Hostname: page-component-8448b6f56d-m8qmq Total loading time: 0 Render date: 2024-04-23T08:47:17.204Z Has data issue: false hasContentIssue false

The developmental environment, epigenetic biomarkers and long-term health

Published online by Cambridge University Press:  28 May 2015

K. M. Godfrey*
Affiliation:
MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
P. M. Costello
Affiliation:
Institute of Developmental Sciences, Academic Unit of Human Development and Health, University of Southampton, Southampton, UK
K. A. Lillycrop
Affiliation:
Centre for Biological Sciences, University of Southampton, Southampton, UK
*
*Address for correspondence: Professor K. Godfrey, MRC Lifecourse Epidemiology Unit, NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Southampton, SO16 6YD, UK. (Email kmg@mrc.soton.ac.uk)

Abstract

Evidence from both human and animal studies has shown that the prenatal and early postnatal environments influence susceptibility to chronic disease in later life and suggests that epigenetic processes are an important mechanism by which the environment alters long-term disease risk. Epigenetic processes, including DNA methylation, histone modification and non-coding RNAs, play a central role in regulating gene expression. The epigenome is highly sensitive to environmental factors in early life, such as nutrition, stress, endocrine disruption and pollution, and changes in the epigenome can induce long-term changes in gene expression and phenotype. In this review we focus on how the early life nutritional environment can alter the epigenome leading to an altered susceptibility to disease in later life.

Type
Review
Copyright
© Cambridge University Press and the International Society for Developmental Origins of Health and Disease 2015 

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

1. Ramachandran, A, Snehalatha, C. Rising burden of obesity in Asia. J Obes. 2010; 868573.Google ScholarPubMed
2. Morris, AP, Voight, BF, Teslovich, TM, et al. Large-scale association analysis provides insights into the genetic architecture and pathophysiology of type 2 diabetes. Nat Genet. 2012; 44, 981990.Google ScholarPubMed
3. Godfrey, KM, Inskip, HM, Hanson, MA. The long term effects of prenatal development on growth and metabolism. Semin Reprod Med. 2011; 29, 257265.CrossRefGoogle Scholar
4. 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, 9195.Google ScholarPubMed
5. Barker, DJ, Winter, PD, Osmond, C, Margetts, B, Simmonds, SJ. Weight in infancy and death from ischaemic heart disease. Lancet. 1989; 2, 577580.CrossRefGoogle ScholarPubMed
6. Pettitt, D, Jovanovic, L. Birth weight as a predictor of type 2 diabetes mellitus: the U-shaped curve. Curr Diab Rep. 2001; 1, 7881.CrossRefGoogle ScholarPubMed
7. Ong, KK. Size at birth, postnatal growth and risk of obesity. Horm Res. 2006; 65(Suppl. 3), 6569.Google ScholarPubMed
8. Hanson, MA, Gluckman, PD. Developmental processes and the induction of cardiovascular function: conceptual aspects. J Physiol. 2005; 565, 2734.CrossRefGoogle ScholarPubMed
9. Roseboom, T, de Rooij, S, Painter, R. The Dutch famine and its long-term consequences for adult health. Early Hum Dev. 2006; 82, 485491.CrossRefGoogle ScholarPubMed
10. Painter, RC, Roseboom, TJ, Bleker, OP. Prenatal exposure to the Dutch famine and disease in later life: an overview. Reprod Toxicol. 2005; 20, 345352.CrossRefGoogle Scholar
11. Hoile, SP, Irvine, NA, Kelsall, CJ, et al. Maternal fat intake in rats alters 20:4n-6 and 22:6n-3 status and the epigenetic regulation of Fads2 in offspring liver. J Nutr Biochem. 2013; 24, 12131220.CrossRefGoogle Scholar
12. Lillycrop, KA, Slater-Jefferies, JL, Hanson, MA, et al. Induction of altered epigenetic regulation of the hepatic glucocorticoid receptor in the offspring of rats fed a protein-restricted diet during pregnancy suggests that reduced DNA methyltransferase-1 expression is involved in impaired DNA methylation and changes in histone modifications. Br J Nutr. 2007; 97, 10641073.CrossRefGoogle ScholarPubMed
13. Cleal, JK, Poore, KR, Boullin, JP, et al. Mismatched pre- and postnatal nutrition leads to cardiovascular dysfunction and altered renal function in adulthood. Proc Natl Acad Sci U S A. 2007; 104, 95299533.CrossRefGoogle ScholarPubMed
14. Desai, M, Jellyman, JK, Han, G, et al. Maternal obesity and high-fat diet program offspring metabolic syndrome. Am J Obstet Gynecol. 2014; 211, 237, e231–237.e213.CrossRefGoogle ScholarPubMed
15. Howie, GJ, Sloboda, DM, Reynolds, CM, Vickers, MH. Timing of maternal exposure to a high fat diet and development of obesity and hyperinsulinemia in male rat offspring: same metabolic phenotype, different developmental pathways? J Nutr Metabol. 2013; 2013, 517384.CrossRefGoogle ScholarPubMed
16. Bird, A, Macleod, D. Reading the DNA methylation signal. Cold Spring Harb Symp Quant Biol. 2004; 69, 113118.CrossRefGoogle ScholarPubMed
17. Fraga, MF, Ballestar, E, Paz, MF, et al. Epigenetic differences arise during the lifetime of monozygotic twins. Proc Natl Acad Sci U S A. 2005; 102, 1060410609.CrossRefGoogle ScholarPubMed
18. Gluckman, PD, Hanson, MA, Spencer, HG. Predictive adaptive responses and human evolution. Trends Ecol Evol. 2005; 20, 527533.CrossRefGoogle ScholarPubMed
19. Kumar, S, Cheng, X, Klimasauskas, S, et al. The DNA (cytosine-5) methyltransferases. Nucleic Acids Res. 1994; 22, 110.CrossRefGoogle ScholarPubMed
20. Ramsahoye, BH, Biniszkiewicz, D, Lyko, F, et al. Non-CpG methylation is prevalent in embryonic stem cells and may be mediated by DNA methyltransferase 3a. Proc Natl Acad Sci U S A. 2000; 97, 52375242.CrossRefGoogle ScholarPubMed
21. Bird, A. DNA methylation patterns and epigenetic memory. Genes Dev. 2002; 16, 621.CrossRefGoogle ScholarPubMed
22. Irizarry, RA, Ladd-Acosta, C, Wen, B, et al. The human colon cancer methylome shows similar hypo- and hypermethylation at conserved tissue-specific CpG island shores. Nat Genet. 2009; 41, 178186.CrossRefGoogle ScholarPubMed
23. Song, F, Smith, JF, Kimura, MT, et al. Association of tissue-specific differentially methylated regions (TDMs) with differential gene expression. Proc Nat Acad Sci U S A. 2005; 102, 33363341.CrossRefGoogle ScholarPubMed
24. Fuks, F, Hurd, PJ, Wolf, D, et al. The methyl-CpG-binding protein MeCP2 links DNA methylation to histone methylation. J Biol Chem. 2003; 278, 40354040.CrossRefGoogle ScholarPubMed
25. Santos, F, Hendrich, B, Reik, W, Dean, W. Dynamic reprogramming of DNA methylation in the early mouse embryo. Dev Biol. 2002; 241, 172182.CrossRefGoogle ScholarPubMed
26. Okano, M, Bell, DW, Haber, DA, Li, E. DNA methyltransferases DNMT3a and DNMT3b are essential for de novo methylation and mammalian development. Cell. 1999; 99, 247257.CrossRefGoogle Scholar
27. Bacolla, A, Pradhan, S, Roberts, RJ, Wells, RD. Recombinant human DNA (cytosine-5) methyltransferase. II. Steady-state kinetics reveal allosteric activation by methylated dna. J Biol Chem. 1999; 274, 3301133019.CrossRefGoogle ScholarPubMed
28. Tahiliani, M, Koh, KP, Shen, Y, et al. Conversion of 5-methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1. Science. 2009; 324, 930935.CrossRefGoogle ScholarPubMed
29. Guibert, S, Weber, M. Chapter two – functions of DNA methylation and hydroxymethylation in mammalian development. In Current Topics in Developmental Biology (ed. Edith H), 2013; pp. 4783. Academic Press: New York, USA.Google Scholar
30. Santiago, M, Antunes, C, Guedes, M, Sousa, N, Marques, CJ. TET enzymes and DNA hydroxymethylation in neural development and function – how critical are they? Genomics. 2014; 104, 334340.CrossRefGoogle Scholar
31. Venkatesh, S, Workman, JL. Histone exchange, chromatin structure and the regulation of transcription. Nat Rev Mol Cell Biol. 2015; 16, 178189.CrossRefGoogle ScholarPubMed
32. Turner, BM. Histone acetylation and an epigenetic code. Bioessays. 2000; 22, 836845.3.0.CO;2-X>CrossRefGoogle Scholar
33. Cedar, H, Bergman, Y. Linking DNA methylation and histone modification: patterns and paradigms. Nat Rev Genet. 2009; 10, 295304.CrossRefGoogle ScholarPubMed
34. Djebali, S, Davis, CA, Merkel, A, et al. Landscape of transcription in human cells. Nature. 2012; 489, 101108.CrossRefGoogle ScholarPubMed
35. Morris, KV, Mattick, JS. The rise of regulatory RNA. Nat Rev Genet. 2014; 15, 423437.CrossRefGoogle ScholarPubMed
36. Hawkins, PG, Santoso, S, Adams, C, Anest, V, Morris, KV. Promoter targeted small RNAs induce long-term transcriptional gene silencing in human cells. Nucleic Acids Res. 2009; 37, 29842995.CrossRefGoogle ScholarPubMed
37. Reik, W, Lewis, A. Co-evolution of X-chromosome inactivation and imprinting in mammals. Nat Rev Genet. 2005; 6, 403410.CrossRefGoogle ScholarPubMed
38. Mercer, TR, Dinger, ME, Mattick, JS. Long non-coding RNAs: insights into functions. Nat Rev Genet. 2009; 10, 155159.CrossRefGoogle ScholarPubMed
39. Kucharski, R, Maleszka, J, Foret, S, Maleszka, R. Nutritional control of reproductive status in honeybees via DNA methylation. Science. 2008; 319, 18271830.CrossRefGoogle ScholarPubMed
40. Morgan, HD, Sutherland, HG, Martin, DI, Whitelaw, E. Epigenetic inheritance at the agouti locus in the mouse. Nat Genet. 1999; 23, 314318.CrossRefGoogle ScholarPubMed
41. Waterland, RA, Jirtle, RL. Transposable elements: targets for early nutritional effects on epigenetic gene regulation. Mol Cell Biol. 2003; 23, 52935300.CrossRefGoogle ScholarPubMed
42. Lillycrop, KA, Phillips, ES, Jackson, AA, Hanson, MA, Burdge, GC. Dietary protein restriction of pregnant rats induces and folic acid supplementation prevents epigenetic modification of hepatic gene expression in the offspring. J Nutr. 2005; 135, 13821386.CrossRefGoogle ScholarPubMed
43. Lillycrop, KA, Phillips, ES, Torrens, C, et al. Feeding pregnant rats a protein-restricted diet persistently alters the methylation of specific cytosines in the hepatic PPAR alpha promoter of the offspring. Br J Nutr. 2008; 100, 278282.CrossRefGoogle ScholarPubMed
44. Gluckman, PD, Lillycrop, KA, Vickers, MH, et al. Metabolic plasticity during mammalian development is directionally dependent on early nutritional status. Proc Natl Acad Sci U S A. 2007; 104, 1279612800.CrossRefGoogle ScholarPubMed
45. Li, CCY, Young, PE, Maloney, CA, et al. Maternal obesity and diabetes induces latent metabolic defects and widespread epigenetic changes in isogenic mice. Epigenetics. 2013; 8, 602611.CrossRefGoogle ScholarPubMed
46. Plagemann, A, Harder, T, Brunn, M, et al. Hypothalamic proopiomelanocortin promoter methylation becomes altered by early overfeeding: an epigenetic model of obesity and the metabolic syndrome. J Physiol. 2009; 587(Pt 20), 49634976.CrossRefGoogle ScholarPubMed
47. Burdge, GC, Lillycrop, KA, Phillips, ES, et al. Folic acid supplementation during the juvenile-pubertal period in rats modifies the phenotype and epigenotype induced by prenatal nutrition. J Nutr. 2009; 139, 10541060.CrossRefGoogle ScholarPubMed
48. Ly, A, Lee, H, Chen, J, et al. Effect of maternal and postweaning folic acid supplementation on mammary tumor risk in the offspring. Cancer Res. 2011; 71, 988997.CrossRefGoogle ScholarPubMed
49. Christman, JK, Sheikhnejad, G, Dizik, M, Abileah, S, Wainfan, E. Reversibility of changes in nucleic acid methylation and gene expression induced in rat liver by severe dietary methyl deficiency. Carcinogenesis. 1993; 14, 551557.CrossRefGoogle ScholarPubMed
50. Aagaard-Tillery, KM, Grove, K, Bishop, J, et al. Developmental origins of disease and determinants of chromatin structure: maternal diet modifies the primate fetal epigenome. J Mol Endocrinol. 2008; 41, 91102.CrossRefGoogle ScholarPubMed
51. Zheng, S, Rollet, M, Pan, Y-X. Maternal protein restriction during pregnancy induces CCAAT/enhancer-binding protein (C/EBPβ) expression through the regulation of histone modification at its promoter region in female offspring rat skeletal muscle. Epigenetics. 2011; 6, 161170.CrossRefGoogle ScholarPubMed
52. Lie, S, Morrison, JL, Williams-Wyss, O, et al. Impact of embryo number and maternal undernutrition around the time of conception on insulin signaling and gluconeogenic factors and microRNAs in the liver of fetal sheep. Am J Physiol Endocrinol Metab. 2014; 9, E10131024.Google Scholar
53. Lie, S, Morrison, JL, Williams-Wyss, O, et al. Periconceptional undernutrition programs changes in insulin-signaling molecules and microRNAs in skeletal muscle in singleton and twin fetal sheep. Biol Reprod. 2014; 90, 5.CrossRefGoogle ScholarPubMed
54. Carone, BR, Fauquier, L, Habib, N, et al. Paternally induced transgenerational environmental reprogramming of metabolic gene expression in mammals. Cell. 2010; 143, 10841096.CrossRefGoogle ScholarPubMed
55. Ng, S-F, Lin, RCY, Laybutt, DR, et al. Chronic high-fat diet in fathers programs [bgr]-cell dysfunction in female rat offspring. Nature. 2010; 467, 963966.CrossRefGoogle ScholarPubMed
56. Öst, A, Lempradl, A, Casas, E, et al. Paternal diet defines offspring chromatin state and intergenerational obesity. Cell. 2014; 159, 13521364.CrossRefGoogle ScholarPubMed
57. Lange, UC, Schneider, R. What an epigenome remembers. Bio Essays. 2010; 32, 659668.Google ScholarPubMed
58. Desai, M, Jellyman, JK, Ross, MG. Epigenomics gestational programming and risk of metabolic syndrome. Int J Obes. 2015; 39, 633641.CrossRefGoogle ScholarPubMed
59. Benyshek, DC, Johnston, CS, Martin, JF. Glucose metabolism is altered in the adequately-nourished grand-offspring (F3 generation) of rats malnourished during gestation and perinatal life. Diabetologia. 2006; 49, 11171119.CrossRefGoogle ScholarPubMed
60. Gregory, AD, Tracy, LB. Maternal high-fat diet effects on third-generation female body size via the paternal lineage. Endocrinology. 2011; 152, 22282236.Google Scholar
61. Tobi, EW, Lumey, LH, Talens, RP, et al. DNA methylation differences after exposure to prenatal famine are common and timing- and sex-specific. Hum Mol Genet. 2009; 18, 40464053.CrossRefGoogle ScholarPubMed
62. Steegers-Theunissen, RP, Obermann-Borst, SA, Kremer, D, et al. Periconceptional maternal folic acid use of 400 microg per day is related to increased methylation of the IGF2 gene in the very young child. PLoS One. 2009; 4, e7845.CrossRefGoogle Scholar
63. Jacobsen, SC, Brøns, C, Bork-Jensen, J, et al. Effects of short-term high-fat overfeeding on genome-wide DNA methylation in the skeletal muscle of healthy young men. Diabetologia. 2012; 55, 33413349.CrossRefGoogle ScholarPubMed
64. Godfrey, KM, Sheppard, A, Gluckman, PD, et al. Epigenetic gene promoter methylation at birth is associated with child’s later adiposity. Diabetes. 2011; 60, 15281534.CrossRefGoogle ScholarPubMed
65. Ollikainen, M, Smith, KR, EJ-H, Joo, et al. DNA methylation analysis of multiple tissues from newborn twins reveals both genetic and intrauterine components to variation in the human neonatal epigenome. Hum Mol Genet. 2010; 19, 41764188.CrossRefGoogle ScholarPubMed
66. Murphy, SK, Huang, Z, Hoyo, C. Differentially methylated regions of imprinted genes in prenatal, perinatal and postnatal human tissues. PLoS One. 2012; 7, e40924.CrossRefGoogle ScholarPubMed
67. Talens, RP, Boomsma, DI, Tobi, EW, et al. Variation, patterns, and temporal stability of DNA methylation: considerations for epigenetic epidemiology. FASEB J. 2010; 24, 31353144.CrossRefGoogle ScholarPubMed
68. Burdge, GC, Hanson, MA, Slater-Jefferies, JL, Lillycrop, KA. Epigenetic regulation of transcription: a mechanism for inducing variations in phenotype (fetal programming) by differences in nutrition during early life? Br J Nutr. 2007; 97, 10361046.CrossRefGoogle ScholarPubMed
69. Barres, R, Yan, J, Egan, B, et al. Acute exercise remodels promoter methylation in human skeletal muscle. Cell Metab. 2012; 15, 405411.CrossRefGoogle ScholarPubMed
70. Wong, CCY, Caspi, A, Williams, B, et al. A longitudinal study of epigenetic variation in twins. Epigenetics. 2010; 5, 516526.CrossRefGoogle ScholarPubMed
71. Clarke-Harris, R, Wilkin, TJ, Hosking, J, et al. PGC1α promoter methylation in blood at 5–7 years predicts adiposity from 9 to 14 years (EarlyBird 50). Diabetes. 2014; 63, 25282537.CrossRefGoogle ScholarPubMed