (International Criteria of <strong>Disease</strong>), 132Immunity, 205e206. See also Allergicdiseases; Autoimmune disordersag<strong>in</strong>g process, 522development of, 371e374, 374fImmunodeficiency-centromeric<strong>in</strong>stability-facial anomalies(ICF) syndrome, 159Impr<strong>in</strong>ted genes, 257e263, 508e509assisted reproductive technologies and,559e560def<strong>in</strong>ition of, 131, 195, 257DNA methylation, 154, 195, 260evolution of, 25811p15 growth-regulatory region,260e261, 261f, 263, 278hypomethylation at multiple loci,268e269impr<strong>in</strong>ted doma<strong>in</strong>s, 261e263loci, 258, 259e260tmutations <strong>in</strong>, 264parental conflict theory of, 258, 276e277stem cell programm<strong>in</strong>g and, 512e513Impr<strong>in</strong>t<strong>in</strong>g centers, 258, 264Impr<strong>in</strong>t<strong>in</strong>g disorders, 4, 263e269. See alsospecific disorderchromosomes, 265e268mutations, 264e265neurodevelopmental, 168e169,195e196, 196f<strong>in</strong> obesity, 275e279, 276t, 287e288phenotypes, 265, 266te267tstem cell epigenetics, 492t, 493e494uniparental disomy, 263e264, 277Induced pluripotent stem cells (iPSCs),487e491disease models us<strong>in</strong>g, 493e495transcription factor network, 505f,505e506Infectious diseases, 5, 415e432. See alsospecific diseaseepigenetic modificationsbacterial, 417e421, 422ttrematode, 432viral, 31e33, 422e431immune development and, 374e376Inflammation<strong>in</strong> ag<strong>in</strong>g process, 533corticosteroids for, 389e391, 391f<strong>in</strong> diabetes pathogenesis, 342e343DNA methylation alterations <strong>in</strong>, 31e33epigenetic drugs for, 235e236Inflammatory bowel diseases (IBD)epigenetic deregulation <strong>in</strong>, 207t,216e217treatment of, 234e235Infliximab, 226Inheritance, transgenerational, 197e198,309e310Inhibitor kappa B k<strong>in</strong>ase complex (IKK),342e343Insul<strong>in</strong>, 322, 336e337, 341Insul<strong>in</strong>-like growth factor (IGF),261e262, 550e551Insul<strong>in</strong> resistance, 283e284, 325e326,342Insul<strong>in</strong> secretion, 322e324International <strong>Human</strong> EpigenomeConsortium (IHEC), 14, 20, 47Intrauter<strong>in</strong>e environment.See Development; MaternalnutritionIntrauter<strong>in</strong>e growth restriction (IUGR)epigenetic consequences of, 547e548,549fHPA axis reprogramm<strong>in</strong>g, 550obesity and, 283e284, 302e305, 324Intrauter<strong>in</strong>e system (IUS), drugconta<strong>in</strong><strong>in</strong>g,459In vitro fertilization (IVF), 307, 559e560JJumonji (JmjC) doma<strong>in</strong>-conta<strong>in</strong><strong>in</strong>gprote<strong>in</strong>s, 60, 116e117, 404, 452,488Juvenile diabetes mellitus (type 1), 207t,217, 321. See also DiabetesKKabuki syndrome, 166K-acetyltransferases (KATs), 333e336,335t, 342e346, 345b. See alsoHistone acetyltransferases573


INDEX574Kaposi’s sarcoma (KS), 424e425, 425tK-demethylases (KDMs), 453t. See alsoHistone demethylasesK<strong>in</strong>ases, 61e62, 62tcancer, 70f, 73e74K-methyltransferases (KMTs). See alsoProte<strong>in</strong> lys<strong>in</strong>emethyltransferases (PKMTs)compounds target<strong>in</strong>g, 350diabetes, 333, 336endometriosis, 451e452KvDMR1, 263LLatent autoimmune diabetes <strong>in</strong> adults(LADA), 321LEARn (latent early-life associatedregulation) model, 529Lept<strong>in</strong>diabetes, 347, 349e350obesity, 275, 285, 286f, 309,311e312,553Lifecourse strategies, 313f, 313e314Life expectancy, 520e521. See also Ag<strong>in</strong>gL<strong>in</strong>eage commitment, 373, 506e508, 507fListeria monocytogenes, 418Lithium, 198Liver fluke (Opistorchis viverr<strong>in</strong>i), 432Locked nucleic acid (LNA), 13Longevity genes, 522Long <strong>in</strong>tergenic ncRNAs (l<strong>in</strong>cRNA), 512Lucas, Alan, 546Lymphocyte changes, <strong>in</strong> neurobehavioraldiseases, 142Lys<strong>in</strong>e demethylases. See HistonedemethylasesLys<strong>in</strong>e methyltransferases. See HistonemethyltransferasesMMacrolides, 391Marie-Strümpell disease, 217Massively parallel short-read DNAsequenc<strong>in</strong>g (MPSS), 19Mass spectrometry, 10e12Maternal duplications, <strong>in</strong>neurodevelopmental disorders,169e170Maternal factorsdiet. See Maternal nutritionimmune system, 371e374, 374f, 379smok<strong>in</strong>g, 377, 557e558substance use, 557e558Maternally <strong>in</strong>herited diabetes anddeafness (MIDD), 321Maternal nutrition, 547e548allergic diseases and, 376e377diabetes and, 324f, 324e326epigenetic consequences of, 547e548,549f, 552, 555fam<strong>in</strong>es, 282e283, 298, 302, 326, 554fetal macrosomia, 548e549, 550fobesity and, 281e282, 298e303animal models, 303e305, 308e312epigenetic modifications, 307e312fetal plasticity, 282e283, 305e307human studies, 312hypernutrition, 284e285postnatal environmental mismatch,283e284, 302e303Maternal UPD 14-like phenotype, 266t,267e268, 276tMatrix-assisted laser desorption ionizationtime-of-flight mass spectrometry(MALDI-TOF), 10e11Mattick, John, 509Maturity-onset diabetes of the young(MODY), 321, 324, 334, 335tMelanocort<strong>in</strong>, 275Membrane-bound catechol-O-methyltransferase (MB-COMT), 142Memory function, 184e187Mendelian childhood-onset extremeobesity syndromes, 274t,274e275Merkel cell polyomavirus (MCPyV), 431Metabolic memory, 401e402Metabolic syndrome, 287e288, 326, 342Metabolic thrift, 553Metabolismag<strong>in</strong>g process, 533e536, 534f, 554e555Alzheimer’s disease, 177f, 177e183, 187early <strong>in</strong>terventions, 551neurobehavioral diseases, 143e145obesity, 275, 278e279, 282, 285e286,286fMetal exposure, 182Metastable alleles, 286e287Metastasis (cancer), 93Methotrexate, 244MethylationDNA. See DNA methylationhistone. See Histone methylationMethylation-sensitive s<strong>in</strong>gle-nucleotideprimer extension (MS-SNuPE),9e10Methylation-specific PCR (MSP), 475Methyl-b<strong>in</strong>d<strong>in</strong>g doma<strong>in</strong>s (MBDs)autoimmune disorders, 236cardiovascular disease, 400neurodevelopmental disorders, 155,156f, 159e161Methyl-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s (MBPs), 155Methyl CpG-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> (MeCP2),508ag<strong>in</strong>g, 524, 527biological role of, 155diabetes, 332e333endometrial cancer, 473endometriosis, 448<strong>in</strong>fectious disease, 419e420, 426e428,432neurobiological disorders, 195e196,196fneurodevelopmental disorders, 159e161,160f, 164e165, 492e493obesity, 306stem cells and, 483, 489e490Methyl donors, prenatal exposure to,554e556MethyLight technology, 10Methylome, 8, 22, 288, 526Microarray analysis, 9, 17tDNA replication tim<strong>in</strong>g, 14e15microRNA, 13Microbial exposures. See also Infectiousdiseasesimmune development and, 374e376MicroRNA (miRNA), 89<strong>in</strong> ag<strong>in</strong>g process, 528<strong>in</strong> allergic diseases, 373<strong>in</strong> Alzheimer’s disease, 185e186, 186t,186fanalysis methods, 12e14, 17t, 102<strong>in</strong> autoimmune disorders, 237e240,241t, 243, 245e246biogenesis, 89e91, 90f<strong>in</strong> cancer, 92e103, 420. See also Cancer<strong>in</strong> cardiovascular disease, 408e409<strong>in</strong> diabetes, 334, 339e341, 352e353<strong>in</strong> endometrial cancer, 474e475<strong>in</strong> endometriosis, 446<strong>in</strong> <strong>in</strong>fectious disease, 427e428<strong>in</strong> stem cell programm<strong>in</strong>g, 510e512, 511ftarget gene <strong>in</strong>teractions, 91e92MicroRNA antagonists (antagomirs),240e243MicroRNA-<strong>in</strong>duced silenc<strong>in</strong>g complexes(miRISCs), 237Microsatellite <strong>in</strong>stability (MSI), 473Milk consumption, 551M<strong>in</strong>or groove b<strong>in</strong>der (MGB)technology, 10Mismatch, postnatal environmental,283e284, 306Mismatch repair (MMR) deficiency, 473Multicentric Castleman’s disease (MCD),424Multiple sclerosis (MS)epigenetic deregulation <strong>in</strong>, 207t, 213treatment of, 235, 240Mutations. See also specific gene or disorderag<strong>in</strong>g process, 421Alzheimer’s disease, 176DNMTs, 34e35<strong>in</strong> epigenomic programm<strong>in</strong>g, 131e132epimutations, 170, 264, 513e514,545e546impr<strong>in</strong>t<strong>in</strong>g, 264e265neurodevelopmental disorders,162e166, 170Myel<strong>in</strong> basic prote<strong>in</strong> (MBP), 213Myelocytomatosis oncogene (c-Myc), 118,122, 510e511NNanog promoter, 483e495, 511e512Neel, James, 552Neurobehavioral diseases, 3, 127e147.See also specific diseasediagnosis of, 132e136environmental factors, 138e139genetics, 136e138genomic <strong>in</strong>stability, 139e142metabolism, 143e145RNA epigenomics, 143


INDEXstem cell epigenetics, 491e492, 492ttherapy, 145e146transcriptional dysregulation, 142e143Neurobiological disorders, 193e200.See also specific disordercongenital, 195e196environmental factors, 194e196, 197f,199e200epigenetic therapies, 198e199stem cell epigenetics, 491e492, 492ttransgenerational <strong>in</strong>heritance, 197e198Neurodegeneration, <strong>in</strong> ag<strong>in</strong>g process,526e529, 535e536Neurodevelopmental disorders, 3,153e170. See also specific disorderepigenetic processes <strong>in</strong>, 153e154, 193aberrant patterns, 166e169mach<strong>in</strong>ery defects, 156e166stem cell epigenetics, 491e493, 492tNeuroendocr<strong>in</strong>e theory of ag<strong>in</strong>g, 522Neurofibrillary tangles (NFT), 175e176Next-generation sequenc<strong>in</strong>g (NGS), 11,17t, 19Nicot<strong>in</strong>amide, 347e348Non-cod<strong>in</strong>g RNAs (ncRNA), 12e14, 257,334, 509e510. See alsoMicroRNA<strong>in</strong> stem cell programm<strong>in</strong>g, 510e513, 511fNon-communicable diseases (NCD), 297.See also specific diseaselifecourse strategies, 313f, 313e314Non-histone prote<strong>in</strong>s, 257acetylation of, 118e122, 388e391Non-nucleoside <strong>in</strong>hibitors, 117e118,120te121tNorthern blot analysis, 12e13Nortriptyl<strong>in</strong>e, 145, 391Nuclear effector (NUE), 421Nuclear factor kappa light cha<strong>in</strong> enhancerof activated B cells (NFkB)airway disease, 388e389autoimmune diseases, 228cancer, 118, 122diabetes, 342e343, 347e348endometriosis, 458Nucleoside <strong>in</strong>hibitors, 117e118, 119te120tNucleosome, 54, 111, 130, 256, 343, 482,508Nutritionag<strong>in</strong>g and, 532Alzheimer’s disease, 177e183, 187diabetes therapy, 350e352endocr<strong>in</strong>e-metabolic disturbances, 551endometriosis and, 454maternal. See Maternal nutritionneurological diseases and, 138e139,145e146, 198obesity, 273e274, 279f, 279e280OObesity, 4, 273e289animal models, 279f, 279e280developmental orig<strong>in</strong>s of, 298e303.See also Maternal nutritionanimal models, 303e305, 308e312epigenetic modifications, 307e312fetal plasticity, 305e307diabetes and, 341environmental factors, 273e274,280e284, 298e303, 310epidemiology of, 297e298epigenetic marks, 275, 307, 309e310,312e314epigenetic therapies, 297e314epigenome-wide studies, 284, 288e289future research, 289genome-wide studies, 275, 287e288, 303,310histone epigenomic modifiers, 285e286impr<strong>in</strong>t<strong>in</strong>g disorders <strong>in</strong>, 275e279, 276t,287e288lept<strong>in</strong>, 275, 285, 286f, 309,311e312, 553Mendelian childhood-onset syndromes,274t, 274e275metastable alleles, 286e287nutrition, 273e274, 279f, 279e280phenotypes, 298, 299te301ttransgenerational effects, 309e310Obesogenic environment, 273e274,280e281, 326Oct4 promoter, 483e495, 511e512“Omni-seq” approach, 289OncogenesmicroRNA as, 31, 94e95replication tim<strong>in</strong>g assays, 15, 16fOncomirs, 93e95One-carbon metabolismag<strong>in</strong>g process, 533e536, 534f, 554e555Alzheimer’s disease, 177f, 177e183,187Opistorchis viverr<strong>in</strong>i (liver fluke), 432Outdoor pollutants, 378e379“Overlap” syndrome, 238Overnutrition, 284e285, 302e305Oxidative phosphorylation (OXPHOS)genes, 323, 325Oxidative stress theory of ag<strong>in</strong>g, 522Oxytoc<strong>in</strong>, 198Oxytoc<strong>in</strong> receptor (OTR), 458PPancreatic islets, 323, 340e341, 347e348Paraff<strong>in</strong>-embedded tissue samples(PAT-CHIP-seq), 75Parental genome conflict, 258, 276e277Parent-of-orig<strong>in</strong>-specific genes.See Impr<strong>in</strong>ted genesPark<strong>in</strong>son’s disease, 127, 193Partially methylated doma<strong>in</strong>s (PMDs), 131Paternal duplications, <strong>in</strong>neurodevelopmental disorders,169e170Paternal UPD 14-like phenotype, 266t,267e268Patho-epigenetics, 416, 432. See alsoInfectious diseasesPCNA (proliferat<strong>in</strong>g cell nuclear antigen)-b<strong>in</strong>d<strong>in</strong>g doma<strong>in</strong>, 30PCR amplification, 9Pelizaeus-Merzbacher disease, 193Periodontal disease, 417e418Peroxisome Proliferator-ActivatedReceptor g, coactivator 1(PPARCG1A), 280, 323, 325Peroxisome proliferator-activated receptorgamma (PPARg), 337e338Personalized medic<strong>in</strong>e, cancer, 44e47Pervasive developmental disorder (PDD),133e134Phenotypesimpr<strong>in</strong>t<strong>in</strong>g disorders, 265, 266te267tobesity-related, 277e279obesity, 298, 299te301tPhenylbutazone, 233Phenylbutyrate (PBA), 185, 349e350Phosphatases, 62, 62tPhosphorylationhistone, 61e62, 62toxidative, 323, 325tau, 182e183Placental impr<strong>in</strong>t<strong>in</strong>g, 276e277Plasticitydevelopmental, 282e283, 305e306, 546neuronal, 527Pluripotency transcription factor network,505f, 505e506, 510e512, 511fPluripotent stem cells, 504e505. See alsoStem cellsPollutants, 378e379, 558e559Polycomb group (PcG) prote<strong>in</strong>s, 257<strong>in</strong>fectious disease, 421, 431stem cells and, 486Polycyclic aromatic hydrocarbons (PAH),378, 559Polycystic ovarian syndrome (PCOS), 459Polymerase cha<strong>in</strong> reaction (PCR), 475,475fPolymyositis, 240Porphyromonas g<strong>in</strong>givalis, 417Posttranslational modifications (PTMs),11, 20, 53e54, 333Prader-Willi syndrome (PWS), 168e169epigenetic mechanisms, 195, 268obesity related to, 275, 276t, 277stem cells and, 493e494PR-B promoter, 447e448PR doma<strong>in</strong>-conta<strong>in</strong><strong>in</strong>g prote<strong>in</strong>s (PRDMs),58Precancerous conditions, 31e33Predictive adaptive response (PAR),305e306Pregnancy. See Development; Maternalfactors; Maternal nutritionPremature ag<strong>in</strong>g, 494e495Presenil<strong>in</strong>1 (PSEN1) gene, 526Primary biliary cirrhosis (PBC), 207t, 215,240Primary effusion lymphoma (PEL), 424Proca<strong>in</strong>amide, 236Process<strong>in</strong>g alterations, microRNA, 98e99Prognostic <strong>in</strong>dicators. See also Biomarkersautoimmune disorders, 243, 246cancer, 44e47, 45e46t, 102endometriosis, 460Progressive systemic sclerosis (PSS), 207t,214, 240575


INDEX576Prote<strong>in</strong> arg<strong>in</strong><strong>in</strong>e methyltransferases(PRMTs), 58e60, 59tAlzheimer’s disease, 183cancer, 70f, 71e73, 115Prote<strong>in</strong> code, 333, 342, 509Prote<strong>in</strong> lys<strong>in</strong>e methyltransferases(PKMTs), 115, 183. See alsoK-methyltransferasesProte<strong>in</strong> modification theory of ag<strong>in</strong>g, 522Prote<strong>in</strong>-restricted (PR) diet, maternal,303e304, 308, 311, 326Proteomic analysis, 238Pseudohypoparathyroidism, 267t, 268,276t, 278Psoriasis, 207t, 214e215Puberty, epigenomic <strong>in</strong>stability dur<strong>in</strong>g,311PWS/AS region, duplications <strong>in</strong>, 169e170Pyrosequenc<strong>in</strong>g, 10QQuantitative analysis of methylated alleles(QAMA), 10Quantitative bisulfite sequenc<strong>in</strong>g us<strong>in</strong>gpyrosequenc<strong>in</strong>g technology(QBSUPT), 10RRadiation therapy, 100e102“Reader/effector” modules, 453Receptor Activator for Nuclear actor kBLigand (RANKL), 233e234Regulatory T cells (Treg), 230, 371e374Reproductive disorders, 5. See alsoEndometrial cancer;EndometriosisRestriction enzymes, methylationsensitive,9Resveratrol, 351e352Ret<strong>in</strong>oid X receptor alpha (RXRA)promoter, 313Retroviruses, 426e428, 428tRett syndrome (RTT), 127, 159e161environmental factors, 199genetics, 132, 164pathogenesis of, 195e196, 199, 199fstem cell epigenetics, 493Reversibility of epigenetic modifications,47, 75, 199e200, 200f, 228, 514,560Rheumatoid arthritis (RA)epigenetic modifications, 207t, 210e213,211ftreatment of, 226, 233, 234b, 239e240,242tRheumatoid arthritis synovial fibroblasts(RASF), 210e211Rh<strong>in</strong>itis, 370. See also Allergic diseasesRNAmicro. See MicroRNAnon-cod<strong>in</strong>g, 12e14, 257, 334, 509e510small nucleolar, 268RNA edit<strong>in</strong>g, 99, 143RNA-<strong>in</strong>duced transcriptional silenc<strong>in</strong>g(RITS), 167Roberts and SC phocomelia syndromes(RS/SC), 163Romideps<strong>in</strong>, 455Royal jelly, 198, 459Rubenste<strong>in</strong>-Taybi syndrome, 165SS-adenosylmethion<strong>in</strong>e (SAM), 74,555e556S-adenosylmethion<strong>in</strong>e/homocyste<strong>in</strong>e(SAM/HCY) metabolism,535e536. See also One-carbonmetabolismSalicylates, 347Schistosoma haematobium, 432Schizophrenia (SZ), 127classification of, 134b, 134e135diagnosis of, 132e133, 133t, 142DNMTs, 157environmental factors, 137f, 138e139epigenomic programm<strong>in</strong>g, 131genetics, 136e138, 137fgenomic <strong>in</strong>stability, 141e142maternal duplications, 169outcomes of, 135f, 135e136transcriptional dysregulation, 142e143Scleroderma, 207t, 214, 240Senile plaques (SP), 175e176Sequenc<strong>in</strong>g analyses, 9Sex-specific differencesautoimmune disorders, 216, 226epigenomic complexity, 131, 195e196genome-wide demethylation, 256Shigella flexneri, 418Signal pathways, DNA methylation status,35e36Silver-Russell syndrome, 265, 266t, 494S<strong>in</strong>gle-molecule DNA sequencer, 11S<strong>in</strong>gle nucleotide polymorphisms (SNPs),136, 275S<strong>in</strong>gle-nucleotide primer extension,methylation-sensitive, 9e10S<strong>in</strong>gle-strand conformationpolymorphism (SSCP), 9Sirtu<strong>in</strong>s (SIRTs), 55e57, 56tcancer, 70f, 70e71cardiovascular disease, 397, 401diabetes, 335e336, 339e341,346e347Sirtu<strong>in</strong> <strong>in</strong>hibitors, 350Sjögrens syndrome (SjS), 207t, 213, 240“Small baby syndrome hypothesis,”548e549Small nucleolar RNAs (snoRNAs), 268Smok<strong>in</strong>gallergic diseases and, 377DNA methylation alterations <strong>in</strong>, 31e33maternal, 377, 557e558Smooth muscle cells (SMCs), 396homeostasis, 401e404, 403f, 406tSomatic cell mosaicism, 513e514Splic<strong>in</strong>g alterationscardiovascular disease, 402, 403fDNMTs, 34e35neurobehavioral diseases, 143Staple isotope label<strong>in</strong>g with am<strong>in</strong>o acids<strong>in</strong> cell culture (SILAC), 238Stem cells (SCs), 5e6, 481e495chromat<strong>in</strong> bivalency, 486e487diabetes, 348e349endothelial cell differentiation, 401epigenetics, 482e485, 484fhuman disease, 491e493, 492tprogramm<strong>in</strong>g, 510e514epimutations, 513e514, 545e546future research, 495histone variants and, 485e486human mesenchymal, 494<strong>in</strong>duced pluripotent. See Inducedpluripotent stem cellsregulatory networks, 506e508, 507frenewal, 505f, 505e506types of, 504f, 504e506Steroidogenic factor-1 (SF-1), 448Stochastic modelsag<strong>in</strong>g, 521e522, 545e546iPSC generation, 489, 489fStressag<strong>in</strong>g and, 532diabetes and, 343e346neurobehavioral diseases and, 138e139,197e198prenatal, 556e557Suberoylanilide hydroxamic acid (SAHA),231e232, 235e236, 490tSubstance use, maternal, 557e558Substrate recognition, 65Sucrose nonferment<strong>in</strong>g 2 (SNF2) family ofchromat<strong>in</strong> remodel<strong>in</strong>g prote<strong>in</strong>s,162Sulforaphane, 351, 391Sulpiride, 145, 198Systemic lupus erythematosus (SLE)epigenetic deregulation <strong>in</strong>, 207t,207e210, 209f, 228treatment of, 232e233, 239TTau phosphorylation, 182e183T-cell differentiation, 372e374Telomerase, 524Telomeres, 533TET (Ten-eleven-translocation) prote<strong>in</strong>s,310, 527Theophyll<strong>in</strong>e, 390e392, 391fTherapy. See also specific disease or drugautoimmune disorders, 226, 233, 242tcancer, 47, 99e102diabetes, 350e352endometriosis, 444e445epigenetic. See Epigenetic therapiesneurobehavioral diseases, 145e146Thiazolid<strong>in</strong>ediones (TZDs), 337e338Thompson, James, 505Thrifty epigenotype hypothesis, 553Thrifty phenotype hypothesis, 282e283,302, 552e553Thyroid diseases, 207t, 215e216Tobacco smoke. See Smok<strong>in</strong>gTocilizumab, 244


INDEXToll-like receptor (TLR) signal<strong>in</strong>g,228e229, 417Totipotent stem cells, 504. See also StemcellsTranscriptional control, histoneacetylation <strong>in</strong>, 57Transcriptional dysregulation<strong>in</strong>fectious disease, 427e428microRNA, 96e97neurobehavioral diseases, 142e143Transcriptional silenc<strong>in</strong>g. See Genesilenc<strong>in</strong>gTranscription factors<strong>in</strong> diabetes, 337e338, 340pluripotency network, 505f, 505e506,510e512, 511fstem cell differentiation, 483, 484fTransgenerational epigenetic <strong>in</strong>heritance,197e198, 309e310Trans mechanism, 63e64Transmembrane prote<strong>in</strong>s, 423e424Transporters, <strong>in</strong> diabetes, 338e339Trematode <strong>in</strong>fections, 432Trichostat<strong>in</strong> A (TSA)allergic diseases, 372autoimmune disorders, 212e213,232e233, 235e236cardiovascular disease, 397, 402, 405cellular reprogramm<strong>in</strong>g, 490tdiabetes, 341endometriosis, 455e457Tr<strong>in</strong>ucleotide repeat diseases, 139e141,140tTrithorax system, 257, 486Tumorigenesis. See also CancerDNA methylation status dur<strong>in</strong>g, 35e36Tumor necrosis factor alpha <strong>in</strong>hibitors,226Tumor suppressorsendometrial cancer, 473, 475e476microRNA as, 31, 93e94, 420Type 1 diabetes mellitus (T1DM), 207t,217, 321. See also DiabetesUUbiquitilases, 62t, 62e63alterations <strong>in</strong> cancer, 70f, 73e74Ubiquit<strong>in</strong>ation, histone, 62t, 62e63<strong>in</strong> cancer, 70f, 73e74Ulcerative colitis (UC)epigenetic deregulation <strong>in</strong>, 207t,216e217treatment of, 234e235Uniparental disomy (UPD), 263e264,277Unmethylated sequences, bisulfiteconversion of, 9U.S. National Institute of Mental Health(NIMH), 127, 132VValproic acid (VPA)autoimmune disorders, 231cardiovascular disease, 401cellular reprogramm<strong>in</strong>g, 490t, 491diabetes, 347, 349endometriosis, 455e459<strong>in</strong>fectious disease, 427neurobehavioral disorders, 130e131,145, 198Ventricular remodel<strong>in</strong>g, 406e407Vidaza, 117Viral <strong>in</strong>fection. See also Infectious diseases;specific diseaseepigenetic modifications, 31e33,422e431Vitam<strong>in</strong> B deficiency, 181, 187, 534, 536Vitam<strong>in</strong> D, 376Vitiligo, 214Vor<strong>in</strong>ostat, 347, 350WWadd<strong>in</strong>gton, Conrad, 128, 193, 254, 482,488, 519XX chromosome, epigenomicprogramm<strong>in</strong>g, 131, 195e196X chromosome <strong>in</strong>activation (XCI), 216,226, 493, 513X-l<strong>in</strong>ked mental retardation (XLMR), 162X prote<strong>in</strong> (HBx or pX), 429YYamanaka, Sh<strong>in</strong>ya, 505Y chromosome, epigenomicprogramm<strong>in</strong>g, 131, 195e196577


This page <strong>in</strong>tentionally left blank


Color PlatesFIGURE 3.1DNA methylation profiles <strong>in</strong> precancerous conditions and renal cell carc<strong>in</strong>omas (RCCs). See p. 39 for details.FIGURE 3.2Hierarchical cluster<strong>in</strong>g analysis of urothelial carc<strong>in</strong>omas (UCs) based on array comparative genomic hybridization (CGH) data. See p. 40 for details.


FIGURE 3.3Diagnostic criteria based on DNA methylation profiles for ductal adenocarc<strong>in</strong>omas of the pancreas. See p. 42 for details.H3N-terH2AN-terS1PK5AcK9AcK12AcK15P Me P PT3 R8 AcS10 T11K4K18 MeR2K9 K14 R17 R26MeMe Ac Me AcAcK23PMeMe AcS28K27K36Me AcMe AcAcAcK36H2AC-terMeK99Ub MeK119T120PPPK79Y41 T45 K56 MeK37 Me AcMeMe AcAcAcK5Ac K12Ac Me PPAc MeK15K108 K85 K43 S36 S32 K28 K27S14K20PUb AcUb AcK120AcK116H2BC-terMe AcR92AcK20MeMeK77AcK91S47K12K79 K59K5AcP K16 K8 AcAc MeAcAcR3MeS1PN-terH2BH4N-terFIGURE 4.1Mammalian core histone modifications. See p. 55 for details.Chromat<strong>in</strong> Residues modified Function regulatedModificationAcetylation Lys<strong>in</strong>e Transcription, DNA repair,replication and condensationMethylation (Lys<strong>in</strong>e) Lys<strong>in</strong>e me1, me2, me3 Transcription, DNA repairMethylation (Arg<strong>in</strong><strong>in</strong>e) Arg<strong>in</strong><strong>in</strong>e-me1, Arg<strong>in</strong><strong>in</strong>e-me2a TranscriptionArg<strong>in</strong><strong>in</strong>e-me2sPhosphorylation Ser<strong>in</strong>e, Threon<strong>in</strong>e, Tyros<strong>in</strong>e Transcription, DNA repair andcondensationUbiqut<strong>in</strong>ationLys<strong>in</strong>eTranscription, DNA repairSumoylation Lys<strong>in</strong>e TranscriptionADP ribosylation Glutamic TranscriptionDeim<strong>in</strong>ation Arg<strong>in</strong><strong>in</strong>e TranscriptionProl<strong>in</strong>e isomerization P-cis, P-trans TranscriptionDifferent classes of histone modificationsAcMePUbAcetylationMethylationPhosphorylationUbiqut<strong>in</strong>ation


KDM5A/JARID2AKDM5B/JARID2BKDM5C/JARID2CKDM5D/JARID2DKDM1A/LSD1KDM1B/LSD2H3MeK4KDM1A/LSD1KDM1B/LSD2KDM3A/JHDM2AKDM3B/JHDM2BKDM4A/JMJD2AKDM4B/JMJD2BKDM4C/JMJD2CKDM4D/JMJD2DKDM7SMYD3KMT7/Set7-9KMT2A/MLL1KMT2B/MLL2KMT2C/MLL3KMT2D/MLL4KMT2E/MLL5KMT2F/hSET1AKMT2G/hSet1BKMT2H/ASH1K9MeKMT1A/SUV39H1KMT1B/SUV39H2KMT1C/G9aKMT1D/GLPKMT1E/SETDB1KMT1F/CLL8KMT8/RIZ1KDM7KDM6A/UTXKDM6B/JMJD3MeK27KDM2A/JHDM1AKDM2B/JHDM1BKDM5A/JARID2AKDM5B/JARID2BKDM5C/JARID2CKDM8EZH1EZH2K36MeK79MeKMT3B/NSD1NSD2SMYD2/KTM3CKMT4/DOT1LPHF8K20MeKMT5B/SUV4-20H1KMT5C/SUV4-20H2KMT5A/Pr-Set7/8Histone lys<strong>in</strong>e methyltransferasesH4Histone lys<strong>in</strong>e demethylasesFIGURE 4.2Histone lys<strong>in</strong>e methylases and demethylases. See p. 60 for details.CategoriesGenesCategoriesGenesHATsHDACsp300CBPMYST familyHDAC1,HDAC2,HDAC3,HDAC5,HDAC6,HDAC10Sirtun 1,2,3,7HATsAcAcHDACsPPPPK<strong>in</strong>aseK<strong>in</strong>asesAURORA-BJAK2MSK1MST1RSKPI3KsCategoriesGenesCategoriesGenesHMTsKTM1A/Suv39h1KTM2A/MLL1KTM2Z/MLL3SMYD2,SMYD3,SMYD4KTM1C/G9AKTM6B/EZH2NSD1,NSD2,NSD3DOT1LHDMsMeMeHMTsUb E3DubsRNF20USP22USP7PRMTsHDMsPRMT1PRMT4/CARM1PRMT6KDM1AKDM2A,KDM2BKDM4A,B,CKDM5A,B,CKDM6A,KDM6BKDM8DubsUbUbUb E3sFIGURE 4.3Histone-modify<strong>in</strong>g enzymes and cancer. Selection of histone modify<strong>in</strong>g enzymes altered <strong>in</strong> human cancer.


FIGURE 5.1miRNA biogenesis and the mechanism of gene silenc<strong>in</strong>g. See p. 90 for details.


FIGURE 6.1Effects of acetylation and methylation on histone residues. The red circle represents acetyl groups, the yellow circle symbolizes methylation and the greensymbolizes methylation of arg<strong>in</strong><strong>in</strong>e residue. See p. 112 for details.FIGURE 7.2Epigenomic programm<strong>in</strong>g of chromat<strong>in</strong>.


FIGURE 7.3Outcome of schizophrenia diagnosis. (adapted from http://www.science.org.au).FIGURE 7.4Odds ratio for schizophrenia as a function of genetic and environmental risk factors (adapted from [73]).


FIGURE 7.6The FMTD hub: Metabolic l<strong>in</strong>k between DNA and RNA synthesis, energy production, epigenomic programm<strong>in</strong>g, oxidative stress responses, <strong>in</strong>flammation, anddopam<strong>in</strong>e metabolism (not all metabolic steps are shown).(A)(B)(C)FIGURE 8.1Establish<strong>in</strong>g and ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g DNA methylation <strong>in</strong> mammals. (A) Stepwise progression from unmethylated based-paired CpG d<strong>in</strong>ucleotides (1) to fully methylatedDNA (2), and its ma<strong>in</strong>tenance follow<strong>in</strong>g DNA replication. Transition between (1) and (2) is de novo methylation. Ma<strong>in</strong>tenance requires an obligatehemimethylated <strong>in</strong>termediate (3). (B) De novo methylation of DNA occurs largely <strong>in</strong> the early stages of gametogenesis and ma<strong>in</strong>tenance methylation is primarilya post-fertilization process. (C) Differentially methylated doma<strong>in</strong>s (DMDs) of impr<strong>in</strong>ted genes are created through a process <strong>in</strong> which only one parental alleleundergoes de novo methylation <strong>in</strong> the germ l<strong>in</strong>eage, and follow<strong>in</strong>g its ma<strong>in</strong>tenance <strong>in</strong> the embryo, a difference <strong>in</strong> methylation of the two parental alleles is seen.Snurf/Snrpn is a maternally methylated DMD and Igf2/H19 is a paternally methylated DMD.


FIGURE 10.1Location of the molecules <strong>in</strong> the synapse, which are associated with the pathogenesis of autism (references [25] and [45]).FIGURE 10.2Schematic representation of epigenetic control of the gene expression. Yellow circle: MeCP2, orange circle: prote<strong>in</strong> associatedwith histone modification, red circle: methyl-residue, green square: chromosomal histone prote<strong>in</strong>, arrow: transcription start site.See p. 196 for details.FIGURE 10.3DNA methylation mechanisms can be affected by environmental factors.


FIGURE 10.4Pathogenesis of Rett syndrome and putative pathogenesis of autism.FIGURE 10.5Overview of the DNA methylation changes and environmental factors. See p. 200 for details.RA DNA METHYLATIONHDACi EFFECT IN RACELL ADHESIONEPHRINBEPHRINBIL6T-CELLGlobal DNAHypomethylationRASFAPOPTOSISDR3METMAPK13miR203IL6MMP1INFLAMMATIONHDACiT-CELLRASFNr AUTO-REACTIVE Th1Nr TregNr Th2• APOPTOSIS• ANGIOGENESIS• PROLIFERATION• INFLAMMATION• MATRIXDEGRADATIONMATRIXDEGRADATIONMONOCYTEOSTEOCLASTFIGURE 11.2Epigenetic changes <strong>in</strong> RA. See p. 211 for details.


FIGURE 12.2Molecular targets for epigenetic treatment of autoimmune diseases.FIGURE 12.3Potential effects of epigenetic changes <strong>in</strong> autoimmune diseases.


(A)(B)FIGURE 13.1Changes <strong>in</strong> the overall level of DNA methylation dur<strong>in</strong>g mammalian development. (A) Changes <strong>in</strong> DNA methylation <strong>in</strong> germ cells. (B) Changes <strong>in</strong> DNA methylationfollow<strong>in</strong>g fertilization. In both panels the level of DNA methylation is shown on the vertical axis and developmental time on the horizontal axis. Adapted from [16].


FIGURE 14.1Arcuate nucleus control of central energybalance between food <strong>in</strong>take and energyexpenditure. See p. 274 for details.FIGURE 14.3Methylation DNA immunoprecipitation (MeDIP) graphical result for the Lept<strong>in</strong> Promoter CpG Island. Slid<strong>in</strong>g scale for methylation level: yellow ¼ low, green ¼mid, blue ¼ high. Hypomethylation seen over the CpG Island and no significant difference seen between T2D cases (f<strong>in</strong>al_avg_diabetes) versus controls(f<strong>in</strong>al_ave_control).


FIGURE 15.1Non-communicable diseases (NCDs) do not fit the medicalmodel <strong>in</strong> which an <strong>in</strong>dividual is healthy until they contract thedisease. See p. 313 for details.(A)(B)(D)(C)FIGURE 17.1Overview of epigenetic mechanisms. See p. 333 fordetails.FIGURE 17.3Simplified overview of how HDACi could target diabetes.See p. 343 for details.


FIGURE 18.1Environmental <strong>in</strong>fluences on develop<strong>in</strong>g immune systemSee p. 374 for details.FIGURE 19.1Post-translational histone modification. See p. 388 for details.FIGURE 19.2Acetylation of the glucocorticoid receptor (GR). See p. 389 for details.


FIGURE 19.3Mechanisms for decreased histone deacetylase(HDAC)2 <strong>in</strong> COPD and its reversal. See p. 391 for details.FIGURE 20.1A schematic illustration shows the role of HDAC7 <strong>in</strong> controll<strong>in</strong>g EC growth. See p. 399 for details.FIGURE 20.2A schematic illustration shows the role of HDAC7 splic<strong>in</strong>g <strong>in</strong> controll<strong>in</strong>g SMC differentiation and proliferation. See p. 403 fordetails.


FIGURE 22.1Schematic illustration of potential therapeutic effects of HDACIs <strong>in</strong> endometriosis. See p. 458 for details.


FIGURE 24.1The pluripotency of ES cells and their differentiation rely largely on transcription factor circuitry and chromat<strong>in</strong> modifications. See p. 484 for details.


FIGURE 24.2The stochastic model proposed by Yamanaka for iPS cell generation is based on the Wadd<strong>in</strong>gton model. See p. 489 for details.FIGURE 25.1Schematic for the general maturation of stem cells and loss of differentiation potential.


FIGURE 25.2Schematic for the <strong>in</strong>duction of pluripotency <strong>in</strong> somatic cells by the pluripotency network of transcription factors.


FIGURE 25.3(A) Schematic of the compet<strong>in</strong>g network of transcription factors and signal<strong>in</strong>g molecules that direct stem cells down specific mesoderm or ectoderm l<strong>in</strong>eages.Green boxes <strong>in</strong>dicate core transcription factors necessary for osteoblast and adipocyte l<strong>in</strong>eage specific differentiation. (B) Overlay of the epigenetic/ncRNAregulatory network. This schematic depicts examples of ncRNAs (blue boxes) and chromat<strong>in</strong> modification factors (p<strong>in</strong>k boxes) that control the balance betweensignal<strong>in</strong>g molecules and transcription factors, to direct stem cell differentiation.


FIGURE 25.4Example of <strong>in</strong>teractions between a pluripotency factor, Myc, and the ncRNA/epigenetics network. See p. 511 for details.


FIGURE 26.1Schematic representation of the DNA methylation reactions: ma<strong>in</strong>tenance methylation, passive demethylation, de novo methylation, and active demethylation.See p. 525 for details.


FIGURE 26.2Many environmental factors can <strong>in</strong>terferewith the organism, <strong>in</strong>duc<strong>in</strong>g epigeneticmodifications. See p. 531 for details.FIGURE 26.3Schematic representation of the most relevantreactions comprises of the “one-carbonmetabolism”, emphasiz<strong>in</strong>g a vitam<strong>in</strong> cofactorscycle (remethylation) and SAM-dependent DNAmethylation (transmethylation). See p. 534 fordetails.


This page <strong>in</strong>tentionally left blank

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

Saved successfully!

Ooh no, something went wrong!