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:

Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals

Abstract

Cells of a multicellular organism are genetically homogeneous but structurally and functionally heterogeneous owing to the differential expression of genes. Many of these differences in gene expression arise during development and are subsequently retained through mitosis. Stable alterations of this kind are said to be 'epigenetic', because they are heritable in the short term but do not involve mutations of the DNA itself. Research over the past few years has focused on two molecular mechanisms that mediate epigenetic phenomena: DNA methylation and histone modifications. Here, we review advances in the understanding of the mechanism and role of DNA methylation in biological processes. Epigenetic effects by means of DNA methylation have an important role in development but can also arise stochastically as animals age. Identification of proteins that mediate these effects has provided insight into this complex process and diseases that occur when it is perturbed. External influences on epigenetic processes are seen in the effects of diet on long-term diseases such as cancer. Thus, epigenetic mechanisms seem to allow an organism to respond to the environment through changes in gene expression. The extent to which environmental effects can provoke epigenetic responses represents an exciting area of future research.

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

Access options

Buy this article

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

Figure 1: Polarization of active and silent chromatin states owing to feedback loops between intermediate states.
Figure 2: Epigenetic effects on mouse coat color.
Figure 3: Transitions between silent and transcriptionally competent chromatin states are dynamic and depend on a balance between factors that sustain a silent state, such as HDACs, and those that promote a transcriptionally active state, such as HATs175.

Similar content being viewed by others

References

  1. Waddington, C. The genetic control of wing development in Drosophila. J. Genet. 41, 75–80 (1940).

    Article  Google Scholar 

  2. Issa, J.P. CpG-island methylation in aging and cancer. Curr. Top. Microbiol. Immunol. 249, 101–118 (2000).

    CAS  PubMed  Google Scholar 

  3. Jähner, D. et al. De novo methylation and expression of retroviral genomes during mouse embryogenesis. Nature 298, 623–628 (1982).

    Article  PubMed  Google Scholar 

  4. Riggs, A.D. X inactivation, differentiation, and DNA methylation. Cytogenet. Cell Genet. 14, 9–25 (1975).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  7. Urnov, F.D. & Wolffe, A.P. Above and within the genome: epigenetics past and present. J. Mammary Gland Biol. Neoplasia 6, 153–167 (2001).

    Article  CAS  PubMed  Google Scholar 

  8. Jones, P.A. & Takai, D. The role of DNA methylation in mammalian epigenetics. Science 293, 1068–1070 (2001).

    Article  CAS  PubMed  Google Scholar 

  9. Ferguson-Smith, A.C. & Surani, M.A. Imprinting and the epigenetic asymmetry between parental genomes. Science 293, 1086–1089 (2001).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  11. Bird, A. DNA methylation patterns and epigenetic memory. Genes Dev. 16, 6–21 (2002).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  13. Jaenisch, R. DNA methylation and imprinting: why bother? Trends Genet. 13, 323–329 (1997).

    Article  CAS  PubMed  Google Scholar 

  14. Mayer, W., Niveleau, A., Walter, J., Fundele, R. & Haaf, T. Demethylation of the zygotic paternal genome. Nature 403, 501–502 (2000).

    Article  CAS  PubMed  Google Scholar 

  15. Oswald, J. et al. Active demethylation of the paternal genome in the mouse zygote. Curr. Biol. 10, 475–478 (2000).

    Article  CAS  PubMed  Google Scholar 

  16. Ehrlich, M. et al. Amount and distribution of 5-methylcytosine in human DNA from different types of tissues of cells. Nucleic Acids Res. 10, 2709–2721 (1982).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Jones, P. et al. De novo methylation of the MyoD1 CpG island during the establisment of immortal cell lines. Proc. Natl. Acad. Sci. USA 87, 6117–6121 (1990).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Kawai, J. et al. Comparison of DNA methylation patterns among mouse cell lines by restriction landmark genomic scanning. Mol. Cell. Biol. 14, 7421–7427 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Bestor, T.H. The DNA methyltransferases of mammals. Hum. Mol. Genet. 9, 2395–2402 (2000).

    Article  CAS  PubMed  Google Scholar 

  20. Li, E., Bestor, T.H. & Jaenisch, R. Targeted mutation of the DNA methyltransferase gene results in embryonic lethality. Cell 69, 915–926 (1992).

    Article  CAS  PubMed  Google Scholar 

  21. Lei, H. et al. De novo DNA cytosine methyltransferase activities in mouse embryonic stem cells. Development 122, 3195–3205 (1996).

    CAS  PubMed  Google Scholar 

  22. Stancheva, I. & Meehan, R.R. Transient depletion of xDnmt1 leads to premature gene activation in Xenopus embryos. Genes Dev. 14, 313–327 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Howell, C. et al. Genomic imprinting disrupted by a maternal effect mutation in the Dnmt1 gene. Cell 104, 829–838 (2001).

    Article  CAS  PubMed  Google Scholar 

  24. Okano, M., Bell, D.W., Haber, D.A. & Li, E. DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell 99, 247–257 (1999).

    Article  CAS  PubMed  Google Scholar 

  25. Lyko, F. et al. Mammalian (cytosine-5) methyltransferases cause genomic DNA methylation and lethality in Drosophila. Nat. Genet. 23, 363–366 (1999).

    Article  CAS  PubMed  Google Scholar 

  26. Bourc'his, D., Xu, G.L., Lin, C.S., Bollman, B. & Bestor, T.H. Dnmt3L and the establishment of maternal genomic imprints. Science 294, 2536–2539 (2001).

    Article  CAS  PubMed  Google Scholar 

  27. Hata, K., Okano, M., Lei, H. & Li, E. Dnmt3L cooperates with the Dnmt3 family of de novo DNA methyltransferases to establish maternal imprints in mice. Development 129, 1983–1993 (2002).

    CAS  PubMed  Google Scholar 

  28. Okano, M., Xie, S. & Li, E. Dnmt2 is not required for de novo and maintenance methylation of viral DNA in embryonic stem cells. Nucleic Acids Res. 26, 2536–2540 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Lyko, F., Ramsahoye, B.H. & Jaenisch, R. DNA methylation in Drosophila melanogaster. Nature 408, 538–540 (2000).

    Article  CAS  PubMed  Google Scholar 

  30. Gowher, H., Leismann, O. & Jeltsch, A. DNA of Drosophila melanogaster contains 5-methylcytosine. EMBO J. 19, 6918–6923 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Panning, B. & Jaenisch, R. DNA hypomethylation can activate Xist expression and silence X-linked genes. Genes Dev. 10, 1991–2002 (1996).

    Article  CAS  PubMed  Google Scholar 

  32. Stancheva, I., Hensey, C. & Meehan, R.R. Loss of the maintenance methyltransferase, xDnmt1, induces apoptosis in Xenopus embryos. EMBO J. 20, 1963–1973 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Jackson-Grusby, L. et al. Loss of genomic methylation causes p53-dependent apoptosis and epigenetic deregulation. Nat. Genet. 27, 31–39 (2001).

    Article  CAS  PubMed  Google Scholar 

  34. Rhee, I. et al. DNMT1 and DNMT3b cooperate to silence genes in human cancer cells. Nature 416, 552–556 (2002).

    Article  CAS  PubMed  Google Scholar 

  35. Walsh, C.P., Chaillet, J.R. & Bestor, T.H. Transcription of IAP endogenous retroviruses is constrained by cytosine methylation. Nat. Genet. 20, 116–117 (1998).

    Article  CAS  PubMed  Google Scholar 

  36. Fan, G. et al. DNA hypomethylation perturbs the function and survival of CNS neurons in postnatal animals. J. Neurosci. 21, 788–797 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Walsh, C.P. & Bestor, T.H. Cytosine methylation and mammalian development. Genes Dev. 13, 26–34 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Bird, A. CpG islands as gene markers in the vertebrate nucleus. Trends Genet. 3, 342–346 (1987).

    Article  CAS  Google Scholar 

  39. Bird, A.P. CpG-rich islands and the function of DNA methylation. Nature 321, 209–213 (1986).

    Article  CAS  PubMed  Google Scholar 

  40. Gardiner-Garden, M. & Frommer, M. CpG islands in vertebrate genomes. J. Mol. Biol. 196, 261–282 (1987).

    Article  CAS  PubMed  Google Scholar 

  41. Larsen, F., Gundersen, G., Lopez, R. & Prydz, H. CpG islands as gene markers in the human genome. Genomics 13, 1095–1107 (1992).

    Article  CAS  PubMed  Google Scholar 

  42. Macleod, D., Ali, R.R. & Bird, A. An alternative promoter in the mouse major histocompatibility complex class II I-Aβ gene: implications for the origin of CpG islands. Mol. Cell. Biol. 18, 4433–4443 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Adachi, N. & Lieber, M.R. Bidirectional gene organization: a common architectural feature of the human genome. Cell 109, 807–809 (2002).

    Article  CAS  PubMed  Google Scholar 

  44. Voo, K.S., Carlone, D.L., Jacobsen, B.M., Flodin, A. & Skalnik, D.G. Cloning of a mammalian transcriptional activator that binds unmethylated CpG motifs and shares a CXXC domain with DNA methyltransferase, human trithorax, and methyl-CpG binding domain protein 1. Mol. Cell. Biol. 20, 2108–2121 (2000).

    Article  CAS  PubMed  Google Scholar 

  45. Lee, J.H., Voo, K.S. & Skalnik, D.G. Identification and characterization of the DNA binding domain of CpG-binding protein. J. Biol. Chem. 276, 44669–44676 (2001).

    Article  CAS  PubMed  Google Scholar 

  46. Mohandas, T., Sparkes, R.S. & Shapiro, L.J. Reactivation of an inactive human X chromosome: evidence for X inactivation by DNA methylation. Science 211, 393–396 (1981).

    Article  CAS  PubMed  Google Scholar 

  47. Wolf, S.F., Jolly, D.J., Lunnen, K.D., Friedmann, T. & Migeon, B.R. Methylation of the hypoxanthine phosphoribosyltransferase locus on the human X chromosome: implications for X-chromosome inactivation. Proc. Natl. Acad. Sci. USA 81, 2806–2810 (1984).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Riggs, A.D., Xiong, Z., Wang, L. & LeBon, J.M. Methylation dynamics, epigenetic fidelity and X chromosome structure. Novartis Found. Symp. 214, 214–232 (1998).

    CAS  PubMed  Google Scholar 

  49. Brandeis, M. et al. Sp1 elements protect a CpG island from de novo methylation. Nature 371, 435–438 (1994).

    Article  CAS  PubMed  Google Scholar 

  50. Macleod, D., Charlton, J., Mullins, J. & Bird, A.P. Sp1 sites in the mouse aprt gene promoter are required to prevent methylation of the CpG island. Genes Dev. 8, 2282–2292 (1994).

    Article  CAS  PubMed  Google Scholar 

  51. Di Croce, L. et al. Methyltransferase recruitment and DNA hypermethylation of target promoters by an oncogenic transcription factor. Science 295, 1079–1082 (2002).

    Article  CAS  PubMed  Google Scholar 

  52. Santoro, R., Li, J. & Grummt, I. The nucleolar remodeling complex NoRC mediates heterochromatin formation and silencing of ribosomal gene transcription. Nat. Genet. 32, 393–396 (2002).

    Article  CAS  PubMed  Google Scholar 

  53. Fuks, F., Burgers, W.A., Godin, N., Kasai, M. & Kouzarides, T. Dnmt3a binds deacetylases and is recruited by a sequence-specific repressor to silence transcription. EMBO J. 20, 2536–2544 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Bachman, K.E., Rountree, M.R. & Baylin, S.B. Dnmt3a and Dnmt3b are transcriptional repressors that exhibit unique localization properties to heterochromatin. J. Biol. Chem. 276, 32282–32287 (2001).

    Article  CAS  PubMed  Google Scholar 

  55. Tamaru, H. & Selker, E.U. A histone H3 methyltransferase controls DNA methylation in Neurospora crassa. Nature 414, 277–283 (2001).

    Article  CAS  PubMed  Google Scholar 

  56. Jackson, J.P., Lindroth, A.M., Cao, X. & Jacobsen, S.E. Control of CpNpG DNA methylation by the KRYPTONITE histone H3 methyltransferase. Nature 416, 556–560 (2002).

    Article  CAS  PubMed  Google Scholar 

  57. Wolffe, A.P. & Guschin, D. Review: chromatin structural features and targets that regulate transcription. J. Struct. Biol. 129, 102–122 (2000).

    Article  CAS  PubMed  Google Scholar 

  58. Grewal, S.I. & Elgin, S.C. Heterochromatin: new possibilities for the inheritance of structure. Curr. Opin. Genet. Dev. 12, 178–187 (2002).

    Article  CAS  PubMed  Google Scholar 

  59. Watt, F. & Molloy, P. Cytosine methylation prevents binding to DNA of a HeLa cell transcription factor required for optimal expression of the adenovirus major late promoter. Genes Dev. 2, 1136–1143 (1988).

    Article  CAS  PubMed  Google Scholar 

  60. Bell, A.C., West, A.G. & Felsenfeld, G. The protein CTCF is required for the enhancer blocking activity of vertebrate insulators. Cell 98, 387–396 (1999).

    Article  CAS  PubMed  Google Scholar 

  61. Ohlsson, R., Renkawitz, R. & Lobanenkov, V. CTCF is a uniquely versatile transcription regulator linked to epigenetics and disease. Trends Genet. 17, 520–527 (2001).

    Article  CAS  PubMed  Google Scholar 

  62. Hark, A.T. et al. CTCF mediates methylation-sensitive enhancer-blocking activity at the H19/Igf2 locus. Nature 405, 486–489 (2000).

    Article  CAS  PubMed  Google Scholar 

  63. Tate, P.H. & Bird, A.P. Effects of DNA methylation on DNA-binding proteins and gene expression. Curr. Opin. Genet. Dev. 3, 226–231 (1993).

    Article  CAS  PubMed  Google Scholar 

  64. Lewis, J.D. et al. Purification, sequence, and cellular localization of a novel chromosomal protein that binds to methylated DNA. Cell 69, 905–914 (1992).

    Article  CAS  PubMed  Google Scholar 

  65. Hendrich, B. & Bird, A. Identification and characterization of a family of mammalian methyl-CpG binding proteins. Mol. Cell. Biol. 18, 6538–6547 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Prokhortchouk, A. et al. The p120 catenin partner Kaiso is a DNA methylation-dependent transcriptional repressor. Genes Dev. 15, 1613–1618 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Nan, X., Meehan, R.R. & Bird, A. Dissection of the methyl-CpG binding domain from the chromosomal protein MeCP2. Nucleic Acids Res. 21, 4886–4892 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Billard, L.M., Magdinier, F., Lenoir, G.M., Frappart, L. & Dante, R. MeCP2 and MBD2 expression during normal and pathological growth of the human mammary gland. Oncogene 21, 2704–2712 (2002).

    Article  CAS  PubMed  Google Scholar 

  69. Rietveld, L.E., Caldenhoven, E. & Stunnenberg, H.G. In vivo repression of an erythroid-specific gene by distinct corepressor complexes. EMBO J. 21, 1389–1397 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. El-Osta, A., Kantharidis, P., Zalcberg, J.R. & Wolffe, A.P. Precipitous release of methyl-CpG binding protein 2 and histone deacetylase 1 from the methylated human multidrug resistance gene (MDR1) on activation. Mol. Cell. Biol. 22, 1844–1857 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Boyes, J. & Bird, A. DNA methylation inhibits transcription indirectly via a methyl-CpG binding protein. Cell 64, 1123–1134 (1991).

    Article  CAS  PubMed  Google Scholar 

  72. Jones, P.L. et al. Methylated DNA and MeCP2 recruit histone deacetylase to repress transcription. Nat. Genet. 19, 187–191 (1998).

    Article  CAS  PubMed  Google Scholar 

  73. Nan, X. et al. Transcriptional repression by the methyl-CpG-binding protein MeCP2 involves a histone deacetylase complex. Nature 393, 386–389 (1998).

    Article  CAS  PubMed  Google Scholar 

  74. Ng, H. et al. MBD2 is a transcriptional repressor belonging to the MeCP1 histone deacetylase complex. Nat. Genet. 23, 58–61 (1999).

    Article  CAS  PubMed  Google Scholar 

  75. Feng, Q. & Zhang, Y. The MeCP1 complex represses transcription through preferential binding, remodeling, and deacetylating methylated nucleosomes. Genes Dev. 15, 827–832 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Hendrich, B., Guy, J., Ramsahoye, B., Wilson, V.A. & Bird, A. Closely related proteins MBD2 and MBD3 play distinctive but interacting roles in mouse development. Genes Dev. 15, 710–723 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Zhang, Y. et al. Analysis of the NuRD subunits reveals a histone deacetylase core complex and a connection with DNA methylation. Genes Dev. 13, 1924–1935 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Hutchins, A.S. et al. Gene silencing quantitatively controls the function of a developmental trans-activator. Mol. Cell 10, 81–91 (2002).

    Article  CAS  PubMed  Google Scholar 

  79. Curradi, M., Izzo, A., Badaracco, G. & Landsberger, N. Molecular mechanisms of gene silencing mediated by DNA methylation. Mol. Cell. Biol. 22, 3157–3173 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Guy, J., Hendrich, B., Holmes, M., Martin, J.E. & Bird, A. A mouse Mecp2-null mutation causes neurological symptoms that mimic Rett syndrome. Nat. Genet. 27, 322–326 (2001).

    Article  CAS  PubMed  Google Scholar 

  81. Amir, R.E. et al. Rett syndrome is caused by mutations in X-linked MECP2, encoding methyl-CpG-binding protein 2. Nat. Genet. 23, 185–188 (1999).

    Article  CAS  PubMed  Google Scholar 

  82. Yusufzai, T.M. & Wolffe, A.P. Functional consequences of Rett syndrome mutations on human MeCP2. Nucleic Acids Res. 28, 4172–4179 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Free, A. et al. DNA recognition by the methyl-CpG binding domain of MeCP2. J. Biol. Chem. 276, 3353–3360 (2001).

    Article  CAS  PubMed  Google Scholar 

  84. Chen, R., Akbarian, S., Tudor, M. & Jaenisch, R. Deficiency of methyl-CpG binding protein-2 in CNS neurons results in a Rett-like phenotype in mice. Nat. Genet. 27, 327–331 (2001).

    Article  CAS  PubMed  Google Scholar 

  85. Shahbazian, M. et al. Mice with truncated MeCP2 recapitulate many Rett syndrome features and display hyperacetylation of histone H3. Neuron 35, 243–254 (2002).

    Article  CAS  PubMed  Google Scholar 

  86. Tudor, M., Akbarian, S., Chen, R.Z. & Jaenisch, R. Transcriptional profiling of a mouse model for Rett syndrome reveals subtle transcriptional changes in the brain. Proc. Natl. Acad. Sci. USA 99, 15536–15541 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Futscher, B.W. et al. Role for DNA methylation in the control of cell type specific maspin expression. Nat. Genet. 31, 175–179 (2002).

    Article  CAS  PubMed  Google Scholar 

  88. Stancheva, I., El-Maarri, O., Walter, J., Niveleau, A. & Meehan, R.R. DNA methylation at promoter regions regulates the timing of gene activation in Xenopus laevis embryos. Dev. Biol. 243, 155–165 (2002).

    Article  CAS  PubMed  Google Scholar 

  89. Avner, P. & Heard, E. X-chromosome inactivation: counting, choice and initiation. Nat. Rev. Genet. 2, 59–67 (2001).

    Article  CAS  PubMed  Google Scholar 

  90. Jones, B.K., Levorse, J.M. & Tilghman, S.M. Igf2 imprinting does not require its own DNA methylation or H19 RNA. Genes Dev. 12, 2200–2207 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Cohen, D.E. & Lee, J.T. X-chromosome inactivation and the search for chromosome-wide silencers. Curr. Opin. Genet. Dev. 12, 219–224 (2002).

    Article  CAS  PubMed  Google Scholar 

  92. Lyle, R. et al. The imprinted antisense RNA at the Igf2r locus overlaps but does not imprint Mas1. Nat. Genet. 25, 19–21 (2000).

    Article  CAS  PubMed  Google Scholar 

  93. Chao, W., Huynh, K.D., Spencer, R.J., Davidow, L.S. & Lee, J.T. CTCF, a candidate trans-acting factor for X-inactivation choice. Science 295, 345–347 (2002).

    Article  CAS  PubMed  Google Scholar 

  94. Brannan, C.I. & Bartolomei, M.S. Mechanisms of genomic imprinting. Curr. Opin. Genet. Dev. 9, 164–170 (1999).

    Article  CAS  PubMed  Google Scholar 

  95. Li, E., Beard, C. & Jaenisch, R. Role for DNA methylation in genomic imprinting. Nature 366, 362–385 (1993).

    Article  CAS  PubMed  Google Scholar 

  96. Beard, C., Li, E. & Jaenisch, R. Loss of methylation activates Xist in somatic but not in embryonic cells. Genes Dev. 9, 2325–2334 (1995).

    Article  CAS  PubMed  Google Scholar 

  97. Lee, J. et al. Erasing genomic imprinting memory in mouse clone embryos produced from day 11.5 primordial germ cells. Development 129, 1807–1817 (2002).

    Article  CAS  PubMed  Google Scholar 

  98. Hajkova, P. et al. Epigenetic reprogramming in mouse primordial germ cells. Mech. Dev. 117, 15 (2002).

    Article  CAS  PubMed  Google Scholar 

  99. Tucker, K. et al. Germ-line passage is required for establishment of methylation and expression patterns of imprinted but not of non-imprinted genes. Genes Dev. 10, 1008–1020 (1996).

    Article  CAS  PubMed  Google Scholar 

  100. Keohane, A.M., O'Neill, L.P., Belyaev, N.D., Lavender, J.S. & Turner, B.M. X-inactivation and histone H4 acetylation in embryonic stem cells. Dev. Biol. 180, 618–630 (1996).

    Article  CAS  PubMed  Google Scholar 

  101. Wutz, A. & Jaenisch, R. A shift from reversible to irreversible X inactivation is triggered during ES cell differentiation. Mol. Cell 5, 695–705 (2000).

    Article  CAS  PubMed  Google Scholar 

  102. Csankovszki, G., Nagy, A. & Jaenisch, R. Synergism of Xist RNA, DNA methylation, and histone hypoacetylation in maintaining X chromosome inactivation. J. Cell Biol. 153, 773–784 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Rideout, W.M., Eggan, K. & Jaenisch, R. Nuclear cloning and epigenetic reprogramming of the genome. Science 293, 1093–1098 (2001).

    Article  CAS  PubMed  Google Scholar 

  104. Young, L.E., Sinclair, K.D. & Wilmut, I. Large offspring syndrome in cattle and sheep. Rev. Reprod. 3, 155–163 (1998).

    Article  CAS  PubMed  Google Scholar 

  105. Ogonuki, N. et al. Early death of mice cloned from somatic cells. Nat. Genet. 30, 253–254 (2002).

    Article  CAS  PubMed  Google Scholar 

  106. Tamashiro, K.L. et al. Cloned mice have an obese phenotype not transmitted to their offspring. Nat. Med. 8, 262–267 (2002).

    Article  CAS  PubMed  Google Scholar 

  107. Hochedlinger, K. & Jaenisch, R. Nuclear transplantation: lessons from frogs and mice. Curr. Opin. Cell Biol. 14, 741–748 (2002).

    Article  CAS  PubMed  Google Scholar 

  108. Dean, W. et al. Conservation of methylation reprogramming in mammalian development: aberrant reprogramming in cloned embryos. Proc. Natl. Acad. Sci. USA 98, 13734–13738 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Kang, Y. et al. Aberrant methylation of donor genome in cloned bovine embryos. Nat. Genet. 28, 173–177 (2001).

    Article  CAS  PubMed  Google Scholar 

  110. Bourc'his, D. et al. Delayed and incomplete reprogramming of chromosome methylation patterns in bovine cloned embryos. Curr. Biol. 11, 1542–1546 (2001).

    Article  CAS  PubMed  Google Scholar 

  111. Boiani, M., Eckardt, S., Scholer, H.R. & McLaughlin, K.J. Oct4 distribution and level in mouse clones: consequences for pluripotency. Genes Dev. 16, 1209–1219 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Bortvin, A. et al. Incomplete reactivation of Oct4-related gene in mouse embryos cloned from somatic nuclei. Development (in the press).

  113. Nichols, J. et al. Formation of pluripotent stem cells in the mammalian embryo depends on the POU transcription factor Oct4. Cell 95, 379–391 (1998).

    Article  CAS  PubMed  Google Scholar 

  114. Humpherys, D. et al. Epigenetic instability in ES cells and cloned mice. Science 293, 95–97 (2001).

    Article  CAS  PubMed  Google Scholar 

  115. Humpherys, D. et al. Abnormal gene expression in cloned mice derived from ES cell and cumulus cell nuclei. Proc. Natl. Acad. Sci. USA 99, 12889–12894 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Inoue, K. et al. Faithful expression of imprinted genes in cloned mice. Science 295, 297 (2002).

    Article  CAS  PubMed  Google Scholar 

  117. Eggan, K. et al. X-chromosome inactivation in cloned mouse embryos. Science 290, 1578–1581 (2000).

    Article  CAS  PubMed  Google Scholar 

  118. Wakayama, T. et al. Cloning of mice to six generations. Nature 407, 318–319 (2000).

    Article  CAS  PubMed  Google Scholar 

  119. Lanza, R. et al. Extension of cell life-span and telomere length in animals cloned from senescent somatic cells. Science 288, 665–669 (2000).

    Article  CAS  PubMed  Google Scholar 

  120. Betts, D. et al. Reprogramming of telomerase activity and rebuilding of telomere length in cloned cattle. Proc. Natl. Acad. Sci. USA 98, 1077–1082 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Rideout, W.M. et al. Generation of mice from wild-type and targeted ES cells by nuclear cloning. Nat. Genet. 24, 109–110 (2000).

    Article  CAS  PubMed  Google Scholar 

  122. Wakayama, T. & Yanagimachi, R. Mouse cloning with nucleus donor cells of different age and type. Mol. Reprod. Dev. 58, 376–383 (2001).

    Article  CAS  PubMed  Google Scholar 

  123. Eggan, K. et al. Hybrid vigor, fetal overgrowth, and viability of mice derived by nuclear cloning and tetraploid embryo complementation. Proc. Natl. Acad. Sci. USA 98, 6209–6214 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Hochedlinger, K. & Jaenisch, R. Monoclonal mice generated by nuclear transfer from mature B and T donor cells. Nature 415, 1035–1038 (2002).

    Article  CAS  PubMed  Google Scholar 

  125. Di Berardino, M.A. Genetic stability and modulation of metazoan nuclei transplanted into eggs and oocytes. Differentiation 17, 17–30 (1980).

    Article  CAS  Google Scholar 

  126. Gurdon, J.B. Genetic reprogramming following nuclear transplantation in Amphibia. Semin. Cell Dev. Biol. 10, 239–243 (1999).

    Article  CAS  PubMed  Google Scholar 

  127. Jaenisch, R. & Wilmut, I. Developmental biology. Don't clone humans! Science 291, 2552 (2001).

    Article  CAS  PubMed  Google Scholar 

  128. Rideout, W.M. 3rd, Hochedlinger, K., Kyba, M., Daley, G.Q. & Jaenisch, R. Correction of a genetic defect by nuclear transplantation and combined cell and gene therapy. Cell 109, 17–27 (2002).

    Article  CAS  PubMed  Google Scholar 

  129. Jones, P.A. & Baylin, S.B. The fundamental role of epigenetic events in cancer. Nat. Rev. Genet. 3, 415–428 (2002).

    Article  CAS  PubMed  Google Scholar 

  130. Esteller, M. & Herman, J.G. Cancer as an epigenetic disease: DNA methylation and chromatin alterations in human tumours. J. Pathol. 196, 1–7 (2002).

    Article  CAS  PubMed  Google Scholar 

  131. Feinberg, A.P. & Vogelstein, B. Hypomethylation distinguishes genes of some human cancers from their normal counterparts. Nature 301, 89–92 (1983).

    Article  CAS  PubMed  Google Scholar 

  132. Gama-Sosa, M.A. et al. The 5-methylcytosine content of DNA from human tumors. Nucleic Acids Res. 11, 6883–6894 (1983).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Jones, P.A. DNA methylation and cancer. Oncogene 21, 5358–5360 (2002).

    Article  CAS  PubMed  Google Scholar 

  134. Gonzalgo, M. & Jones, P. Mutagenic and epigenetic effects of DNA methylation. Mutat. Res. 386, 107–118 (1997).

    Article  CAS  PubMed  Google Scholar 

  135. Jones, P. & Laird, P. Cancer epigenetics comes of age. Nat. Genet. 21, 163–167 (1999).

    Article  CAS  PubMed  Google Scholar 

  136. Xu, G.-L. et al. Chromosome instability and immunodeficiency syndrome caused by mutations in a DNA methyltransferase gene. Nature 402, 187–191 (1999).

    Article  CAS  PubMed  Google Scholar 

  137. Ehrlich, M. et al. High frequencies of ICF syndrome-like pericentromeric heterochromatin decondensation and breakage in chromosome 1 in a chorionic villus sample. J. Med. Genet. 38, 882–884 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Chen, R.Z., Pettersson, U., Beard, C., Jackson-Grusby, L. & Jaenisch, R. DNA hypomethylation leads to elevated mutation rates. Nature 395, 89–93 (1998).

    Article  CAS  PubMed  Google Scholar 

  139. Chan, M.F. et al. Reduced rates of gene loss, gene silencing, and gene mutation in Dnmt1-deficient embryonic stem cells. Mol. Cell. Biol. 21, 7587–7600 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Turker, M.S. Gene silencing in mammalian cells and the spread of DNA methylation. Oncogene 21, 5388–5393 (2002).

    Article  CAS  PubMed  Google Scholar 

  141. Biniszkiewicz, D. et al. Dnmt1 overexpression causes genomic hypermethylation, loss of imprinting, and embryonic lethality. Mol. Cell. Biol. 22, 2124–2135 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Sheldon, C.C. et al. The control of flowering by vernalization. Curr. Opin. Plant Biol. 3, 418–422 (2000).

    Article  CAS  PubMed  Google Scholar 

  143. Sheldon, C.C., Rouse, D.T., Finnegan, E.J., Peacock, W.J. & Dennis, E.S. The molecular basis of vernalization: the central role of FLOWERING LOCUS C (FLC). Proc. Natl. Acad. Sci. USA 97, 3753–3758 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Sheldon, C.C. et al. The FLF MADS box gene: a repressor of flowering in Arabidopsis regulated by vernalization and methylation. Plant Cell 11, 445–458 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Gendall, A.R., Levy, Y.Y., Wilson, A. & Dean, C. The VERNALIZATION 2 gene mediates the epigenetic regulation of vernalization in Arabidopsis. Cell 107, 525–535 (2001).

    Article  CAS  PubMed  Google Scholar 

  146. Brock, H.W. & van Lohuizen, M. The Polycomb group—no longer an exclusive club? Curr. Opin. Genet. Dev. 11, 175–181 (2001).

    Article  CAS  PubMed  Google Scholar 

  147. Wilson, V.L. & Jones, P.A. DNA methylation decreases in aging but not in immortal cells. Science 220, 1055–1057 (1983).

    Article  CAS  PubMed  Google Scholar 

  148. Mays-Hoopes, L., Chao, W., Butcher, H.C. & Huang, R.C. Decreased methylation of the major mouse long interspersed repeated DNA during aging and in myeloma cells. Dev. Genet. 7, 65–73 (1986).

    Article  CAS  PubMed  Google Scholar 

  149. Wilson, V.L., Smith, R.A., Ma, S. & Cutler, R.G. Genomic 5-methyldeoxycytidine decreases with age. J. Biol. Chem. 262, 9948–9951 (1987).

    CAS  PubMed  Google Scholar 

  150. Bestor, T.H. & Tycko, B. Creation of genomic methylation patterns. Nat. Genet. 12, 363–367 (1996).

    Article  CAS  PubMed  Google Scholar 

  151. Barbot, W., Dupressoir, A., Lazar, V. & Heidmann, T. Epigenetic regulation of an IAP retrotransposon in the aging mouse: progressive demethylation and de-silencing of the element by its repetitive induction. Nucleic Acids Res. 30, 2365–2373 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Issa, J.P. et al. Methylation of the oestrogen receptor CpG island links aging and neoplasia in human colon. Nat. Genet. 7, 536–540 (1994).

    Article  CAS  PubMed  Google Scholar 

  153. Toyota, M. et al. CpG island methylator phenotype in colorectal cancer. Proc. Natl. Acad. Sci. USA 96, 8681–8686 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Toyota, M. & Issa, J.P. CpG island methylator phenotypes in aging and cancer. Semin. Cancer Biol. 9, 349–357 (1999).

    Article  CAS  PubMed  Google Scholar 

  155. Van Den Veyver, I.B. Genetic effects of methylation diets. Annu. Rev. Nutr. 22, 255–282 (2002).

    Article  CAS  PubMed  Google Scholar 

  156. Giovannucci, E. et al. Folate, methionine, and alcohol intake and risk of colorectal adenoma. J. Natl. Cancer Inst. 85, 875–884 (1993).

    Article  CAS  PubMed  Google Scholar 

  157. Blount, B.C. et al. Folate deficiency causes uracil misincorporation into human DNA and chromosome breakage: implications for cancer and neuronal damage. Proc. Natl. Acad. Sci. USA 94, 3290–3295 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Jacob, R.A. The role of micronutrients in DNA synthesis and maintenance. Adv. Exp. Med. Biol. 472, 101–113 (1999).

    Article  CAS  PubMed  Google Scholar 

  159. Group, M.V.S.R. Prevention of neural tube defects: results of the Medical Research Council Vitamin Study. MRC Vitamin Study Research Group. Lancet 338, 131–137 (1991).

    Article  Google Scholar 

  160. Friso, S. et al. A common mutation in the 5,10-methylenetetrahydrofolate reductase gene affects genomic DNA methylation through an interaction with folate status. Proc. Natl. Acad. Sci. USA 99, 5606–5611 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Dizik, M., Christman, J.K. & Wainfan, E. Alterations in expression and methylation of specific genes in livers of rats fed a cancer promoting methyl-deficient diet. Carcinogenesis 12, 1307–1312 (1991).

    Article  CAS  PubMed  Google Scholar 

  162. Poirier, L., Zapisek, W. & Lyon-Cook, B. Physiological methylation in carcinogenesis. In Mutation and the Environment Part D, 97–112 (Willey-Liss, 1990).

    Google Scholar 

  163. Wainfan, E. & Poirier, L.A. Methyl groups in carcinogenesis: effects on DNA methylation and gene expression. Cancer Res. 52, 2071s–2077s (1992).

    CAS  PubMed  Google Scholar 

  164. Hoal-van Helden, E.G. & van Helden, P.D. Age-related methylation changes in DNA may reflect the proliferative potential of organs. Mutat. Res. 219, 263–266 (1989).

    Article  CAS  PubMed  Google Scholar 

  165. Christman, J.K., 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 14, 551–557 (1993).

    Article  CAS  PubMed  Google Scholar 

  166. Michaud, E.J. et al. Differential expression of a new dominant agouti allele (Aiapy) is correlated with methylation state and is influenced by parental lineage. Genes Dev. 8, 1463–1472 (1994).

    Article  CAS  PubMed  Google Scholar 

  167. Millar, S.E., Miller, M.W., Stevens, M.E. & Barsh, G.S. Expression and transgenic studies of the mouse agouti gene provide insight into the mechanisms by which mammalian coat color patterns are generated. Development 121, 3223–3232 (1995).

    CAS  PubMed  Google Scholar 

  168. Siracusa, L.D. et al. Hypervariable yellow (Ahvy), a new murine agouti mutation: Ahvy displays the largest variation in coat color phenotypes of all known agouti alleles. J. Hered. 86, 121–128 (1995).

    Article  CAS  PubMed  Google Scholar 

  169. 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 

  170. 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 

  171. Morgan, H.D., Sutherland, H.G.E., Martin, D.I.K. & Whitelaw, E. Epigenetic inheritance at the agouti locus in the mouse. Nat. Genet. 23, 314–318 (1999).

    Article  CAS  PubMed  Google Scholar 

  172. Petronis, A. Human morbid genetics revisited: relevance of epigenetics. Trends Genet. 17, 142–146 (2001).

    Article  CAS  PubMed  Google Scholar 

  173. Tremolizzo, L. et al. An epigenetic mouse model for molecular and behavioral neuropathologies related to schizophrenia vulnerability. Proc. Natl. Acad. Sci. USA 99, 17095–17100 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Adorjan, P. et al. Tumour class prediction and discovery by microarray-based DNA methylation analysis. Nucleic Acids Res. 30, e21 (2002).

    Article  PubMed  PubMed Central  Google Scholar 

  175. Jenuwein, T. & Allis, C.D. Translating the histone code. Science 293, 1074–1080 (2001).

    Article  CAS  PubMed  Google Scholar 

  176. Santini, V., Kantarjian, H.M. & Issa, J.P. Changes in DNA methylation in neoplasia: pathophysiology and therapeutic implications. Ann. Intern. Med. 134, 573–586 (2001).

    Article  CAS  PubMed  Google Scholar 

  177. Juttermann, R., Li, E. & Jaenisch, R. Toxicity of 5-aza-2′-deoxycytidine to mammalian cells is mediated primarily by covalent trapping of DNA methyltransferase rather than DNA demethylation. Proc. Natl. Acad. Sci. USA 91, 11797–11801 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Cameron, E.E., Bachman, K.E., Myohanen, S., Herman, J.G. & Baylin, S.B. Synergy of demethylation and histone deacetylase inhibition in the re-expression of genes silenced in cancer. Nat. Genet 21, 103–107 (1999).

    Article  CAS  PubMed  Google Scholar 

  179. Jaenisch, R., Schnieke, A. & Harbers, K. Treatment of mice with 5-azacytidine efficiently activates silent retroviral genomes in different tissues. Proc. Natl. Acad. Sci. USA 82, 1451–1455 (1985).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Millar, C.B. et al. Enhanced CpG mutability and tumorigenesis in MBD4-deficient mice. Science 297, 403–405 (2002).

    Article  CAS  PubMed  Google Scholar 

  181. Lagger, G. et al. Essential function of histone deacetylase 1 in proliferation control and CDK inhibitor repression. EMBO J. 21, 2672–2681 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Peters, A.H. et al. Loss of the Suv39h histone methyltransferases impairs mammalian heterochromatin and genome stability. Cell 107, 323–337 (2001).

    Article  CAS  PubMed  Google Scholar 

  183. Baylin, S.B. & Herman, J.G. DNA hypermethylation in tumorigenesis: epigenetics joins genetics. Trends Genet. 16, 168–174 (2000).

    Article  CAS  PubMed  Google Scholar 

  184. Laird, P.W. et al. Suppression of intestinal neoplasia by DNA hypomethylation. Cell 81, 197–205 (1995).

    Article  CAS  PubMed  Google Scholar 

  185. Fuks, F. et al. The methyl-CpG-binding protein MeCP2 links DNA methylation to histone methylation. J. Biol. Chem. (2002); advanced online publication 9 November 2002 (doi: 10.1074/jbc.M210256200).

  186. Argeson, A., Nelson, K. & Siracusa, L. Molecular basis of the pleiotropic phenotype of mice carrying the hypervariable yellow (Ahvy) mutation at the agouti locus. Genetics 142, 557–567 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank A. Chess, H. McQueen, H. Jørgensen and all members of the Jaenisch laboratory for critical comments on this manuscript.

Author information

Authors and Affiliations

Authors

Rights and permissions

Reprints and permissions

About this article

Cite this article

Jaenisch, R., Bird, A. Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals. Nat Genet 33 (Suppl 3), 245–254 (2003). https://doi.org/10.1038/ng1089

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng1089

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