Abstract
In mammals, 5-methylcytosine (5mC) and Polycomb repressive complex 2 (PRC2)-deposited histone 3 lysine 27 trimethylation (H3K27me3) are generally mutually exclusive at CpG-rich regions. As mouse embryonic stem cells exit the naive pluripotent state, there is massive gain of 5mC concomitantly with restriction of broad H3K27me3 to 5mC-free, CpG-rich regions. To formally assess how 5mC shapes the H3K27me3 landscape, we profiled the epigenome of naive and differentiated cells in the presence and absence of the DNA methylation machinery. Surprisingly, we found that 5mC accumulation is not required to restrict most H3K27me3 domains. Instead, this 5mC-independent H3K27me3 restriction is mediated by aberrant expression of the PRC2 antagonist Ezhip (encoding EZH inhibitory protein). At the subset of regions where 5mC appears to genuinely supplant H3K27me3, we identified 163 candidate genes that appeared to require 5mC deposition and/or H3K27me3 depletion for their activation in differentiated cells. Using site-directed epigenome editing to directly modulate 5mC levels, we demonstrated that 5mC deposition is sufficient to antagonize H3K27me3 deposition and confer gene activation at individual candidates. Altogether, we systematically measured the antagonistic interplay between 5mC and H3K27me3 in a system that recapitulates early embryonic dynamics. Our results suggest that H3K27me3 restraint depends on 5mC, both directly and indirectly. Our study also implies a noncanonical role of 5mC in gene activation, which may be important not only for normal development but also for cancer progression, as oncogenic cells frequently exhibit dynamic replacement of 5mC for H3K27me3 and vice versa.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$32.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
$259.00 per year
only $21.58 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout






Similar content being viewed by others
Data availability
Datasets generated in this study were uploaded to the NCBI Gene Expression Omnibus under accession number GSE242201. Additional data can be found in the Supplementary Information. Source data are provided with this paper.
Code availability
Scripts used to generate the figures presented are available under an GNU General Public License v3.0 on GitHub (https://github.com/julienrichardalbert/5mC-H3K27me3/releases/tag/v0.2).
References
Boyes, J. & Bird, A. Repression of genes by DNA methylation depends on CpG density and promoter strength: evidence for involvement of a methyl-CpG binding protein. EMBO J. 11, 327–333 (1992).
Weber, M. et al. Distribution, silencing potential and evolutionary impact of promoter DNA methylation in the human genome. Nat. Genet. 39, 457–466 (2007).
Mikkelsen, T. S. et al. Genome-wide maps of chromatin state in pluripotent and lineage-committed cells. Nature 448, 553–560 (2007).
Hawkins, R. D. et al. Distinct epigenomic landscapes of pluripotent and lineage-committed human cells. Cell Stem Cell 6, 479–491 (2010).
Reddington, J. P. et al. Redistribution of H3K27me3 upon DNA hypomethylation results in de-repression of Polycomb target genes. Genome Biol. 14, R25 (2013).
Holoch, D. et al.A cis-acting mechanism mediates transcriptional memory at Polycomb target genes in mammals. Nat. Genet. 53, 1686–1697 (2021).
Li, E., Bestor, T. H. & Jaenisch, R. Targeted mutation of the DNA methyltransferase gene results in embryonic lethality. Cell 69, 915–926 (1992).
Faust, C., Schumacher, A., Holdener, B. & Magnuson, T. The eed mutation disrupts anterior mesoderm production in mice. Development 121, 273–285 (1995).
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).
O’Carroll, D. et al. The Polycomb-group gene Ezh2 is required for early mouse development. Mol. Cell. Biol. 21, 4330–4336 (2001).
Grosswendt, S. et al. Epigenetic regulator function through mouse gastrulation. Nature 584, 102–108 (2020).
Reddington, J. P., Sproul, D. & Meehan, R. R. DNA methylation reprogramming in cancer: does it act by re-configuring the binding landscape of Polycomb repressive complexes? Bioessays 36, 134–140 (2013).
Wassef, M. & Margueron, R. The multiple facets of PRC2 alterations in cancers. J. Mol. Biol. 429, 1978–1993 (2017).
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).
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).
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).
Statham, A. L. et al. Bisulfite sequencing of chromatin immunoprecipitated DNA (BisChIP-seq) directly informs methylation status of histone-modified DNA. Genome Res. 22, 1120–1127 (2012).
Bird, A., Taggart, M., Frommer, M., Miller, O. J. & Macleod, D. A fraction of the mouse genome that is derived from islands of nonmethylated, CpG-rich DNA. Cell 40, 91–99 (1985).
Wang, L. et al. Programming and inheritance of parental DNA methylomes in mammals. Cell 15, 979–991 (2014).
Zheng, H. et al. Resetting epigenetic memory by reprogramming of histone modifications in mammals. Mol. Cell 63, 1066–1079 (2016).
Liu, X. et al. Distinct features of H3K4me3 and H3K27me3 chromatin domains in pre-implantation embryos. Nature 537, 558–562 (2016).
Cooper, S. et al. Targeting Polycomb to pericentric heterochromatin in embryonic stem cells reveals a role for H2AK119u1 in PRC2 recruitment. Cell Rep. 7, 1456–1470 (2014).
Domcke, S. et al. Competition between DNA methylation and transcription factors determines binding of NRF1. Nature 528, 575–579 (2015).
Chen, H. et al. H3K36 dimethylation shapes the epigenetic interaction landscape by directing repressive chromatin modifications in embryonic stem cells. Genome Res. 32, 825–837 (2022).
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).
Montgomery, N. D. et al. The murine Polycomb group protein Eed is required for global histone H3 lysine-27 methylation. Curr. Biol. 15, 942–947 (2005).
Smith, A. Formative pluripotency: the executive phase in a developmental continuum. Development 144, 365–373 (2017).
Kolodziejczyk, A. A. et al. Single cell RNA-sequencing of pluripotent states unlocks modular transcriptional variation. Cell Stem Cell 17, 471–485 (2015).
Hayashi, K., Ohta, H., Kurimoto, K., Aramaki, S. & Saitou, M. Reconstitution of the mouse germ cell specification pathway in culture by pluripotent stem cells. Cell 146, 519–532 (2011).
Marks, H. et al. The transcriptional and epigenomic foundations of ground state pluripotency. Cell 149, 590–604 (2012).
Habibi, E. et al. Whole-genome bisulfite sequencing of two distinct interconvertible DNA methylomes of mouse embryonic stem cells. Cell Stem Cell 13, 360–369 (2013).
Greenberg, M. V. C. et al. Transient transcription in the early embryo sets an epigenetic state that programs postnatal growth. Nat. Genet. 49, 110–118 (2017).
Greenberg, M., Teissandier, A., Walter, M., Noordermeer, D. & Bourc’his, D. Dynamic enhancer partitioning instructs activation of a growth-related gene during exit from naïve pluripotency. eLife 8, e44057 (2019).
Schulz, M. et al. DNA methylation restricts coordinated germline and neural fates in embryonic stem cell differentiation. Nat. Struct. Mol. Biol. 31, 102–114 (2024).
Kaya-Okur, H. S., Janssens, D. H., Henikoff, J. G., Ahmad, K. & Henikoff, S. Efficient low-cost chromatin profiling with CUT&Tag. Nat. Protoc. 15, 3264–3283 (2020).
Blaschke, K. et al. Vitamin C induces Tet-dependent DNA demethylation and a blastocyst-like state in ES cells. Nature 500, 222–226 (2013).
Dubois, A. et al. H3K9 tri-methylation at Nanog times differentiation commitment and enables the acquisition of primitive endoderm fate. Development 149, dev201074 (2022).
Hughes, A. L., Kelley, J. R. & Klose, R. J. Understanding the interplay between CpG island-associated gene promoters and H3K4 methylation. Biochim. Biophys. Acta Gene Regul. Mech. 1863, 194567 (2020).
Holoch, D. & Margueron, R. Mechanisms regulating PRC2 recruitment and enzymatic activity. Trends Biochem. Sci. 42, 531–542 (2017).
Schmitges, F. W. et al. Histone methylation by PRC2 is inhibited by active chromatin marks. Mol. Cell 42, 330–341 (2011).
Finogenova, K. et al. Structural basis for PRC2 decoding of active histone methylation marks H3K36me2/3. eLife 9, e61964 (2020).
Xu, Q. et al. SETD2 regulates the maternal epigenome, genomic imprinting and embryonic development. Nat. Genet. 51, 844–856 (2019).
Baubec, T. et al. Genomic profiling of DNA methyltransferases reveals a role for DNMT3B in genic methylation. Nature 520, 243–247 (2015).
Weinberg, D. N. et al. The histone mark H3K36me2 recruits DNMT3A and shapes the intergenic DNA methylation landscape. Nature 573, 281–286 (2019).
Yano, S. et al. Histone H3K36me2 and H3K36me3 form a chromatin platform essential for DNMT3A-dependent DNA methylation in mouse oocytes. Nat. Commun. 13, 4440 (2022).
Streubel, G. et al. The H3K36me2 methyltransferase Nsd1 demarcates PRC2-mediated H3K27me2 and H3K27me3 domains in embryonic stem cells. Mol. Cell 70, 371–379 (2018).
Pajtler, K. W. et al. Molecular heterogeneity and CXorf67 alterations in posterior fossa group A (PFA) ependymomas. Acta Neuropathol. 136, 211–226 (2018).
Jain, S. U. et al. PFA ependymoma-associated protein EZHIP inhibits PRC2 activity through a H3 K27M-like mechanism. Nat. Commun. 10, 2146 (2019).
Piunti, A. et al. CATACOMB: an endogenous inducible gene that antagonizes H3K27 methylation activity of Polycomb repressive complex 2 via an H3K27M-like mechanism. Sci. Adv. 5, eaax2887 (2019).
Ragazzini, R. et al. EZHIP constrains Polycomb repressive complex 2 activity in germ cells. Nat. Commun. 10, 3858 (2019).
Jessa, S. et al. K27M in canonical and noncanonical H3 variants occurs in distinct oligodendroglial cell lineages in brain midline gliomas. Nat. Genet. 54, 1865–1880 (2022).
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 (2019).
van Mierlo, G. et al. Integrative proteomic profiling reveals PRC2-dependent epigenetic crosstalk maintains ground-state pluripotency. Cell Stem Cell 24, 123–137 (2019).
Ugur, E. et al. Comprehensive chromatin proteomics resolves functional phases of pluripotency and identifies changes in regulatory components. Nucleic Acids Res. 51, 2671–2690 (2023).
Dahlet, T. et al. Genome-wide analysis in the mouse embryo reveals the importance of DNA methylation for transcription integrity. Nat. Commun. 11, 1–14 (2020).
Scelfo, A. et al. Functional landscape of PCGF proteins reveals both RING1A/B-dependent- and RING1A/B-independent-specific activities. Mol. Cell 74, 1037–1052 (2019).
Tanenbaum, M. E., Gilbert, L. A., Qi, L. S., Weissman, J. S. & Vale, R. D. A protein-tagging system for signal amplification in gene expression and fluorescence imaging. Cell 159, 635–646 (2014).
Banaszynski, L. A., Chen, L.-c, Maynard-Smith, L. A., Ooi, A. G. L. & Wandless, T. J. A rapid, reversible, and tunable method to regulate protein function in living cells using synthetic small molecules. Cell 126, 995–1004 (2006).
Senturk, S. et al. Rapid and tunable method to temporally control gene editing based on conditional Cas9 stabilization. Nat. Commun. 8, 14370 (2017).
Walter, M., Teissandier, A., Pérez-Palacios, R. & Bourc’his, D. An epigenetic switch ensures transposon repression upon dynamic loss of DNA methylation in embryonic stem cells. eLife 5, e11418 (2016).
Carlini, V., Policarpi, C. & Hackett, J. A. Epigenetic inheritance is gated by naïve pluripotency and Dppa2. EMBO J. 41, e108677 (2022).
Skene, P. J. & Henikoff, S. An efficient targeted nuclease strategy for high-resolution mapping of DNA binding sites. eLife 6, e21856 (2017).
Monteagudo-Sánchez, A., Noordermeer, D. & Greenberg, M. V. C. The impact of DNA methylation on CTCF-mediated 3D genome organization. Nat. Struct. Mol. Biol. 31, 404–412 (2024).
Morita, S. et al. Targeted DNA demethylation in vivo using dCas9–peptide repeat and scFv–TET1 catalytic domain fusions. Nat. Biotechnol. 34, 1060–1065 (2016).
Horii, T., Morita, S., Kimura, M. & Hatada, I. Efficient generation of epigenetic disease model mice by epigenome editing using the piggyBac transposon system. Epigenetics Chromatin 15, 40 (2022).
Zukher, I., Dujardin, G., Sousa-Luís, R. & Proudfoot, N. J. Elongation roadblocks mediated by dCas9 across human genes modulate transcription and nascent RNA processing. Nat. Struct. Mol. Biol. 30, 1536–1548 (2023).
Sapozhnikov, D. M. & Szyf, M. Unraveling the functional role of DNA demethylation at specific promoters by targeted steric blockage of DNA methyltransferase with CRISPR/dCas9. Nat. Commun. 12, 5711 (2021).
Seisenberger, S. et al. The dynamics of genome-wide DNA methylation reprogramming in mouse primordial germ cells. Mol. Cell 48, 849–862 (2012).
Hajkova, P. et al. Chromatin dynamics during epigenetic reprogramming in the mouse germ line. Nature 452, 877–881 (2008).
Shen, X. et al. EZH1 mediates methylation on histone H3 lysine 27 and complements EZH2 in maintaining stem cell identity and executing pluripotency. Mol. Cell 32, 491–502 (2008).
Shan, Y. et al. PRC2 specifies ectoderm lineages and maintains pluripotency in primed but not naïve ESCs. Nat. Commun. 8, 672 (2017).
Lienert, F. et al. Identification of genetic elements that autonomously determine DNA methylation states. Nat. Genet. 43, 1091–1097 (2011).
Krebs, A. R., Dessus-Babus, S., Burger, L. & Schübeler, D. High-throughput engineering of a mammalian genome reveals building principles of methylation states at CG rich regions. eLife 3, e04094 (2014).
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).
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).
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).
Guo, X. et al. Structural insight into autoinhibition and histone H3-induced activation of DNMT3A. Nature 517, 640–644 (2015).
Manzo, M. et al. Isoform-specific localization of DNMT3A regulates DNA methylation fidelity at bivalent CpG islands. EMBO J. 36, 3421–3434 (2017).
Gu, T. et al. DNMT3A and TET1 cooperate to regulate promoter epigenetic landscapes in mouse embryonic stem cells. Genome Biol. 19, 88 (2018).
Li, H. et al. Polycomb-like proteins link the PRC2 complex to CpG islands. Nature 549, 287–291 (2017).
Perino, M. et al. MTF2 recruits Polycomb repressive complex 2 by helical-shape-selective DNA binding. Nat. Genet. 50, 1002–1010 (2018).
Mochizuki, K. et al. Repression of germline genes by PRC1.6 and SETDB1 in the early embryo precedes DNA methylation-mediated silencing. Nat. Commun. 12, 7020 (2021).
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).
Farcas, A. M. et al. KDM2B links the Polycomb repressive complex 1 (PRC1) to recognition of CpG islands. eLife 1, 6233–6226 (2012).
Yin, Y. et al. Impact of cytosine methylation on DNA binding specificities of human transcription factors. Science 356, eaaj2239 (2017).
Dahlet, T. et al. E2F6 initiates stable epigenetic silencing of germline genes during embryonic development. Nat. Commun. 12, 3582 (2021).
Al Adhami, H. et al. Systematic identification of factors involved in the silencing of germline genes in mouse embryonic stem cells. Nucleic Acids Res. 51, 3130–3149 (2023).
Policarpi, C., Munafò, M., Tsagkris, S., Carlini, V. & Hackett, J. A.Systematic epigenome editing captures the context-dependent instructive function of chromatin modifications. Nat. Genet. 56, 1168–1180 (2024).
von Meyenn, F. et al. Impairment of DNA methylation maintenance is the main cause of global demethylation in naive embryonic stem cells. Mol. Cell 62, 848–861 (2016).
Rowe, H. M. & Trono, D. Dynamic control of endogenous retroviruses during development. Virology 411, 273–287 (2011).
Kojima, S. et al. Epigenome editing reveals core DNA methylation for imprinting control in the Dlk1–Dio3 imprinted domain. Nucleic Acids Res. 50, 5080–5094 (2022).
Shima, Y. et al. Differential expression of the seven-pass transmembrane cadherin genes Celsr1–3 and distribution of the Celsr2 protein during mouse development. Dev. Dyn. 223, 321–332 (2002).
Qu, Y. et al. Atypical cadherins Celsr1–3 differentially regulate migration of facial branchiomotor neurons in mice. J. Neurosci. 30, 9392–9401 (2010).
Li, C. et al. Planar cell polarity protein Celsr2 maintains structural and functional integrity of adult cortical synapses. Prog. Neurobiol. 219, 102352 (2022).
GTEx, C. Human genomics. The Genotype-Tissue Expression (GTEx) pilot analysis: multitissue gene regulation in humans. Science 348, 648–660 (2015).
Lindsay, S. J. et al. HDBR expression: a unique resource for global and individual gene expression studies during early human brain development. Front. Neuroanat. 10, 86 (2016).
Tissir, F. et al. Lack of cadherins Celsr2 and Celsr3 impairs ependymal ciliogenesis, leading to fatal hydrocephalus. Nat. Neurosci. 13, 700–707 (2010).
Wen, Q. et al. Inactivating Celsr2 promotes motor axon fasciculation and regeneration in mouse and human. Brain 145, 670–683 (2022).
He, Y. et al. Spatiotemporal DNA methylome dynamics of the developing mouse fetus. Nature 583, 752–759 (2020).
Spicer, Z. et al. Stomachs of mice lacking the gastric H,K-ATPase α-subunit have achlorhydria, abnormal parietal cells, and ciliated metaplasia. J. Biol. Chem. 275, 21555–21565 (2000).
Horii, T. et al. Successful generation of epigenetic disease model mice by targeted demethylation of the epigenome. Genome Biol. 21, 77 (2020).
Reddington, J. P., Pennings, S. & Meehan, R. R. Non-canonical functions of the DNA methylome in gene regulation. Biochem. J. 451, 13–23 (2013).
Guo, H. et al. DNA hypomethylation silences anti-tumor immune genes in early prostate cancer and CTCs. Cell 186, 2765–2782 (2023).
Huang, Y. H. et al. DNA epigenome editing using CRISPR–Cas SunTag-directed DNMT3A. Genome Biol. 18, 176 (2017).
Chen, B. et al. Dynamic imaging of genomic loci in living human cells by an optimized CRISPR/Cas system. Cell 155, 1479–1491 (2013).
Vidigal, J. A. & Ventura, A. Rapid and efficient one-step generation of paired gRNA CRISPR–Cas9 libraries. Nat. Commun. 6, 8083 (2015).
Leitão, E. et al. Locus-specific DNA methylation analysis by targeted deep bisulfite sequencing. Methods Mol. Biol. 1767, 351–366 (2018).
Kaya-Okur, H. S. & Henikoff, S. Bench top CUT&Tag v2. protocols.io https://doi.org/10.17504/protocols.io.z6hf9b6 (2019).
Buenrostro, J. D. et al. Single-cell chromatin accessibility reveals principles of regulatory variation. Nature 523, 486–490 (2015).
Andrews, S. FastQC: a quality control tool for high throughput sequence data. Available online at http://www.bioinformatics.babraham.ac.uk/projects/fastqc (2010).
Bushnell, B. BBMap: a fast, accurate, splice-aware aligner. Joint Genome Institute (2014).
Bolger, A. M., Lohse, M. & Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014).
Krueger, F. & Andrews, S. R. Bismark: a flexible aligner and methylation caller for Bisulfite-Seq applications. Bioinformatics 27, 1571–1572 (2011).
Neph, S. et al. BEDOPS: high-performance genomic feature operations. Bioinformatics 28, 1919–1920 (2012).
Younesy, H., Möller, T., Lorincz, M. C., Karimi, M. M. & Jones, S. J. M. VisRseq: R-based visual framework for analysis of sequencing data. BMC Bioinformatics 16, S2 (2015).
Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).
Pertea, M. et al. StringTie enables improved reconstruction of a transcriptome from RNA-seq reads. Nat. Biotechnol. 33, 290–295 (2015).
Behdenna, A. et al. pyComBat, a Python tool for batch effects correction in high-throughput molecular data using empirical Bayes methods. BMC Bioinformatics 24, 459 (2023).
Pedregosa, F. et al. Scikit-learn: machine learning in Python. J. Mach. Learn. Res. 12, 2825–2830 (2011).
McKinney, W. Data structures for statistical computing in python. In Proceedings of the 9th Python in Science Conference (eds van der Walt, S. & Millman, J.) (SciPy, 2012).
Reback, J. et al. pandas-dev/pandas: Pandas 1.0.5. Zenodo https://doi.org/10.5281/zenodo.3509134 (2021).
Harris, C. R. et al. Array programming with NumPy. Nature 585, 357–362 (2020).
Hunter, J. D.Matplotlib: a 2D graphics environment. Comput. Sci. Eng. 9, 90–95 (2007).
Ari, N. & Ustazhanov, M. Matplotlib in Python. In Proceedings of the 11th International Conference on Electronics, Computer and Computation (eds Mahamat, M., Adeshina, S. A. & Arreytambe, T.) (IEEE, 2014).
Waskom, M. seaborn: statistical data visualization. J. Open Source Softw. 6, 3021 (2021).
Ramírez, F. et al. deepTools2: a next generation web server for deep-sequencing data analysis. Nucleic Acids Res. 44, W160–W165 (2016).
Ritchie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).
Langmead, B., Wilks, C., Antonescu, V. & Charles, R. Scaling read aligners to hundreds of threads on general-purpose processors. Bioinformatics 35, 421–432 (2019).
Meers, M. P., Tenenbaum, D. & Henikoff, S. Peak calling by sparse enrichment analysis for CUT&RUN chromatin profiling. Epigenetics Chromatin 12, 42 (2019).
Quinlan, A. R. & Hall, I. M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842 (2010).
Bailey, T. L. et al. MEME SUITE: tools for motif discovery and searching. Nucleic Acids Res. 37, W202–W208 (2009).
Thomas-Chollier, M. et al. RSAT peak-motifs: motif analysis in full-size ChIP-seq datasets. Nucleic Acids Res. 40, e31 (2012).
Thomas-Chollier, M. et al. A complete workflow for the analysis of full-size ChIP-seq (and similar) data sets using peak-motifs. Nat. Protoc. 7, 1551–1568 (2012).
Kent, W. J., Zweig, A. S., Barber, G., Hinrichs, A. S. & Karolchik, D. BigWig and BigBed: enabling browsing of large distributed datasets. Bioinformatics 26, 2204–2207 (2010).
Kent, W. J. et al. The human genome browser at UCSC. Genome Res. 12, 996–1006 (2002).
Raney, B. J. et al. Track data hubs enable visualization of user-defined genome-wide annotations on the UCSC Genome Browser. Bioinformatics 30, 1003–1005 (2014).
Acknowledgements
We thank T. Horii and I. Hatada (Gunma University, Japan) for providing the constitutive dCas9–SunTag/TET1CD epigenome-editing construct, B. Guichard (Institut Jacques Monod (IJM), France) and M. Möckel (IMB Mainz, Germany) for purifying the pA-Tn5 protein, C. McQuillen and H. Fanlo Ucar for technical assistance, D. Bourc’his for her insights and support, J. Barau, M. Leeb, A. Bogutz, S. Janssen and M. Pitasi for useful discussions, M. Lorincz and D. Holoch for critical reading, J. Marchand for installing and maintaining computational resources, N. Valentin and the IJM FACS facility. Work in the M.V.C.G. group is supported by the European Research Council (ERC-StG-2019 DyNAmecs) and by a Laboratoire d’Excellence Who Am I? (11-LABX-0071) emerging teams grant. This research was also funded by the Fondation pour la Recherche Médicale (FRM) postdoc France fellowship (SPF202110014238) to J.R.A. and the FRM (SPF202004011789) and Fondation ARC (ARCPDF12020070002563) postdoctoral fellowships to A.M.-S. The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.
Author information
Authors and Affiliations
Contributions
J.R.A., conceptualization, methodology, formal analysis, data curation, visualization, writing—original draft and writing—review and editing; T.U., investigation, visualization, writing—original draft and writing—review and editing; A.M.-S., investigation; A.L.B., investigation; A.S., investigation; A.D., investigation; M. Scarpa, investigation; M. Schulz, investigation; M.V.C.G., conceptualization, methodology, investigation, writing—review and editing, supervision, project administration and funding acquisition.
Corresponding author
Ethics declarations
Competing interests
The authors declare no competing interests.
Peer review
Peer review information
Nature Structural & Molecular Biology thanks Gabriella Ficz and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available. Primary Handling Editor: Dimitris Typas, in collaboration with the Nature Structural & Molecular Biology team.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Extended data
Extended Data Fig. 1 In vitro and in vivo H3K27me3 dynamics during the exit from naïve pluripotency.
a. 2D scatterplot showing H3K27me3 enrichment over genome-wide 10kb bins during in vitro differentiation of WT ESCs to EpiLCs (D7). Datapoints are coloured by 5mC levels in WT EpiLCs. 10kb bins with ≥10 CpGs covered by ≥5 reads are shown (n=252,559), and the density of datapoints is included. b. 2D scatterplot showing H3K27me3 enrichment over genome-wide 10kb bins during in vivo differentiation of E3.5 inner cell mass (ICM) cells of the blastocyst into the E7.5 epiblast. Datapoints are coloured by 5mC levels in E7.5 epiblasts. 10kb bins with ≥10 CpGs covered by ≥5 reads are shown (n=254,190), and the density of datapoints is included. c. Scatterplot showing H3K27me3 levels over CpG islands (CGIs) in WT ESCs (D0) and EpiLCs (D7). Datapoints are coloured by average 5mC levels in WT EpiLCs. CGIs with ≥10 CpGs covered by ≥5 reads are shown (n=15,714/16,009). CGI promoters of specific genes are indicated, contours are drawn at iso-proportions of the density of datapoints. d. UCSC Genome Browser screenshots of Polycomb-group target genes. WGBS and H3K27me3 CUT&Tag data derived from WT ESCs and EpiLCs (day 7) is shown. Refseq genes, CpG density, CGIs and individual CpGs are included. H3K27me3 peak calls are shown as rectangles. Promoter regions are highlighted in yellow. Coordinates: Hoxd12 chr2:74,672,861-74,678,432, Pax7 chr4:139,712,945-139,857,645, Pax2 chr19:44,712,108-44,849,206. e. Heatmap showing expression levels (RPKM) of Polycomb-group (PcG) target genes Hoxd12, Pax7 and Pax2 in vitro and in vivo (inner cell mass cells of embryonic day (E) 3.5 and E4.0 blastocysts, and E5.5 and E6.5 epiblasts. For each in vitro and in vivo dataset, the scale bar for each gene is set independently, either to the maximum expression level or 1, whichever is higher. f. UCSC Genome Browser screenshots of EpiLC-specific genes. Data as in d. Coordinates: Zhx2 chr15:57,676,294-57,758,534, Gpc6 chr14:116,904,924-116,947,162. g. Heatmap showing expression levels (RPKM) of EpiLC-specific genes Zhx2 and Gpc6, as in e. h. UCSC Genome Browser screenshots of EpiLC-silenced genes. Data as in d. in vivo data derived from the inner cell mass (ICM) of E3.5 blastocysts and E7.5 epiblasts is shown. Coordinates: Klf4 chr4:55,521,819-55,537,790, Prdm14 chr1:13,104,889-13,135,730, Tbx3 chr5:119,635,577-119,698,095. i. Heatmap showing expression levels (RPKM) of EpiLC-silenced genes Klf4, Prdm14 and Tbx3, as in e.
Extended Data Fig. 2 The transcriptomic landscape of TKO EpiLCs and genomic composition of SWRs.
a. Heatmaps showing expression levels (RPKM) of naïve (Klf4, Tbx3, Zfp42/Rex1, Fgf4 and Prdm14) and formative/primed (Pou3f1, Sox3, Etv5, Cdh2 and Fgf5) pluripotency markers in vitro and in vivo (inner cell mass cells of E3.5 and E4.0 blastocysts, and E5.5 and E6.5 epiblasts). For each in vitro and in vivo dataset, the scale bar for each gene is set independently to the maximum expression of that gene in WT and TKO samples. b. Principal Component Analysis (PCA) plot showing transcriptomic similarities between naïve ESCs with the inner cell mass cells of the blastocyst, and between EpiLCs with epiblasts. Both groups are arbitrarily circled by a dotted line. D0: ESCs; D2, D4, D7: EpiLCs; Oo.: oocyte; 1C: zygote; E2C: early 2-cell embryo; 2C: 2-cell embryo; L2C: late 2-cell embryo; 4C: 4-cell embryo; 8C: 8-cell embryo; Mo.: morula-stage embryo; ICM: inner cell mass cells of the blastocyst; TE: trophectoderm cells of the blastocyst; E5.5-, 6.5-, 7.5-: embryonic days; Epi: epiblast; ExE: extraembryonic ectoderm; Endo.: endoderm; Meso.: mesoderm; Ecto.: ectoderm; Prim. Str.: primitive streak. c. Pie charts showing the genomic distribution of H3K27me3 domain classes SWitch Regions (SWRs), ESC-Specific Regions (ESRs) and COnstitutive Regions (CORs). A fourth pie chart showing the total area of each genomic compartment is included. d. Motif analysis showing enriched motifs in the putative PRC binding sites within SWRs using ESRs as control sequences and two motif analysis algorithms.
Extended Data Fig. 3 H3K36me3 deposition is not strongly impacted in absence of DNA methylation.
a. 2D scatterplot showing H3K27me3 and H3K36me3 enrichment over genome-wide 10kb bins in WT ESCs. Data points are coloured by 5mC levels in WT ESCs. 10kb bins with ≥10 CpGs covered by ≥5 reads are shown (n=252,812), and the density of datapoints is included. b. 2D scatterplot showing H3K27me3 and H3K36me3 enrichment over genome-wide 10kb bins in WT EpiLCs. Data points are coloured by 5mC levels in WT EpiLCs. 10kb bins with ≥10 CpGs covered by ≥5 reads are shown (n=252,559), and the density of datapoints is included. c. 2D scatterplot showing H3K36me3 enrichment over genome-wide 10kb bins in WT and TKO ESCs. Data points are coloured by 5mC levels in WT ESCs as in a, and density of datapoints is included. d. 2D scatterplot showing H3K36me3 enrichment over genome-wide 10kb bins in WT and TKO EpiLCs. Data points are coloured by 5mC levels in WT EpiLCs as in b, and density of data points is included. e. 2D scatterplot showing H3K27me3 enrichment over genome-wide 10kb bins in WT and TKO EpiLCs, as in Fig. 1c. Data points are coloured by the relative change in H3K36me3 enrichment in TKO versus WT EpiLCs, and density of datapoints is included. f. Violin plot showing the distribution of H3K36me3 level change in TKO EpiLCs compared to WT EpiLCs. 10kb bins are categorized based on H3K27me3 dynamics in WT and TKO EpiLCs; No H3K27me3: insufficient H3K27me3 levels (CPM<1) in either WT or TKO EpiLCs, not DE: H3K27me3 levels do not significantly change between WT and TKO EpiLCs, Down TKO: relative loss of H3K27me3 in TKO EpiLCs, Up TKO: relative enrichment of H3K27me3 in TKO EpiLCs compared to WT EpiLCs.
Extended Data Fig. 4 TKO+Ezhip KO (QKO) ESCs differentiate to EpiLCs.
a. Heatmap showing Ezhip expression levels (RPKM) in WT and Suz12 KO and Eed KO ESCs. The value of the highest expression level in all four conditions is indicated. b. Representative brightfield images of EpiLCs. Scale bar = 1mm. Pictures were taken of D5 EpiLCs. c. Bar plots showing the expression level of naïve (Rex1/Zfp42, Nanog), general (Oct4), formative, (Otx2, Fgf5) and primed (Brachyury/T) pluripotency markers in D7 EpiLCs. Bars represent the mean, error bars the standard deviation, and individual data points are shown for each independent experiment (n=4). No statistical tests were performed. d. Heatmap showing relative enrichment of PRC2 subunit proteins on the chromatin of WT naïve and primed epiblast stem cells (EpiSCs). Data from Ugur et al.54. e. Heatmap depicting PRC2 subunit gene expression levels (RPKM) during EpiLC differentiation in WT and TKO cells.
Extended Data Fig. 5 Examples of SWR regions with maintenance of gene repression or activation.
a. Schema showing a hypothetical H3K27me3-to-5mC SWitch Region (SWR) that overlaps a genic promoter. In this case, dense methylation of the promoter region results in the maintained repression of expression. b. UCSC Genome Browser screenshot of an example SWR overlapping the Piwil1 germline gene promoter. The Refseq gene annotation, CpG density, individual CpGs, CpG islands and scale bar are included. Promoter region is highlighted in yellow. Note the anticorrelated enrichment of 5mC and H3K27me3 in WT cells, and the aberrant maintenance of H3K27me3 in TKO EpiLCs. Coordinates: chr5:128,733,977-128,756,805. c. Heatmap of Piwil1 expression levels (RPKM) in vitro and in vivo differentiation. ESCs: D0; EpiLCs: D2, D4, D7; E3.5-, E4.5-: embryonic days 3.5 and 4.5 inner cell mass cells; E5.5-, E6.5: embryonic days 5.5 and 6.5 epiblasts. d. UCSC Genome Browser screenshots of SWRs that overlap genes activated in WT EpiLCs (see schema, Fig. 3b). Coordinates: Pdyn chr2:129,673,296-129,713,102, Krt80 chr15:101,345,735-101,394,892, Pga5 chr19:10,666,677-10,680,350.
Extended Data Fig. 6 Implementation of an epigenome editing system for site-specific 5mC deposition at candidate SWRs.
a. Schema showing inducible constructs integrated into the Rosa26 locus in ESCs. One allele contains a construct composed of the TetOn array promoter for Doxycycline-dependent activation; DD: FKBP12-derived destabilizing domain, stabilized by the addition of the Shield-1 ligand; dCas9: catalytically dead Cas9, where two point mutations abrogate nucleolytic activity; 10xGCN4: an array of ten GCN4 epitopes (dCas9-SunTag). The homologous allele encodes a TetOn promoter, a destabilizing DD domain, a single-chain antibody (scFv) that recognizes the GCN4 epitope, green fluorescent protein (GFP) and the catalytic domain of DNMT3A (3ACD). Control cells contain an identical construct that encodes a catalytically inactive form of DNMT3A (d3ACD). A separate plasmid containing single guide RNAs (sgRNAs) was randomly integrated by piggyBac transposition. Resistance genes driven by an independent promoter are indicated. b. Western blot validating the inducible dCas9-SunTag/scFv-GFP-3ACD cell line. The two proteins (dCas9-SunTag and scFv-GFP-3ACD) encoded by the two inducible constructs are not detected in the absence of Dox, detected at intermediate levels after Dox induction, and robust expression after the addition of both Dox and Shield-1. c. UCSC Genome Browser screenshots of candidate SWR regions adjacent to the genes: Zdbf2, Celsr2, Atp4a, Arsi, Pga5, Pdyn, and Krt80. The target site for 5mC editing using 3ACD and TET1 is shown; gRNAs are indicated in red (forward strand) or blue (reverse strand). H3K27me3 CUT&Tag in TKO EpiLCs is included. The location of H3K27me3 CUT&RUN quantitative PCR and BS-pyro amplicons are also shown. CpG density, CpG islands (CGIs) and Refseq gene annotations have been added for reference. Coordinates: Zdbf2 chr1:63,246,946-63,275,173; Celsr2 chr3:108,401,129-108,424,638; Atp4a: chr7:30,703,888-30,733,844; Arsi chr18:60,899,912-60,924,326; Pga5 chr19:10,660,312-10,683,383; Pdyn chr2:129,683,316-129,707,017; Krt80 chr15:101,366,622-101,390,127.
Extended Data Fig. 7 Targeted 5mC deposition and concomitant H3K27me3 loss at the Zdbf2 SWR and site-directed 5mC editing at other candidate SWRs.
a. BS-pyro of the Zdbf2 SWR showing 5mC deposition in induced cells (+Dox +Shield-1, green) expressing dCas9 and the 3ACD construct compared to uninduced control cells (-Dox -Shield-1, gray). Cells were collected 7 days after the addition of Dox and Shield-1. 9 replicates are shown, and each replicate represents the average 5mC level across 4 CpGs. b. CUT&RUN-qPCR showing lower average levels of H3K27me3 in induced cells (green) over the Zdbf2 SWR. Data represent mean of n=8 and n=9 replicates for induced and uninduced samples, respectively. c. CUT&RUN-qPCR showing similar levels of H3K27me3 enrichment at the positive control Pax5 promoter locus relative to a negative (background) control, β-actin. Data represents mean of replicates, with each replicate indicated as a dot (d3ACD + Dox n=5, 3ACD +Dox n=8, 3ACD -Dox n=9, 3ACD +Dox n=8). d. BS-pyro results at the Zdbf2, Celsr2, Pdyn, Pga5 and Krt80 candidate SWRs. Each data point represents the average 5mC level over the pyrosequencing amplicon for each replicate, and bars represent the mean of independent experiments (n=2, except Zdbf2 g1 n=10). a, c-d Data are represented as mean ± standard error of independent experiments. Replicates and their mean are shown. P-values were calculated by two-tailed unpaired t-test assuming equal variance.
Extended Data Fig. 8 Targeted 5mC deposition in TKO + Dnmt1 ESCs at the Zdbf2 and Celsr2 SWRs.
a. Western Blot showing DNMT1 protein levels in TKO + Dnmt1 ESCs. Clone B5 was selected for subsequent experiments. b. LUminometric Methylation Assay (LUMA) showing global 5mC levels in ESCs grown in serum conditions. Data points represent technical replicates (n=2). N.D.: not detected. c. Schema of constitutive epigenome editing constructs randomly integrated into ESCs grown in serum conditions. d. BS-pyro of the Zdbf2 and Celsr2 SWRs. Data points represent the average 5mC level over the amplicon for each replicate. e. Bar plot showing expression levels of Zdbf2 and Celsr2 by RT-qPCR. Levels were normalized to the average Ct of two housekeeping genes (Rplp0 and Rrm2) (ΔCt method). d-e Data are represented as mean ± standard error of independent experiments, Replicates and their mean are shown (n=3, except for d3ACD Zdbf2 gRNA n=2). P-values were calculated by two-tailed unpaired t-test assuming equal variance.
Extended Data Fig. 9 Targeted 5mC demethylation of candidate SWRs using the constitutively expressed dCas9-SunTag/scFv-GFP-TET system.
a. BS-pyro results from EpiLCs (day 4) at candidate SWRs Pdyn (gRNA 1-2), Pga5 (gRNA 1-2), Krt80 (gRNA 3-4) and Arsi (gRNA 1-2). All cells constitutively express the dCas9-SunTag/scFv-GFP-TET1CD construct. Cells expressing target gRNAs are in purple, and control cells expressing scrambled gRNAs are in grey. TKO EpiLCs were used as a negative control (orange, n=1, except for Arsi, n=2). Each data point represents the average 5mC level over the pyrosequencing amplicon (3-9 CpGs: 5 for Pdyn, 5 for Pga5, 3 for Krt80, and 9 for Arsi) for each independent experiment (n=4, except TET1 Arsi gRNA, n=3). TKO samples were not considered for statistical analysis. b. CUT&RUN-qPCR results showing similar enrichment of H3K27me3 at the positive control Pax5 locus across cell lines relative to β-actin (negative control locus). Data represent mean of n=6 independent experiments. No significant differences between target and scrambled gRNA samples were detected. c. CUT&RUN qPCR showing H3K27me3 levels normalized to the positive control Pax5 CGI promoter (ΔCt method) for the Zdbf2 (left) and Celsr2 (right) SWRs. Data represent n=6 independent experiments for each line. a-c. Data are represented as mean ± standard error of independent experiments, replicates and their mean are shown. P-values were calculated by two-tailed unpaired t-test assuming equal variance. d. 2D scatterplot showing H3K27me3 levels over genome-wide 10kb bins (n=273,121) in epigenome-edited cells targeting the Zdbf2 and Celsr2 SWRs. The 10kb bins overlapping SWRs are indicated. e. 2D scatterplot showing H3K27me3 levels over genome-wide 10kb bins (n=273,121) in epigenome-edited cells targeting the Arsi and Atp4a SWRs. The 10kb bin overlapping the Atp4a SWR is indicated.
Extended Data Fig. 10 dTET1 epigenome editing to intermediate effects in EpiLCs.
a. BS-pyro of the Zdbf2 and Celsr2 SWRs treated with: TET1 with no target gRNA (scrambled), a catalytically inactive TET1 (dTET1) and target gRNA, TET1 and target gRNA, and in TKO EpiLCs. Each data point represents the average 5mC level over the pyrosequencing amplicon (4-5 CpGs) for each independent experiment (n=3, except TKO n=2). b. MA plot showing H3K27me3 enrichment level changes in EpiLCs targeting TET1 (left) or dTET1 (right) to the Zdbf2 SWR compared to the Celsr2 SWR. c. UCSC Genome browser screenshots of the Zdbf2 and Celsr2 SWR regions showing H3K27me3 level enrichment by CUT&RUN in WT ESCs and epigenome-edited EpiLCs. d. Bar plot showing expression levels of Zdbf2 and Celsr2 by RT-qPCR. Levels were normalized to the average Ct of two housekeeping genes (Rplp0 and Rrm2) (ΔCt method). n=3 independent experiments were conducted (except Zdbf2 TKO, n=2). a,d: Data are represented as mean ± standard error of independent experiments, Replicates and their mean are shown. P-values were calculated by two-tailed unpaired t-test assuming equal variance. TKO EpiLCs datasets were not included in the statistical tests.
Supplementary information
Supplementary Information
Supplementary Figs. 1–9.
Supplementary Tables
Supplementary Table 1: Datasets generated and mined. Supplementary Table 2: Oligonucleotide sequences. Supplementary Table 3: Statistical source data underlying supplementary figures.
Source data
Source Data Figs. 2d and Extended Data Figs. 6b and 8a
Unprocessed western blots.
Source Data Figs. 4b–d and 5b–d and Extended Data Figs. 4c, 7a–d, 8b,d,e, 9a–c and 10a,d.
Statistical source data.
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) 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.
About this article
Cite this article
Richard Albert, J., Urli, T., Monteagudo-Sánchez, A. et al. DNA methylation shapes the Polycomb landscape during the exit from naive pluripotency. Nat Struct Mol Biol 32, 346–357 (2025). https://doi.org/10.1038/s41594-024-01405-4
Received:
Accepted:
Published:
Issue date:
DOI: https://doi.org/10.1038/s41594-024-01405-4