Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Environmental epigenomics and disease susceptibility

Key Points

  • Human epidemiological studies and animal investigations provide compelling evidence that prenatal and early postnatal environmental factors influence the adult risk of developing various chronic diseases, such as cancer, cardiovascular disease, diabetes, obesity and behavioural disorders such as schizophrenia.

  • The developmental origins of adult-onset disease hypothesis proposes that the evolution of developmental plasticity, which enables an organism to adapt to environmental signals during early life, can also increase the risk of developing chronic diseases when there is a mismatch between the perceived environment and that which is encountered in adulthood.

  • Epigenetics is the study of alterations in gene expression that occur not by changing the DNA sequence, but by modifying DNA methylation and remodelling chromatin structure.

  • Prenatal and postnatal environmental exposures could be linked to phenotypic changes later in life through the alteration of the epigenetic marks that regulate the functional output of the information that is stored in the genome.

  • In support of this postulate, maternal methyl-donor supplementation during pregnancy with folic acid, vitamin B12, choline and betaine was shown to effect the phenotype of the Avy (viable yellow agouti) offspring by directly altering the epigenome.

  • Studies with the fungicide vinclozolin demonstrate that heritable environmentally induced epigenetic modifications can also underlie transgenerational alterations in phenotype.

  • Novel genome-wide experimental and bioinformatic techniques are now being used to identify epigenetically labile genes in humans. Such approaches will hopefully allow for the development of unique epigenetic-based diagnostic, prevention and therapeutic strategies for human diseases.

Abstract

Epidemiological evidence increasingly suggests that environmental exposures early in development have a role in susceptibility to disease in later life. In addition, some of these environmental effects seem to be passed on through subsequent generations. Epigenetic modifications provide a plausible link between the environment and alterations in gene expression that might lead to disease phenotypes. An increasing body of evidence from animal studies supports the role of environmental epigenetics in disease susceptibility. Furthermore, recent studies have demonstrated for the first time that heritable environmentally induced epigenetic modifications underlie reversible transgenerational alterations in phenotype. Methods are now becoming available to investigate the relevance of these phenomena to human disease.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Epigenetic regulation of metastable epialleles.
Figure 2: Effect of maternal dietary supplementation on the phenotype and epigenotype of Avy/a offspring.
Figure 3: Epigenetic regulation of imprinted alleles.
Figure 4: Alterations in methylation status during development.
Figure 5: Germline transmission of epigenetically regulated transgenerational phenotypes.
Figure 6: A model for endocrine-disruptor-induced epigenetic transgenerational disease.

Similar content being viewed by others

References

  1. Yajnik, C. S. Early life origins of insulin resistance and type 2 diabetes in India and other Asian countries. J. Nutr. 134, 205–210 (2004).

    Article  CAS  PubMed  Google Scholar 

  2. Barker, D. J. The developmental origins of chronic adult disease. Acta Paediatr. Suppl. 93, 26–33 (2004).

    Article  CAS  PubMed  Google Scholar 

  3. Painter, R. C., Roseboom, T. J. & Bleker, O. P. Prenatal exposure to the Dutch famine and disease in later life: an overview. Reprod. Toxicol. 20, 345–352 (2005).

    Article  CAS  PubMed  Google Scholar 

  4. Gluckman, P. D., Hanson, M. A. & Beedle, A. S. Early life events and their consequences for later disease: a life history and evolutionary perspective. Am. J. Hum. Biol. 19, 1–19 (2007). References 1–4 discuss evidence for the early origins of the adult disease susceptibility hypothesis.

    Article  PubMed  Google Scholar 

  5. St Clair, D. et al. Rates of adult schizophrenia following prenatal exposure to the Chinese famine of 1959–1961. JAMA 294, 557–562 (2005).

    Article  CAS  PubMed  Google Scholar 

  6. van Os, J. & Selten, J. P. Prenatal exposure to maternal stress and subsequent schizophrenia. The May 1940 invasion of The Netherlands. Br. J. Psychiatry 172, 324–326 (1998). References 5 and 6 discuss epidemiological evidence that the adult incidence of schizophrenia is significantly increased in humans who were exposed prenatally to famine conditions.

    Article  CAS  PubMed  Google Scholar 

  7. Li, E. Chromatin modification and epigenetic reprogramming in mammalian development. Nature Rev. Genet. 3, 662–673 (2002).

    Article  CAS  PubMed  Google Scholar 

  8. Klose, R. J. & Bird, A. P. Genomic DNA methylation: the mark and its mediators. Trends Biochem. Sci. 31, 89–97 (2006).

    Article  CAS  PubMed  Google Scholar 

  9. Talbert, P. B. & Henikoff, S. Spreading of silent chromatin: inaction at a distance. Nature Rev. Genet. 7, 793–803 (2006).

    Article  CAS  PubMed  Google Scholar 

  10. Richards, E. J. Inherited epigenetic variation — revisiting soft inheritance. Nature Rev. Genet. 7, 395–401 (2006).

    Article  CAS  PubMed  Google Scholar 

  11. Thorvaldsen, J. L., Verona, R. I. & Bartolomei, M. S. X-tra! X-tra! News from the mouse X chromosome. Dev. Biol. 298, 344–353 (2006).

    Article  CAS  PubMed  Google Scholar 

  12. Huynh, K. D. & Lee, J. T. X-chromosome inactivation: a hypothesis linking ontogeny and phylogeny. Nature Rev. Genet. 6, 410–418 (2005).

    Article  CAS  PubMed  Google Scholar 

  13. Lewis, A. & Reik, W. How imprinting centres work. Cytogenet. Genome Res. 113, 81–89 (2006).

    Article  CAS  PubMed  Google Scholar 

  14. Reik, W. & Walter, J. Genomic imprinting: parental influence on the genome. Nature Rev. Genet. 2, 21–32 (2001).

    Article  CAS  PubMed  Google Scholar 

  15. Falls, J. G., Pulford, D. J., Wylie, A. A. & Jirtle, R. L. Genomic imprinting: implications for human disease. Am. J. Pathol. 154, 635–647 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Murphy, S. K. & Jirtle, R. L. Imprinting evolution and the price of silence. BioEssays 25, 577–588 (2003).

    Article  CAS  PubMed  Google Scholar 

  17. Slotkin, R. K. & Martienssen, R. Transposable elements and the epigenetic regulation of the genome. Nature Rev. Genet. 8, 272–285 (2007).

    Article  CAS  PubMed  Google Scholar 

  18. Wolff, G. L., Kodell, R. L., Moore, S. R. & Cooney, C. A. Maternal epigenetics and methyl supplements affect agouti gene expression in Avy/a mice. FASEB J. 12, 949–957 (1998).

    Article  CAS  PubMed  Google Scholar 

  19. Waterland, R. A. & Jirtle, R. L. Transposable elements: targets for early nutritional effects on epigenetic gene regulation. Mol. Cell. Biol. 23, 5293–5300 (2003). This study demonstrates that maternal methyl donor supplementation during gestation can alter offspring phenotype by methylating the epigenome.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Dolinoy, D. C., Weidman, J. R., Waterland, R. A. & Jirtle, R. L. Maternal genistein alters coat color and protects Avy mouse offspring from obesity by modifying the fetal epigenome. Environ. Health Perspect. 114, 567–572 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Waterland, R. A. et al. Maternal methyl supplements increase offspring DNA methylation at Axin fused. Genesis 44, 401–406 (2006).

    Article  CAS  PubMed  Google Scholar 

  22. Waterland, R. A., Lin, J. R., Smith, C. A. & Jirtle, R. L. Post-weaning diet affects genomic imprinting at the insulin-like growth factor 2 (IGF2) locus. Hum. Mol. Genet. 15, 705–716 (2006).

    Article  CAS  PubMed  Google Scholar 

  23. Li, S. et al. Neonatal diethylstilbestrol exposure induces persistent elevation of c-fos expression and hypomethylation in its exon-4 in mouse uterus. Mol. Carcinog. 38, 78–84 (2003).

    Article  CAS  PubMed  Google Scholar 

  24. Ho, S. M., Tang, W. Y., Belmonte de Frausto, J. & Prins, G. S. Developmental exposure to estradiol and bisphenol A increases susceptibility to prostate carcinogenesis and epigenetically regulates phosphodiesterase type 4 variant 4. Cancer Res. 66, 5624–5632 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Anway, M. D. & Skinner, M. K. Epigenetic transgenerational actions of endocrine disruptors. Endocrinology 147, S43–S49 (2006).

    Article  CAS  PubMed  Google Scholar 

  26. Weaver, I. C. G. et al. Epigenetic programming by maternal behavior. Nature Neurosci. 7, 847–854 (2004).

    Article  CAS  PubMed  Google Scholar 

  27. Weaver, I. C. et al. Reversal of maternal programming of stress responses in adult offspring through methyl supplementation: altering epigenetic marking later in life. J. Neurosci. 25, 11045–11054 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Niemitz, E. L. & Feinberg, A. P. Epigenetics and assisted reproductive technology: a call for investigation. Am. J. Hum. Genet. 74, 599–609 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Rossignol, S. et al. The epigenetic imprinting defect of patients with Beckwith–Wiedemann syndrome born after assisted reproductive technology is not restricted to the 11p15 region. J. Med. Genet. 43, 902–907 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Koturbash, I. et al. Epigenetic dysregulation underlies radiation-induced transgenerational genome instability in vivo. Int. J. Radiat. Oncol. Biol. Phys. 66, 327–330 (2006).

    Article  CAS  PubMed  Google Scholar 

  31. Morgan, H. D., Sutherland, H. G. E., Martin, D. I. K. & Whitelaw, E. Epigenetic inheritance at the agouti locus in the mouse. Nature Genet. 23, 314–318 (1999). This study demonstrates the maternal inheritance of an epigenetic modification at the agouti locus in mice.

    Article  CAS  PubMed  Google Scholar 

  32. Lane, N. et al. Resistance of IAPs to methylation reprogramming may provide a mechanism for epigenetic inheritance in the mouse. Genesis 35, 88–93 (2003).

    Article  CAS  PubMed  Google Scholar 

  33. Rakyan, V. K. et al. Transgenerational inheritance of epigenetic states at the murine Axin(Fu) allele occurs after maternal and paternal transmission. Proc. Natl Acad. Sci. USA 100, 2538–2543 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Anway, M. D., Cupp, A. S., Uzumcu, M. & Skinner, M. K. Epigenetic transgenerational actions of endocrine disruptors and male fertility. Science 308, 1466–1469 (2005). This study demonstrates the ability of environmental factors to induce an epigenetic transgenerational disease phenotype for four generations.

    Article  CAS  PubMed  Google Scholar 

  35. Pembrey, M. E. et al. Sex-specific, male-line transgenerational responses in humans. Eur. J. Hum. Genet. 14, 159–166 (2006). This study demonstrates an inherited disease phenotype in humans that is potentially induced by an epigenetic phenomena.

    Article  PubMed  Google Scholar 

  36. Vasicek, T. J. et al. Two dominant mutations in the mouse fused gene are the result of transposon insertions. Genetics 147, 777–786 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Suter, C. M., Martin, D. I. & Ward, R. L. Germline epimutation of MLH1 in individuals with multiple cancers. Nature Genet. 36, 497–501 (2004).

    Article  CAS  PubMed  Google Scholar 

  38. Chan, T. L. et al. Heritable germline epimutation of MSH2 in a family with hereditary nonpolyposis colorectal cancer. Nature Genet. 38, 1178–1183 (2006).

    Article  CAS  PubMed  Google Scholar 

  39. Esteller, M. Epigenetics provides a new generation of oncogenes and tumour-suppressor genes. Br. J. Cancer 94, 179–183 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Yoo, C. B. & Jones, P. A. Epigenetic therapy of cancer: past, present and future. Nature Rev. Drug Discov. 5, 37–50 (2006).

    Article  CAS  Google Scholar 

  41. Baylin, S. B. & Ohm, J. E. Epigenetic gene silencing in cancer — a mechanism for early oncogenic pathway addiction? Nature Rev. Cancer 6, 107–116 (2006).

    Article  CAS  Google Scholar 

  42. Duhl, D. M., Vrieling, H., Miller, K. A., Wolff, G. L. & Barsh, G. S. Neomorphic agouti mutations in obese yellow mice. Nature Genet. 8, 59–65 (1994). These authors show that the Avy allele results from the insertion of an intracisternal A particle upstream of the agouti gene.

    Article  CAS  PubMed  Google Scholar 

  43. Druker, R., Bruxner, T. J., Lehrbach, N. J. & Whitelaw, E. Complex patterns of transcription at the insertion site of a retrotransposon in the mouse. Nucl. Acids Res. 32, 5800–5808 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Rakyan, V. K., Blewitt, M. E., Druker, R., Preis, J. I. & Whitelaw, E. Metastable epialleles in mammals. Trends Genet. 18, 348–351 (2002).

    Article  CAS  PubMed  Google Scholar 

  45. Druker, R. & Whitelaw, E. Retrotransposon-derived elements in the mammalian genome: a potential source of disease. Inherit. Metab. Dis. 27, 319–330 (2004).

    Article  CAS  Google Scholar 

  46. Miltenberger, R. J., Mynatt, R. L., Wilkinson, J. E. & Woychik, R. P. The role of the agouti gene in the Yellow Obese Syndrome. J. Nutr. 127, 1902S–1907S (1997).

    Article  CAS  PubMed  Google Scholar 

  47. Cooney, C. A., Dave, A. A. & Wolff, G. L. Maternal methyl supplements in mice affect epigenetic variation and DNA methylation of offspring. J. Nutr. 132, 2393S–2400S (2002).

    Article  CAS  PubMed  Google Scholar 

  48. Cropley, J. E., Suter, C. M., Beckman, K. B. & Martin, D. I. Germ-line epigenetic modification of the murine Avy allele by nutritional supplementation. Proc. Natl Acad. Sci. USA 103, 17308–17312 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Haig, D. & Graham, C. Genomic imprinting and the strange case of the insulin-like growth factor II receptor. Cell 64, 1045–1046 (1991). These authors propose that genomic imprinting evolved because of a parental genetic battle to control the amount of nutrients that is extracted from the mother by the offspring.

    Article  CAS  PubMed  Google Scholar 

  50. Wilkins, J. F. & Haig, D. What good is genomic imprinting: the function of parent-specific gene expression. Nature Rev. Genet. 4, 359–368 (2003).

    Article  CAS  PubMed  Google Scholar 

  51. DeChiara, T. M., Robertson, E. & Efstratiadis, A. Parental imprinting of the mouse insulin-like growth factor II gene. Cell 64, 849–859 (1991).

    Article  CAS  PubMed  Google Scholar 

  52. Barlow, D. P., Stoger, R., Herrmann, B. G., Saito, K. & Schweifer, N. The mouse insulin-like growth factor type-2 receptor is imprinted and closely linked to the Tme locus. Nature 349, 84–87 (1991). References 51 and 52 report the first-identified imprinted genes.

    Article  CAS  PubMed  Google Scholar 

  53. Killian, J. K. et al. M6p/IGF2R imprinting evolution in mammals. Mol. Cell 5, 707–716 (2000). This paper demonstrates that genomic imprinting evolved approximately 180 million years ago with the advent of live birth in therian mammals.

    Article  CAS  PubMed  Google Scholar 

  54. Evans, H. K., Weidman, J. R., Cowley, D. O. & Jirtle, R. L. Comparative phylogenetic analysis of Blcap/Nnat reveals eutherian-specific imprinted gene. Mol. Biol. Evol. 22, 1740–1748 (2005).

    Article  CAS  PubMed  Google Scholar 

  55. Weidman, J. R., Maloney, K. A. & Jirtle, R. L. Comparative phylogenetic analysis reveals multiple non-imprinted isoforms of opossum DLK1. Mamm. Genome 17, 157–167 (2006).

    Article  CAS  PubMed  Google Scholar 

  56. Suzuki, S. et al. Genomic imprinting of IGF2, p57(KIP2) and PEG1/MEST in a marsupial, the tammar wallaby. Mech. Dev. 122, 213–222 (2005).

    Article  CAS  PubMed  Google Scholar 

  57. Killian, J. K. et al. Divergent evolution in M6P/IGF2R imprinting from the Jurassic to the Quaternary. Hum. Mol. Genet. 10, 1721–1728 (2001).

    Article  CAS  PubMed  Google Scholar 

  58. De Souza, A. T., Hankins, G. R., Washington, M. K., Orton, T. C. & Jirtle, R. L. M6P/IGF2R gene is mutated in human hepatocellular carcinomas with loss of heterozygosity. Nature Genet. 11, 447–449 (1995).

    Article  CAS  PubMed  Google Scholar 

  59. Weksberg, R., Shuman, C. & Smith, A. C. Beckwith–Wiedemann syndrome. Am. J. Med. Genet. C Semin. Med. Genet. 137, 12–23 (2005).

    Article  Google Scholar 

  60. Kantor, B., Shemer, R. & Razin, A. The Prader-Willi–Angelman imprinted domain and its control center. Cytogenet. Genome Res. 113, 300–305 (2006).

    Article  CAS  PubMed  Google Scholar 

  61. Badcock, C. & Crespi, B. Imbalanced genomic imprinting in brain development: an evolutionary basis for the aetiology of autism. J. Evol. Biol. 19, 1007–1032 (2006). These authors propose that human neurological disorders, such as autism, result from an imbalanced expression of imprinted genes during development.

    Article  CAS  PubMed  Google Scholar 

  62. Morison, I. M., Ramsay, J. P. & Spencer, H. G. A census of mammalian imprinting. Trends Genet. 21, 457–465 (2005).

    Article  CAS  PubMed  Google Scholar 

  63. Feinberg, A. P., Ohlsson, R. & Henikoff, S. The epigenetic progenitor origin of human cancer. Nature Rev. Genet. 7, 21–33 (2006).

    Article  CAS  PubMed  Google Scholar 

  64. Feinberg, A. P. A genetic approach to cancer epigenetics. Cold Spring Harb. Symp. Quant. Biol. 70, 335–341 (2005).

    Article  CAS  PubMed  Google Scholar 

  65. Knudson, A. G. Two genetic hits (more or less) to cancer. Nature Rev. Cancer 1, 157–162 (2001).

    Article  CAS  Google Scholar 

  66. Cui, H. et al. Loss of IGF2 imprinting: a potential marker of colorectal cancer risk. Science 299, 1753–1755 (2003). This paper reports that some humans have IGF2 LOI in peripheral lymphocytes, which is correlated with biallelic expression in normal colonic mucosa and a personal history of colorectal cancer.

    Article  CAS  PubMed  Google Scholar 

  67. Cruz-Correa, M. et al. Loss of imprinting of insulin growth factor II gene: a potential heritable biomarker for colon neoplasia predisposition. Gastroenterology 126, 964–970 (2004).

    Article  CAS  PubMed  Google Scholar 

  68. Jirtle, R. L. IGF2 loss of imprinting: a potential heritable risk factor for colorectal cancer. Gastroenterology 126, 1190–1193 (2004).

    Article  CAS  PubMed  Google Scholar 

  69. Oates, N. A. et al. Increased DNA methylation at the AXIN1 gene in a monozygotic twin from a pair discordant for a caudal duplication anomaly. Am. J. Hum. Genet. 79, 155–162 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Ikeda, M., Tamura, M., Yamashita, J., Suzuki, C. & Tomita, T. Repeated in utero and lactational 2,3,7,8-tetrachlorodibenzo-p-dioxin exposure affects male gonads in offspring, leading to sex ratio changes in F2 progeny. Toxicol. Appl. Pharmacol. 206, 351–355 (2005).

    Article  CAS  PubMed  Google Scholar 

  71. Blatt, J., Van Le, L., Weiner, T. & Sailer, S. Ovarian carcinoma in an adolescent with transgenerational exposure to diethylstilbestrol. J. Pediatr. Hematol. Oncol. 25, 635–636 (2003).

    Article  PubMed  Google Scholar 

  72. Dubrova, Y. E. Radiation-induced transgenerational instability. Oncogene 22, 7087–7093 (2003).

    Article  CAS  PubMed  Google Scholar 

  73. Cheng, R. Y., Hockman, T., Crawford, E., Anderson, L. M. & Shiao, Y. H. Epigenetic and gene expression changes related to transgenerational carcinogenesis. Mol. Carcinog. 40, 1–11 (2004).

    Article  CAS  PubMed  Google Scholar 

  74. Hemmings, D. G., Veerareddy, S., Baker, P. N. & Davidge, S. T. Increased myogenic responses in uterine but not mesenteric arteries from pregnant offspring of diet-restricted rat dams. Biol. Reprod. 72, 997–1003 (2005).

    Article  CAS  PubMed  Google Scholar 

  75. Ferguson, L. R., Karunasinghe, N. & Philpott, M. Epigenetic events and protection from colon cancer in New Zealand. Environ. Mol. Mutagen. 44, 36–43 (2004).

    Article  CAS  PubMed  Google Scholar 

  76. Csaba, G. & Karabelyos, C. Transgenerational effect of a single neonatal benzpyrene treatment (imprinting) on the sexual behavior of adult female rats. Hum. Exp. Toxicol. 16, 553–556 (1997).

    Article  CAS  PubMed  Google Scholar 

  77. Fujii, T. Transgenerational effects of maternal exposure to chemicals on the functional development of the brain in the offspring. Cancer Causes Control 8, 524–528 (1997).

    Article  CAS  PubMed  Google Scholar 

  78. Brucker-Davis, F. Effects of environmental synthetic chemicals on thyroid function. Thyroid 8, 827–856 (1998).

    Article  CAS  PubMed  Google Scholar 

  79. Giusti, R. M., Iwamoto, K. & Hatch, E. E. Diethylstilbestrol revisited: a review of the long-term health effects. Ann. Intern. Med. 122, 778–788 (1995).

    Article  CAS  PubMed  Google Scholar 

  80. Klip, H. et al. Hypospadias in sons of women exposed to diethylstilbestrol in utero: a cohort study. Lancet 359, 1102–1107 (2002).

    Article  CAS  PubMed  Google Scholar 

  81. Parks, L. G. et al. The plasticizer diethylhexyl phthalate induces malformations by decreasing fetal testosterone synthesis during sexual differentiation in the male rat. Toxicol. Sci. 58, 339–349 (2000).

    Article  CAS  PubMed  Google Scholar 

  82. Steinhardt, G. F. Endocrine disruption and hypospadias. Adv. Exp. Med. Biol. 545, 203–215 (2004).

    Article  PubMed  Google Scholar 

  83. Ruden, D. M., Xiao, L., Garfinkel, M. D. & Lu, X. Hsp90 and environmental impacts on epigenetic states: a model for the trans-generational effects of diethylstibesterol on uterine development and cancer. Hum. Mol. Genet. 14, R149–R155 (2005).

  84. Matta, M. B., Linse, J., Cairncross, C., Francendese, L. & Kocan, R. M. Reproductive and transgenerational effects of methylmercury or Aroclor 1268 on Fundulus heteroclitus. Environ. Toxicol. Chem. 20, 327–335 (2001).

    Article  CAS  PubMed  Google Scholar 

  85. Omholt, S. W. & Amdam, G. V. Epigenetic regulation of aging in honeybee workers. Sci. Aging Knowledge Environ. 2004, pe28 (2004).

  86. Ottinger, M. A. et al. Assessing the consequences of the pesticide methoxychlor: neuroendocrine and behavioral measures as indicators of biological impact of an estrogenic environmental chemical. Brain Res. Bull. 65, 199–209 (2005).

    Article  CAS  PubMed  Google Scholar 

  87. Seidl, M. D., Paul, R. J. & Pirow, R. Effects of hypoxia acclimation on morpho-physiological traits over three generations of Daphnia magna. J. Exp. Biol. 208, 2165–2175 (2005).

    Article  CAS  PubMed  Google Scholar 

  88. Foran, C. M., Peterson, B. N. & Benson, W. H. Transgenerational and developmental exposure of Japanese medaka (Oryzias latipes) to ethinylestradiol results in endocrine and reproductive differences in the response to ethinylestradiol as adults. Toxicol. Sci. 68, 389–402 (2002).

    Article  CAS  PubMed  Google Scholar 

  89. Anderson, C. M., Lopez, F., Zimmer, A. & Benoit, J. N. Placental insufficiency leads to developmental hypertension and mesenteric artery dysfunction in two generations of Sprague–Dawley rat offspring. Biol. Reprod. 74, 538–544 (2006).

    Article  CAS  PubMed  Google Scholar 

  90. Csaba, G. & Inczefi-Gonda, A. Transgenerational effect of a single neonatal benzpyrene treatment on the glucocorticoid receptor of the rat thymus. Hum. Exp. Toxicol. 17, 88–92 (1998).

    Article  CAS  PubMed  Google Scholar 

  91. Newbold, R. R., Padilla-Banks, E. & Jefferson, W. N. Adverse effects of the model environmental estrogen diethylstilbestrol are transmitted to subsequent generations. Endocrinology 147, S11–S17 (2006).

    Article  CAS  PubMed  Google Scholar 

  92. Popova, N. V. Transgenerational effect of orthoaminoasotoluol in mice. Cancer Lett. 46, 203–206 (1989).

    Article  CAS  PubMed  Google Scholar 

  93. Zambrano, E. et al. Sex differences in transgenerational alterations of growth and metabolism in progeny (F2) of female offspring (F1) of rats fed a low protein diet during pregnancy and lactation. J. Physiol. 566, 225–236 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Cesani, M. F. et al. Effect of undernutrition on the cranial growth of the rat. An intergenerational study. Cells Tissues Organs 174, 129–135 (2003).

    Article  PubMed  Google Scholar 

  95. Turusov, V. S., Nikonova, T. V. & Parfenov, Y. Increased multiplicity of lung adenomas in five generations of mice treated with benz(a)pyrene when pregnant. Cancer Lett. 55, 227–231 (1990).

    Article  CAS  PubMed  Google Scholar 

  96. Anway, M. D., Leathers, C. & Skinner, M. K. Endocrine disruptor vinclozolin induced epigenetic transgenerational adult-onset disease. Endocrinology 147, 5515–5523 (2006).

    Article  CAS  PubMed  Google Scholar 

  97. Chang, H. S., Anway, M. D., Rekow, S. S. & Skinner, M. K. Transgenerational epigenetic imprinting of the male germline by endocrine disruptor exposure during gonadal sex determination. Endocrinology 147, 5524–5541 (2006). This report demonstrates the ability of vinclozolin to induce the reprogramming of the germ line, and the formation of genes and DNA sequences that contain paternal-allele alterations in DNA methylation associated with transgenerational disease.

    Article  CAS  PubMed  Google Scholar 

  98. Durcova-Hills, G. et al. Influence of sex chromosome constitution on the genomic imprinting of germ cells. Proc. Natl Acad. Sci. USA 103, 11184–11188 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Forum, T. C. News and Information. J. Radiol. Prot. 25, 499–502 (2005).

    Article  Google Scholar 

  100. Allegrucci, C., Thurston, A., Lucas, E. & Young, L. Epigenetics and the germline. Reproduction 129, 137–149 (2005).

    Article  CAS  PubMed  Google Scholar 

  101. McCarrey, J. R., Geyer, C. B. & Yoshioka, H. Epigenetic regulation of testis-specific gene expression. Ann. NY Acad. Sci. 1061, 226–242 (2005).

    Article  CAS  PubMed  Google Scholar 

  102. Trasler, J. M. Origin and roles of genomic methylation patterns in male germ cells. Semin. Cell Dev. Biol. 9, 467–474 (1998).

    Article  CAS  PubMed  Google Scholar 

  103. Weaver, I. C., Meaney, M. J. & Szyf, M. Maternal care effects on the hippocampal transcriptome and anxiety-mediated behaviors in the offspring that are reversible in adulthood. Proc. Natl Acad. Sci. USA 103, 3480–3485 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Hurst, G. D. & Werren, J. H. The role of selfish genetic elements in eukaryotic evolution. Nature Rev. Genet. 2, 597–606 (2001).

    Article  CAS  PubMed  Google Scholar 

  105. Bestor, T. H. Cytosine methylation mediates sexual conflict. Trends Genet. 19, 185–190 (2003).

    Article  CAS  PubMed  Google Scholar 

  106. Luedi, P. P., Hartemink, A. J. & Jirtle, R. L. Genome-wide prediction of imprinted murine genes. Genome Res. 15, 875–884 (2005). These authors demonstrate that imprinted genes and their parental expression bias can be predicted genome-wide with the use of machine learning algorithms.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Waddington, C. H. Organisers and Genes (Cambridge Univ. Press, Cambridge, 1940).

    Google Scholar 

  108. Holliday, R. & Pugh, J. E. DNA modification mechanisms and gene activity during development. Science 187, 226–232 (1975).

    Article  CAS  PubMed  Google Scholar 

  109. Willard, H. F., Brown, C. J., Carrel, L., Hendrich, B. & Miller, A. P. Epigenetic and chromosomal control of gene expression: molecular and genetic analysis of X chromosome inactivation. Cold Spring Harb. Symp. Quant. Biol. 58, 315–322 (1993).

    Article  CAS  PubMed  Google Scholar 

  110. Monk, M. Genomic imprinting. Genes Dev. 2, 921–925 (1988).

    Article  CAS  PubMed  Google Scholar 

  111. Wolffe, A. P. & Matzke, M. A. Epigenetics: regulation through repression. Science 286, 481–486 (1999).

    Article  CAS  PubMed  Google Scholar 

  112. Murrell, A., Rakyan, V. K. & Beck, S. From genome to epigenome. Hum. Mol. Genet. 14, R3–R10 (2005).

    Article  CAS  PubMed  Google Scholar 

  113. Kishino, T. Imprinting in neurons. Cytogenet. Genome Res. 113, 209–214 (2006).

    Article  CAS  PubMed  Google Scholar 

  114. Vu, T. H., Jirtle, R. L. & Hoffman, A. R. Cross-species clues of an epigenetic imprinting regulatory code for the IGF2R gene. Cytogenet. Genome Res. 113, 202–208 (2006).

    Article  CAS  PubMed  Google Scholar 

  115. Mayer, W., Niveleau, A., Walter, J., Fundele, R. & Haaf, T. Demethylation of the zygotic paternal genome. Nature 403, 501–552 (2000).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors wish to thank D. Dolinoy for critically reading the manuscript and for her helpful suggestions. We thank J. Griffin for assistance in the preparation of this manuscript. This work was supported by grants from the US Department of Energy and the US National Institutes of Health.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Randy L. Jirtle.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Related links

Related links

FURTHER INFORMATION

Geneimprint

Glossary

Epigenetic

Refers to mitotically or meiotically heritable changes in gene expression that do not involve a change in DNA sequence.

DNA methylation

DNA methylation occurs predominantly in repetitive genomic regions that contain CpG residues. DNA methylation represses transcription directly by inhibiting the binding of specific transcription factors, and indirectly by recruiting methyl-CpG-binding proteins and their associated repressive chromatin-remodelling activities.

Histone modifications

Histones undergo post-translational modifications that alter their interaction with DNA and nuclear proteins. In particular, the tails of histones H3 and H4 can be covalently modified at several residues. Modifications of the tail include methylation, acetylation, phosphorylation and ubiquitination, and influence several biological processes, including gene expression, DNA repair and chromosome condensation.

MicroRNAs

Endogenous small RNAs of 22 nucleotides in length that act as a cellular rheostat for fine-tuning gene expression during development and differentiation. They target the 3′ UTRs of mRNAs with which they share partial sequence complementarity, leading to post-transcriptional gene silencing through translational repression. When a microRNA has complete sequence complementarity with a target mRNA, it instead directs cleavage of the transcript.

X-chromosome inactivation

The process that occurs in female mammals by which gene expression from one of the pair of X chromosomes is downregulated to match the levels of gene expression from the single X chromosome that is present in males. The inactivation process involves a range of epigenetic mechanisms on the inactivated chromosome, including changes in DNA methylation and histone modifications.

Genomic imprinting

The epigenetic marking of a locus on the basis of parental origin, which results in monoallelic gene expression.

Epigenome

The global epigenetic patterns that distinguish or are variable between cell types. These patterns include DNA methylation, histone modifications and chromatin-associated proteins.

Melanocyte

A specialized cell type, lying at the boundary between the dermis and epidermis, in which the pigment melanin is synthesized.

Embryonic stem cell

A type of pluripotent stem cell that is derived from the inner cell mass of the early embryo. Pluripotent cells are capable of generating virtually all cell types of the organism.

Angelman syndrome

A defect that is caused by the loss of expression of a maternally expressed gene, UBE3A, which is imprinted only in the brain and encodes an E3 ubiquitin ligase. Angelman syndrome occurs in 1 in 15,000 births and its main characteristics include mental retardation, speech impairment and behavioural abnormalities.

Prader–Willi syndrome

The molecular defect that causes this syndrome is complex and involves defects that affect an 2 Mb imprinted domain that contains both paternally and maternally expressed genes. Prader–Willi syndrome occurs in 1 in 20,000 births and is characterized by a failure to thrive during infancy, hyperphagia and obesity during early childhood, mental retardation and behavioural problems.

Beckwith–Weidemann syndrome

A predominantly maternally transmitted disorder, involving fetal and postnatal overgrowth and a predisposition to embryonic tumours. The Beckwith–Weidemann syndrome locus includes several imprinted genes, including IGF2, H19 and KCNQ1, and loss of imprinting at IGF2 is seen in 20% of cases.

Bisulphite conversion

A technique that is used to identify methylcytosines. The approach depends on the relative resistance of the conversion of methylcytosine to uracil compared with cytosine. Conversion can be followed by PCR amplification and sequencing of the DNA. The persistence of a cytosine, instead of a thymine, being detected, reflects the methylation of the cytosine in the starting DNA sample.

Machine learning

The ability of a program to learn from experience — that is, to modify its execution on the basis of newly acquired information. In bioinformatics, neural networks and Monte Carlo Markov chains are well-known examples.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Jirtle, R., Skinner, M. Environmental epigenomics and disease susceptibility. Nat Rev Genet 8, 253–262 (2007). https://doi.org/10.1038/nrg2045

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrg2045

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing