Discover millions of ebooks, audiobooks, and so much more with a free trial

Only $11.99/month after trial. Cancel anytime.

The Epigenome and Developmental Origins of Health and Disease
The Epigenome and Developmental Origins of Health and Disease
The Epigenome and Developmental Origins of Health and Disease
Ebook1,523 pages28 hours

The Epigenome and Developmental Origins of Health and Disease

Rating: 0 out of 5 stars

()

Read preview

About this ebook

Winner of 2016 BMA Medical Award for Basic and Clinical Sciences

The Epigenome and Developmental Origins of Health and Disease synthesizes the existing knowledge on how the in utero environment could be the most important environment in shaping later risk for various diseases or to conversely promote the health of the offspring.

The book mines the existing literature from a variety of disciplines from toxicology to nutrition to epigenetics to reveal how contrasting maternal in utero environmental changes might be leading to epigenetic convergence and the resulting deleterious phenotypic and physiological effects in our offspring.

It is increasingly becoming apparent that even subtle changes in the mother’s diet, stress, and exposure to low concentrations of toxic chemicals at levels deemed safe by the EPA and FDA, such as endocrine disrupting compounds (EDC), can dramatically impact the health of our children, possibly leading to metabolic, cardiovascular, immunological, neurobehavioral disorders, and increased risk for cancer to list but a few examples.

  • Informs how everyday choices pregnant women make can impact child development
  • Ties together how in utero environmental changes may be inducing epigenetic changes in the offspring leading to overlapping phenotypes regardless of the initial insult (toxic, nutrition, or stress)
  • Includes a boxed-in area in each chapter for further references and resources to keep up with the field
  • Features video interviews with the authors and other key leaders in the field
LanguageEnglish
Release dateOct 10, 2015
ISBN9780128016725
The Epigenome and Developmental Origins of Health and Disease

Related to The Epigenome and Developmental Origins of Health and Disease

Related ebooks

Medical For You

View More

Related articles

Related categories

Reviews for The Epigenome and Developmental Origins of Health and Disease

Rating: 0 out of 5 stars
0 ratings

0 ratings0 reviews

What did you think?

Tap to rate

Review must be at least 10 words

    Book preview

    The Epigenome and Developmental Origins of Health and Disease - Cheryl S. Rosenfeld

    The Epigenome and Developmental Origins of Health and Disease

    Editor

    Cheryl S. Rosenfeld

    Table of Contents

    Cover image

    Title page

    Copyright

    Dedication

    List of Contributors

    Acknowledgments

    Chapter 1. The Developmental Origins of Health and Disease (DOHaD) Concept: Past, Present, and Future

    Introduction

    The Origins of the Field

    Conceptual Developments and Experimental Observations

    DOHaD and Epigenetics

    The Wider DOHaD Research Agenda

    DOHaD and Public Policy

    Chapter 2. Historical Perspective of Developmental Origins of Health and Disease in Humans

    Introduction

    Socioeconomic Stressors and Infant and Child Health

    Allostasis and Allostatic Load

    Weathering and African American Health Disparities

    Acculturation and the Hispanic Paradox

    A Life Course Approach and the Developmental Origins of Health and Disease

    Epigenetic Regulation and Effects of Psychosocial Stress

    Health Vulnerabilities Can be Heritable

    Conclusions

    Chapter 3. DOHaD and the Periconceptional Period, a Critical Window in Time

    Introduction: DOHaD and the Periconceptional Perspective

    The Periconceptional Period: A Summary of Key Events

    In Vivo Maternal Nutritional Models of Periconceptional Programming

    In Vitro Models of Periconceptional Programming Related to Assisted Reproductive Treatments

    The Paternal Influence on Periconceptional Developmental Programming

    Conclusions

    Chapter 4. Introduction to Epigenetic Mechanisms: The Probable Common Thread for Various Developmental Origins of Health and Diseases Effects

    Introduction

    DNA Methylation

    Posttranslational Histone Modifications

    Noncoding RNA

    Nucleosome Remodeling

    Embryonic Epigenetic Reprogramming

    Primordial Germ Cell Reprogramming

    Conclusion

    Chapter 5. Perinatal Neurohormonal Programming and Endocrine Disruption

    Introduction

    Effects of Sex Steroid Hormones on Brain Sexual Differentiation

    Sex Steroid-induced Epigenetic Regulation of Brain Sexual Differentiation

    Glucocorticoid and Maternal Stress Effects on Brain Development and Sexual Differentiation

    Endocrine Disruption of Normal Brain Programming

    Conclusions

    Glossary

    Chapter 6. Parental Nutrition and Developmental Origins of Health and Disease

    Introduction

    Overnutrition

    Undernutrition

    Epigenetics and Prenatal Programming by Malnutrition

    Conclusions

    Chapter 7. Maternal Prenatal Stress and the Developmental Origins of Mental Health: The Role of Epigenetics

    Introduction

    Epigenetics

    Conclusions

    Chapter 8. Epigenetics in the Developmental Origin of Cardiovascular Disorders

    Introduction

    Periods of Susceptibility to Developmental Programming

    Programming Stimuli

    Mechanisms of DOHaD in CVD

    Concluding Remarks

    Chapter 9. Developmental Effects of Endocrine-Disrupting Chemicals in the Ovary and on Female Fertility

    Introduction

    Ovarian Development and Function

    Effects of Endocrine-Disrupting Chemicals on Female Reproduction and Ovary

    Conclusions

    Chapter 10. Developmental and Epigenetic Origins of Male Reproductive Pathologies

    Introduction

    Environmental Factors Known to Correlate with Male Reproductive Dysfunction

    Developmental Environment and Establishment of Epigenetic Patterns

    Developmental Exposures and the Incidence of Male Reproductive Abnormalities

    Environmentally Induced Transgenerational Epigenetic Effects Related to Male Reproduction

    Germ Line to Somatic Epigenetic Effects: Lesson from the Vinclozolin Transgenerational Model

    Epigenetic Marks in Fertility: DNA Methylation and Histone Modifications

    Epigenetic Marks and Fertility: Small Noncoding RNAs

    Conclusions and Perspectives

    Chapter 11. Developmental Origins of Childhood Asthma and Allergic Conditions—Is There Evidence of Epigenetic Regulation?

    Introduction

    Conclusions

    Chapter 12. Immune Disorders, Epigenetics, and the Developmental Origins of Health and Disease

    Introduction

    Environmental Risk Factors Affecting Immune Function and Individual Vulnerability

    The Immune System, Developmental Origins of Adult Health and Disease, and Public Health Implications

    Immune Programming by the Microbiota

    Interlinkage of Immune Disorders

    Immune Disorders and Communicable Diseases

    Immune Disorders and Noncommunicable Diseases

    Autoimmune Disorders

    Inflammatory Disorders

    Conclusions

    Chapter 13. Neurobehavioral Disorders and Developmental Origins of Health and Disease

    Introduction

    The Risk Associated with Early-Life Insults

    Changes in the Stress System during Pregnancy

    Organogenesis

    Timing of Intrauterine Exposures

    Emotional Regulation

    Cognitive Regulation

    Epidemiological Evidence for Programming Neurobehavioral Outcomes

    Programming the Vulnerable Human Fetal Brain

    Birth Phenotype and Neuronal Consequences

    Neuronal Consequences of Intrauterine Exposures

    Sex Differences and the DOHaD Model

    Conclusions

    Chapter 14. Metabolic Disorders and Developmental Origins of Health and Disease

    Introduction

    Intrauterine Growth Restriction

    Gestational Diabetes in Pregnancy and Metabolic Programming of the Offspring

    Obesity in Pregnancy and Metabolic Programming of the Offspring

    Conclusion

    Chapter 15. The Developmental Origins of Renal Dysfunction

    Introduction

    Impact of the Prenatal Environment on Renal Dysfunction and Chronic Kidney Disease

    Kidney Development and Nephron Endowment

    The Importance of Nephron Endowment: Links to Blood Pressure and Renal Disease

    Exploring the Mechanisms of Renal Programming of Low Nephron Endowment: Evidence from Animal Models

    Why is the Kidney Susceptible to Programming?

    Mechanisms Leading to Impaired Renal Structure and Function

    Conclusions

    Chapter 16. Cancer and Developmental Origins of Health and Disease—Epigenetic Reprogramming as a Mediator

    Introduction

    Epigenetic Regulation in Cancer

    Developmental Origin of Adult Disease

    Developmental Origin of Adult Disease and Breast Cancer

    Developmental Origin of Adult Disease and Prostate Cancer

    Other Cancers

    Epigenetically Active Agents Involved in Carcinogenesis

    Exogenous Factors

    Endogenous Factors

    Conclusions

    Glossary Terms, Acronyms, and Abbreviations

    Chapter 17. Epigenetic Regulation of Gastrointestinal Epithelial Barrier and Developmental Origins of Health and Disease

    Introduction

    Epigenetics and GI Tract Development and Maturation

    The Gut Microbiota and Programming

    Developmental Origins of GI Diseases

    Food and Epigenetics of the GI Epithelium

    Conclusions and Future Directions

    Abbreviations

    Chapter 18. How the Father Might Epigenetically Program the Risk for Developmental Origins of Health and Disease Effects in His Offspring

    Introduction

    Sperm Epigenetics

    Zygotic Epigenetic Reprogramming

    Paternal Aging

    Paternally Driven Environmental Effects

    Transgenerational Epigenetic Inheritance

    Conclusions

    Chapter 19. Linkage between In Utero Environmental Changes and Preterm Birth

    Introduction

    Environmental Exposures

    Chemicals

    Particles/Air Pollution

    Maternal Health

    Conclusions

    Glossary Terms

    Chapter 20. Sexual Dimorphism and DOHaD through the Lens of Epigenetics: Genetic, Ancestral, Developmental, and Environmental Origins from Previous to the Next Generation(s)

    Introduction

    Complex Trajectories due to Sex Specificity to Both the Transmission and Inheritance of Susceptibility

    Mechanisms of Unequal Expression of X- and Y-Chromosome-Linked Genes

    Epigenetics and Gene Expression: Sex- Specific Marks, Mechanisms, and Dynamics

    The Specific Epigenetic Features of Extraembryonic Tissues and Placenta

    Differences between Male/Female Gametogenesis

    Differences in Reprogramming of Maternal/Paternal Genome

    What Levels of Evidence: The Limiting and Confounders Factors

    Conclusions

    Chapter 21. Transgenerational Epigenetic Inheritance: Past Exposures, Future Diseases

    Introduction

    A Crucial Role for the Germline in Epigenetic and Genetic Inheritance

    Evidence for Transgenerational Epigenetic Inheritance in Animal Models

    Evidence for Transgenerational Epigenetic Inheritance in Humans

    Environmental Exposures Affecting the Male Germline: Adult versus Developmental Exposures

    Conclusions and Perspectives

    Chapter 22. The Placenta and Developmental Origins of Health and Disease

    Introduction

    Epidemiology and the Placenta

    Placental Function

    Environmental Influences on Placental Function

    Epigenetics and the Placenta

    The Placental Phenotype, Interventions, and Treatments

    Concluding Remarks and Future Directions

    Chapter 23. The Moral and Legal Relevance of DOHaD Effects for Pregnant Mothers

    Introduction: Developmental Origins of Health and Disease Research and Legislative Interventions

    The Legal Recognition of Fetal Rights

    The Criminalization of the Bad Mother

    Caring for Fetuses versus Caring for Future Persons

    Beneficence, Nonmaleficence, and Future People

    DOHaD Effects Are Not Deterministic

    DOHaD and the Nonidentity Problem

    Do Women Have a Duty to Abort in Order to Avoid Causing Developmental Origins of Health and Disease Harm to Future Persons?

    Is the Criminalization of Mothers Justifiable?

    Conclusions

    Chapter 24. Introduction to Moms in Motion (MIM)

    Abbreviations

    Chapter 25. Reversing Harmful Developmental Origins of Health and Disease Effects

    Introduction

    Method

    Discussion

    Abbreviations

    Chapter 26. Informational Resources for Developmental Origins of Health and Disease Research

    Introduction

    DOHaD-Related Scientific Organizations

    DOHaD-Related Scientific Meetings

    DOHaD-Related Journals, Databases, and Other Informative Resources

    Social Media Resources Reporting Developmental Origins of Health and Disease Findings

    Judging the Reliability of Developmental Origins of Health and Disease Informational Resources

    Conclusions

    Glossary

    Index

    Copyright

    Academic Press is an imprint of Elsevier

    125 London Wall, London EC2Y 5AS, UK

    525 B Street, Suite 1800, San Diego, CA 92101-4495, USA

    225 Wyman Street, Waltham, MA 02451, USA

    The Boulevard, Langford Lane, Kidlington, Oxford OX5 1GB, UK

    Copyright © 2016 Elsevier Inc. All rights reserved.

    No part of this publication may be reproduced or transmitted in any form or by any means, electronic or mechanical, including photocopying, recording, or any information storage and retrieval system, without permission in writing from the publisher. Details on how to seek permission, further information about the Publisher’s permissions policies and our arrangements with organizations such as the Copyright Clearance Center and the Copyright Licensing Agency, can be found at our website: www.elsevier.com/permissions.

    This book and the individual contributions contained in it are protected under copyright by the Publisher (other than as may be noted herein).

    Notices

    Knowledge and best practice in this field are constantly changing. As new research and experience broaden our understanding, changes in research methods, professional practices, or medical treatment may become necessary.

    Practitioners and researchers must always rely on their own experience and knowledge in evaluating and using any information, methods, compounds, or experiments described herein. In using such information or methods they should be mindful of their own safety and the safety of others, including parties for whom they have a professional responsibility.

    To the fullest extent of the law, neither the Publisher nor the authors, contributors, or editors, assume any liability for any injury and/or damage to persons or property as a matter of products liability, negligence or otherwise, or from any use or operation of any methods, products, instructions, or ideas contained in the material herein.

    ISBN: 978-0-12-801383-0

    British Library Cataloguing-in-Publication Data

    A catalogue record for this book is available from the British Library

    Library of Congress Cataloging-in-Publication Data

    A catalog record for this book is available from the Library of Congress

    For information on all Academic Press publications visit our website at http://store.elsevier.com/

    Typeset by TNQ Books and Journals

    www.tnq.co.in

    Printed and bound in the United States of America

    Dedication

    I dedicate this book to my father, Robert L. Rosenfeld, who passed away on February 5, 2005. From early childhood onwards, he encouraged me to pursue my interests in science and medicine and taught me that a good start and supportive environment can last a lifetime.

    List of Contributors

    Roger Brown,     School of Nursing, University of Wisconsin-Madison, Madison, WI, USA

    Tatjana Buklijas,     Liggins Institute, The University of Auckland, Auckland, New Zealand

    Douglas T. Carrell,     Department of Surgery (Urology), University of Utah School of Medicine, Salt Lake City, UT, USA

    Mei-Wei Chang,     Michigan State University College of Nursing, East Lansing, MI, USA

    Ana Cheong

    Department of Environmental Health,University of Cincinnati College of Medicine,Cincinnati,OH,USA

    Center for Environmental Genetics,University of Cincinnati Medical Center,Cincinnati,OH,USA

    Quetzal A. Class,     Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA

    Jane K. Cleal,     Institute of Developmental Sciences, University of Southampton, Southampton, UK

    James S.M. Cuffe,     School of Biomedical Science, The University of Queensland, St Lucia, QLD, Australia

    Elysia Poggi Davis

    Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA

    Department of Psychology, University of Denver, Denver, CO, USA

    Rodney R. Dietert,     Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA

    M Jean Brancheau Egan,     Michigan Department of Community Health, WIC Division, Lansing, MI, USA

    Kobra Eghtedary,     Michigan Department of Community Health, WIC Division, Lansing, MI, USA

    Tom P. Fleming,     Centre for Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK

    Sara Fneich,     INRA, UMR1198 Biologie du Développement et Reproduction, Jouy-en-Josas, France

    Anne Gabory,     INRA, UMR1198 Biologie du Développement et Reproduction, Jouy-en-Josas, France

    Jeffrey S. Gilbert,     Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN, USA

    Vivette Glover,     Institute of Reproductive and Development Biology, Imperial College London, London, UK

    Peter D. Gluckman,     Liggins Institute, The University of Auckland, Auckland, New Zealand

    Laura M. Glynn

    Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA

    Department of Psychology, Chapman University, Orange, CA, USA

    Amrie C. Grammer,     University of Virginia Research Park, VA, USA

    Carlos Guerrero-Bosagna,     Avian Behavioral Genomics and Physiology Group, IFM Biology, Linköping University, Linköping, Sweden

    Mark A. Hanson,     Institute of Developmental Sciences, University of Southampton and NIHR Nutrition Biomedical Research Centre, University Hospital Southampton, Southampton, UK

    Shuk-Mei Ho

    Department of Environmental Health,University of Cincinnati College of Medicine,Cincinnati,OH,USA

    Center for Environmental Genetics,University of Cincinnati Medical Center,Cincinnati,OH, USA

    Cincinnati Cancer Center,Cincinnati,OH,USA

    Cincinnati Veteran Affairs Medical Center, Cincinnati,OH,USA

    Vinothini Janakiram

    Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH, USA

    Center for Environmental Genetics, University of Cincinnati Medical Center, Cincinnati, OH, USA

    Timothy G. Jenkins,     Department of Surgery (Urology), University of Utah School of Medicine, Salt Lake City, UT, USA

    Claudine Junien,     INRA, UMR1198 Biologie du Développement et Reproduction, Jouy-en-Josas, France

    J.P. Lallès

    Institut National de la Recherche Agronomique, UR1341 ADNC, Saint Gilles, France

    Centre de Recherche en Nutrition Humaine-Ouest, Nantes, France

    Yuet-Kin Leung

    Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH, USA

    Center for Environmental Genetics, University of Cincinnati Medical Center, Cincinnati, OH, USA

    Cincinnati Cancer Center, Cincinnati, OH, USA

    Rohan M. Lewis,     Institute of Developmental Sciences, University of Southampton, Southampton, UK

    Michele Loi,     Centro de Estudos Humanísticos, Universidade do Minho, Campus de Gualtar, Braga, Portugal

    C. Michel

    Centre de Recherche en Nutrition Humaine-Ouest, Nantes, France

    Institut National de la Recherche Agronomique/Université de Nantes, UMR1280, Nantes, France

    Institut des Maladies de l’Appareil Digestif, Nantes, France

    Karen M. Moritz,     School of Biomedical Science, The University of Queensland, St Lucia, QLD, Australia

    Kristin E. Murphy,     Department of Surgery (Urology), University of Utah School of Medicine, Salt Lake City, UT, USA

    Susan Nitzke,     Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA

    Marianna Nobile,     Universita’ degli Studi di Milano-Bicocca, Dipartimento dei Sistemi Giuridici, Milano, Italy

    Kieran J. O’Donnell,     The Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental University Institute, McGill University, Montreal, QC, Canada

    Polina Panchenko,     INRA, UMR1198 Biologie du Développement et Reproduction, Jouy-en-Josas, France

    Sara E. Pinney,     Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Division of Endocrinology and Diabetes, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA

    Ken Resnicow,     University of Michigan School of Public Health, University of Michigan, Ann Arbor, MI, USA

    Lynette K. Rogers,     Center for Perinatal Research,The Research Institute at Nationwide Children’s Hospital,Columbus,Ohio

    Cheryl S. Rosenfeld,     Department of Bond Life Sciences Center, Department of Biomedical Sciences, Genetics Area Program, Thompson Center for Autism and Neurobehavioral Disorders, University of Missouri, Columbia, MO, USA

    Lewis P. Rubin,     Department of Pediatrics, Texas Tech University Health Sciences Center El Paso, Paul L. Foster School of Medicine, El Paso, TX, USA

    Curt A. Sandman,     Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA

    J.P. Segain

    Centre de Recherche en Nutrition Humaine-Ouest, Nantes, France

    Institut National de la Recherche Agronomique/Université de Nantes, UMR1280, Nantes, France

    Institut des Maladies de l’Appareil Digestif, Nantes, France

    Congshan Sun,     Centre for Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK

    Martha Susiarjo,     Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA

    Pheruza Tarapore

    Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH, USA

    Center for Environmental Genetics, University of Cincinnati Medical Center, Cincinnati, OH, USA

    Cincinnati Cancer Center, Cincinnati, OH, USA

    V. Theodorou,     Institut National de la Recherche Agronomique, UMR Toxalim, Toulouse, France

    Sarah To,     Department of Environmental Health,University of Cincinnati College of Medicine,Cincinnati,OH,USA

    Steve Turner,     Child Health, Royal Aberdeen Children’s Hospital, Aberdeen, UK

    Mehmet Uzumcu,     Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA

    Miguel A. Velazquez,     Centre for Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK

    Markus Velten,     Department of Anesthesiology and Intensive Care Medicine,Rheinische Friedrich-Wilhelms-University Bonn,Germany

    Sarah Voisin,     INRA, UMR1198 Biologie du Développement et Reproduction, Jouy-en-Josas, France

    Sarah L. Walton,     School of Biomedical Science, The University of Queensland, St Lucia, QLD, Australia

    Aparna Mahakali Zama,     Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA

    Acknowledgments

    This book would not have been possible without all of the coauthors who kindly shared their knowledge and passion for the various areas. Ms Lisa Eppich and Ms Catherine A. Van Der Laan at Elsevier were integral in making this book come to life.

    The concept of developmental origins of health and disease (DOHaD) was first clearly articulated by the late Sir David Barker. It was thus appropriate that it was originally termed the Barker hypothesis but subsequently changed to fetal origin of adult disease (FOAD), and most recently to developmental origins of adult health and disease. Since its conception, the DOHaD concept has gained currency and led to paradigm shifts in how scientists and clinicians view a variety of noncommunicable diseases. Correspondingly, it has paved the way for new avenues of diagnosis, prevention, and treatment strategies.

    I am grateful to the many teachers, mentors, colleagues, and friends who along the way fostered my interests and curiosity in science and medicine. In particular, I am grateful to the late Mrs Patricia Murphy whose enthusiasm and wonderment for biology were contagious. My PhD mentor, Dr Dennis Lubahn, was incredibly supportive of me and my research ideas. The lessons I learned in his laboratory have stayed with me all of these years. Most of all, I am thankful to Dr R. Michael Roberts. For over 20  years, he has been a wonderful mentor, colleague, and friend.

    I am thankful to Dr Deborah Wagner, my friend and former classmate, for allowing me over various holidays to serve as a relief veterinarian at her animal hospital. As veterinary students, we made a Forrest Gump pact that if she were to ever open her animal hospital, I would be her first mate. It has been rewarding to be able to indulge my veterinary interests and keep in touch with advances in clinical medicine. These experiences have helped shape my thinking and research directions.

    Finally, I am grateful to my mother, sister, brother, nieces and nephews, and other family members who walk on two and four legs.

    Cheryl S. Rosenfeld

    Chapter 1

    The Developmental Origins of Health and Disease (DOHaD) Concept

    Past, Present, and Future

    Peter D. Gluckman¹, Tatjana Buklijas¹,  and Mark A. Hanson²     ¹Liggins Institute, The University of Auckland, Auckland, New Zealand     ²Institute of Developmental Sciences, University of Southampton and NIHR Nutrition Biomedical Research Centre, University Hospital Southampton, Southampton, UK

    Abstract

    The developmental origins of health and disease (DOHaD) is a concept that has emerged over the past 50  years, linking the state of health and risk from disease in later childhood and adult life with the environmental conditions of the early life. Originally based on epidemiologic observations, the concept has given rise to a field that brings together clinical studies in a range of specialties, public and global health, experimental physiology, molecular biology (especially epigenetics), developmental biology, anthropology, the social sciences, and evolutionary biology. This chapter examines the evolution of thinking about the relationship between developmental influences and later-life health and disease; examines the establishment of DOHaD as a conceptual framework and a research field in its own right; discusses criticisms of DOHaD and barriers to its acceptance within the broader research community as well as to its recent integration into public health policy; and, finally, considers future directions that the field may take.

    Keywords

    Development; Developmental origins of health and disease; Disease; Epigenetics; Fetal physiology; History; Life course

    Outline

    Introduction 1

    The Origins of the Field 2

    Conceptual Developments and Experimental Observations 5

    DOHaD and Epigenetics 8

    The Wider DOHaD Research Agenda 9

    DOHaD and Public Policy 10

    References 11

    Introduction

    The overarching argument of the conceptual paradigm and the research field of developmental origins of health and disease (DOHaD) is that the state of health and risk from disease in later childhood and adult life is significantly affected by environmental factors acting during the preconceptional, prenatal, and/or early postnatal periods. The emphasis has been on obesity, type 2 diabetes mellitus, and cardiovascular disease, but a significant body of work has also been focused on endocrine cancers, osteoporosis and frailty in the elderly, mental health, cognitive function, respiratory disease, immune function, and allergy. While the field as currently constructed is just over two decades old, it is based on research that goes back to the 1930s. In this chapter, we bring together research traditions, concepts, and approaches that, over the last 80  years, have explored the question of prenatal and early postnatal environmental influences that impact health and disease in later life and look forward to emergent areas of attention and application of the concept.

    The Origins of the Field

    The idea that experiences in early life influence health in later life may be found throughout the history of Western medicine: well into the 1800s, it was believed that anything that a mother saw, touched, ate, or even imagined—collectively known as maternal impressions—had a capacity to permanently influence the developing organism [1,2]. In the early 1800s, the common view was that a new organism was created in a process called generation, out of maternal and paternal contributions as well as various experiences that the mother had during (and even before) pregnancy [3]. But in the nineteenth century, generation was replaced with reproduction, built on the new idea of heredity. The noun heredity was first used in the 1830s, to describe the transmission of parental qualities during conception, and at the same time to make a distinction between those inherited qualities and the properties that emerged during development [4]. Scientists studied where heredity resided within the cell; how hereditary particles were distributed among cells and their quantity was prevented from doubling in each successive generation; and to what extent were hereditary elements in the cells sensitive to developmental influences [5]. August Weismann’s work provided the conceptual basis for new thinking about heredity and development: while germ cells produced somatic cells, he argued, they were not affected by anything that somatic cells acquired or learned [6]. It followed that each individual was born with a certain predisposition toward disease; no environmental modifications could improve one’s outlook. The best one could do to improve one’s offspring’s chances was to reproduce with a person of better heredity.

    The early 1900s were the heyday of hard heredity, exemplified by the emergence of genetics, an experimental discipline concerned with mechanisms of heredity, and the dominance of the social program of eugenics, seeking to reform society through rationalizing human reproduction [7]. But the deep economic crisis of the 1930s made it obvious that environmental conditions had a strong influence on the emergence and prevalence of disease [8]. New epidemiological work suggested that the conditions of early life played a role at least as important as heredity. A landmark paper by the Scottish epidemiologist William Ogilvy Kermack and colleagues in 1934 argued that the data behaved as if the expectation of life was determined by the conditions existing during the years 0–15 (…) the health of the man is determined preponderantly by the physical constitution which the child has built up [9]. The recognition of the importance of environmental conditions and the apparent demise of eugenics did not, however, mean the fall of the genetic model, which remained dominant through the twentieth century [8].

    The Second World War was a pivotal event in the making of the developmental approach to the study of health and disease, conceptualized in the early twenty-first century as DOHaD. Even before the war, physiologists, teratologists, and agricultural scientists collected experimental evidence showing that manipulating the life conditions of pregnant animals permanently affected the patterns of growth and the phenotype of their offspring [10–12]. Interventions in humans were, for obvious reasons, too subtle to produce substantial differences, and they also focused on maternal mortality and morbidity rather than longer-term outcomes in the offspring [13]. But wartime famines provided rare natural experiments by exposing thousands of women to periods of severe undernutrition, in some cases sharply delineated [14,15]. The longest and the most severe famine took place in Leningrad under German siege (between September 1941 and January 1944) [14], but the clearest data came from Rotterdam and The Hague, two cities in northwestern Holland that had suffered food shortages during German reprisals from September 1944 to May 1945, in what became known as the Dutch Winter Famine [15,16]. Data collected showed that starvation in the last trimester of pregnancy caused a reduction in the birth weight of the offspring, while famine around conception increased the chance of miscarriage and malformation. Postwar Germany provided an opportunity for the British team working on the intersection of physiology, nutritional science, and pediatrics (Robert McCance, Elsie Widdowson, and Rex Dean) to study how low food rations and lack of food variety influenced lactation in new mothers, infant birth weight, and childhood growth [17]. Back in their Cambridge laboratories, Widdowson and McCance tested their clinical findings in animal experiments and demonstrated that the size of the litter and the rate of offspring growth depended on maternal nutrition. Interestingly, prenatal and early postnatal (preweaning) nutrition did not affect just the weight that the pups attained by adulthood: it also influenced susceptibility to infections, body proportions, and timing of reproductive maturation, as well as behavior [18]. Their results supported the theory of critical or sensitive periods, popular across disciplines as diverse as ethology (behavioral studies), linguistics, child psychology, and physiology, according to which each organ or tissue has a distinctive period of critical differentiation as well as a period of maximum growth, during which these organs and tissues are highly sensitive to injury [11].

    But, as the world recovered from the wartime trauma, in the 1960s and 1970s interest in the relationship between prenatal and perinatal influences and later health and disease waned. In this period, fetal physiologists largely focused on questions that emphasized fetal autonomy rather than interplay between the environment and the developing organism. They studied, for example, fetal respiratory movements, fetal endocrine growth mechanisms, or fetal control of the onset of labor [19]. It was mostly researchers with a strong interest in socioeconomic determinants of health inequalities who pursued questions of the interaction between environment and development. At the University of Birmingham’s Department of Social Medicine, under Professor Thomas McKeown, a young David Barker completed his PhD thesis on Prenatal influences and subnormal intelligence (1966) [20]. He found that children of all levels of subnormal intelligence (classified as IQ under 75) had a birth weight lower than expected. Interestingly, the normal siblings of all but the most severely subnormal children (IQ less than 50) were born at low birth weight too. These low birth weights of both normal and subnormal children, Barker suggested, reflected influences which affect the intra-uterine lives of all children in their families.

    At the same time, a pair of South African political immigrants to the United States (via the University of Manchester’s Department of Social Medicine), the Columbia University epidemiologists Zena Stein and Mervyn Susser, undertook a large program of study of the influence of maternal nutrition on mental competence [21,22]. One part of their research was an intervention study of providing food supplements to pregnant women drawn from a population with a high frequency of low birth weight; the other, an observational study based on the Dutch Winter Famine cohort. While both of these studies produced negative results, they rekindled interest in the Dutch Winter Famine cohort, which from then onwards would play a key role in the study of developmental, as well as transgenerational, influences upon adult health.

    The first study (by Stein, Susser, and the Amsterdam researcher Anita Ravelli) to associate undernutrition during gestation in the Dutch Winter Famine cohort with manifestations of metabolic disease was published in 1976, showing that young men exposed in early pregnancy had significantly higher rates of obesity, while men exposed in late pregnancy (and first months of infancy) had lower obesity rates [23]. Women who suffered famine during first and second trimester in utero had offspring whose birth weight was lower than the birth weight of offspring born to women who had not been exposed to famine as fetuses; but the offspring of women exposed during the third trimester showed no reduction in birth weight [24]. Overall, from the mid-1970s onwards multiple historical and prospective epidemiological studies studied the relationship between maternal morbidity, infant mortality and birth weight, and morbidity and mortality from cardiovascular disease [25–28]. In North America, Millicent Higgins was the first to employ a long-term formal birth cohort to examine later-life outcomes, reporting in 1980 that the male offspring of women with toxemia developed significantly higher blood pressures by 20  years of age. By the mid-1980s, reports linking low birth weight and later hypertension started to merge [27,28].

    Innovative conceptual developments took place in Eastern Germany, where the Berlin endocrinologist Günther Dörner studied differences in phenotypes between men born before, during, and after the Second World War and showed association between nutrition and later prevalence of metabolic and cardiovascular disease [29–31]. He became particularly interested in the effects of maternal stress and gestational diabetes on the offspring [32,33]. Dörner argued that the concentrations of hormones, neurotransmitters, and metabolites during the early development preprogrammed feedback loops control reproduction and development [34,35]. Inappropriately set feedback loops could then trigger disease. Dörner’s "functional teratology," as he termed it, may be compared to the proposal by another endocrinologist in this period, Norbert Freinkel, who spoke of metabolic teratogenesis when describing the effect of gestational diabetes on the next generation [36]. Around the same time, Frans Van Assche and colleagues used experimental models of gestational diabetes (in which part of the pancreatic β cells were chemically destroyed) to show that prenatal diabetogenic conditions permanently affected the adult offspring’s ability to handle situations stressing their glucose metabolism, such as pregnancy, and also had similar transgenerational effects [37].

    It was, however, the work of David Barker in the 1980s, by then at the University of Southampton’s MRC Environmental Epidemiology Unit, that prompted this diverse collection of observations to coalesce into a field. In 1985, Barker’s unit produced a compendium of maps that used color gradation to show differences in mortality from selected diseases across the counties of England and Wales [38]. The mortality from cardiovascular disease appeared highest in poor and lowest in rich areas, a finding that opposed the view established by the Framingham Heart Study, according to which affluence caused the modern epidemic of heart diseases [39]. Barker, who since his Birmingham days with McKeown had been interested in prenatal influences especially nutrition, observed a strong geographical relationship between the areas of high infant mortality in the early twentieth century and the areas of high cardiovascular mortality between 1968 and 1978 [40]. Barker’s best-known study used the largest and most detailed set of records on infant welfare that could be found in the United Kingdom, those from the county of Hertfordshire from 1911 onwards. These records contained information about birth weight and early growth and development, and became the basis of a large study cohort. The follow-up of the Hertfordshire infants showed that death rates for cardiovascular disease fell progressively from those weighing 2.5  kg or less at birth to those weighing 4.3  kg, with a slight increase in the heaviest group (above 4.3  kg) [41]. Blood pressure and cholesterol levels showed equivalent trends.

    Barker’s work attracted wide interest. The Southampton team began collaboration with the Amsterdam group studying the Dutch Winter Famine cohort. Other studies, both retrospective and prospective, followed worldwide. Early on, Barker’s work came to the attention of the doyen of fetal physiology Geoffrey Dawes, who was near retirement [42]. In Dawes’s view, Barker’s findings opened up new research opportunities. Dawes introduced Barker to his colleagues working in fetal physiology at a workshop meeting near La Spezia, Italy, in 1989. The proceedings, published under the title Fetal Autonomy and Adaptation, make interesting reading: the physiologists were clearly skeptical about Barker’s observations, but resolved to test them experimentally. The first workshop on fetal origins of adult disease took place in Sydney in 1994, and it brought a broader group of perinatal scientists in contact with Barker’s group [43]. This meeting launched a fast-growing series of annual workshops. Under David Barker’s leadership, foremost investigators of that period created the Council for the Fetal Origins of Adult Disease (FOAD) with John Challis as its first chair. The first global congress on FOAD was held in Mumbai, India, in 2001. At the second congress, in Brighton, UK, in 2003, it was decided the council would be reformed into an academic society, Developmental Origins of Health and Disease (DOHaD), with Peter Gluckman as the founding president and Mark Hanson as secretary.

    Conceptual Developments and Experimental Observations

    A major conceptual development in the DOHaD field took place in 1992 when David Barker and Nicholas Hales proposed their thrifty phenotype hypothesis [44]. Thrifty phenotype was a developmental alternative to the thrifty genotype explanation of the modern epidemic of noncommunicable diseases (NCDs), proposed in the 1960s by a medical geneticist James Neel [45,46] interested in the evolution of contemporary human populations. Neel argued that in the past thrifty genes had been selected because they provided advantage in the time of famine; but in the affluent contemporary world they only increased disease risk. The thrifty phenotype, by contrast, placed an emphasis on development, arguing that nutritionally inadequate conditions in pregnancy not only affected fetal growth but also induced permanent changes in insulin secretory capacity and in glucose metabolism. While well adapted for famine, in a nutritionally rich postnatal environment the individual would have a higher risk of metabolic disease.

    The initial focus of the field—in David Barker’s work and in studies of the Dutch Winter Famine cohort, as well as, earlier, in postwar undernutrition studies—was on the consequences of low birth weight. For this reason, experimental studies induced intrauterine growth restriction (IUGR) in animal models, while clinical studies compared the outcomes of normal-weight and IUGR offspring [47–50]. The centrality of low birth weight to Barker’s hypothesis, however, became a problem. First, the supposed link between low birth weight and adult NCD contradicted the observed ecological trends: post–Second World War, the incidence of NCDs increased precisely in those countries that also had high average birth weights, such as Norway, Finland, and Scotland [51]. Second, disciplinary divisions hindered recognition of this problem. Most early epidemiological work studied processes associated with suboptimal nutritional conditions or excessive maternal stress and later disease risk; at the same time, the class of phenomena highly relevant for the modern, developed world, associated with gestational diabetes, maternal obesity, and infant overfeeding, was being studied by endocrinologists and obstetricians. The relative importance of these pathways clearly differed in different populations and at different stages of the nutritional transition. Yet the two disciplinary groups, epidemiologists and clinicians, presented their results at different venues and rarely talked to each other. Third, many epidemiological studies produced data inconsistent with or even contrary to Barker’s hypothesis [52]. Some even argued that the observed differences did not exist, i.e., that they were results of errors, confounding, and inappropriate study design [53]. Even the studies of historical famines did not at all support the thrifty phenotype: for instance, studies on survivors of the Leningrad siege found no difference in glucose tolerance, lipid concentrations, hypertension, or cardiovascular disease rates between those groups exposed and those who were not exposed to famine during development [54]. A Finnish historical study found no effect of famine exposure upon survival in adulthood [55]. Even studies based on the Dutch Winter Famine cohorts produced results that contradicted previous findings and the main hypothesis: for example, that glucose tolerance was lower in participants exposed in mid- to late gestation rather than early to mid-gestation [56,57]. Critics maintained that Barker’s hypothesis was neither precisely formulated nor consistent, with regard to the timing of critical events at the stage of gestation when undernutrition might have been expected to produce the specific impact [58]. They emphasized the need to move from epidemiological to mechanistic studies, because retrospective studies rarely provided sufficiently detailed data, prospective studies took too long, and neither could explain how, for example, a modification in nutrition in mid-gestation may lead to a change in blood pressure decades later [57].

    Rather than undermining the field, these challenges to the DOHaD concept encouraged further research and inspired conceptual thinking. Researchers recognized that there was indeed an excessive emphasis on birth weight, while in reality early life events informed later disease outcomes in multiple ways, depending on the type and timing of the insult [59]. Birth weight was only one of many possible proxies for variable intrauterine effects [60]. Programming (a term introduced by Alan Lucas [61], a decade and a half after Dörner’s preprogramming [34,35], which, while popular, is problematic for its inference of a predetermined developmental course rather than a plastic one) [62] could, and did, operate in the absence of effects on birth weight. For example, one study showed that variation in maternal nutrition influenced childhood carotid intima media thickness independently of birth weight [63]. Insulin resistance, predicted by the thrifty phenotype hypothesis to be a major adaptation to adverse prenatal nutritional conditions, only appeared some years after birth. In fact, growth-retarded babies tended to have greater insulin sensitivity [64]. A more sophisticated understanding of the developmental events was therefore required.

    In the early 2000s, the Cambridge ethologist Patrick Bateson, first alone [65] and then together with the authors and others [66], proposed a comprehensive hypothesis that placed the DOHaD phenomenon firmly within the evolutionary framework of developmental plasticity and allowed for multiple pathways of induction. We argued that, within the normal range, environmental cues received during development influenced the developmental path taken by the organism. The path was adaptive if the later postnatal environment matched the prenatal one, but if the environment changed or the prediction proved to be wrong, it would turn out to be pathological. The model was gradually refined to take into account the observations that the environments that mattered were those of later childhood, after weaning and up to the age of peak reproductive fitness at the end of the second decade of life [62,67]. This concept, termed the predictive adaptive response, could account for findings previously deemed paradoxical or inexplicable [68,69]. For instance, infants born during the Leningrad famine grew up in a nutritional environment much poorer than the Dutch Winter Famine babies; because their nutritional forecast was correct, the lack of increase in the incidence of cardiovascular or metabolic disease was not surprising. In Jamaica, children born small were more likely to respond to malnutrition with marasmus/wasting, while those born larger responded with kwashiorkor (syndrome characterized by altered protein, amino acid, and lipid metabolism in addition to wasting). Kwashiorkor is a more severe condition, associated with higher mortality than marasmus, so it may be argued that larger babies forecast a more plentiful nutritional environment during their development and were maladapted to low nutritional planes [70]. Although this heuristic model has received criticisms, these have largely been addressed [67].

    We now argue that there are two categories of pathways by which developmental factors induce later disease risk [62,71]. The induction by normative exposures involving maternal stress or reduced fetal nutrition could be interpreted using the predictive adaptive model and is thus best framed in terms of an evolved adaptive response, ultimately disadvantageous in the mismatched modern obesogenic environment. However, maternal obesity, gestational diabetes, and infant overfeeding could be interpreted as evolutionary novel exposures, likely inducing long-term effects through alternative mechanisms including the adipogenic effect of fetal hyperinsulinemia [72].

    Theoretical work has both fed off and inspired further expansion and refinement of molecular human and experimental animal research. Experimental studies demonstrated how intrauterine challenges could lead to metabolic disease in adult animals [73,74]. The early models used a variety of maternal nutritional challenges in rodents [75,76]. In general, these models showed that maternal undernutrition led to offspring obesity, hypertension, and insulin resistance. A feature of these studies is that they induced an integrated, relatively stereotypic phenotype in the offspring, with common features including hyperphagia, altered energy expenditure [77], fat preference in the diet [78,79], and altered timing of puberty [80], as well as the endothelial dysfunction, hypertension, insulin resistance, and obesity [73]. Importantly, Vickers, then others, showed that long-term effects of maternal undernutrition could be reversed by neonatal leptin treatment [81]. This finding was interpreted as experimental support of the predictive adaptive response model, a conclusion reinforced when other approaches to reversal were also demonstrated to be effective [82]. As it became clear clinically that there were at least two major pathways to long-term developmental effects, researchers started to study offspring of experimental animals exposed to high fat [83,84] or high fructose concentrations [85,86]. These experiments confirmed that the offspring of mothers under such exposures developed obesity and insulin resistance as adults. A limited amount of work was also done in large-animal models [87,88].

    At the same time, a considerable number of experimental studies examined the impact of maternal stress and care on the offspring phenotype. Pregnant animals were exposed to dexamethasone, to mimic maternal stress; their offspring exhibited phenotypic outcomes similar to those seen with maternal nutritional manipulation [89]. Jonathan Seckl, Michael Meaney, Frances Champagne, and colleagues examined the impact of maternal care upon their offspring’s neuroendocrine and behavioral development using the highly influential model of rat dams that exhibited either high licking/grooming (LG) or low LG behavior [90,91]. Importantly, they showed that the maternal behavior changed the expression of a glucocorticoid receptor gene (through epigenetic modification), and thus the offspring’s response to stress, and that the difference persisted into adulthood, yet could be eliminated by cross-fostering, to high from a low-LG or to low from a high-LG mother [91]. The phenotype produced by different levels of stress exposure could also be reversed by manipulation of the epigenetic change (in the glucocorticoid receptor gene) using a histone deacetylase inhibitor. Together, these studies pointed to the long-term effects of maternal nutritional and hormonal signals upon the offspring phenotype.

    DOHaD and Epigenetics

    The success of DOHaD over the past decade was in no small part related to the application of epigenetics to explain the relationship between developmental exposure and later risk from disease in molecular terms. Epigenetics is today usually defined as the study of mitotically and/or meiotically heritable changes in gene function that cannot be explained by changes in DNA section [92]. Although the name epigenetics goes back to the mid–twentieth century and the work of Conrad Waddington (who used it to describe the study of causal mechanisms by which the genes bring about phenotypic effects), the discipline started growing from the mid-1990s onwards [93]. Today the discipline is well established though somewhat controversial: while mitotic heritability of epigenetic marks is widely accepted, the existence and role of transgenerational epigenetic inheritance, well documented in some species and especially in plants [94], remains contentious, with some experts doubting its significance.

    Paradoxically, it was the publication of the human genome and the rise of genomics in the 1990s and 2000s that gave epigenetics a boost. Throughout the century of the gene [95], it was believed that once the DNA sequence was revealed, the variation in disease risk at the population level would be explained. But a decade later and following many genome-wide association studies as well other genomic and genetic studies, it is clear that, while some single nucleotide polymorphisms are indeed associated with the risk of NCDs, those linked to large effects in individuals are rare in the population [96,97]. The quest for common variants that contribute smaller effects continues, but the interest in other explanations of disease causation—in particular, developmental ones—has increased.

    Epigenetics provided DOHaD with tools to show precisely how the developmental environment modulates gene transcription, producing a long-term effect on gene expression and on phenotypic outcome. For epigenetics, DOHaD offered a repository of clinically important research problems. While DOHaD studies in the 1990s largely focused on explaining the effects of modifications in developmental environment in functional terms (e.g., reduced renal nephron number [98], altered hormonal sensitivity [99], altered endothelial function [100], or altered hepatic metabolic activity [101]), by the mid-2000s, and particularly after the seminal work on the LG/HG mice [91], attention shifted to molecular epigenetics.

    While there is a large range of epigenetic modifications, DNA methylation and histone modification are the best studied. The exposures that received the most research attention have been maternal stress and maternal nutrition. Studies in rats have shown, for example, that a change in the maternal diet altered DNA methylation and histone modification in the 5′ regulatory regions of specific nonimprinted genes [102–105]. Induced changes could be prevented by nutritional interventions in pregnancy [102] or changed by hormonal modifications in the juvenile period [106].

    In contrast to good experimental evidence from animal models, data supporting this argument for humans have been scarcer. Perhaps the most definitive and influential study was that of Godfrey and colleagues [107] on two independent cohorts of children aged 6 or 9  years. Using DNA obtained from the umbilical cord at birth and taking a relatively unbiased discovery approach, the authors found a strong association, consistent across cohorts, between the methylation of a particular site in the RXRα promoter region and the degree of adiposity 6–9  years later. They also demonstrated that this epigenetic change was associated with maternal carbohydrate intake in early pregnancy: mothers with lower carbohydrate intake had higher methylation in the RXRα promoter region. Later, the group showed in vitro that epigenetic changes induced nutritionally at this region of the promoter did not affect adipocyte differentiation, but they did alter insulin sensitivity and glucose metabolism in these same adipocytes when they were fully differentiated [108]. Since then, others, e.g., Harvey et al. [109], have linked other epigenetic changes at birth to other later phenotypic effects. The discussed two studies have suggested that, independent of birth weight, a sizable portion of phenotypic variance in body composition in late childhood is determined by normative in utero exposures, especially those operating in early pregnancy. Such observations, together with epidemiological analysis of periconceptional nutrition [110] and experimental observations [87,111] of periconceptional undernutrition, are shifting attention to the periconceptional and preconceptional period.

    The Wider DOHaD Research Agenda

    When thinking about the present and future DOHaD research, three major research avenues come to mind: (1) broadening and refining the epigenetic approach: studying a range of epigenetic marks; studying epigenetic inheritance through the male line as well as inheritance across multiple generations; (2) considering preventative interventions, targeted at epigenetic modifications; (3) expanding the range of studied exposures from nutrition and stress to include a variety of environmental variables. These avenues are based on recent research. For example, nongenetic inheritance through the male line has become an intense area of study. A limited number of experimental [112,113] and epidemiological [114] studies suggest that paternal factors may influence offspring health too [115]. Such studies are in their infancy, but there is growing evidence showing that epigenetic marks are transmitted across generations via the sperm. Intergenerational maternal effects have been more clearly documented and there are multiple mechanisms by which such transduction might occur [116]. Epigenetic marks can be induced de novo (under persisting environmental influences) in each generation [117] or can be transmitted directly to the offspring [118].

    The experimental data and, particularly, recent human data linking maternal state to the offspring’s epigenetic state [119] have allowed investigators to consider preventative interventions before and during pregnancy. An increasing number of clinical studies have commenced in which maternal nutrition is manipulated to explore the impact of nutritional variations on the offspring. Animal studies continue to explore potentially applicable techniques to reverse conditioning in human infants. The applicability of such interventions will rely heavily on validation of epigenetic biomarkers to monitor effects, given the time it will take for the phenotype to emerge.

    Such studies are all highly experimental and preliminary, but they point to the direction in which DOHaD research will progress. There will be ongoing expansion of epigenetic methodologies to inform both experimental studies and clinical human studies. The skill set of investigators engaged in DOHaD research is thus likely to shift significantly in the next decade.

    Regarding the range of the studied exposures, DOHaD had its early origins in the study of teratogenesis; more recently, environmental toxicologists have embraced the DOHaD paradigm [120]. They, too, have seen that subtle levels of toxins such as air pollutants, or endocrine disruptors such as bisphenol A, can affect fetal epigenetic states and, at least experimentally, induce longer-term phenotypic change not dissimilar to that found in the more traditional DOHaD domain [121]. The interaction between epigenetic processes and the environment is of much current interest [122].

    Globally, concern has been increasing over the effects of air pollution, as well as many industrial and agricultural chemicals, on the fetus. While their effects on the fetal epigenetic state are undisputed, the clinical significance of such observations is less clear. This lack of clarity stresses the importance of new prospective clinical cohorts, preferably starting before conception, providing biological samples to measure the exposome of mother and infant, measuring the epigenetic state of the infant as well as its emergent phenotype over time. Such studies are complex and expensive but important [123,124].

    DOHaD and Public Policy

    From its origins, DOHaD was focused on the causes of NCDs, especially cardiovascular and metabolic disease. Today NCDs—especially diabetes, cardiovascular disease, chronic lung disease, and common forms of cancer—account for around 60% of all deaths worldwide [125]. A high proportion of these diseases occurs in Asia, especially China and India, but a rise in the near future in parts of the world poorly equipped to deal with them—e.g., sub-Saharan Africa—has been predicted [126]. While the rise in prevalence has to do with the Western lifestyle, urbanization, and greater economic prosperity, the newly emerging epidemic is caused not by these factors as such, but by the magnitude of the recent change in behavior and environment. Scholars in DOHaD from Barker [127] onward [128] have long argued that a life course approach is important in understanding both the origins and prevention of the obesity and NCD epidemics.

    However, despite the wealth of experimental, epidemiological, and clinical data supporting the DOHaD concept, the latter has gained essentially no traction within the public health community. The reasons for this failure have been discussed above and in other articles [62], and they include the overemphasis on low birth weight, lack of a conceptual framework, and a lack of underlying mechanisms. But the evidence of multiple pathways, operating independently of birth weight; the construction of a conceptual framework; and the elaboration of the epigenetic mechanisms have all addressed these concerns. Perhaps the most difficult problem was obtaining an estimate of the weight of the developmental effect. But the calculations of Barker and colleagues suggest that, using the proxy of low birth weight as a marker of a poor developmental environment, the risk of cardiovascular disease is increased by a factor of about sevenfold in adults with birth weight at the low end of the range. However, studies such as Godfrey’s showed that this pathway was not pathological and exceptional but rather normative and ubiquitous, operating in uncomplicated pregnancies across the normal developmental range. The rising rates of maternal obesity and gestational diabetes point to the prevalence of nonadaptive responses, no longer restricted to the developed world, but of high importance in the developing world, where the double burden of unbalanced nutrition is a growing concern. The normative nature of the exposures pointed to the overlap with the reproductive–maternal–neonatal child health agenda, which the World Health Organisation (WHO) and other organizations recognize as linked to the millennium development goals [129].

    In 2011, a landmark event occurred when the United Nations General Assembly adopted the resolution titled Political declaration of the high-level meeting of the General Assembly on the prevention and control of non-communicable diseases (document A/66/L.1). For the first time, the influence of early life course events was recognized by the international community and a specific clause explained the DOHaD concept (see Box 1). This declaration led the WHO and its regional divisions to start considering life course biology and its import in greater depth. With an increased attention, nongovernmental organizations interested in NCDs have started taking notice. Private- as well as public-sector research is increasingly engaged.

    In 2014, the Director General of the WHO announced the establishment of a Commission to End Childhood Obesity. The background paper made it clear that the life course approach, of which DOHaD forms a part, made an important basis of this initiative. The announcement recognized the longer-term benefits of the primary prevention of childhood obesity. The commission and its working groups include many members with a depth of experience in DOHaD-related research. The DOHaD research is at last poised to influence public health.

    Box 1

    Clause 26 of the Political Declaration of the High-Level Meeting of the General Assembly on the Prevention and Control of Noncommunicable Diseases (Document A/66/L.1). UN General Assembly, 66th Session, Follow-Up to the Outcome of the Millennium Summit, September 16, 2011 (http://www.un.org/ga/search/view_doc.asp?symbol=A/66/L.1)

    (We) note also with concern that maternal and child health is inextricably linked with NCDs and their risk factors, specifically as prenatal malnutrition and low birth weight create a predisposition to obesity, high blood pressure, heart disease and diabetes later in life; and that pregnancy conditions, such as maternal obesity and gestational diabetes, are associated with similar risks in both the mother and her offspring.

    References

    [1] Shildrick M. Maternal imagination: reconceiving first impressions. Rethink Hist. 2000;4(3):243–260.

    [2] Hanson C. A cultural history of pregnancy. Basingstoke (UK): Palgrave; 2004.

    [3] Hopwood N. Embryology. In: Bowler P.J, Pickstone J.V, eds. The Cambridge history of scienceThe modern biological and earth sciences. vol. 6. Cambridge: Cambridge University Press; 2009 p. 285–315.

    [4] López-Beltrán C. The medical origins of heredity. In: Müller-Wille S, Rheinberger H.-J, eds. Heredity produced: at the crossroads of biology, politics and culture, 1500–1870. Cambridge (MA): Massachusetts Institute of Technology; 2007 p. 105–32.

    [5] Farley J. Gametes and spores: ideas about sexual reproduction, 1750–1914. Baltimore (MD): Johns Hopkins University Press; 1982.

    [6] Müller-Wille S, Rheinberger H.-J. A cultural history of heredity. Chicago: University of Chicago Press; 2012.

    [7] Paul D.B. Controlling human heredity: 1865 to the present. Atlantic Highlands (NJ): Humanities Press; 1995.

    [8] Kevles D. In the name of eugenics: genetics and the uses of human heredity. New York: Alfred Knopf; 1985.

    [9] Smith G.D, Kuh D, Kermack W.O. Commentary: William Ogilvy Kermack and the childhood origins of adult health and disease. Int J Epidemiol. 2001;30(4):696–703.

    [10] Walton A.J, Hammond J. The maternal effects on growth and conformation in Shire horses-Shetland pony crosses. Proc R Soc B Biol Sci. 1938;125:311–335.

    [11] Kalter H. Teratology in the 20th century: environmental causes of congenital malformations in humans and how they were established. Neurotoxicol Teratol. 2003;25:131–282.

    [12] Barcroft J. Researches on pre-natal life. Oxford: Blackwell; 1946.

    [13] Williams S. Relief and research: the nutrition work of the National Birthday Trust Fund, 1935–9. In: Smith D.F, ed. Nutrition in Britain: science, scientists and politics in the twentieth century. London: Routledge; 1997:99–122.

    [14] Antonov A.N. Children born during the siege of Leningrad in 1942. J Pediatr. 1947;30(3):250–259.

    [15] Smith C.A. Effects of maternal undernutrition upon the newborn infant in Holland (1944–45). J Pediatr. 1947;30(3):229–243.

    [16] Burger G.C.E, Drummond J.C, Sandstead H.R. Nutrition and starvation in the Western Netherlands. The Hague: General State Printing Office; 1948.

    [17] Dean R.F.A. Studies of undernutrition Wuppertal 1946-9. XXVIII. The size of the baby at birth and the yield of breast milk. Spec Rep Ser Med Res Counc (G B). 1951;275:346–378.

    [18] McCance R.A, Widdowson E.M. The determinants of growth and form. Proc R Soc Lond Ser B Containing Papers Biol Character. 1974;185(78):1–17.

    [19] Gluckman P.D, Buklijas T. Sir Graham Collingwood (Mont) Liggins. 24 June 1926–24 August 2010. Biogr Mem Fellows R Soc. 2013;59:195–214.

    [20] Barker D.J. Low intelligence. Its relation to length of gestation and rate of foetal growth. Br J Prev Soc Med. 1966;20(2):58–66.

    [21] Neugebauer R, Paneth N. Epidemiology and the wider world: celebrating Zena Stein and Mervyn Susser. Paediatr Perinat Epidemiol. 1992;6(2):122–132.

    [22] Stein Z, Susser M, Saenger G, Marolla F. Famine and human development: the Dutch Winter Famine of 1944–1945. New York: Oxford University Press; 1975.

    [23] Ravelli G.P, Stein Z.A, Susser M.W. Obesity in young men after famine exposure in utero and early infancy. N Engl J Med. 1976;295(7):349–353.

    [24] Lumey L.H. Decreased birthweights in infants after maternal in utero exposure to the Dutch famine of 1944–1945. Paediatr Perinat Epidemiol. 1992;6(2):240–253.

    [25] Forsdahl A. Are poor living conditions in childhood and adolescence an important risk factor for arteriosclerotic heart disease? Br J Prev Soc Med. 1977;31(2):91–95.

    [26] Higgins M, Keller J, Moore F, Ostrander L, Metzner H, Stock L. Studies of blood pressure in Tecumseh, Michigan. I. Blood pressure in young people and its relationship to personal and familial characteristics and complications of pregnancy in mothers. Am J Epidemiol. 1980;111:142–155.

    [27] Gennser G, Rymark P, Isberg P.E. Low birth weight and risk of high blood pressure in adulthood. Br Med J Clin Res Ed. 1988;296(6635):1498–1500.

    [28] Wadsworth M.E, Cripps H.A, Midwinter R.E, Colley J.R. Blood pressure in a national birth cohort at the age of 36 related to social and familial factors, smoking, and body mass. Br Med J Clin Res Ed. 1985;291(6508):1534–1538.

    [29] Dörner G. Die mögliche Bedeutung der prä- und/oder perinatalen Ernährung für die Pathogenese der Obesitas. Acta biologica medica Ger. 1973;30:19–22.

    [30] Dörner G, Haller K, Leonhardt M. Zur möglichen Bedeutung der prä- und/oder früh postnatalen Ernährung für die Pathogenese der Arterioskleroze. Acta Biol Med Ger. 1973;31:31–35.

    [31] Dörner G, Mohnike A. Zur möglichen Bedeutung der prä- und/oder frühpostnatalen Ernährung für die Pathogenese der Diabetes Mellitus. Acta Biol Med Ger. 1973;31:7–10.

    [32] Rohde W, Ohkawa T, Dobashi K, Arai K, Okinaga S, Dörner G. Acute effects of maternal stress on fetal blood catecholamines and hypothalamic LH-RH content. Exp Clin Endocrinol. 1983;82(3):268–274.

    [33] Dörner G, Plagemann A, Rückert J, et al. Teratogenetic maternofoetal transmission and prevention of diabetes susceptibility. Exp Clin Endocrinol. 1988;91(3):247–258.

    [34] Koletzko B. Developmental origins of adult disease: Barker’s or Dörner’s hypothesis? Am J Hum Biol. 2005;17(3):381–382.

    [35] Dörner G. Perinatal hormone levels and brain organization. In: Stumpf W, Grant L.D, eds. Anatomical neuroendocrinology. Basel: Karger; 1975 p. 245–52.

    [36] Freinkel N. Banting Lecture 1980. Of pregnancy and progeny. Diabetes. 1980;29(12):1023–1035.

    [37] Van Assche F.A, Holemans K, Aerts L. Long-term consequences for offspring of diabetes during pregnancy. Br Med Bull. 2001;60:173–182.

    [38] Gardner M, Winter P, Barker D. Atlas of mortality from selected diseases in England and Wales, 1968–1978. Chichester: Wiley; 1984.

    [39] Levy D, Brink S. A change of heart: how the Framingham heart study helped unravel the mysteries of cardiovascular disease. 1st ed. New York: Knopf; 2005.

    [40] Barker D.J, Osmond C. Infant mortality, childhood nutrition, and ischaemic heart disease in England and Wales. Lancet. 1986;1(8489):1077–1081.

    [41] Barker D.J.P. Mothers, babies and disease in later life. London: BMJ Publishing Group; 1994.

    [42] Dawes G.S, Borruto F, Zacutti A, eds. Fetal autonomy and adaptation. Chichester: Wiley; 1990.

    [43] Barker D.J, Gluckman P.D, Robinson J.S. Conference report: fetal origins of adult disease–report of the first international study group, Sydney, 29–30 October 1994. Placenta. 1995;16(3):317–320.

    [44] Hales C.N, Barker D.J. Type 2 (non-insulin-dependent) diabetes mellitus: the thrifty phenotype hypothesis. Diabetologia. 1992;35(7):595–601.

    [45] Neel J.V. Diabetes mellitus: a thrifty genotype rendered detrimental by progressAm J Hum Genet. 1962;14(4):353–362.

    [46] Neel J.V. The study of natural selection in primitive and civilized populations. Hum Biol. 1958;30(1):43–72.

    [47] De Prins F.A, Van Assche F.A. Intrauterine growth retardation and development of endocrine pancreas in the experimental rat. Biol Neonat. 1982;41:16–21.

    [48] Cogswell M.E, Yip R. The influence of fetal and maternal factors on the distribution of birthweight. Semin Perinatol. 1995;19(3):222–240.

    [49] Curhan G.C, Chertow G.M, Willett W.C, et al. Birth weight and adult hypertension and obesity in women. Circulation. 1996;94(6):1310–1315.

    [50] Curhan G.C, Willett W.C, Rimm E.B, Spiegelman D, Ascherio A.L, Stampfer M.J. Birth weight and adult hypertension, diabetes mellitus, and obesity in US men. Circulation. 1996;94(12):3246–3250.

    [51] Kramer M.S, Joseph K.S. Enigma of fetal/infant-origins hypothesis. Lancet. 1996;348(9037):1254–1255.

    [52] Paneth N, Susser M. Early origin of coronary heart disease (the Barker hypothesis). Br Med J. 1995;310:411–412.

    [53] Huxley R, Neil A, Collins R. Unravelling the fetal origins hypothesis: is there really an inverse association between birthweight and subsequent blood pressure? Lancet. 2002;360(9334):659–665.

    [54] Stanner S.A, Bulmer K, Andres C, et al. Does malnutrition in utero determine diabetes and coronary heart disease in adulthood? Results from the Leningrad siege study, a cross sectional study. BMJ. 1997;315(7119):1342–1348.

    [55] Kannisto V, Christensen K, Vaupel J.W. No increased mortality in later life for cohorts born during famine. Am J Epidemiol. 1997;145(11):987–994.

    [56] Ravelli A.C, Van der Meulen J.H.P, Michels R.P.J, et al. Glucose tolerance in adults after prenatal exposure to famine. Lancet. 1998;351:173–177.

    [57] Lumey L.H. Glucose tolerance in adults after prenatal exposure to famine. Lancet. 2001;357(9254):472–473.

    [58] Editorial. An overstretched hypothesis? Lancet. 2001;357(9254):405.

    [59] Gluckman P.D, Pinal C. Comment on An overstretched hypothesis? [Lancet 2001]. Lancet. 2001;357(9270) 1798.

    [60] Kuzawa C.W, Gluckman P.D, Hanson M.A. Developmental perspectives on the origin of obesity. In: Fantuzzi G, Mazzone T, eds. Adipose tissue and adipokines in health and disease. Totowa (NJ): Humana Press; 2007:207–219.

    [61] Lucas A. Programming by early nutrition in man. Ciba Found Symp. 1991;156:38–50.

    [62] Hanson M.A, Gluckman P.D. Early developmental conditioning of later health and disease: physiology or pathophysiology? Physiol Rev. 2014;94:1027–1076.

    [63] Gale C.R, Jiang B, Robinson S.M, Godfrey K.M, Law C.M, Martyn C.N. Maternal diet during pregnancy and carotid intima-media thickness in children. Arterioscler Thromb Vasc Biol. 2006;26:1877–1882.

    [64] Mericq V, Ong K.K, Bazaes R.A, et al. Longitudinal changes in insulin sensitivity and secretion from birth to age three years in small- and appropriate-for-gestational-age children. Diabetologia. 2005;48:2609–2614.

    [65] Bateson P. Fetal experience and good adult design. Int J Epidemiol. 2001;30(5):928–934.

    [66] Bateson P, Barker D, Clutton-Brock T, et al. Developmental plasticity and human health. Nature. 2004;430:419–421.

    [67] Bateson P, Gluckman P, Hanson M. The biology of developmental plasticity and the Predictive Adaptive Response hypothesis. J Physiol. 2014;592(11):2357–2368.

    [68] Hanson M, Gluckman P. The human camel: the concept of predictive adaptive responses and the obesity epidemic. Pract Diabetes Int. 2003;20(8):267–268.

    [69] Gluckman P.D, Hanson M.A, Spencer H.G. Predictive adaptive responses and human evolution. Trends Ecol Evol. 2005;20(10):527–533.

    [70] Forrester T.E, Badaloo A.V, Boyne M.S, et al. Prenatal factors contribute to emergence of kwashiorkor or marasmus in response to severe undernutrition: evidence for the predictive adaptation model. PLoS One. 2012;7(4):e35907.

    [71] Kuzawa C.W, Gluckman P.D, Hanson M.A, Beedle A.S, Stearns S.C, Koella J.C. Evolution, developmental plasticity, and metabolic disease. In: Stearns S.C, Koella J.C, eds. Evolution in health and disease. 2nd ed. Oxford: Oxford University Press; 2007 p. 253–64.

    [72] Hanson M.A, Gluckman P.D, Ma R.C.W, Matzen P, Biesma R.G. Early life opportunities for prevention of diabetes in low and middle income countries. BMC Public Health. 2012;12:1025.

    [73] Vickers M.H, Breier B.H, Cutfield W.S, Hofman P.L, Gluckman P.D. Fetal origins of hyperphagia, obesity, and hypertension and postnatal amplification by hypercaloric nutrition. Am J Physiol. 2000;279:E83–E87.

    [74] McMillen I.C, Robinson J.S. Developmental origins of the metabolic syndrome: prediction, plasticity, and programming. Physiol Rev. 2005;85:571–633.

    [75] Langley S.C, Jackson A.A. Increased systolic blood pressure in adult rats induced by fetal exposure to maternal low protein diets. Clin Sci. 1994;86:217–222.

    [76] Woodall S.M, Johnston B.M, Breier B.H, Gluckman P.D. Chronic maternal undernutrition in the rat leads to delayed postnatal growth and elevated blood pressure of offspring. Pediatr Res. 1996;40:438–443.

    [77] Vickers M.H, Breier B.H, McCarthy D, Gluckman P.D. Sedentary behavior during postnatal life is determined by the prenatal environment and exacerbated by postnatal hypercaloric nutrition. Am J Physiol. 2003;285(1):R271–R273.

    [78] Bellinger L, Lilley C, Langley-Evans S.C. Prenatal exposure to a low protein diet programmes a preference for high-fat foods in the young adult rat. Br J Nutr. 2004;92:513–520.

    [79] Bayol S.A, Farrington S.J, Stickland N.C. A maternal ‘junk food’ diet in pregnancy and lactation promotes an exacerbated taste for ‘junk food’ and a greater propensity for obesity in rat offspring. Br J Nutr. 2007;98(4):843–851.

    [80] Sloboda D.M, Howie G.J, Pleasants A, Gluckman P.D, Vickers M.H. Pre- and postnatal nutritional histories influence reproductive maturation and ovarian function in the rat. PLoS One. 2009;4(8):e6744.

    [81] Vickers M.H, Gluckman P.D, Coveny A.H, et al. Neonatal leptin treatment reverses developmental programming. Endocrinology. 2005;146:4211–4216.

    [82] Gray C, Li M, Reynolds C.M, Vickers M.H. Pre-weaning growth hormone treatment reverses hypertension and endothelial dysfunction in adult male offspring of mothers undernourished during pregnancy. PLoS One. 2013;8(1):e53505.

    [83] Howie G.J, Sloboda D.M, Kamal T, Vickers M.H. Maternal nutritional history predicts obesity in adult offspring independent of postnatal diet. J Physiol. 2009;587(4):905–915.

    [84] Elahi M.M, Cagampang F.R, Mukhter D, Anthony F.W, Ohri S.K, Hanson M.A. Long-term maternal high-fat feeding from weaning through pregnancy and lactation predisposes offspring to hypertension, raised plasma lipids and fatty liver in mice. Br J Nutr. 2009;102:514–519.

    [85] Vickers M.H, Clayton Z.E, Yap C, Sloboda D.M. Maternal fructose intake during pregnancy and lactation alters placental growth and leads to sex-specific changes in fetal and neonatal endocrine function. Endocrinology. 2011;152(4):1378–1387.

    [86] Flynn E.R, Alexander B.T, Lee J, Hutchens Z.M, Maric-Bilkan C. High-fat/fructose feeding during prenatal and postnatal development in female rats increases susceptibility to renal and metabolic injury later in life. Am J Physiol. Regul Integr Comp Physiol. 2013;304(4):R278–R285.

    [87] Todd S.E, Oliver M, Jaquiery A.L, Bloomfield F.H, Harding J.E. Periconceptional undernutrition of ewes impairs glucose tolerance in their adult offspring. Pediatr Res. 2009;65:409–413.

    [88] Cleal J.K, Poore K.R, Boullin J.P, et al. Mismatched pre- and postnatal nutrition leads to cardiovascular dysfunction and altered renal function in adulthood. Proc Natl Acad Sci USA. 2007;104:9529–9533.

    [89] Seckl J.R, Cleasby M, Nyirenda M.J. Glucocorticoids, 11β-hydroxysteroid dehydrogenase, and fetal programming. Kidney Int. 2000;57:1412–1417.

    [90] Meaney M.J. Maternal care, gene expression, and the transmission of individual differences in stress reactivity across generations. Annu Rev Neurosci. 2001;24:1161–1192.

    [91] Weaver I.C.G, Cervoni N, Champagne F.A, et al. Epigenetic programming by maternal behavior. Nat Neurosci. 2004;7(8):847–854.

    [92] Riggs A.D, Martienssen R.A, Russo V.E.A. Introduction. In: Russo V.E.A, Martienssen R.A, Riggs A.D, eds.Epigenetic mechanisms of gene regulation. Cold Spring Harbor: Cold Spring Harbor Laboratory Press; 1996 p. 1–4.

    [93] Haig D. The (dual) origin of epigenetics. Cold Spring Harb Symp Quant Biol. 2004;69:67–70.

    [94] Herman J.J, Sultan S.E. Adaptive transgenerational plasticity in plants: case studies, mechanisms, and implications for natural populations. Front Plant Sci. 2011;2.

    [95] Fox Keller E. Century of the gene. Cambridge (MA): Harvard University Press; 2000.

    [96] Maher B. Personal genomes: the

    Enjoying the preview?
    Page 1 of 1