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:

DNA methylation: roles in mammalian development

Key Points

  • DNA methylation is essential for mammalian development and has notable roles in gene silencing, protection against spurious repetitive element activity, genomic stability during mitosis and parent-of-origin imprinting.

  • DNA methylation functions to coordinately silence some promoter and enhancer classes that are typified by low overall CpG density. By contrast, many CpG islands, particularly those at promoters, remain generally and constitutively protected from DNA methylation through numerous opposing cis- and trans-based mechanisms.

  • Embryonic stem cells (ESCs) remain one of the most highly studied in vitro models for dissecting epigenetic mechanisms during cellular differentiation. They are unique in their ability to retain the molecular characteristics of their cellular state in the absence of DNA methylation but cannot differentiate into embryonic lineages while simultaneously gaining extra-embryonic potential.

  • ESCs are also unique in their heterogeneous ability to maintain repetitive element silencing through DNA methyltransferase 1 (DNMT1) maintenance activity alone and rely on the DNMT3 enzymes to re-silence these elements continuously de novo. The pre-implantation stage of development is one of the few stages at which endogeneous transposable elements are actively regulated.

  • Promoters of germline-specific genes are coordinately repressed by DNMT3B in embryonic lineages and are reactivated during the global demethylation that accompanies primordial germ cell (PGC) specification.

  • Haematopoiesis has served as a benchmark model for the function of DNA methylation in adult stem cells and in later lineage specification. Several notable regulatory principles for DNA methylation were originally or comprehensively described in this system, including the gating of lymphoid versus myeloid fates, the dynamics of intragenic CpG island methylation in transcript selection and demethylation as a lineage stabilizer by providing a mitotically heritable memory of transcription factor binding.

  • DNA methylation is globally erased during specification of the germ line and during fertilization, at which point the paternal genome is specifically demethylated. These two key events have numerous related cascades of chromatin re-organization that suggest similar epigenetic mechanisms for erasure and re-establishment of global nuclear organization, although the relationship between these events remains incompletely defined.

  • Although they have remained mysterious for more than 2 decades, the targets and mechanisms for DNA methylation erasure in the paternal genome as well as for protection of the maternal genome during fertilization are becoming clearer. For example, there are distinct temporal windows for global hydroxymethylation followed by erasure to unmodified cytosines occurring later in zygotic progression. The epigenetic determinants that retain silencing at repetitive element classes and imprints are less clear, but they are likely to be discovered as the dynamics and relationships of other epigenetic regulators are refined.

Abstract

DNA methylation is among the best studied epigenetic modifications and is essential to mammalian development. Although the methylation status of most CpG dinucleotides in the genome is stably propagated through mitosis, improvements to methods for measuring methylation have identified numerous regions in which it is dynamically regulated. In this Review, we discuss key concepts in the function of DNA methylation in mammals, stemming from more than two decades of research, including many recent studies that have elucidated when and where DNA methylation has a regulatory role in the genome. We include insights from early development, embryonic stem cells and adult lineages, particularly haematopoiesis, to highlight the general features of this modification as it participates in both global and localized epigenetic regulation.

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: DNA methylation and its targets in mouse embryonic stem cells.
Figure 2: Lineage restriction and renewal of embryonic and adult stem cells.
Figure 3: Local demethylation in support of cellular memory.
Figure 4: Epigenetic events during global DNA demethylation in primordial germ cells.
Figure 5: Epigenetic events during global DNA demethylation in the zygote.

Similar content being viewed by others

References

  1. Feng, S., Jacobsen, S. E. & Reik, W. Epigenetic reprogramming in plant and animal development. Science 330, 622–627 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Ramsahoye, B. H. et al. Non-CpG methylation is prevalent in embryonic stem cells and may be mediated by DNA methyltransferase 3a. Proc. Natl Acad. Sci. USA 97, 5237–5242 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Ziller, M. J. et al. Genomic distribution and inter-sample variation of non-CpG methylation across human cell types. PLoS Genet. 7, e1002389 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. 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 

  5. 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 

  6. Deaton, A. M. & Bird, A. CpG islands and the regulation of transcription. Genes Dev. 25, 1010–1022 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  9. Laird, P. W. Principles and challenges of genomewide DNA methylation analysis. Nature Rev. Genet. 11, 191–203 (2010).

    Article  CAS  PubMed  Google Scholar 

  10. Smith, Z. D. et al. A unique regulatory phase of DNA methylation in the early mammalian embryo. Nature 484, 339–344 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Illingworth, R. S. et al. Orphan CpG islands identify numerous conserved promoters in the mammalian genome. PLoS Genet. 6, e1001134 (2010). This paper reconciles the apparent dissimilarities in CpG content between the mouse and human genome and shows that functional enrichment for unmethylated regions reveals an almost equivalent compliment of CpG islands. Moreover, it confirms that half of the CpG islands do not occur at known promoter regions and are more frequently methylated than those at promoters.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. 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 

  13. Brandeis, M. et al. SP1 elements protect a CpG island from de novo methylation. Nature 371, 435–438 (1994).

    Article  CAS  PubMed  Google Scholar 

  14. Lienert, F. et al. Identification of genetic elements that autonomously determine DNA methylation states. Nature Genet. 43, 1091–1097 (2011). This study extends to numerous different sites within pluripotent cells, the observations of References 12 and 13, which had first demonstrated that transcription factor binding instructs local demethylation.

    Article  CAS  PubMed  Google Scholar 

  15. Stadler, M. B. et al. DNA-binding factors shape the mouse methylome at distal regulatory regions. Nature 480, 490–495 (2011).

    CAS  PubMed  Google Scholar 

  16. Thomson, J. P. et al. CpG islands influence chromatin structure via the CpG-binding protein CFP1. Nature 464, 1082–1086 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Clouaire, T. et al. CFP1 integrates both CpG content and gene activity for accurate H3K4me3 deposition in embryonic stem cells. Genes Dev. 26, 1714–1728 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Erfurth, F. E. et al. MLL protects CpG clusters from methylation within the Hoxa9 gene, maintaining transcript expression. Proc. Natl Acad. Sci. USA 105, 7517–7522 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Otani, J. et al. Structural basis for recognition of H3K4 methylation status by the DNA methyltransferase 3A ATRX–DNMT3–DNMT3L domain. EMBO Rep. 10, 1235–1241 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Ooi, S. K. et al. DNMT3L connects unmethylated lysine 4 of histone H3 to de novo methylation of DNA. Nature 448, 714–717 (2007). This work highlights the important relationship between DNMT3 enzyme binding and local chromatin context. Particularly, it showed that recruitment of DNA methyltransferases requires histone tail templates that lack euchromatic H3K4 methylation.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Conerly, M. L. et al. Changes in H2A.Z occupancy & DNA methylation during B-cell lymphomagenesis. Genome Res. 20, 1383–1390 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Yang, X. et al. Gene reactivation by 5-aza-2′-deoxycytidine-induced demethylation requires SRCAP-mediated H2A.Z insertion to establish nucleosome depleted regions. PLoS Genet. 8, e1002604 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Brinkman, A. B. et al. Sequential ChIP-bisulfite sequencing enables direct genome-scale investigation of chromatin and DNA methylation cross-talk. Genome Res. 22, 1128–1138 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Bartke, T. et al. Nucleosome-interacting proteins regulated by DNA and histone methylation. Cell 143, 470–484 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Lindroth, A. M. et al. Antagonism between DNA and H3K27 methylation at the imprinted Rasgrf1 locus. PLoS Genet. 4, e1000145 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Jones, P. A. Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nature Rev. Genet. 13, 484–492 (2012).

    Article  CAS  PubMed  Google Scholar 

  27. Ginno, P. A., Lott, P. L., Christensen, H. C., Korf, I. & Chedin, F. R-loop formation is a distinctive characteristic of unmethylated human CpG island promoters. Mol. Cell 45, 814–825 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Jeltsch, A. On the enzymatic properties of DNMT1: specificity, processivity, mechanism of linear diffusion and allosteric regulation of the enzyme. Epigenetics 1, 63–66 (2006).

    Article  PubMed  Google Scholar 

  29. Song, J., Rechkoblit, O., Bestor, T. H. & Patel, D. J. Structure of DNMT1–DNA complex reveals a role for autoinhibition in maintenance DNA methylation. Science 331, 1036–1040 (2011).

    Article  CAS  PubMed  Google Scholar 

  30. Schoorlemmer, J. et al. Characterization of a negative retinoic acid response element in the murine OCT4 promoter. Mol. Cell. Biol. 14, 1122–1136 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Dennis, K., Fan, T., Geiman, T., Yan, Q. & Muegge, K. LSH, a member of the SNF2 family, is required for genome-wide methylation. Genes Dev. 15, 2940–2944 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Zhu, H. et al. LSH is involved in de novo methylation of DNA. EMBO J. 25, 335–345 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Myant, K. & Stancheva, I. LSH cooperates with DNA methyltransferases to repress transcription. Mol. Cell. Biol. 28, 215–226 (2008).

    Article  CAS  PubMed  Google Scholar 

  34. Myant, K. et al. LSH and G9A/GLP complex are required for developmentally programmed DNA methylation. Genome Res. 21, 83–94 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Epsztejn-Litman, S. et al. De novo DNA methylation promoted by G9A prevents reprogramming of embryonically silenced genes. Nature Struct. Mol. Biol. 15, 1176–1183 (2008).

    Article  CAS  Google Scholar 

  36. Dong, K. B. et al. DNA methylation in ES cells requires the lysine methyltransferase G9A but not its catalytic activity. EMBO J. 27, 2691–2701 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Ayyanathan, K. et al. Regulated recruitment of HP1 to a euchromatic gene induces mitotically heritable, epigenetic gene silencing: a mammalian cell culture model of gene variegation. Genes Dev. 17, 1855–1869 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Lehnertz, B. et al. SUV39H-mediated histone H3 lysine 9 methylation directs DNA methylation to major satellite repeats at pericentric heterochromatin. Curr. Biol. 13, 1192–1200 (2003).

    Article  CAS  PubMed  Google Scholar 

  39. 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 

  40. Chen, T., Tsujimoto, N. & Li, E. The PWWP domain of DNMT3A and DNMT3B is required for directing DNA methylation to the major satellite repeats at pericentric heterochromatin. Mol. Cell. Biol. 24, 9048–9058 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Gopalakrishnan, S., Sullivan, B. A., Trazzi, S., Della Valle, G. & Robertson, K. D. DNMT3B interacts with constitutive centromere protein CENP-C to modulate DNA methylation and the histone code at centromeric regions. Hum. Mol. Genet. 18, 3178–3193 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Okada, T. et al. CENP-B controls centromere formation depending on the chromatin context. Cell 131, 1287–1300 (2007).

    Article  CAS  PubMed  Google Scholar 

  43. Waterston, R. H. et al. Initial sequencing and comparative analysis of the mouse genome. Nature 420, 520–562 (2002).

    Article  CAS  PubMed  Google Scholar 

  44. Liang, G. et al. Cooperativity between DNA methyltransferases in the maintenance methylation of repetitive elements. Mol. Cell. Biol. 22, 480–491 (2002). By investigating the levels of demethylation in Dnmt1 -knockout compared with Dnmt3a - or Dnmt3b -knockout mouse ESCs across different genomic features, Liang et al . demonstrate selective demethylation within particular repetitive element classes in the absence of de novo methyltransferase activity.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Wolf, D. & Goff, S. P. Embryonic stem cells use ZFP809 to silence retroviral DNAs. Nature 458, 1201–1204 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Rowe, H. M. et al. KAP1 controls endogenous retroviruses in embryonic stem cells. Nature 463, 237–240 (2010).

    Article  CAS  PubMed  Google Scholar 

  47. Wolf, D. & Goff, S. P. TRIM28 mediates primer binding site-targeted silencing of murine leukemia virus in embryonic cells. Cell 131, 46–57 (2007).

    Article  CAS  PubMed  Google Scholar 

  48. Wiznerowicz, M. et al. The Kruppel-associated box repressor domain can trigger de novo promoter methylation during mouse early embryogenesis. J. Biol. Chem. 282, 34535–34541 (2007).

    Article  CAS  PubMed  Google Scholar 

  49. Matsui, T. et al. Proviral silencing in embryonic stem cells requires the histone methyltransferase ESET. Nature 464, 927–931 (2010).

    Article  CAS  PubMed  Google Scholar 

  50. Karimi, M. M. et al. DNA methylation and SETDB1/H3K9me3 regulate predominantly distinct sets of genes, retroelements, and chimeric transcripts in mESCs. Cell Stem Cell 8, 676–687 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Leung, D. C. et al. Lysine methyltransferase G9a is required for de novo DNA methylation and the establishment, but not the maintenance, of proviral silencing. Proc. Natl Acad. Sci. USA 108, 5718–5723 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Ooi, S. K. et al. Dynamic instability of genomic methylation patterns in pluripotent stem cells. Epigenet. Chromat. 3, 17 (2010).

    Article  CAS  Google Scholar 

  53. Muotri, A. R. et al. L1 retrotransposition in neurons is modulated by MeCP2. Nature 468, 443–446 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  55. 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 

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

    Article  CAS  PubMed  Google Scholar 

  57. Stewart, C. L., Stuhlmann, H., Jahner, D. & Jaenisch, R. De novo methylation, expression, and infectivity of retroviral genomes introduced into embryonal carcinoma cells. Proc. Natl Acad. Sci. USA 79, 4098–4102 (1982).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Smallwood, S. A. & Kelsey, G. De novo DNA methylation: a germ cell perspective. Trends Genet. 28, 33–42 (2012).

    Article  CAS  PubMed  Google Scholar 

  59. Lucifero, D., Mann, M. R., Bartolomei, M. S. & Trasler, J. M. Gene-specific timing and epigenetic memory in oocyte imprinting. Hum. Mol. Genet. 13, 839–849 (2004).

    Article  CAS  PubMed  Google Scholar 

  60. Davis, T. L., Yang, G. J., McCarrey, J. R. & Bartolomei, M. S. The H19 methylation imprint is erased and re-established differentially on the parental alleles during male germ cell development. Hum. Mol. Genet. 9, 2885–2894 (2000).

    Article  CAS  PubMed  Google Scholar 

  61. Lee, D. H. et al. CTCF-dependent chromatin bias constitutes transient epigenetic memory of the mother at the H19-IGF2 imprinting control region in prospermatogonia. PLoS Genet. 6, e1001224 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Henckel, A. et al. Histone methylation is mechanistically linked to DNA methylation at imprinting control regions in mammals. Hum. Mol. Genet. 18, 3375–3383 (2009).

    Article  CAS  PubMed  Google Scholar 

  63. Messerschmidt, D. M. et al. TRIM28 is required for epigenetic stability during mouse oocyte to embryo transition. Science 335, 1499–1502 (2012).

    Article  CAS  PubMed  Google Scholar 

  64. Li, X. et al. A maternal-zygotic effect gene, ZFP57, maintains both maternal and paternal imprints. Dev. Cell 15, 547–557 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Quenneville, S. et al. In embryonic stem cells, ZFP57/KAP1 recognize a methylated hexanucleotide to affect chromatin and DNA methylation of imprinting control regions. Mol. Cell 44, 361–372 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Tsumura, A. et al. Maintenance of self-renewal ability of mouse embryonic stem cells in the absence of DNA methyltransferases DNMT1, DNMT3A and DNMT3B. Genes Cells 11, 805–814 (2006).

    Article  CAS  PubMed  Google Scholar 

  67. Chen, T., Ueda, Y., Dodge, J. E., Wang, Z. & Li, E. Establishment and maintenance of genomic methylation patterns in mouse embryonic stem cells by DNMT3A and DNMT3B. Mol. Cell. Biol. 23, 5594–5605 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Jackson, M. et al. Severe global DNA hypomethylation blocks differentiation and induces histone hyperacetylation in embryonic stem cells. Mol. Cell. Biol. 24, 8862–8871 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  70. Ng, R. K. et al. Epigenetic restriction of embryonic cell lineage fate by methylation of ELF5. Nature Cell Biol. 10, 1280–1290 (2008).

    Article  CAS  PubMed  Google Scholar 

  71. Nakanishi, M. O. et al. Trophoblast-specific DNA methylation occurs after the segregation of the trophectoderm and inner cell mass in the mouse periimplantation embryo. Epigenetics 7, 173–182 (2012).

    Article  CAS  PubMed  Google Scholar 

  72. Arand, J. et al. In vivo control of CpG and non-CpG DNA methylation by DNA methyltransferases. PLoS Genet. 8, e1002750 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Hu, Y. G. et al. Regulation of DNA methylation activity through DNMT3L promoter methylation by DNMT3 enzymes in embryonic development. Hum. Mol. Genet. 17, 2654–2664 (2008).

    Article  CAS  PubMed  Google Scholar 

  74. Ficz, G. et al. Dynamic regulation of 5-hydroxymethylcytosine in mouse ES cells and during differentiation. Nature 473, 398–402 (2011).

    Article  CAS  PubMed  Google Scholar 

  75. Booth, M. J. et al. Quantitative sequencing of 5-methylcytosine and 5-hydroxymethylcytosine at single-base resolution. Science 336, 934–937 (2012).

    Article  CAS  PubMed  Google Scholar 

  76. Macfarlan, T. S. et al. Embryonic stem cell potency fluctuates with endogenous retrovirus activity. Nature 487, 57–63 (2012). This paper presents compelling evidence that epigenetically unstable endogenous retroelement activity corresponds with extra-embryonic potential in pluripotent stem cell populations.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Feldman, N. et al. G9A-mediated irreversible epigenetic inactivation of Oct-3/4 during early embryogenesis. Nature Cell Biol. 8, 188–194 (2006).

    Article  CAS  PubMed  Google Scholar 

  78. Athanasiadou, R. et al. Targeting of de novo DNA methylation throughout the Oct-4 gene regulatory region in differentiating embryonic stem cells. PLoS ONE 5, e9937 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. You, J. S. et al. OCT4 establishes and maintains nucleosome-depleted regions that provide additional layers of epigenetic regulation of its target genes. Proc. Natl Acad. Sci. USA 108, 14497–14502 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Hathaway, N. A. et al. Dynamics and memory of heterochromatin in living cells. Cell 149, 1447–1460 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Zhang, Y. et al. Histone H1 depletion impairs embryonic stem cell differentiation. PLoS Genet. 8, e1002691 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Maatouk, D. M. et al. DNA methylation is a primary mechanism for silencing postmigratory primordial germ cell genes in both germ cell and somatic cell lineages. Development 133, 3411–3418 (2006).

    Article  CAS  PubMed  Google Scholar 

  83. Pohlers, M. et al. A role for E2F6 in the restriction of male-germ-cell-specific gene expression. Curr. Biol. 15, 1051–1057 (2005).

    Article  CAS  PubMed  Google Scholar 

  84. Velasco, G. et al. DNMT3B recruitment through E2F6 transcriptional repressor mediates germ-line gene silencing in murine somatic tissues. Proc. Natl Acad. Sci. USA 107, 9281–9286 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  85. Borgel, J. et al. Targets and dynamics of promoter DNA methylation during early mouse development. Nature Genet. 42, 1093–1100 (2010). In addition to presenting the first study of genome-scale remethylation kinetics during mammalian implantation, this paper confirms that DNMT3B is specifically necessary to silence the germline gene promoters, which is one of the best established instances of direct, coordinated silencing by DNA methylation.

    Article  CAS  PubMed  Google Scholar 

  86. Hackett, J. A. et al. Promoter DNA methylation couples genome-defence mechanisms to epigenetic reprogramming in the mouse germline. Development 139, 3623–3632 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Chan, M. M., Smith, Z. D., Egli, D., Regev, A. & Meissner, A. Mouse ooplasm confers context-specific reprogramming capacity. Nature Genet. 44, 978–980 (2012).

    Article  CAS  PubMed  Google Scholar 

  88. Hackett, J. A. et al. Germline DNA demethylation dynamics and imprint erasure through 5-hydroxymethylcytosine. Science 6 Dec 2012 (doi:10.1126/science.1229277).

    Article  CAS  PubMed  Google Scholar 

  89. Williams, K. et al. TET1 and hydroxymethylcytosine in transcription and DNA methylation fidelity. Nature 473, 343–348 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Augui, S., Nora, E. P. & Heard, E. Regulation of X-chromosome inactivation by the X-inactivation centre. Nature Rev. Genet. 12, 429–442 (2011).

    Article  CAS  PubMed  Google Scholar 

  91. Gendrel, A. V. et al. SMCHD1-dependent and -independent pathways determine developmental dynamics of CpG island methylation on the inactive X chromosome. Dev. Cell 23, 265–279 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Hodges, E. et al. Directional DNA methylation changes and complex intermediate states accompany lineage specificity in the adult hematopoietic compartment. Mol. Cell 44, 17–28 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Deaton, A. M. et al. Cell type-specific DNA methylation at intragenic CpG islands in the immune system. Genome Res. 21, 1074–1086 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Bock, C. et al. DNA methylation dynamics during in vivo differentiation of blood and skin stem cells. Mol. Cell 47, 633–647 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Trowbridge, J. J., Snow, J. W., Kim, J. & Orkin, S. H. DNA methyltransferase 1 is essential for and uniquely regulates hematopoietic stem and progenitor cells. Cell Stem Cell 5, 442–449 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Broske, A. M. et al. DNA methylation protects hematopoietic stem cell multipotency from myeloerythroid restriction. Nature Genet. 41, 1207–1215 (2009).

    Article  CAS  PubMed  Google Scholar 

  97. Hogart, A. et al. Genome-wide DNA methylation profiles in hematopoietic stem and progenitor cells reveal overrepresentation of ETS transcription factor binding sites. Genome Res. 5, 1407–1418 (2012).

    Article  CAS  Google Scholar 

  98. Ji, H. et al. Comprehensive methylome map of lineage commitment from haematopoietic progenitors. Nature 467, 338–342 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Fan, G. et al. DNA methylation controls the timing of astrogliogenesis through regulation of JAK-STAT signaling. Development 132, 3345–3356 (2005).

    Article  CAS  PubMed  Google Scholar 

  100. Tadokoro, Y., Ema, H., Okano, M., Li, E. & Nakauchi, H. De novo DNA methyltransferase is essential for self-renewal, but not for differentiation, in hematopoietic stem cells. J. Exp. Med. 204, 715–722 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Challen, G. A. et al. DNMT3A is essential for hematopoietic stem cell differentiation. Nature Genet. 44, 23–31 (2012).

    Article  CAS  Google Scholar 

  102. Bocker, M. T. et al. Genome-wide promoter DNA methylation dynamics of human hematopoietic progenitor cells during differentiation and aging. Blood 117, e182–e189 (2011).

    Article  CAS  PubMed  Google Scholar 

  103. Calvanese, V. et al. A promoter DNA demethylation landscape of human hematopoietic differentiation. Nucleic Acids Res. 40, 116–131 (2012).

    Article  CAS  PubMed  Google Scholar 

  104. Figueroa, M. E. et al. Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. Cancer Cell 18, 553–567 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Ko, M. et al. Ten-eleven-translocation 2 (TET2) negatively regulates homeostasis and differentiation of hematopoietic stem cells in mice. Proc. Natl Acad. Sci. USA 108, 14566–14571 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Moran-Crusio, K. et al. TET2 loss leads to increased hematopoietic stem cell self-renewal and myeloid transformation. Cancer Cell 20, 11–24 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Quivoron, C. et al. TET2 inactivation results in pleiotropic hematopoietic abnormalities in mouse and is a recurrent event during human lymphomagenesis. Cancer Cell 20, 25–38 (2011).

    Article  CAS  PubMed  Google Scholar 

  108. Kallin, E. M. et al. TET2 facilitates the derepression of myeloid target genes during CEBPα-induced transdifferentiation of pre-B cells. Mol. Cell 48, 266–276 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Klug, M. et al. Active DNA demethylation in human postmitotic cells correlates with activating histone modifications, but not transcription levels. Genome Biol. 11, R63 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Lee, P. P. et al. A critical role for DNMT1 and DNA methylation in T cell development, function, and survival. Immunity 15, 763–774 (2001).

    Article  CAS  PubMed  Google Scholar 

  111. Chappell, C., Beard, C., Altman, J., Jaenisch, R. & Jacob, J. DNA methylation by DNA methyltransferase 1 is critical for effector CD8 T cell expansion. J. Immunol. 176, 4562–4572 (2006).

    Article  CAS  PubMed  Google Scholar 

  112. Shaknovich, R. et al. DNA methyltransferase 1 and DNA methylation patterning contribute to germinal center B-cell differentiation. Blood 118, 3559–3569 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Shearstone, J. R. et al. Global DNA demethylation during mouse erythropoiesis in vivo. Science 334, 799–802 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Makar, K. W. & Wilson, C. B. DNA methylation is a nonredundant repressor of the Th2 effector program. J. Immunol. 173, 4402–4406 (2004).

    Article  CAS  PubMed  Google Scholar 

  115. Makar, K. W. et al. Active recruitment of DNA methyltransferases regulates interleukin 4 in thymocytes and T cells. Nature Immunol. 4, 1183–1190 (2003).

    Article  CAS  Google Scholar 

  116. Bruniquel, D. & Schwartz, R. H. Selective, stable demethylation of the interleukin-2 gene enhances transcription by an active process. Nature Immunol. 4, 235–240 (2003).

    Article  CAS  Google Scholar 

  117. Murayama, A. et al. A specific CpG site demethylation in the human interleukin 2 gene promoter is an epigenetic memory. EMBO J. 25, 1081–1092 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Zheng, Y. et al. Role of conserved non-coding DNA elements in the Foxp3 gene in regulatory T-cell fate. Nature 463, 808–812 (2010). This study carefully dissects the sequential activation of different enhancers as Foxp3 is activated in regulatory T cells.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Lorincz, M. C., Dickerson, D. R., Schmitt, M. & Groudine, M. Intragenic DNA methylation alters chromatin structure and elongation efficiency in mammalian cells. Nature Struct. Mol. Biol. 11, 1068–1075 (2004).

    Article  CAS  Google Scholar 

  120. Laurent, L. et al. Dynamic changes in the human methylome during differentiation. Genome Res. 20, 320–331 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Shukla, S. et al. CTCF-promoted RNA polymerase II pausing links DNA methylation to splicing. Nature 479, 74–79 (2011). This study mechanistically links CTCF binding at unmethylated regions downstream of generally excluded weak exons to their inclusion within mature mRNA transcripts through local Pol II pausing.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Ohinata, Y. et al. BLIMP1 is a critical determinant of the germ cell lineage in mice. Nature 436, 207–213 (2005).

    Article  CAS  PubMed  Google Scholar 

  123. Ancelin, K. et al. BLIMP1 associates with PRMT5 and directs histone arginine methylation in mouse germ cells. Nature Cell Biol. 8, 623–630 (2006).

    Article  CAS  PubMed  Google Scholar 

  124. Saitou, M. Germ cell specification in mice. Curr. Opin. Genet. Dev. 19, 386–395 (2009).

    Article  CAS  PubMed  Google Scholar 

  125. Okamura, D., Tokitake, Y., Niwa, H. & Matsui, Y. Requirement of OCT3/4 function for germ cell specification. Dev. Biol. 317, 576–584 (2008).

    Article  CAS  PubMed  Google Scholar 

  126. Saitou, M., Barton, S. C. & Surani, M. A. A molecular programme for the specification of germ cell fate in mice. Nature 418, 293–300 (2002).

    Article  CAS  PubMed  Google Scholar 

  127. Sato, M. et al. Identification of PGC7, a new gene expressed specifically in preimplantation embryos and germ cells. Mech. Dev. 113, 91–94 (2002).

    Article  CAS  PubMed  Google Scholar 

  128. Seki, Y. et al. Extensive and orderly reprogramming of genome-wide chromatin modifications associated with specification and early development of germ cells in mice. Dev. Biol. 278, 440–458 (2005).

    Article  CAS  PubMed  Google Scholar 

  129. Hajkova, P. et al. Epigenetic reprogramming in mouse primordial germ cells. Mech. Dev. 117, 15–23 (2002).

    Article  CAS  PubMed  Google Scholar 

  130. Lees-Murdock, D. J., De Felici, M. & Walsh, C. P. Methylation dynamics of repetitive DNA elements in the mouse germ cell lineage. Genomics 82, 230–237 (2003).

    Article  CAS  PubMed  Google Scholar 

  131. 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 

  132. Popp, C. et al. Genome-wide erasure of DNA methylation in mouse primordial germ cells is affected by AID deficiency. Nature 463, 1101–1105 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Guibert, S., Forne, T. & Weber, M. Global profiling of DNA methylation erasure in mouse primordial germ cells. Genome Res. 22, 633–641 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Seisenberger, S. et al. The dynamics of genome-wide DNA methylation reprogramming in mouse primordial germ cells. Mol. Cell 48, 849–862 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Farthing, C. R. et al. Global mapping of DNA methylation in mouse promoters reveals epigenetic reprogramming of pluripotency genes. PLoS Genet. 4, e1000116 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Meissner, A. et al. Genome-scale DNA methylation maps of pluripotent and differentiated cells. Nature 454, 766–770 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Hajkova, P. et al. Genome-wide reprogramming in the mouse germ line entails the base excision repair pathway. Science 329, 78–82 (2010).

    Article  CAS  PubMed  Google Scholar 

  138. Dawlaty, M. M. et al. Tet1 is dispensable for maintaining pluripotency and its loss is compatible with embryonic and postnatal development. Cell Stem Cell 9, 166–175 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Yamaguchi, S. et al. Tet1 controls meiosis by regulating meiotic gene expression. Nature 20 Dec 2012 (doi:10.1038/nature11709).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Kurimoto, K. et al. Complex genome-wide transcription dynamics orchestrated by BLIMP1 for the specification of the germ cell lineage in mice. Genes Dev. 22, 1617–1635 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Kagiwada, S., Kurimoto, K., Hirota, T., Yamaji, M. & Saitou, M. Replication-coupled passive DNA demethylation for the erasure of genome imprints in mice. EMBO J. 14 Dec 2012 (doi:10.1038/emboj.2012.331).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Seki, Y. et al. Cellular dynamics associated with the genome-wide epigenetic reprogramming in migrating primordial germ cells in mice. Development 134, 2627–2638 (2007).

    Article  CAS  PubMed  Google Scholar 

  143. Hajkova, P. et al. Chromatin dynamics during epigenetic reprogramming in the mouse germ line. Nature 452, 877–881 (2008).

    Article  CAS  PubMed  Google Scholar 

  144. Smallwood, S. A. et al. Dynamic CpG island methylation landscape in oocytes and preimplantation embryos. Nature Genet. 43, 811–814 (2011).

    Article  CAS  PubMed  Google Scholar 

  145. Oakes, C. C., La Salle, S., Smiraglia, D. J., Robaire, B. & Trasler, J. M. Developmental acquisition of genome-wide DNA methylation occurs prior to meiosis in male germ cells. Dev. Biol. 307, 368–379 (2007).

    Article  CAS  PubMed  Google Scholar 

  146. 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 

  147. 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 

  148. Kaneda, M. et al. Essential role for de novo DNA methyltransferase DNMT3A in paternal and maternal imprinting. Nature 429, 900–903 (2004).

    Article  CAS  PubMed  Google Scholar 

  149. Kaneda, M. et al. Genetic evidence for DNMT3A-dependent imprinting during oocyte growth obtained by conditional knockout with Zp3-Cre and complete exclusion of DNMT3B by chimera formation. Genes Cells 1 Feb 2012 (doi:10.1111/j.1365-2443.2009.01374.x).

    Article  CAS  PubMed  Google Scholar 

  150. Bourc'his, D. & Bestor, T. H. Meiotic catastrophe and retrotransposon reactivation in male germ cells lacking DNMT3L. Nature 431, 96–99 (2004). This work links the sterile phenotype of Dnmt3l -null males to inappropriate repetitive element activity, thus highlighting the role of de novo methylation in protecting the germ line from spurious retrotransposition.

    Article  CAS  PubMed  Google Scholar 

  151. Kato, Y. et al. Role of the DNMT3 family in de novo methylation of imprinted and repetitive sequences during male germ cell development in the mouse. Hum. Mol. Genet. 16, 2272–2280 (2007).

    Article  CAS  PubMed  Google Scholar 

  152. Kobayashi, H. et al. Contribution of intragenic DNA methylation in mouse gametic DNA methylomes to establish oocyte-specific heritable marks. PLoS Genet. 8, e1002440 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. De La Fuente, R. et al. LSH is required for meiotic chromosome synapsis and retrotransposon silencing in female germ cells. Nature Cell Biol. 8, 1448–1454 (2006).

    Article  CAS  PubMed  Google Scholar 

  154. Howlett, S. K. & Reik, W. Methylation levels of maternal and paternal genomes during preimplantation development. Development 113, 119–127 (1991).

    CAS  PubMed  Google Scholar 

  155. Aravin, A. A. et al. A piRNA pathway primed by individual transposons is linked to de novo DNA methylation in mice. Mol. Cell 31, 785–799 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Watanabe, T. et al. Role for piRNAs and noncoding RNA in de novo DNA methylation of the imprinted mouse Rasgrf1 locus. Science 332, 848–852 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Aravin, A. A., Sachidanandam, R., Girard, A., Fejes-Toth, K. & Hannon, G. J. Developmentally regulated piRNA clusters implicate MILI in transposon control. Science 316, 744–747 (2007).

    Article  CAS  PubMed  Google Scholar 

  158. Kuramochi-Miyagawa, S. et al. DNA methylation of retrotransposon genes is regulated by PIWI family members MILI and MIWI2 in murine fetal testes. Genes Dev. 22, 908–917 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Santos, F., Hendrich, B., Reik, W. & Dean, W. Dynamic reprogramming of DNA methylation in the early mouse embryo. Dev. Biol. 241, 172–182 (2002).

    Article  CAS  PubMed  Google Scholar 

  160. 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 

  161. Kim, S. H. et al. Differential DNA methylation reprogramming of various repetitive sequences in mouse preimplantation embryos. Biochem. Biophys. Res. Commun. 324, 58–63 (2004).

    Article  CAS  PubMed  Google Scholar 

  162. Wossidlo, M. et al. Dynamic link of DNA demethylation, DNA strand breaks and repair in mouse zygotes. EMBO J. 29, 1877–1888 (2010). This work measures demethylation during zygotic progression, emphasizing DNA replication and the activity of DNA repair pathways as the time when demethylation is most robustly observed.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Iqbal, K., Jin, S. G., Pfeifer, G. P. & Szabo, P. E. Reprogramming of the paternal genome upon fertilization involves genome-wide oxidation of 5-methylcytosine. Proc. Natl Acad. Sci. USA 108, 3642–3647 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Wossidlo, M. et al. 5-hydroxymethylcytosine in the mammalian zygote is linked with epigenetic reprogramming. Nature Commun. 2, 241 (2011).

    Article  CAS  Google Scholar 

  165. Gu, T. P. et al. The role of Tet3 DNA dioxygenase in epigenetic reprogramming by oocytes. Nature 477, 606–610 (2011). This work first described oocyte contributed TET3 as the responsible agent in the rapid hydroxymethylation of the paternal genome after fertilization and characterized the deleterious effects of its knockout on embryonic progression.

    Article  CAS  PubMed  Google Scholar 

  166. Okada, Y., Yamagata, K., Hong, K., Wakayama, T. & Zhang, Y. A role for the elongator complex in zygotic paternal genome demethylation. Nature 463, 554–558 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Inoue, A. & Zhang, Y. Replication-dependent loss of 5-hydroxymethylcytosine in mouse preimplantation embryos. Science 334, 194 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Ziegler-Birling, C., Helmrich, A., Tora, L. & Torres-Padilla, M. E. Distribution of p53 binding protein 1 (53BP1) and phosphorylated H2A.X during mouse preimplantation development in the absence of DNA damage. Int. J. Dev. Biol. 53, 1003–1011 (2009).

    Article  CAS  PubMed  Google Scholar 

  169. Burton, A. & Torres-Padilla, M. E. Epigenetic reprogramming and development: a unique heterochromatin organization in the preimplantation mouse embryo. Brief Funct. Genom. 9, 444–454 (2010).

    Article  CAS  Google Scholar 

  170. Puschendorf, M. et al. PRC1 and SUV39H specify parental asymmetry at constitutive heterochromatin in early mouse embryos. Nature Genet. 40, 411–420 (2008).

    Article  CAS  PubMed  Google Scholar 

  171. Santos, F., Peters, A. H., Otte, A. P., Reik, W. & Dean, W. Dynamic chromatin modifications characterise the first cell cycle in mouse embryos. Dev. Biol. 280, 225–236 (2005).

    Article  CAS  PubMed  Google Scholar 

  172. Nakamura, T. et al. PGC7 binds histone H3K9me2 to protect against conversion of 5mC to 5hmC in early embryos. Nature 486, 415–419 (2012). This study clarifies the mechanism through which Stella (also known as PGC7) protects maternally methylated DNA from oxidation or demethylation by recognizing H3K9 methylation in the chromatinized maternal genome.

    Article  CAS  PubMed  Google Scholar 

  173. Nakamura, T. et al. PGC7/Stella protects against DNA demethylation in early embryogenesis. Nature Cell Biol. 9, 64–71 (2007).

    Article  CAS  PubMed  Google Scholar 

  174. Hirasawa, R. et al. Maternal and zygotic DNMT1 are necessary and sufficient for the maintenance of DNA methylation imprints during preimplantation development. Genes Dev. 22, 1607–1616 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Ratnam, S. et al. Dynamics of DNMT1 methyltransferase expression and intracellular localization during oogenesis and preimplantation development. Dev. Biol. 245, 304–314 (2002).

    Article  CAS  PubMed  Google Scholar 

  176. Hashimoto, H. et al. Recognition and potential mechanisms for replication and erasure of cytosine hydroxymethylation. Nucleic Acids Res. 40, 4841–4849 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Frauer, C. et al. Recognition of 5-hydroxymethylcytosine by the UHRF1 SRA domain. PLoS ONE 6, e21306 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Valinluck, V. & Sowers, L. C. Endogenous cytosine damage products alter the site selectivity of human DNA maintenance methyltransferase DNMT1. Cancer Res. 67, 946–950 (2007).

    Article  CAS  PubMed  Google Scholar 

  179. Reese, K. J., Lin, S., Verona, R. I., Schultz, R. M. & Bartolomei, M. S. Maintenance of paternal methylation and repression of the imprinted H19 gene requires MBD3. PLoS Genet. 3, e137 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Yildirim, O. et al. MDB3/NURD complex regulates expression of 5-hydroxymethylcytosine marked genes in embryonic stem cells. Cell 147, 1498–1510 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Ito, S. et al. Role of TET proteins in 5mC to 5hmC conversion, ES-cell self-renewal and inner cell mass specification. Nature 466, 1129–1133 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. 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 

  183. Kaji, K., Nichols, J. & Hendrich, B. MBD3, a component of the NuRD co-repressor complex, is required for development of pluripotent cells. Development 134, 1123–1132 (2007).

    Article  CAS  PubMed  Google Scholar 

  184. Kishikawa, S., Murata, T., Ugai, H., Yamazaki, T. & Yokoyama, K. K. Control elements of Dnmt1 gene are regulated in cell-cycle dependent manner. Nucleic Acids Res. (Suppl. 3), 307–308 (2003).

  185. Chuang, L. S. et al. Human DNA-(cytosine-5) methyltransferase-PCNA complex as a target for p21WAF1. Science 277, 1996–2000 (1997).

    Article  CAS  PubMed  Google Scholar 

  186. Sharif, J. et al. The SRA protein Np95 mediates epigenetic inheritance by recruiting DNMT1 to methylated DNA. Nature 450, 908–912 (2007).

    Article  CAS  PubMed  Google Scholar 

  187. Bostick, M. et al. UHRF1 plays a role in maintaining DNA methylation in mammalian cells. Science 317, 1760–1764 (2007).

    Article  CAS  PubMed  Google Scholar 

  188. Arita, K., Ariyoshi, M., Tochio, H., Nakamura, Y. & Shirakawa, M. Recognition of hemi-methylated DNA by the SRA protein UHRF1 by a base-flipping mechanism. Nature 455, 818–821 (2008).

    Article  CAS  PubMed  Google Scholar 

  189. Avvakumov, G. V. et al. Structural basis for recognition of hemi-methylated DNA by the SRA domain of human UHRF1. Nature 455, 822–825 (2008).

    Article  CAS  PubMed  Google Scholar 

  190. Du, Z. et al. DNMT1 stability is regulated by proteins coordinating deubiquitination and acetylation-driven ubiquitination. Sci. Signal. 3, ra80 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Esteve, P. O. et al. Regulation of DNMT1 stability through SET7-mediated lysine methylation in mammalian cells. Proc. Natl Acad. Sci. USA 106, 5076–5081 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  192. Robertson, K. D. et al. DNMT1 forms a complex with Rb, E2F1 and HDAC1 and represses transcription from E2F-responsive promoters. Nature Genet. 25, 338–342 (2000).

    Article  CAS  PubMed  Google Scholar 

  193. Wang, J. et al. The lysine demethylase LSD1 (KDM1) is required for maintenance of global DNA methylation. Nature Genet. 41, 125–129 (2009).

    Article  CAS  PubMed  Google Scholar 

  194. Fuks, F., Burgers, W. A., Brehm, A., Hughes-Davies, L. & Kouzarides, T. DNA methyltransferase DNMT1 associates with histone deacetylase activity. Nature Genet. 24, 88–91 (2000).

    Article  CAS  PubMed  Google Scholar 

  195. Rothbart, S. B. et al. Association of UHRF1 with methylated H3K9 directs the maintenance of DNA methylation. Nature Struct. Mol. Biol. 19, 1155–1160 (2012).

    Article  CAS  Google Scholar 

  196. Lorincz, M. C., Schubeler, D., Hutchinson, S. R., Dickerson, D. R. & Groudine, M. DNA methylation density influences the stability of an epigenetic imprint and DNMT3A/B-independent de novo methylation. Mol. Cell. Biol. 22, 7572–7580 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Tahiliani, M. et al. Conversion of 5-methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1. Science 324, 930–935 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Wu, H. et al. Dual functions of TET1 in transcriptional regulation in mouse embryonic stem cells. Nature 473, 389–393 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Cortellino, S. et al. Thymine DNA glycosylase is essential for active DNA demethylation by linked deamination-base excision repair. Cell 146, 67–79 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  200. Cortazar, D. et al. Embryonic lethal phenotype reveals a function of TDG in maintaining epigenetic stability. Nature 470, 419–423 (2011).

    Article  CAS  PubMed  Google Scholar 

  201. Siomi, M. C., Sato, K., Pezic, D. & Aravin, A. A. PIWI-interacting small RNAs: the vanguard of genome defence. Nature Rev. Mol. Cell Biol. 12, 246–258 (2011).

    Article  CAS  Google Scholar 

  202. Frost, R. J. et al. MOV10L1 is necessary for protection of spermatocytes against retrotransposons by PIWI-interacting RNAs. Proc. Natl Acad. Sci. USA 107, 11847–11852 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Reuter, M. et al. Loss of the MILI-interacting Tudor domain-containing protein-1 activates transposons and alters the MILI-associated small RNA profile. Nature Struct. Mol. Biol. 16, 639–646 (2009).

    Article  CAS  Google Scholar 

  204. Shoji, M. et al. The TDRD9–MIWI2 complex is essential for piRNA-mediated retrotransposon silencing in the mouse male germline. Dev. Cell 17, 775–787 (2009).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank members of the Meissner laboratory as well as M. M. Chan, A. Regev, T. S. Mikkelsen, R. P. Koche, H. Gu, A. Gnirke, and P. Boyle for discussion and insight. A.M. is supported by the Pew Charitable Trusts, US National Institutes of Health (NIH) grants (U01ES017155 and P01GM099117) and is a New York Stem Cell Foundation (NYSCF) Robertson Investigator.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Alexander Meissner.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Related links

Related links

FURTHER INFORMATION

Alexander Meissner's homepage

Glossary

CpG islands

Regions of several hundred to approximately two thousand base pairs that are frequently found at promoters and that exhibit strong enrichment for CpG dinucleotides. Those CpG islands at promoters are predominantly unmethylated across cell types.

Symmetrically methylated CpGs

The presence of a methyl group on carbon 5 of the cytosine base is typically found on both bases within the palindromic CpGs of opposing DNA strands, reflecting successful maintenance methylation during DNA synthesis.

CXXC

A conserved Cys–X–X–Cys domain that is frequently found in developmental epigenetic regulators that bind unmethylated CpG-containing DNA and often instruct the modification of histones in ways that oppose DNA methylation.

H2A.Z

A histone H2A variant that is incorporated into euchromatic nucleosomes and is believed to contribute to the rapid exchange of histones in active genomic loci.

Azacytidine

A cytosine analogue that incorporates into synthesizing DNA and covalently binds DNA methyltransferases (DNMTs), sequestering and inhibiting their function. It is used in some cancer therapies.

TET dioxygenases

Enzymes that associate with DNA and use α-ketoglutarate and Fe2+ cofactors to mediate the oxidation of methylated cytosine to hydroxy-, formyl- or carboxymethylcytosine, which are potential demethylation intermediates.

Lymphoid-specific helicase

(LSH). A SWI/SNF-like helicase believed to participate in the remodelling of nucleosomes that is necessary to initiate heterochromatin assembly and de novo methylation.

Minor satellite

A repetitive array of AT-rich sequences that instructs the formation of the centromeric nucleosomes to form the kinetocore and to permit sister chromatid pairing. Centromere protein B (CENPB) mediates the assembly of CENPA-containing nucleosomes.

Lagging anaphase bridges

Delayed sister chromatid segregation during mitosis as a consequence of faulty centromere formation or connection to the mitotic spindle; a frequent cause of chromosome loss and aneuploidy.

Long interspersed nuclear elements

(LINEs). A type of repetitive element that encodes the necessary proteins for integration into the genome of its reverse-transcribed RNA transcript. It preferentially integrates within gene-poor regions.

Trophectodermal lineage

Cells that contribute to placental tissues. They are derived from the external component of blastocyst stage embryos; this represents the first restriction in cellular potency during mammalian development.

Intracisternal A-type particles

(IAPs). These are notable class II long terminal repeat (LTR)-containing retroelements that are specific to mice and that retain high methylation levels throughout development.

Hemimethylation

Asymmetric methylation of only one cytosine within opposing CpGs. If not remethylated during S phase by DNA methyltransferase 1 (DNMT1), hemimethylated DNA can lead to loss of mitotic inheritance after a subsequent round of replication.

Hydroxymethylation

Oxidation of the methyl group as mediated by the TET dioxygenases through α-ketoglutarate catalysis. Hydroxymethylated residues can be further oxidized to formyl- and carboxymethylcytosine.

Germline-specific genes

Genes of unique gametogenic function that are tightly regulated outside gametes or the early embryo by DNA methylation. They are specifically demethylated during primordial germ cell specification.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Smith, Z., Meissner, A. DNA methylation: roles in mammalian development. Nat Rev Genet 14, 204–220 (2013). https://doi.org/10.1038/nrg3354

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrg3354

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