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:

Regulation, functions and transmission of bivalent chromatin during mammalian development

Abstract

Cells differentiate and progress through development guided by a dynamic chromatin landscape that mediates gene expression programmes. During development, mammalian cells display a paradoxical chromatin state: histone modifications associated with gene activation (trimethylated histone H3 Lys4 (H3K4me3)) and with gene repression (trimethylated H3 Lys27 (H3K27me3)) co-occur at promoters of developmental genes. This bivalent chromatin modification state is thought to poise important regulatory genes for expression or repression during cell-lineage specification. In this Review, we discuss recent work that has expanded our understanding of the molecular basis of bivalent chromatin and its contributions to mammalian development. We describe the factors that establish bivalency, especially histone-lysine N-methyltransferase 2B (KMT2B) and Polycomb repressive complex 2 (PRC2), and consider evidence indicating that PRC1 shapes bivalency and may contribute to its transmission between generations. We posit that bivalency is a key feature of germline and embryonic stem cells, as well as other types of stem and progenitor cells. Finally, we discuss the relevance of bivalent chromtin to human development and cancer, and outline avenues 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

Fig. 1: Overview of the establishment and maintenance of bivalent chromatin.
Fig. 2: Maintenance versus resolution of bivalency following cell divisions and differentiation.
Fig. 3: Molecular mechanisms regulating bivalent chromatin during early mouse development.

Similar content being viewed by others

References

  1. Bernstein, B. E. et al. A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell 125, 315–326 (2006).

    Article  CAS  Google Scholar 

  2. Piunti, A. & Shilatifard, A. Epigenetic balance of gene expression by Polycomb and COMPASS families. Science 352, aad9780 (2016).

    Article  Google Scholar 

  3. Azuara, V. et al. Chromatin signatures of pluripotent cell lines. Nat. Cell Biol. 8, 532–538 (2006).

    Article  CAS  Google Scholar 

  4. Mikkelsen, T. S. et al. Genome-wide maps of chromatin state in pluripotent and lineage-committed cells. Nature 448, 553–560 (2007).

    Article  CAS  Google Scholar 

  5. Zhao, X. D. et al. Whole-genome mapping of histone H3 Lys4 and 27 trimethylations reveals distinct genomic compartments in human embryonic stem cells. Cell Stem Cell 1, 286–298 (2007).

    Article  CAS  Google Scholar 

  6. Pan, G. et al. Whole-genome analysis of histone H3 lysine 4 and lysine 27 methylation in human embryonic stem cells. Cell Stem Cell 1, 299–312 (2007).

    Article  CAS  Google Scholar 

  7. Rugg-Gunn, P. J., Cox, B. J., Ralston, A. & Rossant, J. Distinct histone modifications in stem cell lines and tissue lineages from the early mouse embryo. Proc. Natl Acad. Sci. USA 107, 10783–10790 (2010).

    Article  CAS  Google Scholar 

  8. Zheng, H. et al. Resetting epigenetic memory by reprogramming of histone modifications in mammals. Mol. Cell 63, 1066–1079 (2016).

    Article  CAS  Google Scholar 

  9. Xiang, Y. et al. Epigenomic analysis of gastrulation identifies a unique chromatin state for primed pluripotency. Nat. Genet. 52, 95–105 (2020).

    Article  CAS  Google Scholar 

  10. Liu, X. et al. Distinct features of H3K4me3 and H3K27me3 chromatin domains in pre-implantation embryos. Nature 537, 558–562 (2016).

    Article  CAS  Google Scholar 

  11. Sachs, M. et al. Bivalent chromatin marks developmental regulatory genes in the mouse embryonic germline in vivo. Cell Rep. 3, 1777–1784 (2013).

    Article  CAS  Google Scholar 

  12. Ng, J.-H. et al. In vivo epigenomic profiling of germ cells reveals germ cell molecular signatures. Dev. Cell 24, 324–333 (2013).

    Article  CAS  Google Scholar 

  13. Lesch, B. J., Dokshin, G. A., Young, R. A., McCarrey, J. R. & Page, D. C. A set of genes critical to development is epigenetically poised in mouse germ cells from fetal stages through completion of meiosis. Proc. Natl Acad. Sci. USA 110, 16061–16066 (2013).

    Article  CAS  Google Scholar 

  14. Mu, W., Starmer, J., Fedoriw, A. M., Yee, D. & Magnuson, T. Repression of the soma-specific transcriptome by Polycomb-repressive complex 2 promotes male germ cell development. Gene Dev. 28, 2056–2069 (2014).

    Article  CAS  Google Scholar 

  15. Roh, T.-Y., Cuddapah, S., Cui, K. & Zhao, K. The genomic landscape of histone modifications in human T cells. Proc. Natl Acad. Sci. USA 103, 15782–15787 (2006).

    Article  CAS  Google Scholar 

  16. Barski, A. et al. High-resolution profiling of histone methylations in the human genome. Cell 129, 823–837 (2007).

    Article  CAS  Google Scholar 

  17. Voigt, P. et al. Asymmetrically modified nucleosomes. Cell 151, 181–193 (2012).

    Article  CAS  Google Scholar 

  18. Sen, S., Block, K. F., Pasini, A., Baylin, S. B. & Easwaran, H. Genome-wide positioning of bivalent mononucleosomes. BMC Med. Genomics 9, 60 (2016).

    Article  Google Scholar 

  19. Voigt, P., Tee, W.-W. & Reinberg, D. A double take on bivalent promoters. Gene Dev. 27, 1318–1338 (2013).

    Article  CAS  Google Scholar 

  20. Lesch, B. J., Silber, S. J., McCarrey, J. R. & Page, D. C. Parallel evolution of male germline epigenetic poising and somatic development in animals. Nat. Genet. 48, 888–894 (2016).

    Article  CAS  Google Scholar 

  21. Dattani, A. et al. Epigenetic analyses of planarian stem cells demonstrate conservation of bivalent histone modifications in animal stem cells. Genome Res. 28, 1543–1554 (2018).

    Article  CAS  Google Scholar 

  22. Vastenhouw, N. L. et al. Chromatin signature of embryonic pluripotency is established during genome activation. Nature 464, 922–926 (2010).

    Article  CAS  Google Scholar 

  23. Kang, H. et al. Bivalent complexes of PRC1 with orthologs of BRD4 and MOZ/MORF target developmental genes in Drosophila. Gene Dev. 31, 1988–2002 (2017).

    Article  CAS  Google Scholar 

  24. Schertel, C. et al. A large-scale, in vivo transcription factor screen defines bivalent chromatin as a key property of regulatory factors mediating Drosophila wing development. Genome Res. 25, 514–523 (2015).

    Article  CAS  Google Scholar 

  25. Akmammedov, A., Geigges, M. & Paro, R. Bivalency in Drosophila embryos is associated with strong inducibility of Polycomb target genes. Fly 13, 42–50 (2019).

    Article  Google Scholar 

  26. Akkers, R. C. et al. A hierarchy of H3K4me3 and H3K27me3 acquisition in spatial gene regulation in xenopus embryos. Dev. Cell 17, 425–434 (2009).

    Article  CAS  Google Scholar 

  27. Yu, J.-R., Lee, C.-H., Oksuz, O., Stafford, J. M. & Reinberg, D. PRC2 is high maintenance. Gene Dev. 33, 903–935 (2019).

    Article  CAS  Google Scholar 

  28. Schuettengruber, B., Bourbon, H.-M., Croce, L. D. & Cavalli, G. Genome regulation by polycomb and trithorax: 70 years and counting. Cell 171, 34–57 (2017).

    Article  CAS  Google Scholar 

  29. Cenik, B. K. & Shilatifard, A. COMPASS and SWI/SNF complexes in development and disease. Nat. Rev. Genet. 22, 38–58 (2021).

    Article  CAS  Google Scholar 

  30. Piunti, A. & Shilatifard, A. The roles of Polycomb repressive complexes in mammalian development and cancer. Nat. Rev. Mol. Cell Biol. 22, 326–345 (2021).

    Article  CAS  Google Scholar 

  31. Blackledge, N. P. & Klose, R. J. The molecular principles of gene regulation by Polycomb repressive complexes. Nat. Rev. Mol. Cell Biol. 22, 815–833 (2021).

    Article  CAS  Google Scholar 

  32. Denissov, S. et al. Mll2 is required for H3K4 trimethylation on bivalent promoters in embryonic stem cells, whereas Mll1 is redundant. Development 141, 526–537 (2014).

    Article  CAS  Google Scholar 

  33. Hu, D. et al. Not All H3K4 methylations are created equal: Mll2/COMPASS dependency in primordial germ cell specification. Mol. Cell 65, 460–475.e6 (2017).

    Article  CAS  Google Scholar 

  34. Hu, D. et al. The Mll2 branch of the COMPASS family regulates bivalent promoters in mouse embryonic stem cells. Nat. Struct. Mol. Biol. 20, 1093–1097 (2013).

    Article  CAS  Google Scholar 

  35. Mas, G. et al. Promoter bivalency favors an open chromatin architecture in embryonic stem cells. Nat. Genet. 50, 1452–1462 (2018).

    Article  CAS  Google Scholar 

  36. Sze, C. C. et al. Coordinated regulation of cellular identity–associated H3K4me3 breadth by the COMPASS family. Sci. Adv. 6, eaaz4764 (2020).

    Article  CAS  Google Scholar 

  37. Douillet, D. et al. Uncoupling histone H3K4 trimethylation from developmental gene expression via an equilibrium of COMPASS, Polycomb and DNA methylation. Nat. Genet. 52, 615–625 (2020).

    Article  CAS  Google Scholar 

  38. Rao, R. C. & Dou, Y. Hijacked in cancer: the KMT2 (MLL) family of methyltransferases. Nat. Rev. Cancer 15, 334–346 (2015).

    Article  CAS  Google Scholar 

  39. Shilatifard, A. The COMPASS family of histone H3K4 methylases: mechanisms of regulation in development and disease pathogenesis. Annu. Rev. Biochem. 81, 65–95 (2012).

    Article  CAS  Google Scholar 

  40. Ferrari, K. J. et al. Polycomb-dependent H3K27me1 and H3K27me2 regulate active transcription and enhancer fidelity. Mol. Cell 53, 49–62 (2014).

    Article  CAS  Google Scholar 

  41. Lee, C.-H. et al. Distinct stimulatory mechanisms regulate the catalytic activity of polycomb repressive complex 2. Mol. Cell 70, 435–448.e5 (2018).

    Article  CAS  Google Scholar 

  42. Lavarone, E., Barbieri, C. M. & Pasini, D. Dissecting the role of H3K27 acetylation and methylation in PRC2 mediated control of cellular identity. Nat. Commun. 10, 1679 (2019).

    Article  Google Scholar 

  43. Cao, R. & Zhang, Y. SUZ12 is required for both the histone methyltransferase activity and the silencing function of the EED-EZH2 complex. Mol. Cell 15, 57–67 (2004).

    Article  CAS  Google Scholar 

  44. Højfeldt, J. W. et al. Accurate H3K27 methylation can be established de novo by SUZ12-directed PRC2. Nat. Struct. Mol. Biol. 25, 225–232 (2018).

    Article  Google Scholar 

  45. Qi, W. et al. An allosteric PRC2 inhibitor targeting the H3K27me3 binding pocket of EED. Nat. Chem. Biol. 13, 381–388 (2017).

    Article  CAS  Google Scholar 

  46. Jiao, L. & Liu, X. Structural basis of histone H3K27 trimethylation by an active polycomb repressive complex 2. Science 350, aac4383 (2015).

    Article  Google Scholar 

  47. Margueron, R. et al. Role of the polycomb protein EED in the propagation of repressive histone marks. Nature 461, 762–767 (2009).

    Article  CAS  Google Scholar 

  48. Mierlo, G., van, Veenstra, G. J. C., Vermeulen, M. & Marks, H. The complexity of PRC2 subcomplexes. Trends Cell Biol. 29, 660–671 (2019).

    Article  Google Scholar 

  49. Li, H. et al. Polycomb-like proteins link the PRC2 complex to CpG islands. Nature 549, 287–291 (2017).

    Article  CAS  Google Scholar 

  50. Choi, J. et al. DNA binding by PHF1 prolongs PRC2 residence time on chromatin and thereby promotes H3K27 methylation. Nat. Struct. Mol. Biol. 24, 1039–1047 (2017).

    Article  CAS  Google Scholar 

  51. Perino, M. et al. MTF2 recruits polycomb repressive complex 2 by helical-shape-selective DNA binding. Nat. Genet. 50, 1002–1010 (2018).

    Article  CAS  Google Scholar 

  52. Hunkapiller, J. et al. Polycomb-like 3 promotes polycomb repressive complex 2 binding to CpG islands and embryonic stem cell self-renewal. PLoS Genet. 8, e1002576 (2012).

    Article  CAS  Google Scholar 

  53. Healy, E. et al. PRC2.1 and PRC2.2 synergize to coordinate H3K27 trimethylation. Mol. Cell 76, 437–452.e6 (2019).

    Article  CAS  Google Scholar 

  54. Højfeldt, J. W. et al. Non-core subunits of the PRC2 complex are collectively required for its target-site specificity. Mol. Cell 76, 423–436.e3 (2019).

    Article  Google Scholar 

  55. Perino, M. et al. Two functional axes of feedback-enforced PRC2 recruitment in mouse embryonic stem cells. Stem Cell Rep. 15, 1287–1300 (2020).

    Article  CAS  Google Scholar 

  56. Blackledge, N. P. et al. Variant PRC1 complex-dependent H2A ubiquitylation drives PRC2 recruitment and polycomb domain formation. Cell 157, 1445–1459 (2014).

    Article  CAS  Google Scholar 

  57. Boyer, L. A. et al. Polycomb complexes repress developmental regulators in murine embryonic stem cells. Nature 441, 349–353 (2006).

    Article  CAS  Google Scholar 

  58. Bracken, A. P., Dietrich, N., Pasini, D., Hansen, K. H. & Helin, K. Genome-wide mapping of Polycomb target genes unravels their roles in cell fate transitions. Gene Dev. 20, 1123–1136 (2006).

    Article  CAS  Google Scholar 

  59. Francis, N. J., Kingston, R. E. & Woodcock, C. L. Chromatin compaction by a Polycomb group protein complex. Science 306, 1574–1577 (2004).

    Article  CAS  Google Scholar 

  60. Wani, A. H. et al. Chromatin topology is coupled to Polycomb group protein subnuclear organization. Nat. Commun. 7, 10291 (2016).

    Article  CAS  Google Scholar 

  61. Geng, Z. & Gao, Z. Mammalian PRC1 complexes: compositional complexity and diverse molecular mechanisms. Int. J. Mol. Sci. 21, 8594 (2020).

    Article  CAS  Google Scholar 

  62. Landeira, D. et al. Jarid2 is a PRC2 component in embryonic stem cells required for multi-lineage differentiation and recruitment of PRC1 and RNA Polymerase II to developmental regulators. Nat. Cell Biol. 12, 618–624 (2010).

    Article  CAS  Google Scholar 

  63. Ku, M. et al. Genomewide analysis of PRC1 and PRC2 occupancy identifies two classes of bivalent domains. PLoS Genet. 4, e1000242 (2008).

    Article  Google Scholar 

  64. Schmitges, F. W. et al. Histone methylation by PRC2 is inhibited by active chromatin marks. Mol. Cell 42, 330–341 (2011).

    Article  CAS  Google Scholar 

  65. Sneppen, K. & Ringrose, L. Theoretical analysis of Polycomb-Trithorax systems predicts that poised chromatin is bistable and not bivalent. Nat. Commun. 10, 2133 (2019).

    Article  Google Scholar 

  66. Steffen, P. A. & Ringrose, L. What are memories made of? How Polycomb and Trithorax proteins mediate epigenetic memory. Nat. Rev. Mol. Cell Biol. 15, 340–356 (2014).

    Article  CAS  Google Scholar 

  67. Lee, T. I. et al. Control of developmental regulators by polycomb in human embryonic stem cells. Cell 125, 301–313 (2006).

    Article  CAS  Google Scholar 

  68. Tanay, A., O’Donnell, A. H., Damelin, M. & Bestor, T. H. Hyperconserved CpG domains underlie Polycomb-binding sites. Proc. Natl Acad. Sci. USA 104, 5521–5526 (2007).

    Article  CAS  Google Scholar 

  69. Mendenhall, E. M. et al. GC-rich sequence elements recruit PRC2 in mammalian ES cells. PLoS Genet. 6, e1001244 (2010).

    Article  Google Scholar 

  70. Wachter, E. et al. Synthetic CpG islands reveal DNA sequence determinants of chromatin structure. Elife 3, e03397 (2014).

    Article  Google Scholar 

  71. Jermann, P., Hoerner, L., Burger, L. & Schübeler, D. Short sequences can efficiently recruit histone H3 lysine 27 trimethylation in the absence of enhancer activity and DNA methylation. Proc. Natl Acad. Sci. USA 111, E3415–E3421 (2014).

    Article  CAS  Google Scholar 

  72. Lynch, M. D. et al. An interspecies analysis reveals a key role for unmethylated CpG dinucleotides in vertebrate Polycomb complex recruitment. EMBO J. 31, 317–329 (2012).

    Article  CAS  Google Scholar 

  73. Ballaré, C. et al. Phf19 links methylated Lys36 of histone H3 to regulation of Polycomb activity. Nat. Struct. Mol. Biol. 19, 1257–1265 (2012).

    Article  Google Scholar 

  74. Brien, G. L. et al. Polycomb PHF19 binds H3K36me3 and recruits PRC2 and demethylase NO66 to embryonic stem cell genes during differentiation. Nat. Struct. Mol. Biol. 19, 1273–1281 (2012).

    Article  CAS  Google Scholar 

  75. Cai, L. et al. An H3K36 methylation-engaging Tudor motif of polycomb-like proteins mediates PRC2 complex targeting. Mol. Cell 49, 571–582 (2013).

    Article  CAS  Google Scholar 

  76. Casanova, M. et al. Polycomblike 2 facilitates the recruitment of PRC2 Polycomb group complexes to the inactive X chromosome and to target loci in embryonic stem cells. Development 138, 1471–1482 (2011).

    Article  CAS  Google Scholar 

  77. Walker, E. et al. Polycomb-like 2 associates with PRC2 and regulates transcriptional networks during mouse embryonic stem cell self-renewal and differentiation. Cell Stem Cell 6, 153–166 (2010).

    Article  CAS  Google Scholar 

  78. Sarma, K., Margueron, R., Ivanov, A., Pirrotta, V. & Reinberg, D. Ezh2 requires PHF1 to efficiently catalyze H3 lysine 27 trimethylation in vivo. Mol. Cell Biol. 28, 2718–2731 (2008).

    Article  CAS  Google Scholar 

  79. van de Lagemaat, L. N. et al. CpG binding protein (CFP1) occupies open chromatin regions of active genes, including enhancers and non-CpG islands. Epigenet Chromatin 11, 59 (2018).

    Article  Google Scholar 

  80. Fouse, S. D. et al. Promoter CpG methylation contributes to ES cell gene regulation in parallel with Oct4/Nanog, PcG complex, and histone H3 K4/K27 trimethylation. Cell Stem Cell 2, 160–169 (2008).

    Article  CAS  Google Scholar 

  81. Hanna, C. W. et al. MLL2 conveys transcription-independent H3K4 trimethylation in oocytes. Nat. Struct. Mol. Biol. 25, 73–82 (2018).

    Article  CAS  Google Scholar 

  82. Howe, F. S., Fischl, H., Murray, S. C. & Mellor, J. Is H3K4me3 instructive for transcription activation? Bioessays 39, 1–12 (2017).

    Article  CAS  Google Scholar 

  83. Morgan, M. A. J. & Shilatifard, A. Reevaluating the roles of histone-modifying enzymes and their associated chromatin modifications in transcriptional regulation. Nat. Genet. 52, 1271–1281 (2020).

    Article  CAS  Google Scholar 

  84. Ladopoulos, V. et al. The histone methyltransferase KMT2B is required for RNA polymerase II association and protection from DNA methylation at the MagohB CpG island promoter. Mol. Cell Biol. 33, 1383–1393 (2013).

    Article  CAS  Google Scholar 

  85. Yan, J., Dutta, B., Hee, Y. T. & Chng, W.-J. Towards understanding of PRC2 binding to RNA. RNA Biol. 16, 176–184 (2019).

    Article  Google Scholar 

  86. Betancur, J. G. Pervasive lncRNA binding by epigenetic modifying complexes — the challenges ahead. Biochim. Biophys. Acta 1859, 93–101 (2016).

    Article  CAS  Google Scholar 

  87. Davidovich, C. et al. Toward a consensus on the binding specificity and promiscuity of PRC2 for RNA. Mol. Cell 57, 552–558 (2015).

    Article  CAS  Google Scholar 

  88. Rosenberg, M. et al. Motif-driven interactions between RNA and PRC2 are rheostats that regulate transcription elongation. Nat. Struct. Mol. Biol. 28, 103–117 (2021).

    Article  CAS  Google Scholar 

  89. Wei, C. et al. RBFox2 binds nascent RNA to globally regulate Polycomb complex 2 targeting in mammalian genomes. Mol. Cell 62, 875–889 (2016).

    Article  CAS  Google Scholar 

  90. Wang, Z. et al. Prediction of histone post-translational modification patterns based on nascent transcription data. Nat. Genet. 54, 295–305 (2022).

    Article  CAS  Google Scholar 

  91. Riising, E. M. et al. Gene silencing triggers Polycomb repressive complex 2 recruitment to CpG islands genome wide. Mol. Cell 55, 347–360 (2014).

    Article  CAS  Google Scholar 

  92. Hosogane, M., Funayama, R., Shirota, M. & Nakayama, K. Lack of transcription triggers H3K27me3 accumulation in the gene body. Cell Rep. 16, 696–706 (2016).

    Article  CAS  Google Scholar 

  93. Long, Y. et al. RNA is essential for PRC2 chromatin occupancy and function in human pluripotent stem cells. Nat. Genet. 52, 931–938 (2020).

    Article  CAS  Google Scholar 

  94. Janssen, S. M. & Lorincz, M. C. Interplay between chromatin marks in development and disease. Nat. Rev. Genet. 23, 137–153 (2022).

    Article  CAS  Google Scholar 

  95. Greenberg, M. V. C. & Bourc’his, D. The diverse roles of DNA methylation in mammalian development and disease. Nat. Rev. Mol. Cell Biol. 20, 590–607 (2019).

    Article  CAS  Google Scholar 

  96. van Mierlo, G. et al. Integrative proteomic profiling reveals PRC2-dependent epigenetic crosstalk maintains ground-state pluripotency. Cell Stem Cell 24, 123–137.e8 (2019).

    Article  Google Scholar 

  97. King, A. D. et al. Reversible regulation of promoter and enhancer histone landscape by DNA methylation in mouse embryonic stem cells. Cell Rep. 17, 289–302 (2016).

    Article  CAS  Google Scholar 

  98. Li, Y. et al. Genome-wide analyses reveal a role of Polycomb in promoting hypomethylation of DNA methylation valleys. Genome Biol. 19, 18 (2018).

    Article  Google Scholar 

  99. 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  Google Scholar 

  100. Reddington, J. P. et al. Redistribution of H3K27me3 upon DNA hypomethylation results in de-repression of Polycomb target genes. Genome Biol. 14, R25–R25 (2013).

    Article  Google Scholar 

  101. McLaughlin, K. et al. DNA methylation directs Polycomb-dependent 3D genome re-organization in naive pluripotency. Cell Rep. 29, 1974–1985.e6 (2019).

    Article  CAS  Google Scholar 

  102. Manzo, M. et al. Isoform-specific localization of DNMT3A regulates DNA methylation fidelity at bivalent CpG islands. EMBO J. 36, 3421–3434 (2017).

    Article  CAS  Google Scholar 

  103. Gu, T. et al. DNMT3A and TET1 cooperate to regulate promoter epigenetic landscapes in mouse embryonic stem cells. Genome Biol. 19, 88 (2018).

    Article  Google Scholar 

  104. Ooi, S. K. T. et al. DNMT3L connects unmethylated lysine 4 of histone H3 to de novo methylation of DNA. Nature 448, 714–717 (2007).

    Article  CAS  Google Scholar 

  105. Guo, X. et al. Structural insight into autoinhibition and histone H3-induced activation of DNMT3A. Nature 517, 640–644 (2015).

    Article  CAS  Google Scholar 

  106. 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  Google Scholar 

  107. Li, B.-Z. et al. Histone tails regulate DNA methylation by allosterically activating de novo methyltransferase. Cell Res. 21, 1172–1181 (2011).

    Article  CAS  Google Scholar 

  108. Zhang, Y. et al. Chromatin methylation activity of Dnmt3a and Dnmt3a/3L is guided by interaction of the ADD domain with the histone H3 tail. Nucleic Acids Res. 38, 4246–4253 (2010).

    Article  CAS  Google Scholar 

  109. Noh, K.-M. et al. Engineering of a histone-recognition domain in Dnmt3a alters the epigenetic landscape and phenotypic features of mouse ESCs. Mol. Cell 59, 89–103 (2015).

    Article  CAS  Google Scholar 

  110. Ross, S. E. & Bogdanovic, O. TET enzymes, DNA demethylation and pluripotency. Biochem. Soc. Trans. 47, 875–885 (2019).

    Article  CAS  Google Scholar 

  111. Neri, F. et al. Genome-wide analysis identifies a functional association of Tet1 and Polycomb repressive complex 2 in mouse embryonic stem cells. Genome Biol. 14, R91 (2013).

    Article  Google Scholar 

  112. Villaseñor, R. et al. ChromID identifies the protein interactome at chromatin marks. Nat. Biotechnol. 38, 728–736 (2020).

    Article  Google Scholar 

  113. Parry, A., Rulands, S. & Reik, W. Active turnover of DNA methylation during cell fate decisions. Nat. Rev. Genet. 22, 59–66 (2021).

    Article  CAS  Google Scholar 

  114. Ginno, P. A. et al. A genome-scale map of DNA methylation turnover identifies site-specific dependencies of DNMT and TET activity. Nat. Commun. 11, 2680 (2020).

    Article  CAS  Google Scholar 

  115. Verma, N. et al. TET proteins safeguard bivalent promoters from de novo methylation in human embryonic stem cells. Nat. Genet. 50, 83–95 (2018).

    Article  CAS  Google Scholar 

  116. Eckersley-Maslin, M. A. Keeping your options open: insights from Dppa2/4 into how epigenetic priming factors promote cell plasticity. Biochem. Soc. Trans. 48, 2891–2902 (2020).

    Article  CAS  Google Scholar 

  117. Kubinyecz, O., Santos, F., Drage, D., Reik, W. & Eckersley-Maslin, M. A. Maternal Dppa2 and Dppa4 are dispensable for zygotic genome activation but important for offspring survival. Development 148, dev200191 (2021).

    Article  CAS  Google Scholar 

  118. Chen, Z., Xie, Z. & Zhang, Y. DPPA2 and DPPA4 are dispensable for mouse zygotic genome activation and preimplantation development. Development 148, dev200178 (2021).

    Article  CAS  Google Scholar 

  119. Eckersley-Maslin, M. A. et al. Epigenetic priming by Dppa2 and 4 in pluripotency facilitates multi-lineage commitment. Nat. Struct. Mol. Biol. 27, 1–10 (2020).

    Article  Google Scholar 

  120. Gretarsson, K. H. & Hackett, J. A. Dppa2 and Dppa4 counteract de novo methylation to establish a permissive epigenome for development. Nat. Struct. Mol. Biol. 27, 1–11 (2020).

    Article  Google Scholar 

  121. Zhang, J. et al. Highly enriched BEND3 prevents the premature activation of bivalent genes during differentiation. Science 375, 1053–1058 (2022).

    Article  CAS  Google Scholar 

  122. Alabert, C. et al. Two distinct modes for propagation of histone PTMs across the cell cycle. Gene Dev. 29, 585–590 (2015).

    Article  CAS  Google Scholar 

  123. Radman-Livaja, M. et al. Patterns and mechanisms of ancestral histone protein inheritance in budding yeast. PLoS Biol. 9, e1001075 (2011).

    Article  CAS  Google Scholar 

  124. Reverón-Gómez, N. et al. Accurate recycling of parental histones reproduces the histone modification landscape during DNA replication. Mol. Cell 72, 239–249.e5 (2018).

    Article  Google Scholar 

  125. Singh, A. M. et al. Cell-cycle control of bivalent epigenetic domains regulates the exit from pluripotency. Stem Cell Rep. 5, 323–336 (2015).

    Article  CAS  Google Scholar 

  126. Asenjo, H. G. et al. Polycomb regulation is coupled to cell cycle transition in pluripotent stem cells. Sci. Adv. 6, eaay4768 (2020).

    Article  CAS  Google Scholar 

  127. Liu, L., Michowski, W., Kolodziejczyk, A. & Sicinski, P. The cell cycle in stem cell proliferation, pluripotency and differentiation. Nat. Cell Biol. 21, 1060–1067 (2019).

    Article  CAS  Google Scholar 

  128. O’Farrell, P. H., Stumpff, J. & Su, T. T. Embryonic cleavage cycles: how is a mouse like a fly? Curr. Biol. 14, R35–R45 (2004).

    Article  Google Scholar 

  129. O’Farrell, P. H. Growing an embryo from a single cell: a hurdle in animal life. CSH Perspect. Biol. 7, a019042 (2015).

    Google Scholar 

  130. Chamberlain, S. J., Yee, D. & Magnuson, T. Polycomb repressive complex 2 is dispensable for maintenance of embryonic stem cell pluripotency. Stem Cell 26, 1496–1505 (2008).

    Article  CAS  Google Scholar 

  131. Shah, R. N. et al. Re-evaluating the role of nucleosomal bivalency in early development. Preprint at bioRxiv https://doi.org/10.1101/2021.09.09.458948 (2021).

    Article  Google Scholar 

  132. Macrae, T. A. & Ramalho-Santos, M. The deubiquitinase Usp9x regulates PRC2-mediated chromatin reprogramming during mouse development. Nat. Commun. 12, 1865 (2021).

    Article  CAS  Google Scholar 

  133. Kumar, D., Cinghu, S., Oldfield, A. J., Yang, P. & Jothi, R. Decoding the function of bivalent chromatin in development and cancer. Genome Res. 31, 2170–2184 (2021).

    Article  Google Scholar 

  134. Stock, J. K. et al. Ring1-mediated ubiquitination of H2A restrains poised RNA polymerase II at bivalent genes in mouse ES cells. Nat. Cell Biol. 9, 1428–1435 (2007).

    Article  CAS  Google Scholar 

  135. Brookes, E. et al. Polycomb associates genome-wide with a specific RNA polymerase II variant, and regulates metabolic genes in ESCs. Cell Stem Cell 10, 157–170 (2012).

    Article  CAS  Google Scholar 

  136. Liu, J., Wu, X., Zhang, H., Pfeifer, G. P. & Lu, Q. Dynamics of RNA polymerase II pausing and bivalent histone H3 methylation during neuronal differentiation in brain development. Cell Rep. 20, 1307–1318 (2017).

    Article  CAS  Google Scholar 

  137. Yoon, S.-J., Foley, J. W. & Baker, J. C. HEB associates with PRC2 and SMAD2/3 to regulate developmental fates. Nat. Commun. 6, 6546 (2015).

    Article  CAS  Google Scholar 

  138. Tee, W.-W., Shen, S. S., Oksuz, O., Narendra, V. & Reinberg, D. Erk1/2 activity promotes chromatin features and RNAPII phosphorylation at developmental promoters in mouse ESCs. Cell 156, 678–690 (2014).

    Article  CAS  Google Scholar 

  139. Compe, E. & Egly, J.-M. TFIIH: when transcription met DNA repair. Nat. Rev. Mol. Cell Biol. 13, 343–354 (2012).

    Article  CAS  Google Scholar 

  140. Alder, O. et al. Ring1B and Suv39h1 delineate distinct chromatin states at bivalent genes during early mouse lineage commitment. Development 137, 2483–2492 (2010).

    Article  CAS  Google Scholar 

  141. Bilodeau, S., Kagey, M. H., Frampton, G. M., Rahl, P. B. & Young, R. A. SetDB1 contributes to repression of genes encoding developmental regulators and maintenance of ES cell state. Gene Dev. 23, 2484–2489 (2009).

    Article  CAS  Google Scholar 

  142. Yuan, P. et al. Eset partners with Oct4 to restrict extraembryonic trophoblast lineage potential in embryonic stem cells. Gene Dev. 23, 2507–2520 (2009).

    Article  CAS  Google Scholar 

  143. Shen, H., Xu, W. & Lan, F. Histone lysine demethylases in mammalian embryonic development. Exp. Mol. Med. 49, e325 (2017).

    Article  CAS  Google Scholar 

  144. Kidder, B. L., Hu, G. & Zhao, K. KDM5B focuses H3K4 methylation near promoters and enhancers during embryonic stem cell self-renewal and differentiation. Genome Biol. 15, R32 (2014).

    Article  Google Scholar 

  145. Dahle, Y., Kumar, A. & Kuehn, M. R. Nodal signaling recruits the histone demethylase Jmjd3 to counteract polycomb-mediated repression at target genes. Sci. Signal. 3, ra48 (2010).

    Article  Google Scholar 

  146. Dhar, S. S. et al. An essential role for UTX in resolution and activation of bivalent promoters. Nucleic Acids Res. 44, 3659–3674 (2016).

    Article  CAS  Google Scholar 

  147. Gao, Y., Gan, H., Lou, Z. & Zhang, Z. Asf1a resolves bivalent chromatin domains for the induction of lineage-specific genes during mouse embryonic stem cell differentiation. Proc. Natl Acad. Sci. USA 115, E6162–E6171 (2018).

    Article  CAS  Google Scholar 

  148. Cruz-Molina, S. et al. PRC2 facilitates the regulatory topology required for poised enhancer function during pluripotent stem cell differentiation. Cell Stem Cell 20, 689–705.e9 (2017).

    Article  CAS  Google Scholar 

  149. Crispatzu, G. et al. The chromatin, topological and regulatory properties of pluripotency-associated poised enhancers are conserved in vivo. Nat. Commun. 12, 4344 (2021).

    Article  CAS  Google Scholar 

  150. Kar, G. et al. Flipping between Polycomb repressed and active transcriptional states introduces noise in gene expression. Nat. Commun. 8, 36 (2017).

    Article  Google Scholar 

  151. Creyghton, M. P. et al. Histone H3K27ac separates active from poised enhancers and predicts developmental state. Proc. Natl Acad. Sci. USA 107, 21931–21936 (2010).

    Article  CAS  Google Scholar 

  152. Rada-Iglesias, A. et al. A unique chromatin signature uncovers early developmental enhancers in humans. Nature 470, 279–283 (2011).

    Article  CAS  Google Scholar 

  153. Zentner, G. E., Tesar, P. J. & Scacheri, P. C. Epigenetic signatures distinguish multiple classes of enhancers with distinct cellular functions. Genome Res. 21, 1273–1283 (2011).

    Article  CAS  Google Scholar 

  154. Calo, E. & Wysocka, J. Modification of enhancer chromatin: what, how, and why? Mol. Cell 49, 825–837 (2013).

    Article  CAS  Google Scholar 

  155. Yang, Z. et al. EBS is a bivalent histone reader that regulates floral phase transition in Arabidopsis. Nat. Genet. 50, 1247–1253 (2018).

    Article  CAS  Google Scholar 

  156. Reinig, J., Ruge, F., Howard, M. & Ringrose, L. A theoretical model of Polycomb/Trithorax action unites stable epigenetic memory and dynamic regulation. Nat. Commun. 11, 4782 (2020).

    Article  CAS  Google Scholar 

  157. Berry, S., Dean, C. & Howard, M. Slow chromatin dynamics allow polycomb target genes to filter fluctuations in transcription factor activity. Cell Syst. 4, 445–457.e8 (2017).

    Article  CAS  Google Scholar 

  158. Lu, X. et al. Evolutionary epigenomic analyses in mammalian early embryos reveal species-specific innovations and conserved principles of imprinting. Sci. Adv. 7, eabi6178 (2021).

    Article  CAS  Google Scholar 

  159. Galonska, C., Ziller, M. J., Karnik, R. & Meissner, A. Ground state conditions induce rapid reorganization of core pluripotency factor binding before global epigenetic reprogramming. Cell Stem Cell 17, 462–470 (2015).

    Article  CAS  Google Scholar 

  160. Marks, H. et al. The transcriptional and epigenomic foundations of ground state pluripotency. Cell 149, 590–604 (2012).

    Article  CAS  Google Scholar 

  161. Tosolini, M. et al. Contrasting epigenetic states of heterochromatin in the different types of mouse pluripotent stem cells. Sci. Rep. 8, 5776 (2018).

    Article  Google Scholar 

  162. Kumar, B. & Elsässer, S. J. Quantitative multiplexed ChIP reveals global alterations that shape promoter bivalency in ground state embryonic stem cells. Cell Rep. 28, 3274–3284.e5 (2019).

    Article  CAS  Google Scholar 

  163. Kurimoto, K. et al. Quantitative dynamics of chromatin remodeling during germ cell specification from mouse embryonic stem cells. Cell Stem Cell 16, 517–532 (2015).

    Article  CAS  Google Scholar 

  164. Glaser, S. et al. Multiple epigenetic maintenance factors implicated by the loss of Mll2 in mouse development. Development 133, 1423–1432 (2006).

    Article  CAS  Google Scholar 

  165. Lubitz, S., Glaser, S., Schaft, J., Stewart, A. F. & Anastassiadis, K. Increased apoptosis and skewed differentiation in mouse embryonic stem cells lacking the histone methyltransferase Mll2. Mol. Biol. Cell 18, 2356–2366 (2007).

    Article  CAS  Google Scholar 

  166. Leeb, M. et al. Polycomb complexes act redundantly to repress genomic repeats and genes. Gene Dev. 24, 265–276 (2010).

    Article  CAS  Google Scholar 

  167. Shan, Y. et al. PRC2 specifies ectoderm lineages and maintains pluripotency in primed but not naïve ESCs. Nat. Commun. 8, 672 (2017).

    Article  Google Scholar 

  168. Moody, J. D. et al. First critical repressive H3K27me3 marks in embryonic stem cells identified using designed protein inhibitor. Proc. Natl Acad. Sci. USA 114, 10125–10130 (2017).

    Article  CAS  Google Scholar 

  169. Collinson, A. et al. Deletion of the polycomb-group protein EZH2 leads to compromised self-renewal and differentiation defects in human embryonic stem cells. Cell Rep. 17, 2700–2714 (2016).

    Article  CAS  Google Scholar 

  170. Pasini, D., Bracken, A. P., Hansen, J. B., Capillo, M. & Helin, K. The Polycomb Group protein Suz12 is required for embryonic stem cell differentiation. Mol. Cell Biol. 27, 3769–3779 (2007).

    Article  CAS  Google Scholar 

  171. Thornton, S. R., Butty, V. L., Levine, S. S. & Boyer, L. A. Polycomb repressive complex 2 regulates lineage fidelity during embryonic stem cell differentiation. PLoS ONE 9, e110498 (2014).

    Article  Google Scholar 

  172. Nishimura, K., Fukagawa, T., Takisawa, H., Kakimoto, T. & Kanemaki, M. An auxin-based degron system for the rapid depletion of proteins in nonplant cells. Nat. Methods 6, 917–922 (2009).

    Article  CAS  Google Scholar 

  173. Petracovici, A. & Bonasio, R. Distinct PRC2 subunits regulate maintenance and establishment of Polycomb repression during differentiation. Mol. Cell 81, 2625–2639.e5 (2021).

    Article  CAS  Google Scholar 

  174. Dobrinić, P., Szczurek, A. T. & Klose, R. J. PRC1 drives Polycomb-mediated gene repression by controlling transcription initiation and burst frequency. Nat. Struct. Mol. Biol. 28, 811–824 (2021).

    Article  Google Scholar 

  175. Nabet, B. et al. The dTAG system for immediate and target-specific protein degradation. Nat. Chem. Biol. 14, 431–441 (2018).

    Article  CAS  Google Scholar 

  176. Zepeda-Martinez, J. A. et al. Parallel PRC2/cPRC1 and vPRC1 pathways silence lineage-specific genes and maintain self-renewal in mouse embryonic stem cells. Sci. Adv. 6, eaax5692 (2020).

    Article  CAS  Google Scholar 

  177. Grosswendt, S. et al. Epigenetic regulator function through mouse gastrulation. Nature 584, 102–108 (2020).

    Article  CAS  Google Scholar 

  178. Jambhekar, A., Dhall, A. & Shi, Y. Roles and regulation of histone methylation in animal development. Nat. Rev. Mol. Cell Biol. 20, 625–641 (2019).

    Article  CAS  Google Scholar 

  179. Faust, C., Schumacher, A., Holdener, B. & Magnuson, T. The eed mutation disrupts anterior mesoderm production in mice. Development 121, 273–285 (1995).

    Article  CAS  Google Scholar 

  180. Pasini, D., Bracken, A. P., Jensen, M. R., Denchi, E. L. & Helin, K. Suz12 is essential for mouse development and for EZH2 histone methyltransferase activity. EMBO J. 23, 4061–4071 (2004).

    Article  CAS  Google Scholar 

  181. O’Carroll, D. et al. The Polycomb-group gene Ezh2 is required for early mouse development. Mol. Cell Biol. 21, 4330–4336 (2001).

    Article  Google Scholar 

  182. Morin-Kensicki, E. M., Faust, C., LaMantia, C. & Magnuson, T. Cell and tissue requirements for the gene eed during mouse gastrulation and organogenesis. Genesis 31, 142–146 (2001).

    Article  CAS  Google Scholar 

  183. Saitou, M. & Yamaji, M. Primordial germ cells in mice. CSH Perspect. Biol. 4, a008375 (2012).

    Google Scholar 

  184. 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  Google Scholar 

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

    Article  CAS  Google Scholar 

  186. Huang, T.-C. et al. Sex-specific chromatin remodelling safeguards transcription in germ cells. Nature 600, 737–742 (2021).

    Article  CAS  Google Scholar 

  187. Kimmins, S. & Sassone-Corsi, P. Chromatin remodelling and epigenetic features of germ cells. Nature 434, 583–589 (2005).

    Article  CAS  Google Scholar 

  188. Andreu-Vieyra, C. V. et al. MLL2 is required in oocytes for bulk histone 3 lysine 4 trimethylation and transcriptional silencing. PLoS Biol. 8, e1000453 (2010).

    Article  Google Scholar 

  189. Dahl, J. A. et al. Broad histone H3K4me3 domains in mouse oocytes modulate maternal-to-zygotic transition. Nature 537, 548–552 (2016).

    Article  CAS  Google Scholar 

  190. Xu, Q. et al. SETD2 regulates the maternal epigenome, genomic imprinting and embryonic development. Nat. Genet. 51, 844–856 (2019).

    Article  CAS  Google Scholar 

  191. Shirane, K., Miura, F., Ito, T. & Lorincz, M. C. NSD1-deposited H3K36me2 directs de novo methylation in the mouse male germline and counteracts Polycomb-associated silencing. Nat. Genet. 52, 1088–1098 (2020).

    Article  CAS  Google Scholar 

  192. Maezawa, S. et al. Polycomb protein SCML2 facilitates H3K27me3 to establish bivalent domains in the male germline. Proc. Natl Acad. Sci. USA 115, 201804512 (2018).

    Article  Google Scholar 

  193. Ragazzini, R. et al. EZHIP constrains Polycomb repressive complex 2 activity in germ cells. Nat. Commun. 10, 3858 (2019).

    Article  Google Scholar 

  194. Wang, T., Gao, H., Li, W. & Liu, C. Essential role of histone replacement and modifications in male fertility. Front. Genet. 10, 962 (2019).

    Article  CAS  Google Scholar 

  195. Brykczynska, U. et al. Repressive and active histone methylation mark distinct promoters in human and mouse spermatozoa. Nat. Struct. Mol. Biol. 17, 679–687 (2010).

    Article  CAS  Google Scholar 

  196. Hammoud, S. S. et al. Distinctive chromatin in human sperm packages genes for embryo development. Nature 460, 473–478 (2009).

    Article  CAS  Google Scholar 

  197. Erkek, S. et al. Molecular determinants of nucleosome retention at CpG-rich sequences in mouse spermatozoa. Nat. Struct. Mol. Biol. 20, 868–875 (2013).

    Article  CAS  Google Scholar 

  198. Yamaguchi, K. et al. Re-evaluating the localization of sperm-retained histones revealed the modification-dependent accumulation in specific genome regions. Cell Rep. 23, 3920–3932 (2018).

    Article  CAS  Google Scholar 

  199. Carone, B. R. et al. High-resolution mapping of chromatin packaging in mouse embryonic stem cells and sperm. Dev. Cell 30, 11–22 (2014).

    Article  CAS  Google Scholar 

  200. Sin, H.-S., Kartashov, A. V., Hasegawa, K., Barski, A. & Namekawa, S. H. Poised chromatin and bivalent domains facilitate the mitosis-to-meiosis transition in the male germline. BMC Biol. 13, 53 (2015).

    Article  Google Scholar 

  201. Lesch, B. J. & Page, D. C. Poised chromatin in the mammalian germ line. Development 141, 3619–3626 (2014).

    Article  CAS  Google Scholar 

  202. van Arensbergen, J. et al. Derepression of Polycomb targets during pancreatic organogenesis allows insulin-producing beta-cells to adopt a neural gene activity program. Genome Res. 20, 722–732 (2010).

    Article  Google Scholar 

  203. Jadhav, U. et al. Acquired tissue-specific promoter bivalency is a basis for PRC2 necessity in adult cells. Cell 165, 1389–1400 (2016).

    Article  CAS  Google Scholar 

  204. McLay, D. & Clarke, H. Remodelling the paternal chromatin at fertilization in mammals. Reproduction 125, 625–633 (2003).

    Article  CAS  Google Scholar 

  205. van der Heijden, G. W. et al. Asymmetry in histone H3 variants and lysine methylation between paternal and maternal chromatin of the early mouse zygote. Mech. Dev. 122, 1008–1022 (2005).

    Article  Google Scholar 

  206. Lepikhov, K. & Walter, J. Differential dynamics of histone H3 methylation at positions K4 and K9 in the mouse zygote. BMC Dev. Biol. 4, 12 (2004).

    Article  Google Scholar 

  207. Sarmento, O. F. et al. Dynamic alterations of specific histone modifications during early murine development. J. Cell Sci. 117, 4449–4459 (2004).

    Article  CAS  Google Scholar 

  208. 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  Google Scholar 

  209. Mei, H. et al. H2AK119ub1 guides maternal inheritance and zygotic deposition of H3K27me3 in mouse embryos. Nat. Genet. 53, 539–550 (2021).

    Article  CAS  Google Scholar 

  210. Shao, G.-B. et al. Dynamic patterns of histone H3 lysine 4 methyltransferases and demethylases during mouse preimplantation development. Vitr. Cell Dev. Biol. Anim. 50, 603–613 (2014).

    Article  CAS  Google Scholar 

  211. Zhang, B. et al. Allelic reprogramming of the histone modification H3K4me3 in early mammalian development. Nature 537, 553–557 (2016).

    Article  CAS  Google Scholar 

  212. Weiner, A. et al. Co-ChIP enables genome-wide mapping of histone mark co-occurrence at single-molecule resolution. Nat. Biotechnol. 34, 953–961 (2016).

    Article  CAS  Google Scholar 

  213. Chen, Z., Djekidel, M. N. & Zhang, Y. Distinct dynamics and functions of H2AK119ub1 and H3K27me3 in mouse preimplantation embryos. Nat. Genet. 53, 551–563 (2021).

    Article  CAS  Google Scholar 

  214. Zhu, Y. et al. Genomewide decoupling of H2AK119ub1 and H3K27me3 in early mouse development. Sci. Bull. 66, 2489–2497 (2021).

    Article  CAS  Google Scholar 

  215. Albert, J. R. & Greenberg, M. V. C. The Polycomb landscape in mouse development. Nat. Genet. 53, 427–429 (2021).

    Article  Google Scholar 

  216. Murphy, P. J., Wu, S. F., James, C. R., Wike, C. L. & Cairns, B. R. Placeholder nucleosomes underlie germline-to-embryo DNA methylation reprogramming. Cell 172, 993–1006.e13 (2018).

    Article  CAS  Google Scholar 

  217. Hickey, G. J. et al. Establishment of developmental gene silencing by ordered polycomb complex recruitment in early zebrafish embryos. Elife 11, e67738 (2022).

    Article  CAS  Google Scholar 

  218. Wagner, E. J. & Carpenter, P. B. Understanding the language of Lys36 methylation at histone H3. Nat. Rev. Mol. Cell Biol. 13, 115–126 (2012).

    Article  CAS  Google Scholar 

  219. Lee, J. T. Epigenetic regulation by long noncoding RNAs. Science 338, 1435–1439 (2012).

    Article  CAS  Google Scholar 

  220. Skvortsova, K., Iovino, N. & Bogdanović, O. Functions and mechanisms of epigenetic inheritance in animals. Nat. Rev. Mol. Cell Biol. 19, 774–790 (2018).

    Article  CAS  Google Scholar 

  221. Puschendorf, M. et al. PRC1 and Suv39h specify parental asymmetry at constitutive heterochromatin in early mouse embryos. Nat. Genet. 40, 411–420 (2008).

    Article  CAS  Google Scholar 

  222. Yang, L. et al. The maternal effect genes UTX and JMJD3 play contrasting roles in Mus musculus preimplantation embryo development. Sci. Rep. 6, 26711 (2016).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  224. Snow, M. H. L. Gastrulation in the mouse: growth and regionalization of the epiblast. Development 42, 293–303 (1977).

    Article  Google Scholar 

  225. Oksuz, O. et al. Capturing the onset of PRC2-mediated repressive domain formation. Mol. Cell 70, 1149–1162.e5 (2018).

    Article  CAS  Google Scholar 

  226. Gifford, C. A. et al. Transcriptional and epigenetic dynamics during specification of human embryonic stem cells. Cell 153, 1149–1163 (2013).

    Article  CAS  Google Scholar 

  227. Xie, W. et al. Epigenomic analysis of multilineage differentiation of human embryonic stem cells. Cell 153, 1134–1148 (2013).

    Article  CAS  Google Scholar 

  228. Weinberger, L., Ayyash, M., Novershtern, N. & Hanna, J. H. Dynamic stem cell states: naive to primed pluripotency in rodents and humans. Nat. Rev. Mol. Cell Biol. 17, 155–169 (2016).

    Article  CAS  Google Scholar 

  229. Clerck, L. D. et al. Untargeted histone profiling during naive conversion uncovers conserved modification markers between mouse and human. Sci. Rep. 9, 17240 (2019).

    Article  Google Scholar 

  230. Gafni, O. et al. Derivation of novel human ground state naive pluripotent stem cells. Nature 504, 282–286 (2013).

    Article  CAS  Google Scholar 

  231. Xia, W. et al. Resetting histone modifications during human parental-to-zygotic transition. Science 365, 353–360 (2019).

    Article  CAS  Google Scholar 

  232. Wu, J. et al. Chromatin analysis in human early development reveals epigenetic transition during ZGA. Nature 557, 256–260 (2018).

    Article  CAS  Google Scholar 

  233. Guo, H. et al. The DNA methylation landscape of human early embryos. Nature 511, 606–610 (2014).

    Article  CAS  Google Scholar 

  234. Deevy, O. & Bracken, A. P. PRC2 functions in development and congenital disorders. Development 146, dev181354 (2019).

    Article  CAS  Google Scholar 

  235. Hammoud, S. S. et al. Genome-wide analysis identifies changes in histone retention and epigenetic modifications at developmental and imprinted gene loci in the sperm of infertile men. Hum. Reprod. 26, 2558–2569 (2011).

    Article  CAS  Google Scholar 

  236. Stringer, J. M. et al. Reduced PRC2 function alters male germline epigenetic programming and paternal inheritance. BMC Biol. 16, 104 (2018).

    Article  Google Scholar 

  237. Tomizawa, S. et al. Kmt2b conveys monovalent and bivalent H3K4me3 in mouse spermatogonial stem cells at germline and embryonic promoters. Development 145, dev169102 (2018).

    Article  Google Scholar 

  238. Lismer, A., Siklenka, K., Lafleur, C., Dumeaux, V. & Kimmins, S. Sperm histone H3 lysine 4 trimethylation is altered in a genetic mouse model of transgenerational epigenetic inheritance. Nucleic Acids Res. 48, 11380–11393 (2020).

    Article  CAS  Google Scholar 

  239. Halstead, M. M., Ma, X., Zhou, C., Schultz, R. M. & Ross, P. J. Chromatin remodeling in bovine embryos indicates species-specific regulation of genome activation. Nat. Commun. 11, 4654 (2020).

    Article  CAS  Google Scholar 

  240. Ikeda, H., Sone, M., Yamanaka, S. & Yamamoto, T. Structural and spatial chromatin features at developmental gene loci in human pluripotent stem cells. Nat. Commun. 8, 1616 (2017).

    Article  Google Scholar 

  241. Delachat, A. M.-F. et al. Engineered multivalent sensors to detect coexisting histone modifications in living stem cells. Cell Chem. Biol. 25, 51–56.e6 (2018).

    Article  CAS  Google Scholar 

  242. Vastenhouw, N. L. & Schier, A. F. Bivalent histone modifications in early embryogenesis. Curr. Opin. Cell Biol. 24, 374–386 (2012).

    Article  CAS  Google Scholar 

  243. Boettiger, A. N. & Levine, M. Synchronous and stochastic patterns of gene activation in the Drosophila embryo. Science 325, 471–473 (2009).

    Article  CAS  Google Scholar 

  244. Zeitlinger, J. et al. RNA polymerase stalling at developmental control genes in the Drosophila melanogaster embryo. Nat. Genet. 39, 1512–1516 (2007).

    Article  CAS  Google Scholar 

  245. Cooper, D. N. & Youssoufian, H. The CpG dinucleotide and human genetic disease. Hum. Genet. 78, 151–155 (1988).

    Article  CAS  Google Scholar 

  246. Holliday, R. & Grigg, G. W. DNA methylation and mutation. Mutat. Res. Fundam. Mol. Mech. Mutagen. 285, 61–67 (1993).

    Article  CAS  Google Scholar 

  247. Mushtaq, A. et al. Role of histone methylation in maintenance of genome integrity. Genes 12, 1000 (2021).

    Article  CAS  Google Scholar 

  248. Aguilera-Castrejon, A. et al. Ex utero mouse embryogenesis from pre-gastrulation to late organogenesis. Nature 593, 119–124 (2021).

    Article  CAS  Google Scholar 

  249. van den Brink, S. C. & van Oudenaarden, A. 3D gastruloids: a novel frontier in stem cell-based in vitro modeling of mammalian gastrulation. Trends Cell Biol. 31, 747–759 (2021).

    Article  Google Scholar 

  250. Simunovic, M. & Brivanlou, A. H. Embryoids, organoids and gastruloids: new approaches to understanding embryogenesis. Development 144, 976–985 (2017).

    Article  CAS  Google Scholar 

  251. Shahbazi, M. N., Siggia, E. D. & Zernicka-Goetz, M. Self-organization of stem cells into embryos: a window on early mammalian development. Science 364, 948–951 (2019).

    Article  CAS  Google Scholar 

  252. Cohen, A. S. A. et al. A novel mutation in EED associated with overgrowth. J. Hum. Genet. 60, 339–342 (2015).

    Article  CAS  Google Scholar 

  253. Smigiel, R. et al. Novel de novo mutation affecting two adjacent aminoacids in the EED gene in a patient with Weaver syndrome. J. Hum. Genet. 63, 517–520 (2018).

    Article  CAS  Google Scholar 

  254. Cooney, E., Bi, W., Schlesinger, A. E., Vinson, S. & Potocki, L. Novel EED mutation in patient with Weaver syndrome. Am. J. Med. Genet. A 173, 541–545 (2017).

    Article  CAS  Google Scholar 

  255. Cyrus, S. S. et al. Rare SUZ12 variants commonly cause an overgrowth phenotype. Am. J. Med. Genet. Part C. Semin. Med. Genet. 181, 532–547 (2019).

    Article  CAS  Google Scholar 

  256. Imagawa, E. et al. Novel SUZ12 mutations in Weaver-like syndrome. Clin. Genet. 94, 461–466 (2018).

    Article  CAS  Google Scholar 

  257. Imagawa, E. et al. Mutations in genes encoding polycomb repressive complex 2 subunits cause Weaver syndrome. Hum. Mutat. 38, 637–648 (2017).

    Article  CAS  Google Scholar 

  258. Lui, J. C. et al. Ezh2 mutations found in the weaver overgrowth syndrome cause a partial loss of H3K27 histone methyltransferase activity. J. Clin. Endocrinol. Metab. 103, 1470–1478 (2017).

    Article  Google Scholar 

  259. Collaboration, T. C. O. et al. Germline mutations in the oncogene EZH2 cause Weaver syndrome and increased human height. Oncotarget 2, 1127–1133 (2011).

    Article  Google Scholar 

  260. Weaver, D. D., Graham, C. B., Thomas, I. T. & Smith, D. W. A new overgrowth syndrome with accelerated skeletal maturation, unusual facies, and camptodactyly. J. Pediatrics 84, 547–552 (1974).

    Article  CAS  Google Scholar 

  261. Gibson, W. T. et al. Mutations in EZH2 cause Weaver syndrome. Am. J. Hum. Genet. 90, 110–118 (2012).

    Article  CAS  Google Scholar 

  262. Miyake, N. et al. Delineation of clinical features in Wiedemann–Steiner syndrome caused by KMT2A mutations. Clin. Genet. 89, 115–119 (2016).

    Article  CAS  Google Scholar 

  263. Mendelsohn, B. A., Pronold, M., Long, R., Smaoui, N. & Slavotinek, A. M. Advanced bone age in a girl with Wiedemann–Steiner syndrome and an exonic deletion in KMT2A (MLL). Am. J. Med. Genet. A 164, 2079–2083 (2014).

    Article  CAS  Google Scholar 

  264. Strom, S. P. et al. De novo variants in the KMT2A (MLL) gene causing atypical Wiedemann-Steiner syndrome in two unrelated individuals identified by clinical exome sequencing. BMC Med. Genet. 15, 49–49 (2014).

    Article  Google Scholar 

  265. Jones, W. D. et al. De novo mutations in MLL cause Wiedemann-Steiner syndrome. Am. J. Hum. Genet. 91, 358–364 (2012).

    Article  CAS  Google Scholar 

  266. Dafsari, H. S. et al. Novel mutations in KMT2B offer pathophysiological insights into childhood-onset progressive dystonia. J. Hum. Genet. 64, 803–813 (2019).

    Article  CAS  Google Scholar 

  267. Consortium, U. et al. Mutations in the histone methyltransferase gene KMT2B cause complex early-onset dystonia. Nat. Genet. 49, 223–237 (2017).

    Article  Google Scholar 

  268. Zech, M. et al. Haploinsufficiency of KMT2B, encoding the lysine-specific histone methyltransferase 2B, results in early-onset generalized dystonia. Am. J. Hum. Genet. 99, 1377–1387 (2016).

    Article  CAS  Google Scholar 

  269. Kleefstra, T. et al. Disruption of an EHMT1-associated chromatin-modification module causes intellectual disability. Am. J. Hum. Genet. 91, 73–82 (2012).

    Article  CAS  Google Scholar 

  270. Koemans, T. S. et al. Functional convergence of histone methyltransferases EHMT1 and KMT2C involved in intellectual disability and autism spectrum disorder. PLoS Genet. 13, e1006864 (2017).

    Article  Google Scholar 

  271. Miyake, N. et al. MLL2 and KDM6A mutations in patients with Kabuki syndrome. Am. J. Med. Genet. A 161, 2234–2243 (2013).

    Article  CAS  Google Scholar 

  272. Laarhoven, P. M. V. et al. Kabuki syndrome genes KMT2D and KDM6A: functional analyses demonstrate critical roles in craniofacial, heart and brain development. Hum. Mol. Genet. 24, 4443–4453 (2015).

    Article  Google Scholar 

  273. Luo, X. et al. Rare deleterious de novo missense variants in Rnf2/Ring2 are associated with a neurodevelopmental disorder with unique clinical features. Hum. Mol. Genet. 30, 1283–1292 (2021).

    Article  CAS  Google Scholar 

  274. Turnpenny, P. D. et al. Missense mutations of the Pro65 residue of PCGF2 cause a recognizable syndrome associated with craniofacial, neurological, cardiovascular, and skeletal features. Am. J. Hum. Genet. 103, 786–793 (2018).

    Article  CAS  Google Scholar 

  275. Faundes, V. et al. Histone lysine methylases and demethylases in the landscape of human developmental disorders. Am. J. Hum. Genet. 102, 175–187 (2018).

    Article  CAS  Google Scholar 

  276. Lebrun, N. et al. Novel KDM5B splice variants identified in patients with developmental disorders: functional consequences. Gene 679, 305–313 (2018).

    Article  CAS  Google Scholar 

  277. Martin, H. C. et al. Quantifying the contribution of recessive coding variation to developmental disorders. Science 362, 1161–1164 (2018).

    Article  CAS  Google Scholar 

  278. Miyake, N. et al. KDM6A point mutations cause Kabuki syndrome. Hum. Mutat. 34, 108–110 (2013).

    Article  CAS  Google Scholar 

  279. Stolerman, E. S. et al. Genetic variants in the KDM6B gene are associated with neurodevelopmental delays and dysmorphic features. Am. J. Med. Genet. A 179, 1276–1286 (2019).

    CAS  Google Scholar 

  280. Beck, D. B. et al. Delineation of a human Mendelian disorder of the DNA demethylation machinery: TET3 deficiency. Am. J. Hum. Genet. 106, 234–245 (2020).

    Article  CAS  Google Scholar 

  281. Heyn, P. et al. Gain-of-function DNMT3A mutations cause microcephalic dwarfism and hypermethylation of Polycomb-regulated regions. Nat. Genet. 51, 96–105 (2019).

    Article  CAS  Google Scholar 

  282. Consortium, C. O. et al. Mutations in the DNA methyltransferase gene, DNMT3A, cause an overgrowth syndrome with intellectual disability. Nat. Genet. 46, 385–388 (2014).

    Article  Google Scholar 

  283. Balci, T. B. et al. Tatton-Brown-Rahman syndrome: six individuals with novel features. Am. J. Med. Genet. A 182, 673–680 (2020).

    Article  CAS  Google Scholar 

  284. Shen, W. et al. The spectrum of DNMT3A variants in Tatton–Brown–Rahman syndrome overlaps with that in hematologic malignancies. Am. J. Med. Genet. A 173, 3022–3028 (2017).

    Article  CAS  Google Scholar 

  285. Sotos, J. F., Dodge, P. R., Muirhead, D., Crawford, J. D. & Talbot, N. B. Cerebral gigantism in childhood — a syndrome of excessively rapid growth with acromegalic features and a nonprogressive neurologic disorder. N. Engl. J. Med. 271, 109–116 (1964).

    Article  CAS  Google Scholar 

  286. Tatton-Brown, K. et al. Genotype-phenotype associations in Sotos syndrome: an analysis of 266 individuals with NSD1 aberrations. Am. J. Hum. Genet. 77, 193–204 (2005).

    Article  CAS  Google Scholar 

  287. Kurotaki, N. et al. Haploinsufficiency of NSD1 causes Sotos syndrome. Nat. Genet. 30, 365–366 (2002).

    Article  CAS  Google Scholar 

  288. Kurotaki, N. et al. Fifty microdeletions among 112 cases of Sotos syndrome: low copy repeats possibly mediate the common deletion. Hum. Mutat. 22, 378–387 (2003).

    Article  CAS  Google Scholar 

  289. Douglas, J. et al. NSD1 mutations are the major cause of Sotos syndrome and occur in some cases of Weaver syndrome but are rare in other overgrowth phenotypes. Am. J. Hum. Genet. 72, 132–143 (2003).

    Article  CAS  Google Scholar 

  290. Choufani, S. et al. NSD1 mutations generate a genome-wide DNA methylation signature. Nat. Commun. 6, 10207 (2015).

    Article  CAS  Google Scholar 

  291. Luscan, A. et al. Mutations in SETD2 cause a novel overgrowth condition. J. Med. Genet. 51, 512 (2014).

    Article  CAS  Google Scholar 

  292. Mantsoki, A., Devailly, G. & Joshi, A. CpG island erosion, polycomb occupancy and sequence motif enrichment at bivalent promoters in mammalian embryonic stem cells. Sci. Rep. 5, 16791 (2015).

    Article  CAS  Google Scholar 

  293. Smith, Z. D. et al. Epigenetic restriction of extraembryonic lineages mirrors the somatic transition to cancer. Nature 549, 543–547 (2017).

    Article  Google Scholar 

  294. Bulut-Karslioglu, A. et al. Inhibition of mTOR induces a paused pluripotent state. Nature 540, 119–123 (2016).

    Article  CAS  Google Scholar 

  295. Laugesen, A., Højfeldt, J. W. & Helin, K. Molecular mechanisms directing PRC2 recruitment and H3K27 methylation. Mol. Cell 74, 8–18 (2019).

    Article  CAS  Google Scholar 

  296. Blanco, E., González-Ramírez, M., Alcaine-Colet, A., Aranda, S. & Croce, L. D. The bivalent genome: characterization, structure, and regulation. Trends Genet. 36, 118–131 (2020).

    Article  CAS  Google Scholar 

  297. Zang, C. et al. A clustering approach for identification of enriched domains from histone modification ChIP-Seq data. Bioinformatics 25, 1952–1958 (2009).

    Article  CAS  Google Scholar 

  298. Zhang, Y. et al. Model-based analysis of ChIP-Seq (MACS). Genome Biol. 9, R137 (2008).

    Article  Google Scholar 

  299. Lun, A. T. L. & Smyth, G. K. csaw: a bioconductor package for differential binding analysis of ChIP-seq data using sliding windows. Nucleic Acids Res. 44, e45–e45 (2016).

    Article  Google Scholar 

  300. Percharde, M., Bulut-Karslioglu, A. & Ramalho-Santos, M. Hypertranscription in development, stem cells, and regeneration. Dev. Cell 40, 9–21 (2017).

    Article  CAS  Google Scholar 

  301. Chen, K. et al. The overlooked fact: fundamental need for spike-in control for virtually all genome-wide analyses. Mol. Cell Biol. 36, 662–667 (2016).

    Article  CAS  Google Scholar 

  302. Albert, M. et al. Epigenome profiling and editing of neocortical progenitor cells during development. EMBO J. 36, 2642–2658 (2017).

    Article  CAS  Google Scholar 

  303. Mohn, F. et al. Lineage-specific polycomb targets and de novo DNA methylation define restriction and potential of neuronal progenitors. Mol. Cell 30, 755–766 (2008).

    Article  CAS  Google Scholar 

  304. Minoux, M. et al. Gene bivalency at Polycomb domains regulates cranial neural crest positional identity. Science 355, eaal2913 (2017).

    Article  Google Scholar 

  305. Liu, L. et al. Chromatin modifications as determinants of muscle stem cell quiescence and chronological aging. Cell Rep. 4, 189–204 (2013).

    Article  CAS  Google Scholar 

  306. Cui, K. et al. Chromatin signatures in multipotent human hematopoietic stem cells indicate the fate of bivalent genes during differentiation. Cell Stem Cell 4, 80–93 (2009).

    Article  CAS  Google Scholar 

  307. Cockburn, K. & Rossant, J. Making the blastocyst: lessons from the mouse. J. Clin. Invest. 120, 995–1003 (2010).

    Article  CAS  Google Scholar 

  308. Dahl, J. A., Reiner, A. H., Klungland, A., Wakayama, T. & Collas, P. Histone H3 lysine 27 methylation asymmetry on developmentally-regulated promoters distinguish the first two lineages in mouse preimplantation embryos. PLoS ONE 5, e9150 (2010).

    Article  Google Scholar 

  309. Erhardt, S. et al. Consequences of the depletion of zygotic and embryonic enhancer of zeste 2 during preimplantation mouse development. Development 130, 4235–4248 (2003).

    Article  CAS  Google Scholar 

  310. Saha, B. et al. EED and KDM6B coordinate the first mammalian cell lineage commitment to ensure embryo implantation. Mol. Cell Biol. 33, 2691–2705 (2013).

    Article  CAS  Google Scholar 

  311. Schoenfelder, S. et al. Divergent wiring of repressive and active chromatin interactions between mouse embryonic and trophoblast lineages. Nat. Commun. 9, 4189 (2018).

    Article  Google Scholar 

  312. Beumer, J. & Clevers, H. Regulation and plasticity of intestinal stem cells during homeostasis and regeneration. Development 143, 3639–3649 (2016).

    Article  CAS  Google Scholar 

  313. Chiacchiera, F., Rossi, A., Jammula, S., Zanotti, M. & Pasini, D. PRC 2 preserves intestinal progenitors and restricts secretory lineage commitment. EMBO J. 35, 2301–2314 (2016).

    Article  CAS  Google Scholar 

  314. Koppens, M. A. J. et al. Deletion of polycomb repressive complex 2 from mouse intestine causes loss of stem cells. Gastroenterology 151, 684–697.e12 (2016).

    Article  CAS  Google Scholar 

  315. Bracken, A. P. et al. The Polycomb group proteins bind throughout the INK4A-ARF locus and are disassociated in senescent cells. Gene Dev. 21, 525–530 (2007).

    Article  CAS  Google Scholar 

  316. Ezhkova, E. et al. EZH1 and EZH2 cogovern histone H3K27 trimethylation and are essential for hair follicle homeostasis and wound repair. Gene Dev. 25, 485–498 (2011).

    Article  CAS  Google Scholar 

  317. Ezhkova, E. et al. Ezh2 orchestrates gene expression for the stepwise differentiation of tissue-specific stem cells. Cell 136, 1122–1135 (2009).

    Article  CAS  Google Scholar 

  318. Chen, H. et al. Polycomb protein Ezh2 regulates pancreatic β-cell Ink4a/Arf expression and regeneration in diabetes mellitus. Gene Dev. 23, 975–985 (2009).

    Article  CAS  Google Scholar 

  319. Jadhav, U. et al. Replicational dilution of H3K27me3 in mammalian cells and the role of poised promoters. Mol. Cell 78, 141–151.e5 (2020).

    Article  CAS  Google Scholar 

  320. Jadhav, U. et al. Extensive recovery of embryonic enhancer and gene memory stored in hypomethylated enhancer DNA. Mol. Cell 74, 542–554.e5 (2019).

    Article  CAS  Google Scholar 

  321. Micalizzi, D. S., Farabaugh, S. M. & Ford, H. L. Epithelial-mesenchymal transition in cancer: parallels between normal development and tumor progression. J. Mammary Gland. Biol. 15, 117–134 (2010).

    Article  Google Scholar 

  322. Lu, T. T.-H. et al. The polycomb-dependent epigenome controls β cell dysfunction, dedifferentiation, and diabetes. Cell Metab. 27, 1294–1308.e7 (2018).

    Article  CAS  Google Scholar 

  323. Alexanian, M., Padmanabhan, A., McKinsey, T. A. & Haldar, S. M. Epigenetic therapies in heart failure. J. Mol. Cell Cardiol. 130, 197–204 (2019).

    Article  CAS  Google Scholar 

  324. Gilsbach, R. et al. Distinct epigenetic programs regulate cardiac myocyte development and disease in the human heart in vivo. Nat. Commun. 9, 391 (2018).

    Article  Google Scholar 

  325. Grindheim, J. M., Nicetto, D., Donahue, G. & Zaret, K. S. Polycomb repressive complex 2 proteins EZH1 and EZH2 regulate timing of postnatal hepatocyte maturation and fibrosis by repressing genes with euchromatic promoters in mice. Gastroenterology 156, 1834–1848 (2019).

    Article  CAS  Google Scholar 

  326. Bae, W. K. et al. The methyltransferases enhancer of zeste homolog (EZH) 1 and EZH2 control hepatocyte homeostasis and regeneration. FASEB J. 29, 1653–1662 (2015).

    Article  CAS  Google Scholar 

  327. Le, H. Q. et al. An EZH2-dependent transcriptional complex promotes aberrant epithelial remodelling after injury. EMBO Rep. 22, e52785 (2021).

    Article  CAS  Google Scholar 

  328. Comet, I., Riising, E. M., Leblanc, B. & Helin, K. Maintaining cell identity: PRC2-mediated regulation of transcription and cancer. Nat. Rev. Cancer 16, 803–810 (2016).

    Article  CAS  Google Scholar 

  329. Wang, G. G. et al. Haematopoietic malignancies caused by dysregulation of a chromatin-binding PHD finger. Nature 459, 847–851 (2009).

    Article  CAS  Google Scholar 

  330. Trowbridge, J. J. et al. Haploinsufficiency of Dnmt1 impairs leukemia stem cell function through derepression of bivalent chromatin domains. Gene Dev. 26, 344–349 (2012).

    Article  CAS  Google Scholar 

  331. Béguelin, W. et al. EZH2 is required for germinal center formation and somatic EZH2 mutations promote lymphoid transformation. Cancer Cell 23, 677–692 (2013).

    Article  Google Scholar 

  332. Clevers, H. The cancer stem cell: premises, promises and challenges. Nat. Med. 17, 313–319 (2011).

    Article  CAS  Google Scholar 

  333. Zaidi, S. K. et al. Bivalent epigenetic control of oncofetal gene expression in cancer. Mol. Cell Biol. 37, e00352-17 (2017).

    Article  Google Scholar 

  334. Messier, T. L. et al. Oncofetal epigenetic bivalency in breast cancer cells: H3K4 and H3K27 tri-methylation as a biomarker for phenotypic plasticity. J. Cell Physiol. 231, 2474–2481 (2016).

    Article  CAS  Google Scholar 

  335. Chaffer, C. L. et al. Poised chromatin at the ZEB1 promoter enables breast cancer cell plasticity and enhances tumorigenicity. Cell 154, 61–74 (2013).

    Article  CAS  Google Scholar 

  336. Terranova, C. J. et al. Reprogramming of bivalent chromatin states in NRAS mutant melanoma suggests PRC2 inhibition as a therapeutic strategy. Cell Rep. 36, 109410 (2021).

    Article  CAS  Google Scholar 

  337. Lima-Fernandes, E. et al. Targeting bivalency de-represses Indian hedgehog and inhibits self-renewal of colorectal cancer-initiating cells. Nat. Commun. 10, 1436 (2019).

    Article  Google Scholar 

  338. Bernhart, S. H. et al. Changes of bivalent chromatin coincide with increased expression of developmental genes in cancer. Sci. Rep. 6, 37393 (2016).

    Article  CAS  Google Scholar 

  339. Easwaran, H. et al. A DNA hypermethylation module for the stem/progenitor cell signature of cancer. Genome Res. 22, 837–849 (2012).

    Article  CAS  Google Scholar 

  340. Ohm, J. E. et al. A stem cell–like chromatin pattern may predispose tumor suppressor genes to DNA hypermethylation and heritable silencing. Nat. Genet. 39, 237–242 (2007).

    Article  CAS  Google Scholar 

  341. Rakyan, V. K. et al. Human aging-associated DNA hypermethylation occurs preferentially at bivalent chromatin domains. Genome Res. 20, 434–439 (2010).

    Article  CAS  Google Scholar 

  342. Valencia, A. M. & Kadoch, C. Chromatin regulatory mechanisms and therapeutic opportunities in cancer. Nat. Cell Biol. 21, 152–161 (2019).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors thank B. Bernstein, R. Klose, B. Lesch, M. Sachs and reviewers for critical reading of the manuscript, as well as members of the Santos laboratory for discussions and feedback (especially B. Cho, G. Furlan and S. McClymont). The authors apologize to authors whose work they did not cite owing to space constraints or oversight on their part. The original Figs. 1 and 2 were created with BioRender. T.A.M. was supported by the UCSF Medical Scientist Training Program during manuscript preparation. J.F.-R. is supported by the Lunenfeld-Tanenbaum Research Institute Studentships at Sinai Health System. Research in the Santos laboratory is supported by the Canada 150 Research Chair in Developmental Epigenetics and project grant 420231 from the Canadian Institutes of Health Research.

Author information

Authors and Affiliations

Authors

Contributions

All authors wrote and edited the manuscript. T.A.M. and J.F.-R. created the original figures. T.A.M. performed data analysis.

Corresponding authors

Correspondence to Trisha A. Macrae or Miguel Ramalho-Santos.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Reviews Molecular Cell Biology thanks Mélanie Eckersley-Maslin and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Glossary

Epiblast

A developmental stage; the source of pluripotent cells. In the mouse, the epiblast arises approximately embryonic day 4.5 (E4.5) and persists to E7.5; in humans, this period corresponds to approximately E7–E9.

Primordial germ cells

(PGCs). Germline cells, precursors of the sex-specific gametes (oocytes and sperm).

Polycomb response elements

Sequences in fruitfly genomes that recruit Polycomb factors. They are also referred to as ‘Polycomb/Trithorax response elements’, as Trithorax proteins tend to concentrate at the same sequences.

Epigenetic priming factors

Proteins involved in establishing chromatin states conducive to transcriptional programmes for later developmental stages.

Zygotic genome activation

(ZGA). A period in early embryogenesis when transcription from the embryonic genome becomes the predominant source of transcripts in the embryo.

Pluripotency

A term that describes the ability of certain cells to generate all germ layers (mesoderm, endoderm, ectoderm and germ line) of the embryo proper. Pluripotency is perpetuated indefinitely in embryonic stem cells in vitro but occurs transiently in the embryo around implantation.

2i

Culture medium that includes inhibitors of MEK and glycogen synthase kinase 3β (GSK3β) together with leukaemia inhibitory factor (LIF), which promote a ‘ground state’ of cell pluripotency correlating with the pre-implantation mouse embryo.

Serum

Culture medium that includes fetal bovine serum and leukaemia inhibitory factor (LIF), which promote a state of pluripotency with beginning stages of lineage gene expression.

Primed

Refers to pluripotent cells (mouse or human) that display transcriptional, chromatin and developmental similarities to the post-implantation embryo.

Totipotency

Refers to the ability of cells to generate all embryonic and extra-embryonic tissues.

Inner cell mass

Cells that cluster in the inner part of the pre-implantation embryo, which give rise to the embryo proper and to fetal contributions to the extra-embryonic tissues.

Implantation

Developmental stage in which the eutherian embryo attaches to the wall of the uterus, roughly corresponding to embryonic day 4.5 in mice and a window beginning at embryonic day 6 in humans.

Naive

Refers to pluripotent cells (mouse or human) that display transcriptional, chromatin and developmental similarities to the pre-implantation embryo.

Polycomb domains

Gene-repressive domains marked by trimethylated histone H3 Lys27 and monoubiquitylated histone H2A Lys119 that tend to be heritable through cell divisions. These domains are bound by chromobox (CBX) proteins (part of canonical Polycomb repressive complex 1) that nucleate chromatin compaction.

Rights and permissions

Springer Nature or its licensor holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Macrae, T.A., Fothergill-Robinson, J. & Ramalho-Santos, M. Regulation, functions and transmission of bivalent chromatin during mammalian development. Nat Rev Mol Cell Biol 24, 6–26 (2023). https://doi.org/10.1038/s41580-022-00518-2

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41580-022-00518-2

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