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:

H2A.Z reinforces maternal H3K4me3 formation and is essential for meiotic progression in mouse oocytes

Abstract

Mammalian oocytes establish a unique landscape of histone modifications, some of which are inherited by early embryos. How histone variants shape the maternal histone landscape remains unknown. Here we map histone H2A variants in mouse fully grown oocytes (FGOs) and find that H2A.Z forms broad domains across intergenic regions, along non-canonical H3K4me3 (ncH3K4me3). During oocyte growth, H2A.Z progressively transitions from an active promoter-rich, canonical distribution to a non-canonical broad distribution (ncH2A.Z). Depletion of H2A.Z in oocytes partially impairs ncH3K4me3 formation and causes severe defects in meiotic progression, which resemble Mll2-knockout oocytes. Conversely, depletion of ncH3K4me3 by Mll2 knockout also causes a reduction of ncH2A.Z in FGOs. Thus, our study suggests that ncH2A.Z and ncH3K4me3 reinforce each other to form functional oocytes.

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: Distribution of H2A variants in FGOs.
Fig. 2: Shifting to ncH2A.Z distribution during oocyte growth.
Fig. 3: H2A.Z is dissociated from bivalent promoters in FGOs.
Fig. 4: H2A.Z contributes to MLL2-dependent ncH3K4me3 formation.
Fig. 5: ncH2A.Z and MLL2-dependent ncH3K4me3 reinforce each other.
Fig. 6: Both H2A.Z-DKO and Mll2-KO oocytes are defective in meiotic maturation.
Fig. 7: Illustration of H2A.Z dynamics in 5-day GOs and FGOs, and histone modification changes in H2A.Z-DKO and Mll2-KO FGOs.

Similar content being viewed by others

Data availability

All the sequencing datasets generated in this study have been deposited in the GEO database under accession numbers GSE262044 and GSE262049. The H2A.Z ChIP–seq datasets of mouse MII oocytes and preimplantation embryos were from GSE188590 (ref. 17). RNA-seq datasets during oogenesis were from GSE70116 (ref. 41). H3K4me3 ChIP–seq datasets of four stages of WT oocytes were from GSE93941 (ref. 7). H3K27ac ChIP–seq datasets of mouse oocytes were from GSE217970 (ref. 33). H3K4me3 ChIP–seq datasets of mouse MII oocytes and preimplantation embryos were from GSE73952 (ref. 6) and GSE72784 (ref. 8), respectively. All other data supporting the findings of this study are available from the corresponding author on reasonable request. Source data are provided with this paper.

Code availability

The software and tools used in our study are all publicly available. Scripts used for data visualization are available upon request.

References

  1. Clift, D. & Schuh, M. Restarting life: fertilization and the transition from meiosis to mitosis. Nat. Rev. Mol. Cell Biol. 14, 549–562 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Zaffagnini, G. et al. Mouse oocytes sequester aggregated proteins in degradative super-organelles. Cell 187, 1109–1126 (2024).

    Article  CAS  PubMed  Google Scholar 

  3. Jentoft, I. M. A. et al. Mammalian oocytes store proteins for the early embryo on cytoplasmic lattices. Cell 186, 5308–5327 (2023).

    Article  CAS  PubMed  Google Scholar 

  4. Staubli, A. & Peters, A. H. Mechanisms of maternal intergenerational epigenetic inheritance. Curr. Opin. Genet. Dev. 67, 151–162 (2021).

    Article  CAS  PubMed  Google Scholar 

  5. Xu, Q. & Xie, W. Epigenome in early mammalian development: inheritance, reprogramming and establishment. Trends Cell Biol. 28, 237–253 (2018).

    Article  CAS  PubMed  Google Scholar 

  6. 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 

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

    Article  PubMed  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  Google Scholar 

  10. 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).

  11. Buschbeck, M. & Hake, S. B. Variants of core histones and their roles in cell fate decisions, development and cancer. Nat. Rev. Mol. Cell Biol. 18, 299–314 (2017).

    Article  CAS  PubMed  Google Scholar 

  12. Martire, S. & Banaszynski, L. A. The roles of histone variants in fine-tuning chromatin organization and function. Nat. Rev. Mol. Cell Biol. 21, 522–541 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Nashun, B. et al. Continuous histone replacement by hira is essential for normal transcriptional regulation and de novo DNA methylation during mouse oogenesis. Mol. Cell 60, 611–625 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Ishiuchi, T. et al. Reprogramming of the histone H3.3 landscape in the early mouse embryo. Nat. Struct. Mol. Biol. 28, 38–49 (2021).

    Article  CAS  PubMed  Google Scholar 

  15. Shinagawa, T. et al. Histone variants enriched in oocytes enhance reprogramming to induced pluripotent stem cells. Cell Stem Cell 14, 217–227 (2014).

    Article  CAS  PubMed  Google Scholar 

  16. Nashun, B., Yukawa, M., Liu, H. L., Akiyama, T. & Aoki, F. Changes in the nuclear deposition of histone H2A variants during pre-implantation development in mice. Development 137, 3785–3794 (2010).

    Article  CAS  PubMed  Google Scholar 

  17. Liu, X. et al. Hierarchical accumulation of histone variant H2A.Z regulates transcriptional states and histone modifications in early mammalian embryos. Adv. Sci. 9, 2200057 (2022).

    Article  CAS  Google Scholar 

  18. Chang, C. C. et al. A maternal store of macroH2A is removed from pronuclei prior to onset of somatic macroH2A expression in preimplantation embryos. Dev. Biol. 278, 367–380 (2005).

    Article  CAS  PubMed  Google Scholar 

  19. Faast, R. et al. Histone variant H2A.Z is required for early mammalian development. Curr. Biol. 11, 1183–1187 (2001).

    Article  CAS  PubMed  Google Scholar 

  20. Celeste, A. et al. Genomic instability in mice lacking histone H2AX. Science 296, 922–927 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Pehrson, J. R., Changolkar, L. N., Costanzi, C. & Leu, N. A. Mice without macroH2A histone variants. Mol. Cell. Biol. 34, 4523–4533 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Mylonas, C., Lee, C., Auld, A. L., Cisse, I. I. & Boyer, L. A. A dual role for H2A.Z.1 in modulating the dynamics of RNA polymerase II initiation and elongation. Nat. Struct. Mol. Biol. 28, 435–442 (2021).

    Article  CAS  PubMed  Google Scholar 

  23. Weber, C. M., Ramachandran, S. & Henikoff, S. Nucleosomes are context-specific, H2A.Z-modulated barriers to RNA polymerase. Mol. Cell 53, 819–830 (2014).

    Article  CAS  PubMed  Google Scholar 

  24. Ku, M. C. et al. H2A.Z landscapes and dual modifications in pluripotent and multipotent stem cells underlie complex genome regulatory functions. Genome Biol. 13, R85 (2012).

  25. Hu, G. Q. et al. H2A.Z facilitates access of active and repressive complexes to chromatin in embryonic stem cell self-renewal and differentiation. Cell Stem Cell 12, 180–192 (2013).

    Article  CAS  PubMed  Google Scholar 

  26. Creyghton, M. P. et al. H2AZ is enriched at polycomb complex target genes in ES cells and is necessary for lineage commitment. Cell 135, 649–661 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Long, H. et al. H2A.Z facilitates licensing and activation of early replication origins. Nature 577, 576–581 (2020).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  Google Scholar 

  29. Matsuwaka, M., Kumon, M. & Inoue, A. H3K27 dimethylation dynamics reveal stepwise establishment of facultative heterochromatin in early mouse embryos. Nat. Cell Biol. 27, 28–38 (2025).

    Article  CAS  PubMed  Google Scholar 

  30. De La Fuente, R. et al. Major chromatin remodeling in the germinal vesicle (GV) of mammalian oocytes is dispensable for global transcriptional silencing but required for centromeric heterochromatin function. Dev. Biol. 275, 447–458 (2004).

    Article  PubMed  Google Scholar 

  31. Wang, C. F. et al. Dynamic nucleosome organization after fertilization reveals regulatory factors for mouse zygotic genome activation. Cell Res. 32, 801–813 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  33. Liu, B. et al. Mapping putative enhancers in mouse oocytes and early embryos reveals TCF3/12 as key folliculogenesis regulators. Nat. Cell Biol. 26, 962–974 (2024).

    Article  CAS  PubMed  Google Scholar 

  34. Bensaude, O. Inhibiting eukaryotic transcription: which compound to choose? How to evaluate its activity?. Transcription 2, 103–108 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  35. 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 

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

    Article  CAS  PubMed  Google Scholar 

  37. de Vries, W. N. et al. Expression of Cre recombinase in mouse oocytes: a means to study maternal effect genes. Genesis 26, 110–112 (2000).

    Article  PubMed  Google Scholar 

  38. 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  PubMed  Google Scholar 

  39. Huang, C. & Zhu, B. Roles of H3K36-specific histone methyltransferases in transcription: antagonizing silencing and safeguarding transcription fidelity. Biophys. Rep. 4, 170–177 (2018).

    Article  CAS  PubMed  Google Scholar 

  40. Peaston, A. et al. Retrotransposons regulate host genes in mouse oocytes and preimplantation embryos. Dev. Cell 7, 597–606 (2004).

    Article  CAS  PubMed  Google Scholar 

  41. Veselovska, L. et al. Deep sequencing and de novo assembly of the mouse oocyte transcriptome define the contribution of transcription to the DNA methylation landscape. Genome Biol. 16, 209 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Huang, L. et al. Biallelic variants in MAD2L1BP (p31(comet)) cause female infertility characterized by oocyte maturation arrest. eLife 12, e85649 (2023).

  43. Hwang, G. et al. SMC5/6 is required for the formation of segregation-competent bivalent chromosomes during meiosis I in mouse oocytes. Development 144, 1648–1660 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Wong, M. M., Cox, L. K. & Chrivia, J. C. The chromatin remodeling protein, SRCAP, is critical for deposition of the histone variant H2A.Z at promoters. J. Biol. Chem. 282, 26132–26139 (2007).

    Article  CAS  PubMed  Google Scholar 

  45. Liang, X. P. et al. Structural basis of H2A.Z recognition by SRCAP chromatin-remodeling subunit YL1. Nat. Struct. Mol. Biol. 23, 317–323 (2016).

    Article  CAS  PubMed  Google Scholar 

  46. Zhao, B. et al. Znhit1 controls intestinal stem cell maintenance by regulating H2A.Z incorporation. Nat. Commun. 10, 1071 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Mao, Z. et al. Anp32e, a higher eukaryotic histone chaperone directs preferential recognition for H2A.Z. Cell Res. 24, 389–399 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Obri, A. et al. ANP32E is a histone chaperone that removes H2A.Z from chromatin. Nature 505, 648–653 (2014).

    Article  PubMed  Google Scholar 

  49. Alatwi, H. E. & Downs, J. A. Removal of H2A.Z by INO80 promotes homologous recombination. EMBO Rep. 16, 986–994 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Greenberg, R. S., Long, H. K., Swigut, T. & Wysocka, J. Single amino acid change underlies distinct roles of H2A.Z subtypes in human syndrome. Cell 178, 1421–1436 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Sales-Gil, R. et al. Non-redundant functions of H2A.Z.1 and H2A.Z.2 in chromosome segregation and cell cycle progression. EMBO Rep. 22, e52061 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Matsuda, R. et al. Identification and characterization of the two isoforms of the vertebrate H2A.Z histone variant. Nucleic Acids Res. 38, 4263–4273 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Lukauskas, S. et al. Decoding chromatin states by proteomic profiling of nucleosome readers. Nature 627, 671–679 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Eckersley-Maslin, M. A. et al. Epigenetic priming by Dppa2 and 4 in pluripotency facilitates multi-lineage commitment. Nat. Struct. Mol. Biol. 27, 696–705 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Wang, M., Chen, Z. & Zhang, Y. CBP/p300 and HDAC activities regulate H3K27 acetylation dynamics and zygotic genome activation in mouse preimplantation embryos. EMBO J. 41, e112012 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Davis, W. Jr. & Schultz, R. M. Developmental change in TATA-box utilization during preimplantation mouse development. Dev. Biol. 218, 275–283 (2000).

    Article  CAS  PubMed  Google Scholar 

  57. Gazdag, E. et al. TBP2 is essential for germ cell development by regulating transcription and chromatin condensation in the oocyte. Genes Dev. 23, 2210–2223 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Hamazaki, N. et al. Reconstitution of the oocyte transcriptional network with transcription factors. Nature 589, 264–269 (2021).

    Article  CAS  PubMed  Google Scholar 

  59. Yu, C. et al. TBPL2/TFIIA complex establishes the maternal transcriptome through oocyte-specific promoter usage. Nat. Commun. 11, 6439 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Du, Z. et al. Polycomb group proteins regulate chromatin architecture in mouse oocytes and early embryos. Mol. Cell 77, 825–839 (2020).

    Article  CAS  PubMed  Google Scholar 

  61. Chen, K. F. et al. Broad H3K4me3 is associated with increased transcription elongation and enhancer activity at tumor-suppressor genes. Nat. Genet. 47, 1149–1157 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  62. Berta, D. G. et al. Deficient H2A.Z deposition is associated with genesis of uterine leiomyoma. Nature 596, 398–403 (2021).

    Article  CAS  PubMed  Google Scholar 

  63. Wang, Y. et al. Histone variants H2A.Z and H3.3 coordinately regulate PRC2-dependent H3K27me3 deposition and gene expression regulation in mES cells. BMC Biol. 16, 107 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  64. Rangasamy, D., Greaves, I. & Tremethick, D. J. RNA interference demonstrates a novel role for H2A.Z in chromosome segregation. Nat. Struct. Mol. Biol. 11, 650–655 (2004).

    Article  CAS  PubMed  Google Scholar 

  65. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. van den Boom, V. et al. Non-canonical PRC1.1 targets active genes independent of H3K27me3 and is essential for leukemogenesis. Cell Rep. 14, 332–346 (2016).

    Article  CAS  PubMed  Google Scholar 

  67. Zhou, Y., Romero-Campero, F. J., Gomez-Zambrano, A., Turck, F. & Calonje, M. H2A monoubiquitination in Arabidopsis thaliana is generally independent of LHP1 and PRC2 activity. Genome Biol. 18, 69 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  68. Loubiere, V., Papadopoulos, G. L., Szabo, Q., Martinez, A.-M. & Cavalli, G. Widespread activation of developmental gene expression characterized by PRC1-dependent chromatin looping. Sci. Adv. 6, eaax4001 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Surface, L. E. et al. H2A.Z.1 monoubiquitylation antagonizes BRD2 to maintain poised chromatin in ESCs. Cell Rep. 14, 1142–1155 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Sarcinella, E., Zuzarte, P. C., Lau, P. N. I., Draker, R. & Cheung, P. Monoubiquitylation of H2A.Z distinguishes its association with euchromatin or facultative heterochromatin. Mol. Cell. Biol. 27, 6457–6468 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. 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  PubMed  PubMed Central  Google Scholar 

  72. 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 

  73. Xu, Q. et al. H2A.Z is essential for oocyte maturation and female fertility in mouse. Nat. Struct. Mol. Biol. https://doi.org/10.1038/s41594-025-01580-y (2025).

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

    Article  CAS  PubMed  Google Scholar 

  75. Concordet, J. P. & Haeussler, M. CRISPOR: intuitive guide selection for CRISPR/Cas9 genome editing experiments and screens. Nucleic Acids Res. 46, W242–W245 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Gu, B., Pósfai, E. & Rossant, J. Efficient generation of targeted large insertions by microinjection into two-cell-stage mouse embryos. Nat. Biotechnol. 36, 632–637 (2018).

    Article  CAS  PubMed  Google Scholar 

  77. Inoue, A., Akiyama, T., Nagata, M. & Aoki, F. The perivitelline space-forming capacity of mouse oocytes is associated with meiotic competence. J. Reprod. Dev. 53, 1043–1052 (2007).

    Article  PubMed  Google Scholar 

  78. Hodges, C. & Hunt, P. Simultaneous analysis of chromosomes and chromosome-associated proteins in mammalian oocytes and embryos. Chromosoma 111, 165–169 (2002).

    Article  CAS  PubMed  Google Scholar 

  79. 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 (2018).

    Article  CAS  PubMed  Google Scholar 

  80. 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 

  81. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Hayashi, R. & Inoue, A. Low-input CUT&RUN for mouse oocytes and preimplantation embryos. Methods Mol. Biol. 2577, 83–92 (2023).

  84. Skene, P. J. & Henikoff, S. An efficient targeted nuclease strategy for high-resolution mapping of DNA binding sites. eLife 6, 576 (2017).

    Article  Google Scholar 

  85. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–U54 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. 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 

  87. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  88. Ramírez, F., Dündar, F., Diehl, S., Grüning, B. A. & Manke, T. deepTools: a flexible platform for exploring deep-sequencing data. Nucleic Acids Res. 42, W187–W191 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  89. Robinson, J. T. et al. Integrative genomics viewer. Nat. Biotechnol. 29, 24–26 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

  91. Quinlan, A. R. & Hall, I. M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  93. Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).

    Article  CAS  PubMed  Google Scholar 

  94. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    Article  CAS  PubMed  Google Scholar 

  95. Li, B. & Dewey, C. N. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinformatics 12, 323 (2011).

  96. Yu, G. C., Wang, L. G. & He, Q. Y. ChIPseeker: an R/Bioconductor package for ChIP peak annotation, comparison and visualization. Bioinformatics 31, 2382–2383 (2015).

    Article  CAS  PubMed  Google Scholar 

  97. Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. McLean, C. Y. et al. GREAT improves functional interpretation of cis-regulatory regions. Nat. Biotechnol. 28, 495–U155 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Zhou, Y. Y. et al. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat. Commun. 10, 1523 (2019).

Download references

Acknowledgements

We thank L. Shen (Zhejiang Univ.) for sharing a detailed protocol for CATCH-seq. We appreciate the Inoue Lab members for critical reading of the paper and D. Noda (Kumamoto Univ.) for advice about the flox knock-in construct. This project was partly supported by the Ministry of Education, Culture, Sports, Science and Technology (MEXT) Leading Initiative for Excellent Young Researchers Grant to A.I., Grant-in-Aid for Scientific Research on Innovative Areas (19H05754 to A.I.), Grant-in-Aid for Transformative Research Areas (A) (25H01355 to A.I.), Grant-in-Aid for Scientific Research (B) (23K27093 to A.I. and 23K28009 to C.K.), Grant-in-Aid for JSPS Fellows (20J21541 to R.H.), Japan Agency for Medical Research and Development PRIME (JP20gm6110012 to A.I.), and intramural grants within RIKEN including the RIKEN Pioneering Project ‘Genome Building from TADs’ (to A.I. and H.K.) and ‘Long-timescale Molecular Chronobiology’ (to A.I.).

Author information

Authors and Affiliations

Authors

Contributions

A.I. conceived of the project. H.M. and A.I. designed the experiments. R.H., C.K., M.K. and A.I. performed the experiments. H.M. analyzed sequence data. H.M. and A.I. interpreted the data. H.K. provided the H2afz and H2afv flox mouse lines. H.M. and A.I. wrote the paper.

Corresponding author

Correspondence to Azusa Inoue.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Structural & Molecular Biology thanks Patrick Murphy and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Dimitris Typas, in collaboration with the Nature Structural and 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 H2A.Z dynamics during oogenesis.

a, Scatter plots showing the correlations between biological replicates of H2A.Z CATCH-seq. b, Heatmaps showing the enrichment of H2A.Z signals at genic regions and expression levels of genes classified into 4 clusters. Actively expressed genes were defined by [RPKM > 1], while lowly expressed genes are [0.1 < RPKM < 1], and inactive genes are [RPKM < 0.1] in all stages, respectively. The remaining genes were defined as variably expressed genes. The RNA-seq datasets were from Veselovska et al.41. c, Box plots showing the H2A.Z signal intensity at the promoters of the 4 clusters. The center lines in the boxes represent median values. The box edges, upper and lower whiskers indicate the interquartile range (IQR, from the 25th to 75th percentile), the maximal value smaller than 1.5 x the IQR above the 75th percentile, and the minimal value larger than 1.5 x the IQR below the 25th percentile, respectively. P-value was calculated by two-sided t-test. d, Transcription factor motifs identified at intergenic ncH2A.Z-H3K27ac dual-marked regions (left). Right bubble plots represent the expression level of TFs in FGOs and the statistical significance of the motif enrichment (P-value of hypergeometric test with one-sided). e, The proportion of the number of merged H2A.Z domains when merging adjacent peaks at each different distance from 1- to 12-kb. After summing the number of merged domains at each different distance from 1- to 12-kb, the number of merged domains at each distance was divided by the total number. f, Heatmap showing the global Pearson correlations between H2A.Z and H3K4me3 in four stages during oogenesis. The bin size is 5 kb. g, Violin plot showing the “skewing distance” of each H2A.Z peak. h, Representative images of 5-ethynyl uridine (EU) incorporation assay to validate transcriptional inhibition. 10d-GOs were first treated with 0.2% DMSO, 100 µM 5,6-dichlorobenzimidazole (DRB), or 100 µg/mL α-amanitin for 2 hrs, incubated with a medium containing EU and the respective inhibitors for additional 2 hrs, and then fixed. Bar plot indicates quantification of the EU signal intensity. The data are representative of a single time experiment using 10 (DMSO), 12 (DRB), and 10 (amanitin) oocytes. Scale bar, 20 µm. i, Genome browser views of H2A.Z and H3K4me3 landscapes in 5d-GO, 10d-GO, 15d-GO, and 28d-FGO oocytes, as well as their dynamic profiles in mouse preimplantation embryos. The H2A.Z ChIP-seq datasets are from Liu et al.17. The H3K4me3 ChIP-seq datasets in oocytes and preimplantation embryos are from Hanna et al.7 and Dahl et al.8, respectively. Broad ncH3K4me3 domains are highlighted.

Source data

Extended Data Fig. 2 Characterization of H2A.Z loss in FGOs.

a, Volcano plots showing differentially modified regions of H2A.Z at two adjacent stages. H2A.Z-lost peaks are highlighted (FDR < 0.05, |FC | > 2). FDR is calculated by Benjamini-Hochberg adjusted p-value (two-sided) of glmLRT in ‘edgeR’. b, Genomic distribution of H2A.Z peaks that are lost or retained in FGOs compared with 15 d GOs. c, Transcription factor motifs identified at H2A.Z-lost promoters (left). Right bubble plots represent the expression level of TFs in FGOs and the statistical significance of the motif enrichment (P-value of hypergeometric test with one-sided). d, Box plots showing expression levels of the genes whose promoters lose or retain H2A.Z in FGOs. The center lines in the boxes represent median values. The box edges, upper and lower whiskers indicate the interquartile range (IQR, from the 25th to 75th percentile), the maximal value smaller than 1.5 x the IQR above the 75th percentile, and the minimal value larger than 1.5 x the IQR below the 25th percentile, respectively (n = 1 as biological replicates were combined). The number of H2A.Z-lost or H2A.Z-retained genes are 1,799 and 10,567, respectively. e, Heatmaps showing the H2A.Z enrichment at bivalent promoters in DMSO-, DRB- and α-amanitin-treated GOs. f, Genome browser views showing the loss of H2A.Z at bivalent promoters in DRB- and α-amanitin-treated GOs.

Source data

Extended Data Fig. 3 CATCH-seq in H2A.Z.1 and H2A.Z.2 double knockout oocytes.

a, Scheme of generation of the H2A.Z.1 and H2A.Z.2 conditional alleles. The location of the genotyping PCR primers is indicated by red arrows. b, Scatter plots showing the correlations between biological replicates of H2A.Z and H3K4me3 CATCH-seq in H2A.Z control and DKO FGOs. c, Volcano plots showing differential H3K4me3 changes between H2A.Z control and DKO FGOs. ‘H3K4me3 loss’ was defined by FDR < 0.05 & |FC | ≥ 2, while ‘Intermediate’ was defined by FDR < 0.05 & 1.5 < |FC | < 2. FDR is calculated by Benjamini-Hochberg adjusted p-value (two-sided) of glmLRT in ‘edgeR’. d, GO biological processes enriched for H3K4me3-lost intergenic regions in H2A.Z DKO FGOs. Adjusted P-value (two-sided) is calculated by a binomial test with FDR correction. e, Pie charts showing the proportion of H2A.Z, H3K4me3, and H3K36me2 modified chromatins in WT FGOs. f, Genome browser views showing the mutually exclusive distribution of H2A.Z and H3K36me2 in mouse FGOs. g, Average signal profiles of H2A.Z, H3K4me3, and H3K36me2 at intergenic H3K4me3 peaks. h, Scatter plots showing the correlations between biological replicates of H3K36me2 CATCH-seq in H2A.Z control and DKO FGOs. i, Density plot showing the change of H3K36me2 over H2A.Z-enriched bins between H2A.Z control and DKO FGOs. j, Genome browser views showing gain of H3K36me2 at H2A.Z/H3K4me3-lost regions in H2A.Z DKO FGOs. k, Box plots showing H3K4me3 levels at H3K36me2-up bins in H2A.Z control and DKO FGOs. The center lines in the boxes represent median values. The box edges, upper and lower whiskers indicate the interquartile range (IQR, from the 25th to 75th percentile), the maximal value smaller than 1.5 x the IQR above the 75th percentile, and the minimal value larger than 1.5 x the IQR below the 25th percentile, respectively (n = 1 as biological replicates were combined). The number of H3K36me2-up bins in CTR and DKO are 15,008. P-value was calculated using a two-sided t-test. l, Bar charts showing the proportion of transcriptionally up- and down-regulated bins among H3K36me2-up & expressed bins (the number of intergenic RNA-seq reads >= 5 in either CTR or DKO).

Source data

Extended Data Fig. 4 CATCH-seq in Mll2 conditional knockout oocytes.

a, Scheme of CRISPR/Cas9-mediated knock-in of the flox cassette at Mll2/Kmt2b. The location of the genotyping PCR primers is indicated by red arrows. b, Scatter plots showing the correlations between biological replicates of H2A.Z and H3K4me3 CATCH-seq in Mll2 WT and KO FGOs. c, Volcano plots showing differential H2A.Z changes between Mll2 WT and KO FGOs. ‘H2A.Z loss’ was defined by FDR < 0.05 & |FC | ≥ 2, while ‘Intermediate’ was defined by FDR < 0.05 & 1.5 < |FC | < 2. FDR is calculated by Benjamini-Hochberg adjusted p-value (two-sided) of glmLRT in ‘edgeR’. d, Scatter plots of genome-wide non-TSS bins of H2A.Z levels in Mll2 WT and KO FGOs (left) and H3K4me3 levels in H2A.Z CTR and DKO FGOs (right). Green and red dots represent H3K4me3-lost bins in the H2A.Z DKO and H2A.Z-lost bins in the Mll2 KO, respectively. Shown are 50,000 randomly selected bins (2-kb).

Source data

Extended Data Fig. 5 Development of H2A.Z DKO oocytes and characterization of differentially expressed genes.

a, Representative images at 24- and 96- hours post-fertilization (hpf), which correspond to the time points of 2-cell and blastocyst stages, respectively, in CTR. Scale bar, 100 µm. Two independent experiments were performed with similar results. b, The ratios of preimplantation development. The total number of zygotes examined in 2 independent experiments was 106 (H2A.Z CTR) and 160 (H2A.Z DKO). c, d, Scatter plots showing the correlations of RNA-seq data between biological duplicates. e, f, MA plot of log2 FC in gene expression of H2A.Z DKO (e) and Mll2 KO (f) FGOs versus WT. Significantly up- and down-regulated genes in statistics (Benjamini-Hochberg-adjusted P-value (two-sided) < 0.01) are highlighted. g, Gene ontology terms enriched for the down- and up-regulated DEGs overlapped between H2A.Z DKO and Mll2 KO FGOs. P-value is calculated by accumulative hypergeometric test (two-sided).

Source data

Extended Data Fig. 6 Effect of H2A.Z DKO in H2AK119ub1 and H3K27me3 in FGOs.

a, b, Scatter plots showing the correlations between biological duplicates of H3K27me3 (a) and H2AK119ub1 (b) CATCH-seq in H2A.Z control and DKO FGOs. c, Heatmaps showing the signal intensity of H2A.Z, H3K4me3, H3K27me3, and H2AK119ub1 at all genes in H2A.Z control and H2A.Z DKO FGOs. Actively expressed genes are defined by [RPKM > 1], while lowly expressed genes are [0.1 < RPKM < 1], and inactive genes are [RPKM < 0.1] in all stages, respectively. The remaining genes are defined as variably expressed genes. The RNA-seq datasets are from Veselovska et al.41. d, Box plots showing the signal intensity of H2A.Z, H3K4me3, H3K27me3, and H2AK119ub1 at promoters in H2A.Z control and DKO FGOs. The center lines in the boxes represent median values. The box edges, upper and lower whiskers indicate the interquartile range (IQR, from the 25th to 75th percentile), the maximal value smaller than 1.5 x the IQR above the 75th percentile, and the minimal value larger than 1.5 x the IQR below the 25th percentile, respectively (n = 1 as biological replicates were combined). P-value was calculated by two-sided t-test. e, Heatmaps (left panel) showing the signal intensity of indicated histone modifications at H2AK119ub1/H3K4me3-harbored promoters. The line plots at right show the average signal profiles of H2K119ub1 and H3K4me3 around TSSs in H2A.Z control and DKO FGOs. f, Genome browser views of representative genes that show a reduction of H2AK119ub1 at active genes in H2A.Z DKO FGOs. g, Heatmaps showing the signal intensity of indicated histone modifications at ‘H3K4me3-lost’, ‘Intermediate’, and ‘H3K4me3-unchanged’ promoters in H2A.Z control and DKO FGOs. h, Box plots showing the signal intensity of H2AK119ub1 and H3K4me3 at ‘H3K4me3-lost’, ‘Intermediate’, and ‘H3K4me3-unchanged’ regions in H2A.Z control and DKO FGOs. The definitions of lines, box edges, whiskers, and n are the same as panel d. P-value was calculated by two-sided t-test.

Source data

Supplementary information

Reporting Summary

Supplementary Tables 1–4

Table 1: List of all genes and DEGs of RNA-seq in H2A.Z CTR and DKO FGOs. Table 2: List of all genes and DEGs of RNA-seq in Mll2 WT and KO FGOs. Table 3: Primers and oligo DNA information. Table 4: Sequence data summary

Source data

Source Data Fig. 1

Numerical source data.

Source Data Fig. 2

Numerical source data.

Source Data Fig. 3

Numerical source data.

Source Data Fig. 4

Numerical source data.

Source Data Fig. 5

Numerical source data.

Source Data Fig. 6

Numerical source data.

Source Data Extended Data Fig. 1

Numerical source data.

Source Data Extended Data Fig. 2

Numerical source data.

Source Data Extended Data Fig. 3

Numerical source data.

Source Data Extended Data Fig. 4

Numerical source data.

Source Data Extended Data Fig. 5

Numerical source data.

Source Data Extended Data Fig. 6

Numerical 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.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mei, H., Hayashi, R., Kozuka, C. et al. H2A.Z reinforces maternal H3K4me3 formation and is essential for meiotic progression in mouse oocytes. Nat Struct Mol Biol (2025). https://doi.org/10.1038/s41594-025-01573-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41594-025-01573-x

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