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.

  • Article
  • Published:

Distinct dynamics and functions of H2AK119ub1 and H3K27me3 in mouse preimplantation embryos

An Author Correction to this article was published on 22 April 2021

This article has been updated

Abstract

Polycomb repressive complexes 1 and 2 (PRC1/2) maintain transcriptional silencing of developmental genes largely by catalyzing the formation of mono-ubiquitinated histone H2A at lysine 119 (H2AK119ub1) and trimethylated histone H3 at lysine 27 (H3K27me3), respectively. How Polycomb domains are reprogrammed during mammalian preimplantation development remains largely unclear. Here we show that, although H2AK119ub1 and H3K27me3 are highly colocalized in gametes, they undergo differential reprogramming dynamics following fertilization. H3K27me3 maintains thousands of maternally biased domains until the blastocyst stage, whereas maternally biased H2AK119ub1 distribution in zygotes is largely equalized at the two-cell stage. Notably, while maternal PRC2 depletion has a limited effect on global H2AK119ub1 in early embryos, it disrupts allelic H2AK119ub1 at H3K27me3 imprinting loci including Xist. By contrast, acute H2AK119ub1 depletion in zygotes does not affect H3K27me3 imprinting maintenance, at least by the four-cell stage. Importantly, loss of H2AK119ub1, but not H3K27me3, causes premature activation of developmental genes during zygotic genome activation (ZGA) and subsequent embryonic arrest. Thus, our study reveals distinct dynamics and functions of H3K27me3 and H2AK119ub1 in mouse preimplantation embryos.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: H2AK119ub1 (H2Aub) in mouse oocytes and early embryos is highly dynamic.
Fig. 2: Loss of parental H2Aub asymmetric distribution during preimplantation development.
Fig. 3: Distinct dynamics of H2Aub and H3K27me3 during preimplantation development.
Fig. 4: Loss of maternal Eed has a limited effect on global H2Aub except for maternal H3K27me3-dependent imprinted genes.
Fig. 5: Acute depletion of H2Aub in zygotes has minimal immediate effects on H3K27me3.
Fig. 6: Acute depletion of H2Aub causes premature activation of developmental genes during ZGA and leads to early embryonic arrest.
Fig. 7: Models of the allelic dynamics of H2Aub and H3K27me3.

Similar content being viewed by others

Data availability

All sequencing data were deposited in the Gene Expression Omnibus under accession number GSE153531.

Code availability

The code for this study is available at https://github.com/YiZhang-lab/H2Aub_K27me3_preimplantation_dynamics.

Change history

References

  1. de Napoles, M. et al. Polycomb group proteins Ring1A/B link ubiquitylation of histone H2A to heritable gene silencing and X inactivation. Dev. Cell 7, 663–676 (2004).

    Article  PubMed  Google Scholar 

  2. Wang, H. et al. Role of histone H2A ubiquitination in Polycomb silencing. Nature 431, 873–878 (2004).

    Article  CAS  PubMed  Google Scholar 

  3. Gao, Z. et al. PCGF homologs, CBX proteins, and RYBP define functionally distinct PRC1 family complexes. Mol. Cell 45, 344–356 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Tavares, L. et al. RYBP–PRC1 complexes mediate H2A ubiquitylation at Polycomb target sites independently of PRC2 and H3K27me3. Cell 148, 664–678 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Blackledge, N. P., Rose, N. R. & Klose, R. J. Targeting Polycomb systems to regulate gene expression: modifications to a complex story. Nat. Rev. Mol. Cell Biol. 16, 643–649 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Cao, R. et al. Role of histone H3 lysine 27 methylation in Polycomb-group silencing. Science 298, 1039–1043 (2002).

    Article  CAS  PubMed  Google Scholar 

  8. Min, J., Zhang, Y. & Xu, R. M. Structural basis for specific binding of Polycomb chromodomain to histone H3 methylated at Lys 27. Genes Dev. 17, 1823–1828 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Wang, L. et al. Hierarchical recruitment of Polycomb group silencing complexes. Mol. Cell 14, 637–646 (2004).

    Article  CAS  PubMed  Google Scholar 

  10. 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  PubMed  PubMed Central  Google Scholar 

  11. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Cooper, S. et al. Jarid2 binds mono-ubiquitylated H2A lysine 119 to mediate crosstalk between Polycomb complexes PRC1 and PRC2. Nat. Commun. 7, 13661 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Kalb, R. et al. Histone H2A monoubiquitination promotes histone H3 methylation in Polycomb repression. Nat. Struct. Mol. Biol. 21, 569–571 (2014).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Cohen, I., Bar, C. & Ezhkova, E. Activity of PRC1 and histone H2AK119 monoubiquitination: revising popular misconceptions. Bioessays 42, e1900192 (2020).

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  19. Inoue, A., Jiang, L., Lu, F. & Zhang, Y. Genomic imprinting of Xist by maternal H3K27me3. Genes Dev. 31, 1927–1932 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Inoue, A., Jiang, L., Lu, F., Suzuki, T. & Zhang, Y. Maternal H3K27me3 controls DNA methylation-independent imprinting. Nature 547, 419–424 (2017).

    Article  CAS  PubMed  Google Scholar 

  21. Chen, Z. & Zhang, Y. Maternal H3K27me3-dependent autosomal and X chromosome imprinting. Nat. Rev. Genet. 22, 555–571 (2020).

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

    Article  CAS  PubMed  Google Scholar 

  23. Skene, P. J. & Henikoff, S. An efficient targeted nuclease strategy for high-resolution mapping of DNA binding sites. eLife 6, e21856 (2017).

  24. Fursova, N. A. et al. Synergy between variant PRC1 complexes defines Polycomb-mediated gene repression. Mol. Cell 74, 1020–1036 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Ernst, J. & Kellis, M. ChromHMM: automating chromatin-state discovery and characterization. Nat. Methods 9, 215–216 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Blackledge, N. P. et al. PRC1 catalytic activity is central to Polycomb system function. Mol. Cell 77, 857–874 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Tamburri, S. et al. Histone H2AK119 mono-ubiquitination is essential for Polycomb-mediated transcriptional repression. Mol. Cell 77, 840–856 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  29. Yuan, W. et al. H3K36 methylation antagonizes PRC2-mediated H3K27 methylation. J. Biol. Chem. 286, 7983–7989 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Yuan, G. et al. Histone H2A ubiquitination inhibits the enzymatic activity of H3 lysine 36 methyltransferases. J. Biol. Chem. 288, 30832–30842 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Matoba, S. et al. Loss of H3K27me3 imprinting in somatic cell nuclear transfer embryos disrupts post-implantation development. Cell Stem Cell 23, 343–354 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Borensztein, M. et al. Xist-dependent imprinted X inactivation and the early developmental consequences of its failure. Nat. Struct. Mol. Biol. 24, 226–233 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Inoue, A., Chen, Z., Yin, Q. & Zhang, Y. Maternal Eed knockout causes loss of H3K27me3 imprinting and random X inactivation in the extraembryonic cells. Genes Dev. 32, 1525–1536 (2018).

  34. Chen, Z., Yin, Q., Inoue, A., Zhang, C. & Zhang, Y. Allelic H3K27me3 to allelic DNA methylation switch maintains noncanonical imprinting in extraembryonic cells. Sci. Adv. 5, eaay7246 (2019).

  35. Almeida, M. et al. PCGF3/5-PRC1 initiates Polycomb recruitment in X chromosome inactivation. Science 356, 1081–1084 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Posfai, E. et al. Polycomb function during oogenesis is required for mouse embryonic development. Genes Dev. 26, 920–932 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Scheuermann, J. C. et al. Histone H2A deubiquitinase activity of the Polycomb repressive complex PR-DUB. Nature 465, 243–247 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Balasubramani, A. et al. Cancer-associated ASXL1 mutations may act as gain-of-function mutations of the ASXL1–BAP1 complex. Nat. Commun. 6, 7307 (2015).

    Article  CAS  PubMed  Google Scholar 

  39. Sahtoe, D. D., van Dijk, W. J., Ekkebus, R., Ovaa, H. & Sixma, T. K. BAP1/ASXL1 recruitment and activation for H2A deubiquitination. Nat. Commun. 7, 10292 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Dobrinić, P., Szczurek, A. & Klose, R. PRC1 drives Polycomb-mediated gene repression by controlling transcription initiation and burst frequency. Preprint at bioRxiv https://doi.org/10.1101/2020.10.09.333294 (2020).

  41. Harris, C. et al. Conversion of random X-inactivation to imprinted X-inactivation by maternal PRC2. eLife 8, e44258 (2019).

  42. Cohen, I. et al. PRC1 fine-tunes gene repression and activation to safeguard skin development and stem cell specification. Cell Stem Cell 22, 726–739 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Du, Z. et al. Polycomb group proteins regulate chromatin architecture in mouse oocytes and early embryos. Mol. Cell 77, 825–839 (2019).

  44. Prokopuk, L. et al. Loss of maternal EED results in postnatal overgrowth. Clin. Epigenetics 10, 95 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Zenk, F. et al. Germ line-inherited H3K27me3 restricts enhancer function during maternal-to-zygotic transition. Science 357, 212–216 (2017).

    Article  CAS  PubMed  Google Scholar 

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

  47. Wang, C. et al. Reprogramming of H3K9me3-dependent heterochromatin during mammalian embryo development. Nat. Cell Biol. 20, 620–631 (2018).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  49. Weinberg, D. N. et al. The histone mark H3K36me2 recruits DNMT3A and shapes the intergenic DNA methylation landscape. Nature 573, 281–286 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Zhang, C. et al. The chromatin remodeler Snf2h is essential for oocyte meiotic cell cycle progression. Genes Dev. 34, 166–178 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Chen, Z. & Zhang, Y. Loss of DUX causes minor defects in zygotic genome activation and is compatible with mouse development. Nat. Genet. 51, 947–951 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Lu, F. et al. Establishing chromatin regulatory landscape during mouse preimplantation development. Cell 165, 1375–1388 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Sugimoto, M. et al. A simple and robust method for establishing homogeneous mouse epiblast stem cell lines by Wnt inhibition. Stem Cell Rep. 4, 744–757 (2015).

    Article  CAS  Google Scholar 

  54. Inoue, A. & Zhang, Y. Nucleosome assembly is required for nuclear pore complex assembly in mouse zygotes. Nat. Struct. Mol. Biol. 21, 609–616 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. 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  PubMed  Google Scholar 

  56. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S. L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 10, R25 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Tarasov, A., Vilella, A. J., Cuppen, E., Nijman, I. J. & Prins, P. Sambamba: fast processing of NGS alignment formats. Bioinformatics 31, 2032–2034 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Krueger, F. & Andrews, S. R. SNPsplit: allele-specific splitting of alignments between genomes with known SNP genotypes. F1000Res 5, 1479 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Ramirez, F., Dundar, F., Diehl, S., Gruning, B. A. & Manke, T. deepTools: a flexible platform for exploring deep-sequencing data. Nucleic Acids Res. 42, W187–W191 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Meers, M. P., Bryson, T. D., Henikoff, J. G. & Henikoff, S. Improved CUT&RUN chromatin profiling tools. eLife 8, e46314 (2019).

  61. Kim, D., Langmead, B. & Salzberg, S. L. HISAT: a fast spliced aligner with low memory requirements. Nat. Methods 12, 357–360 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Pertea, M. et al. StringTie enables improved reconstruction of a transcriptome from RNA-seq reads. Nat. Biotechnol. 33, 290–295 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Jin, Y., Tam, O. H., Paniagua, E. & Hammell, M. TEtranscripts: a package for including transposable elements in differential expression analysis of RNA-seq datasets. Bioinformatics 31, 3593–3599 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Anders, S. & Huber, W. Differential expression analysis for sequence count data. Genome Biol. 11, R106 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Yu, G., Wang, L. G., Han, Y. & He, Q. Y. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS 16, 284–287 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Connell, J. & Højsgaard, S. Hidden semi Markov models for multiple observation sequences: the mhsmm package for R. J. Stat. Softw. 39, 1–22 (2011).

    Article  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  68. Lawrence, M., Gentleman, R. & Carey, V. rtracklayer: an R package for interfacing with genome browsers. Bioinformatics 25, 1841–1842 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Lawrence, M. et al. Software for computing and annotating genomic ranges. PLoS Comput. Biol. 9, e1003118 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Huber, W. et al. Orchestrating high-throughput genomic analysis with Bioconductor. Nat. Methods 12, 115–121 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Gu, Z., Eils, R. & Schlesner, M. Complex heatmaps reveal patterns and correlations in multidimensional genomic data. Bioinformatics 32, 2847–2849 (2016).

    Article  CAS  PubMed  Google Scholar 

  72. Gu, Z., Eils, R., Schlesner, M. & Ishaque, N. EnrichedHeatmap: an R/Bioconductor package for comprehensive visualization of genomic signal associations. BMC Genomics 19, 234 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  73. Kent, W. J. et al. The human genome browser at UCSC. Genome Res. 12, 996–1006 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Wu, J. et al. The landscape of accessible chromatin in mammalian preimplantation embryos. Nature 534, 652–657 (2016).

    Article  CAS  PubMed  Google Scholar 

  75. Andergassen, D. et al. Mapping the mouse Allelome reveals tissue-specific regulation of allelic expression. eLife 6, e25125 (2017).

Download references

Acknowledgements

We thank C. Zhang and W. Zhang for critical reading of the manuscript. This project was supported by the NIH (R01HD092465) and the HHMI. Y.Z. is an Investigator of the Howard Hughes Medical Institute.

Author information

Authors and Affiliations

Authors

Contributions

Y.Z. conceived the project. Z.C. and Y.Z. designed the experiments. Z.C. performed the experiments. M.N.D. performed most of the bioinformatic analyses, and Z.C. analyzed some of the ChIP-seq, CUT&RUN and RNA-seq datasets. All authors were involved in the interpretation of data. Z.C. and Y.Z. wrote the manuscript with M.N.D.’s input.

Corresponding author

Correspondence to Yi Zhang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Genetics thanks Maxim Greenberg and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

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

Extended data

Extended Data Fig. 1 H2Aub dynamics in oocytes and early embryos.

a, Genome browser view of H2Aub profiles in oocytes and early embryos. The H2Aub ChIP-seq data of mESC were obtained from24. b, Bar plots showing the reproducibility of the biological replicates of H2Aub CUT&RUN datasets. The H2Aub enrichment was calculated as reads per kilobase per million reads (RPKM) by 5-kb bin. The Pearson correlation co-efficient are also shown. c, Heatmap with hierarchical clustering showing the correlation of global H2Aub enrichment between oocytes and early embryos. d, Bar plot showing the percentage of genomic regions occupied by H2Aub domains (regions with H2Aub signal determined using ChromHMM) at different developmental stages. e, Bar plot showing the percentage of H2Aub CUT&RUN reads that are mapped to each ChromHMM type at different developmental stages.

Extended Data Fig. 2 Allelic dynamics of H2Aub in preimplantation development.

a, Genome browser views comparing our sperm H3K27me3 STAR ChIP-seq data with publicly available datasets22,55 b, Venn diagram showing that most H3K27me3 domains identified are common between Zheng et al., and our data. c, Genome browser view showing sperm H2Aub STAR ChIP-seq. d, Scatter plot showing the reproducibility of the biological replicates of H2Aub sperm ChIP-seq datasets. The H2Aub enrichment was calculated as reads per kilobase per million reads (RPKM) by 5-kb bin. The Pearson correlation co-efficient is also shown. e, Bar plot showing the number of allelically biased H2Aub regions at different developmental stages. Maternally (Mat) and paternally (Pat) biased H2Aub regions were identified by merging adjacent allelically biased 5kb bins meeting cutoff: fold change (FC) > 2 and adjusted p-value (p-adj) < 0.01. P values were calculated using a binomial exact test of the null hypothesis that both alleles are equally enriched and adjusted using Benjamin & Hochberg method. f, Box plot showing the expression levels of genes within allelically biased H2Aub regions at 1-cell stage. The number of genes within maternally or paternally biased H2Aub domains at 1-cell were 1113 and 15, respectively. The middle lines in the boxes represent medians. Box hinges indicate the twenty-fifth and seventy-fifth percentiles, the whiskers indicate the hinge ± 1.5 × interquartile range, and the dots represent outliers. RNA-seq data were obtained from47. Mat: maternal allele; Pat: paternal allele. g, Heatmaps showing allelic dynamics of the H2Aub regions that are not allelically biased at 1-cell during preimplantation development. The number of regions for each category are indicated. h, Genome browser view of allelic dynamics of the H2Aub regions that are not allelically biased at 1-cell during preimplantation development.

Extended Data Fig. 3 Distinct non-allelic dynamics of H3K27me3 and H2Aub in preimplantation embryos.

a, Genome browser view of H3K27me3, H2Aub and H3K36me348 dynamics in oocytes and preimplantation embryos. b, Venn diagrams showing overlaps of H3K27me3 and H2Aub domains at different developmental stages. The H3K27me3 ChIP-seq data in panels A and B were from17,22. c, Heatmap showing enrichment of H3K27me3, H2Aub, and H3K36me3 at H2Aub domains identified in GV oocytes. The GV H2Aub domains were classified into six groups based on dynamics of these histone modifications during development.

Extended Data Fig. 4 Distinct allelic dynamics of H3K27me3 and H2Aub in preimplantation embryos.

a, Genome browser view of allelic H3K27me3 and H2Aub dynamics at the indicated embryonic stages. Note that maternally biased H2Aub at 1-cell becomes equalized at 2-cell stage by gaining H2Aub on the paternal allele. However, H3K27me3 maintains maternally biased from 1-cell to morula stage. The H3K27me3 ChIP-seq/CUT&RUN data were from22,33. M: maternal; P: paternal. b, Genome browser view of allelic H3K27me3 and H2Aub at Jade1 (also known as Phf17) and Sfmbt2 locus. Note that maternal-biased H2Aub is lost at the imprinted loci in 2-cell embryos then regain during later development. In contrast, maternal biased H3K27me3 is maintained from 1-cell to morula stage. c, Box plots showing allelic bias of H2Aub at gene bodies of the 76 putative maternal H3K27me3-dependent imprinted genes. The left panel represents genes showing transient loss and regain of H2Aub allelic bias in early development, whereas the right panel shows genes that loss of H2Aub allelic bias after 2-cell stage. The middle lines in the boxes represent medians. Box hinges indicate the twenty-fifth and seventy-fifth percentiles, the whiskers indicate the hinge ± 1.5 × interquartile range, and the dots show outliers. Mat: maternal; Pat: paternal.

Extended Data Fig. 5 Loss of maternal Eed has limited effect on global H2Aub except the maternal H3K27me3-dependent imprinted loci.

a, Relative intensity of the H3K27me3 and H2Aub signal in 1-cell, 2-cell, and 4-cell embryos. For 1-cell embryos, only signals on the maternal pronuclei were measured. The average signal intensity of CTR embryos was set as 1.0. The total number of embryos analyzed were 9 (CTR) and 7 (matKO) for 1-cell embryos, 6 (CTR) and 6 (matKO) for 2-cell embryos, and 7 (CTR) and 8 (matKO) for 4-cell embryos, respectively. Center dot and error bars indicate mean and standard deviation, respectively. matKO: maternal knockout. b, Scatter plots showing the reproducibility between biological replicates of H2Aub CUT&RUN datasets. The H2Aub enrichment was calculated as RPKM in 5-kb bins. The Pearson correlation co-efficient are also shown. CKO: conditional knockout. c, Hierarchical clustering of maternal (M) and paternal (P) alleles of preimplantation embryo and GV oocytes based on their global H2Aub enrichment. d, Genome browser view showing comparable enrichment of H2Aub between CTR and CKO in GV oocytes and between CTR and matKO in preimplantation embryos. e, Genome browser view showing H2Aub enrichment at Gab1 and Sfmbt2 loci in CTR and Eed CKO GV oocytes and matKO morula embryos.

Extended Data Fig. 6 Quality control of acute H2Aub depletion and CUT&RUN datasets.

a, Representative zygotes immunostaining images. M: maternal pronucleus; P: paternal pronucleus; h: human; CTR: control; OE: over-expression; PR-DUB: Polycomb repressive deubiquitinase; WT: wild type. scale bar: 20 µm. The quantifications are in panel B) Relative H2Aub signal intensity of zygotes. Only signal on the maternal pronuclei (matPN) was measured. The number of embryos analyzed were 6 for non-injected, 11 for hBAP1 (C91A), 20 for CTR, 10 for hBAP1 (WT), and 22 for OE, respectively. c, Relative H2Aub signal intensity of 2-cell and 4-cell embryos. The number of embryos analyzed were 2 for non-injected 2-cell, 3 for CTR 2-cell, 3 for OE 2-cell, 9 for non-injected 4-cell, 10 for CTR 4-cell, and 9 for OE 4-cell, respectively. For Panel B-C), center dot and error bars indicate mean and standard deviation, respectively. d, Scatter plot comparing H2Aub enrichment between non-injected and CTR 4-cell embryos. Pearson correlation co-efficient is shown. e, Stacked bar plot showing the overlap between the top 1000 bins and RING1B-binding sites identified in mESCs24. f, Box plots showing RING1B signals in mESCs and the H2Aub levels in CTR and OE embryos at rank ordered RING1B-binding sites (n = 8833). The bottom 5 (Bot5%, n = 442), top 5 (Top5%, n = 442), and middle 50 percentile (Mid50%, n = 4416) RING1B sites are indicated. The middle lines in the boxes represent medians. Box hinges indicate the twenty-fifth and seventy-fifth percentiles, the whiskers indicate the hinge ± 1.5 × interquartile range. P-value was calculated by Wilcoxon test (two-sided). g, Bar plots showing reproducibility of the biological replicates of H3K27me3 CUT&RUN datasets. The Pearson correlation co-efficient are shown. h, Genome browser view of H2Aub and H3K27me3 enrichment at the Jade1 and Sfmbt2 loci in 4-cell embryos.

Extended Data Fig. 7 The transcriptional effects of H2Aub and H3K27me3 depletion in early embryos.

a, Heatmap showing the correlation between replicates of PR-DUB RNA-seq samples. b, Left panel shows representative immunostaining images of zygotes. M: maternal pronucleus; P: paternal pronucleus; Pb: polar body. scale bar: 20 µm. Right panel shows quantification of the H3K27me3 signal intensity. Only signal on the maternal pronuclei (matPN) was measured. The number of embryos analyzed were 8 for non-injected, 4 for KDM6b (CTR), and 8 for KDM6b (OE), respectively. Center dot and error bars indicate mean and standard deviation, respectively. C) Heatmap showing the correlation between replicates of Kdm6b and Eed RNA-seq samples. d, Scatter plot comparing gene expression levels of KDM6B (CTR) and (OE) 2-cell and 4-cell embryos. fold change (FC) >2, P-value < 0.05, FPKM > 1. e, Scatter plot comparing gene expression levels of Eed (CTR) and (matKO) 2-cell embryos. fold change (FC) >2, P-value < 0.05, FPKM > 1. f, Left panel, heatmaps of the genes up-regulated by H2Aub removal in 2-cell embryos; Right panel, heatmaps showing expression dynamics of the genes on the left panel at different developmental stages. RNA-seq data in right panel were from17,20,74,75. G) Box plots comparing the expression levels of ZGA and maternal decay genes. The middle lines in the boxes represent medians. Box hinges indicate the twenty-fifth and seventy-fifth percentiles, the whiskers indicate the hinge ± 1.5 × interquartile range. The ZGA (n = 3090) and maternal decay (n = 2343) genes were identified by comparing 2-cell and MII oocytes total RNA-seq data (Supplementary Table 1). Genes up- and down- regulated in 2-cell versus MII oocyte were defined as ZGA and maternal decay genes, respectively. fold change > 5, p-value < 0.05, and RPKM > 1.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

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). https://doi.org/10.1038/s41588-021-00821-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-021-00821-2

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