Clin Nutr Res.  2014 Jan;3(1):1-8. 10.7762/cnr.2014.3.1.1.

Nutrition, Epigenetics, and Diseases

Affiliations
  • 1Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, MA 02115, USA. hjang1@partners.org
  • 2Harvard Medical School, Boston, MA 02115, USA.

Abstract

Increasing epidemiological evidence suggests that maternal nutrition and environmental exposure early in development play an important role in susceptibility to disease in later life. In addition, these disease outcomes seem to pass through subsequent generations. Epigenetic modifications provide a potential link between the nutrition status during critical periods in development and changes in gene expression that may lead to disease phenotypes. An increasing body of evidence from experimental animal studies supports the role of epigenetics in disease susceptibility during critical developmental periods, including periconceptional period, gestation, and early postnatal period. The rapid improvements in genetic and epigenetic technologies will allow comprehensive investigations of the relevance of these epigenetic phenomena in human diseases.

Keyword

Nutrition; Reprogramming; Development; Epigenetic; Disease

MeSH Terms

Animals
Critical Period (Psychology)
Disease Susceptibility
Environmental Exposure
Epigenomics*
Family Characteristics
Gene Expression
Humans
Nutritional Status
Phenotype
Pregnancy

Reference

1. Barker DJ. Fetal origins of coronary heart disease. BMJ. 1995; 311:171–174.
Article
2. Byrne CD, Phillips DI. Fetal origins of adult disease: epidemiology and mechanisms. J Clin Pathol. 2000; 53:822–828.
Article
3. Roseboom TJ, Painter RC, van Abeelen AF, Veenendaal MV, de Rooij SR. Hungry in the womb: what are the consequences? Lessons from the Dutch famine. Maturitas. 2011; 70:141–145.
Article
4. Painter RC, Osmond C, Gluckman P, Hanson M, Phillips DI, Roseboom TJ. Transgenerational effects of prenatal exposure to the Dutch famine on neonatal adiposity and health in later life. BJOG. 2008; 115:1243–1249.
Article
5. Roseboom T, de Rooij S, Painter R. The Dutch famine and its long-term consequences for adult health. Early Hum Dev. 2006; 82:485–491.
Article
6. Painter RC, de Rooij SR, Bossuyt PM, Simmers TA, Osmond C, Barker DJ, Bleker OP, Roseboom TJ. Early onset of coronary artery disease after prenatal exposure to the Dutch famine. Am J Clin Nutr. 2006; 84:322–327.
Article
7. de Rooij SR, Painter RC, Roseboom TJ, Phillips DI, Osmond C, Barker DJ, Tanck MW, Michels RP, Bossuyt PM, Bleker OP. Glucose tolerance at age 58 and the decline of glucose tolerance in comparison with age 50 in people prenatally exposed to the Dutch famine. Diabetologia. 2006; 49:637–643.
Article
8. Painter RC, De Rooij SR, Bossuyt PM, Osmond C, Barker DJ, Bleker OP, Roseboom TJ. A possible link between prenatal exposure to famine and breast cancer: a preliminary study. Am J Hum Biol. 2006; 18:853–856.
Article
9. de Rooij SR, Painter RC, Holleman F, Bossuyt PM, Roseboom TJ. The metabolic syndrome in adults prenatally exposed to the Dutch famine. Am J Clin Nutr. 2007; 86:1219–1224.
Article
10. Bygren LO, Kaati G, Edvinsson S. Longevity determined by paternal ancestors' nutrition during their slow growth period. Acta Biotheor. 2001; 49:53–59.
11. Pembrey ME, Bygren LO, Kaati G, Edvinsson S, Northstone K, Sjöström M, Golding J. ALSPAC Study Team. Sex-specific, male-line transgenerational responses in humans. Eur J Hum Genet. 2006; 14:159–166.
Article
12. Bird A. Perceptions of epigenetics. Nature. 2007; 447:396–398.
Article
13. Wade PA, Archer TK. Epigenetics: environmental instructions for the genome. Environ Health Perspect. 2006; 114:A140–A141.
14. Musselman CA, Lalonde ME, Côté J, Kutateladze TG. Perceiving the epigenetic landscape through histone readers. Nat Struct Mol Biol. 2012; 19:1218–1227.
Article
15. Bird A. DNA methylation patterns and epigenetic memory. Genes Dev. 2002; 16:6–21.
Article
16. Bird AP. CpG-rich islands and the function of DNA methylation. Nature. 1986; 321:209–213.
Article
17. Reik W, Dean W, Walter J. Epigenetic reprogramming in mammalian development. Science. 2001; 293:1089–1093.
Article
18. Li E, Beard C, Jaenisch R. Role for DNA methylation in genomic imprinting. Nature. 1993; 366:362–365.
Article
19. Rainier S, Johnson LA, Dobry CJ, Ping AJ, Grundy PE, Feinberg AP. Relaxation of imprinted genes in human cancer. Nature. 1993; 362:747–749.
Article
20. Gonzalo S, Jaco I, Fraga MF, Chen T, Li E, Esteller M, Blasco MA. DNA methyltransferases control telomere length and telomere recombination in mammalian cells. Nat Cell Biol. 2006; 8:416–424.
Article
21. Luger K, Mäder AW, Richmond RK, Sargent DF, Richmond TJ. Crystal structure of the nucleosome core particle at 2.8 A resolution. Nature. 1997; 389:251–260.
Article
22. Berger SL. An embarrassment of niches: the many covalent modifications of histones in transcriptional regulation. Oncogene. 2001; 20:3007–3013.
Article
23. Jenuwein T, Allis CD. Translating the histone code. Science. 2001; 293:1074–1080.
Article
24. Cole PA. Chemical probes for histone-modifying enzymes. Nat Chem Biol. 2008; 4:590–597.
Article
25. Kim DH, Saetrom P, Snøve O Jr, Rossi JJ. MicroRNA-directed transcriptional gene silencing in mammalian cells. Proc Natl Acad Sci U S A. 2008; 105:16230–16235.
Article
26. Szenthe K, Koroknai A, Banati F, Bathori Z, Lozsa R, Burgyan J, Wolf H, Salamon D, Nagy K, Niller HH, Minarovits J. The 5' regulatory sequences of active miR-146a promoters are hypomethylated and associated with euchromatic histone modification marks in B lymphoid cells. Biochem Biophys Res Commun. 2013; 433:489–495.
Article
27. Li E. Chromatin modification and epigenetic reprogramming in mammalian development. Nat Rev Genet. 2002; 3:662–673.
Article
28. Bernal AJ, Jirtle RL. Epigenomic disruption: the effects of early developmental exposures. Birth Defects Res A Clin Mol Teratol. 2010; 88:938–944.
Article
29. Jirtle RL, Skinner MK. Environmental epigenomics and disease susceptibility. Nat Rev Genet. 2007; 8:253–262.
Article
30. Weaver JR, Susiarjo M, Bartolomei MS. Imprinting and epigenetic changes in the early embryo. Mamm Genome. 2009; 20:532–543.
Article
31. DeBaun MR, Niemitz EL, McNeil DE, Brandenburg SA, Lee MP, Feinberg AP. Epigenetic alterations of H19 and LIT1 distinguish patients with Beckwith-Wiedemann syndrome with cancer and birth defects. Am J Hum Genet. 2002; 70:604–611.
Article
32. Murphy SK, Jirtle RL. Imprinting evolution and the price of silence. Bioessays. 2003; 25:577–588.
Article
33. Sinclair KD, Allegrucci C, Singh R, Gardner DS, Sebastian S, Bispham J, Thurston A, Huntley JF, Rees WD, Maloney CA, Lea RG, Craigon J, McEvoy TG, Young LE. DNA methylation, insulin resistance, and blood pressure in offspring determined by maternal periconceptional B vitamin and methionine status. Proc Natl Acad Sci U S A. 2007; 104:19351–19356.
Article
34. Reik W, Constancia M, Dean W, Davies K, Bowden L, Murrell A, Feil R, Walter J, Kelsey G. Igf2 imprinting in development and disease. Int J Dev Biol. 2000; 44:145–150.
Article
35. Heijmans BT, Tobi EW, Stein AD, Putter H, Blauw GJ, Susser ES, Slagboom PE, Lumey LH. Persistent epigenetic differences associated with prenatal exposure to famine in humans. Proc Natl Acad Sci U S A. 2008; 105:17046–17049.
Article
36. Kwong WY, Miller DJ, Ursell E, Wild AE, Wilkins AP, Osmond C, Anthony FW, Fleming TP. Imprinted gene expression in the rat embryo-fetal axis is altered in response to periconceptional maternal low protein diet. Reproduction. 2006; 132:265–277.
Article
37. Waterland RA, Lin JR, Smith CA, Jirtle RL. Post-weaning diet affects genomic imprinting at the insulin-like growth factor 2 (Igf2) locus. Hum Mol Genet. 2006; 15:705–716.
Article
38. Claycombe KJ, Uthus EO, Roemmich JN, Johnson LK, Johnson WT. Prenatal low-protein and postnatal high-fat diets induce rapid adipose tissue growth by inducing Igf2 expression in Sprague Dawley rat offspring. J Nutr. 2013; 143:1533–1539.
Article
39. Dolinoy DC. The agouti mouse model: an epigenetic biosensor for nutritional and environmental alterations on the fetal epigenome. Nutr Rev. 2008; 66:Suppl 1. S7–S11.
Article
40. Wolff GL, Kodell RL, Moore SR, Cooney CA. Maternal epigenetics and methyl supplements affect agouti gene expression in Avy/a mice. FASEB J. 1998; 12:949–957.
Article
41. Waterland RA, Jirtle RL. Early nutrition, epigenetic changes at transposons and imprinted genes, and enhanced susceptibility to adult chronic diseases. Nutrition. 2004; 20:63–68.
Article
42. Flier JS. Obesity wars: molecular progress confronts an expanding epidemic. Cell. 2004; 116:337–350.
43. Li G, Kohorst JJ, Zhang W, Laritsky E, Kunde-Ramamoorthy G, Baker MS, Fiorotto ML, Waterland RA. Early postnatal nutrition determines adult physical activity and energy expenditure in female mice. Diabetes. 2013; 62:2773–2783.
Article
44. Plagemann A, Harder T, Brunn M, Harder A, Roepke K, Wittrock-Staar M, Ziska T, Schellong K, Rodekamp E, Melchior K, Dudenhausen JW. Hypothalamic proopiomelanocortin promoter methylation becomes altered by early overfeeding: an epigenetic model of obesity and the metabolic syndrome. J Physiol. 2009; 587:4963–4976.
Article
45. Plagemann A, Roepke K, Harder T, Brunn M, Harder A, Wittrock-Staar M, Ziska T, Schellong K, Rodekamp E, Melchior K, Dudenhausen JW. Epigenetic malprogramming of the insulin receptor promoter due to developmental overfeeding. J Perinat Med. 2010; 38:393–400.
Article
46. Vucetic Z, Kimmel J, Totoki K, Hollenbeck E, Reyes TM. Maternal high-fat diet alters methylation and gene expression of dopamine and opioid-related genes. Endocrinology. 2010; 151:4756–4764.
Article
47. Begum G, Stevens A, Smith EB, Connor K, Challis JR, Bloomfield F, White A. Epigenetic changes in fetal hypothalamic energy regulating pathways are associated with maternal undernutrition and twinning. FASEB J. 2012; 26:1694–1703.
Article
48. Coupé B, Amarger V, Grit I, Benani A, Parnet P. Nutritional programming affects hypothalamic organization and early response to leptin. Endocrinology. 2010; 151:702–713.
Article
49. Gluckman PD, Hanson MA, Buklijas T, Low FM, Beedle AS. Epigenetic mechanisms that underpin metabolic and cardiovascular diseases. Nat Rev Endocrinol. 2009; 5:401–408.
Article
50. Watkins AJ, Wilkins A, Cunningham C, Perry VH, Seet MJ, Osmond C, Eckert JJ, Torrens C, Cagampang FR, Cleal J, Gray WP, Hanson MA, Fleming TP. Low protein diet fed exclusively during mouse oocyte maturation leads to behavioural and cardiovascular abnormalities in offspring. J Physiol. 2008; 586:2231–2244.
Article
51. Kwong WY, Wild AE, Roberts P, Willis AC, Fleming TP. Maternal undernutrition during the preimplantation period of rat development causes blastocyst abnormalities and programming of postnatal hypertension. Development. 2000; 127:4195–4202.
Article
52. Oben JA, Mouralidarane A, Samuelsson AM, Matthews PJ, Morgan ML, McKee C, Soeda J, Fernandez-Twinn DS, Martin-Gronert MS, Ozanne SE, Sigala B, Novelli M, Poston L, Taylor PD. Maternal obesity during pregnancy and lactation programs the development of offspring non-alcoholic fatty liver disease in mice. J Hepatol. 2010; 52:913–920.
Article
53. Oben JA, Patel T, Mouralidarane A, Samuelsson AM, Matthews P, Pombo J, Morgan M, McKee C, Soeda J, Novelli M, Poston L, Taylor P. Maternal obesity programmes offspring development of non-alcoholic fatty pancreas disease. Biochem Biophys Res Commun. 2010; 394:24–28.
Article
54. Tamashiro KL, Terrillion CE, Hyun J, Koenig JI, Moran TH. Prenatal stress or high-fat diet increases susceptibility to diet-induced obesity in rat offspring. Diabetes. 2009; 58:1116–1125.
Article
55. Sun B, Purcell RH, Terrillion CE, Yan J, Moran TH, Tamashiro KL. Maternal high-fat diet during gestation or suckling differentially affects offspring leptin sensitivity and obesity. Diabetes. 2012; 61:2833–2841.
Article
56. Martins MR, Vieira AK, de Souza EP, Moura AS. Early overnutrition impairs insulin signaling in the heart of adult Swiss mice. J Endocrinol. 2008; 198:591–598.
Article
57. Aubert R, Suquet JP, Lemonnier D. Long-term morphological and metabolic effects of early under- and over-nutrition in mice. J Nutr. 1980; 110:649–661.
Article
58. Fiorotto ML, Burrin DG, Perez M, Reeds PJ. Intake and use of milk nutrients by rat pups suckled in small, medium, or large litters. Am J Physiol. 1991; 260:R1104–R1113.
Article
59. Rodrigues AL, de Moura EG, Passos MC, Trevenzoli IH, da Conceição EP, Bonono IT, Neto JF, Lisboa PC. Postnatal early overfeeding induces hypothalamic higher SOCS3 expression and lower STAT3 activity in adult rats. J Nutr Biochem. 2011; 22:109–117.
Article
60. You S, Götz F, Rohde W, Dörner G. Early postnatal overfeeding and diabetes susceptibility. Exp Clin Endocrinol. 1990; 96:301–306.
Article
61. Waterland RA, Garza C. Early postnatal nutrition determines adult pancreatic glucose-responsive insulin secretion and islet gene expression in rats. J Nutr. 2002; 132:357–364.
Article
62. Steegers-Theunissen RP, Obermann-Borst SA, Kremer D, Lindemans J, Siebel C, Steegers EA, Slagboom PE, Heijmans BT. 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.
63. Cooney CA, Dave AA, Wolff GL. Maternal methyl supplements in mice affect epigenetic variation and DNA methylation of offspring. J Nutr. 2002; 132:2393S–2400S.
Article
64. Waterland RA, Travisano M, Tahiliani KG. Diet-induced hypermethylation at agouti viable yellow is not inherited transgenerationally through the female. FASEB J. 2007; 21:3380–3385.
Article
65. Waterland RA, Travisano M, Tahiliani KG, Rached MT, Mirza S. Methyl donor supplementation prevents transgenerational amplification of obesity. Int J Obes (Lond). 2008; 32:1373–1379.
Article
66. Dolinoy DC, Weidman JR, Waterland RA, Jirtle RL. Maternal genistein alters coat color and protects Avy mouse offspring from obesity by modifying the fetal epigenome. Environ Health Perspect. 2006; 114:567–572.
Article
67. Dolinoy DC, Huang D, Jirtle RL. Maternal nutrient supplementation counteracts bisphenol A-induced DNA hypomethylation in early development. Proc Natl Acad Sci U S A. 2007; 104:13056–13061.
Article
Full Text Links
  • CNR
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr