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:

Epigenomic analysis of gastrulation identifies a unique chromatin state for primed pluripotency

Abstract

Around implantation, the epiblast (Epi) transits from naïve to primed pluripotency, before giving rise to the three germ layers. How chromatin is reconfigured during this developmental window remains poorly understood. We performed a genome-wide investigation of chromatin landscapes during this period. We find that enhancers in ectoderm are already pre-accessible in embryonic day 6.5 (E6.5) Epi when cells enter a primed pluripotent state. Unexpectedly, strong trimethylation of histone H3 at lysine 4 (H3K4me3) emerges at developmental gene promoters in E6.5 Epi and positively correlates with H3K27me3, thus establishing bivalency. These genes also show enhanced spatial interactions. Both the strong bivalency and spatial clustering are virtually absent in preimplantation embryos and are markedly reduced in fate-committed lineages. Finally, we show that KMT2B is essential for establishing bivalent H3K4me3 at E6.5 but becomes partially dispensable later. Its deficiency leads to impaired activation of developmental genes and subsequent embryonic lethality. Thus, our data characterize lineage-specific chromatin reconfiguration and a unique chromatin state for primed pluripotency.

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: Global view of chromatin states during gastrulation in mouse embryos.
Fig. 2: Epigenetic regulation of lineage-restricted putative enhancers.
Fig. 3: Super bivalency identified in primed pluripotent Epi in vivo.
Fig. 4: Loss of super bivalent H3K4me3 is associated with aberrant activation of developmental genes.
Fig. 5: TET proteins promote super bivalent H3K4me3 in E6.5 Epi.
Fig. 6: Super bivalent genes show strong spatial clustering.
Fig. 7: Dynamic chromatin regulation of developmental genes from naïve pluripotency to committed lineages in vivo.

Similar content being viewed by others

Data availability

All data have been deposited to GEO with the accession number GSE125318. Source data for Extended Data Fig. 6 are available online.

Code availability

Software and code used to analyze these data are listed in the Nature Research Reporting Summary.

References

  1. Rossant, J. & Tam, P. P. Emerging asymmetry and embryonic patterning in early mouse development. Dev. Cell 7, 155–164 (2004).

    CAS  PubMed  Google Scholar 

  2. Zernicka-Goetz, M., Morris, S. A. & Bruce, A. W. Making a firm decision: multifaceted regulation of cell fate in the early mouse embryo. Nat. Rev. Genet. 10, 467–477 (2009).

    CAS  PubMed  Google Scholar 

  3. Arnold, S. J. & Robertson, E. J. Making a commitment: cell lineage allocation and axis patterning in the early mouse embryo. Nat. Rev. Mol. Cell Biol. 10, 91–103 (2009).

    CAS  PubMed  Google Scholar 

  4. Lawson, K. A., Meneses, J. J. & Pedersen, R. A. Clonal analysis of epiblast fate during germ layer formation in the mouse embryo. Development 113, 891–911 (1991).

    CAS  PubMed  Google Scholar 

  5. Bielinska, M., Narita, N. & Wilson, D. B. Distinct roles for visceral endoderm during embryonic mouse development. Int. J. Dev. Biol. 43, 183–205 (1999).

    CAS  PubMed  Google Scholar 

  6. Nichols, J. & Smith, A. Naive and primed pluripotent states. Cell Stem Cell 4, 487–492 (2009).

    CAS  PubMed  Google Scholar 

  7. Marks, H. & Stunnenberg, H. G. Transcription regulation and chromatin structure in the pluripotent ground state. Biochim. Biophys. Acta 1839, 129–137 (2014).

    CAS  PubMed  Google Scholar 

  8. 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).

    CAS  PubMed  Google Scholar 

  9. 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).

    CAS  PubMed  Google Scholar 

  10. Joshi, O. et al. Dynamic reorganization of extremely long-range promoter–promoter interactions between two states of pluripotency. Cell Stem Cell 17, 748–757 (2015).

    CAS  PubMed  Google Scholar 

  11. Kouzarides, T. Chromatin modifications and their function. Cell 128, 693–705 (2007).

    CAS  PubMed  Google Scholar 

  12. Vermeulen, M. & Timmers, H. T. Grasping trimethylation of histone H3 at lysine 4. Epigenomics 2, 395–406 (2010).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  15. Margueron, R. & Reinberg, D. The Polycomb complex PRC2 and its mark in life. Nature 469, 343–349 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  23. Glaser, S. et al. The histone 3 lysine 4 methyltransferase, Mll2, is only required briefly in development and spermatogenesis. Epigenetics Chromatin 2, 5 (2009).

    PubMed  PubMed Central  Google Scholar 

  24. Sze, C. C. et al. Histone H3K4 methylation-dependent and -independent functions of Set1A/COMPASS in embryonic stem cell self-renewal and differentiation. Genes Dev. 31, 1732–1737 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  33. Du, Z. et al. Allelic reprogramming of 3D chromatin architecture during early mammalian development. Nature 547, 232–235 (2017).

    CAS  PubMed  Google Scholar 

  34. Zhang, Y. et al. Dynamic epigenomic landscapes during early lineage specification in mouse embryos. Nat. Genet. 50, 96–105 (2018).

    CAS  PubMed  Google Scholar 

  35. Ren, B. & Yue, F. Transcriptional enhancers: Bridging the genome and phenome. Cold Spring Harb. Symp. Quant. Biol. 80, 17–26 (2015).

    PubMed  Google Scholar 

  36. McLean, C. Y. et al. GREAT improves functional interpretation of cis-regulatory regions. Nat. Biotechnol. 28, 495–501 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Ang, S. L. & Rossant, J. Anterior mesendoderm induces mouse Engrailed genes in explant cultures. Development 118, 139–149 (1993).

    CAS  PubMed  Google Scholar 

  38. Burdsal, C. A., Flannery, M. L. & Pedersen, R. A. FGF-2 alters the fate of mouse epiblast from ectoderm to mesoderm in vitro. Dev. Biol. 198, 231–244 (1998).

    CAS  PubMed  Google Scholar 

  39. Argelaguet, R. et al. Multi-omics profiling of mouse gastrulation at single-cell resolution. Nature https://doi.org/10.1038/s41586-019-1825-8 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Yang, X. et al. Distinct enhancer signatures in the mouse gastrula delineate progressive cell fate continuum during embryo development. Cell Res. 29, 911–926 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Scholer, H. R., Dressler, G. R., Balling, R., Rohdewohld, H. & Gruss, P. Oct-4: a germline-specific transcription factor mapping to the mouse t-complex. EMBO J. 9, 2185–2195 (1990).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Ang, S. L. et al. The formation and maintenance of the definitive endoderm lineage in the mouse: involvement of HNF3/forkhead proteins. Development 119, 1301–1315 (1993).

    CAS  PubMed  Google Scholar 

  44. Clevers, H. Wnt/beta-catenin signaling in development and disease. Cell 127, 469–480 (2006).

    CAS  PubMed  Google Scholar 

  45. Cheung, M. & Briscoe, J. Neural crest development is regulated by the transcription factor Sox9. Development 130, 5681–5693 (2003).

    CAS  PubMed  Google Scholar 

  46. Azcoitia, V., Aracil, M., Martinez, A. C. & Torres, M. The homeodomain protein Meis1 is essential for definitive hematopoiesis and vascular patterning in the mouse embryo. Dev. Biol. 280, 307–320 (2005).

    CAS  PubMed  Google Scholar 

  47. Shen, Y. et al. A map of the cis-regulatory sequences in the mouse genome. Nature 488, 116–120 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Hon, G. C. et al. Epigenetic memory at embryonic enhancers identified in DNA methylation maps from adult mouse tissues. Nat. Genet. 45, 1198–1206 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Brind’Amour, J. et al. An ultra-low-input native ChIP-seq protocol for genome-wide profiling of rare cell populations. Nat. Commun. 6, 6033 (2015).

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

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Jeong, M. et al. Large conserved domains of low DNA methylation maintained by Dnmt3a. Nat. Genet. 46, 17–23 (2014).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Cheutin, T. & Cavalli, G. Polycomb silencing: from linear chromatin domains to 3D chromosome folding. Curr. Opin. Genet. Dev. 25, 30–37 (2014).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Tropepe, V. et al. Direct neural fate specification from embryonic stem cells: a primitive mammalian neural stem cell stage acquired through a default mechanism. Neuron 30, 65–78 (2001).

    CAS  PubMed  Google Scholar 

  57. Darrow, E. M. et al. Deletion of DXZ4 on the human inactive X chromosome alters higher-order genome architecture. Proc. Natl Acad. Sci. USA 113, E4504–E4512 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Delbarre, E. et al. PML protein organizes heterochromatin domains where it regulates histone H3.3 deposition by ATRX/DAXX. Genome Res. 27, 913–921 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Harrison, S. M., Dunwoodie, S. L., Arkell, R. M., Lehrach, H. & Beddington, R. S. Isolation of novel tissue-specific genes from cDNA libraries representing the individual tissue constituents of the gastrulating mouse embryo. Development 121, 2479–2489 (1995).

    CAS  PubMed  Google Scholar 

  60. Nagy, A., Gertsenstein, M., Vintersten, K. & Behringer, R. Separating postimplantation germ layers. CSH Protoc. 2006, https://doi.org/10.1101/pdb.prot4368 (2006).

    PubMed  Google Scholar 

  61. Wang, W., Zhang, Y. & Wang, H. Generating mouse models using zygote electroporation of nucleases (ZEN) Technology with high efficiency and throughput. Methods Mol. Biol. 1605, 219–230 (2017).

    CAS  PubMed  Google Scholar 

  62. Peng, X. et al. TELP, a sensitive and versatile library construction method for next-generation sequencing. Nucleic Acids Res. 43, e35 (2015).

    PubMed  Google Scholar 

  63. Picelli, S. et al. Full-length RNA-seq from single cells using Smart-seq2. Nat. Protoc. 9, 171–181 (2014).

    CAS  PubMed  Google Scholar 

  64. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Trapnell, C. et al. Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nat. Protoc. 7, 562–578 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Guo, W. et al. BS-Seeker2: a versatile aligning pipeline for bisulfite sequencing data. BMC Genomics 14, 774 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Durand, N. C. et al. Juicer provides a one-click system for analyzing loop-resolution hi-C experiments. Cell Syst. 3, 95–98 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

  69. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Dennis, G. Jr. et al. DAVID: Database for annotation, visualization, and integrated discovery. Genome Biol. 4, P3 (2003).

    PubMed  Google Scholar 

  72. Durand, N. C. et al. Juicebox provides a visualization system for hi-C contact maps with unlimited zoom. Cell Systems 3, 99–101 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Yang, X. et al. Silencing of developmental genes by H3K27me3 and DNA methylation reflects the discrepant plasticity of embryonic and extraembryonic lineages. Cell Res. 28, 593–596 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank members of the Xie laboratory for comments during the preparation of the manuscript. We appreciate the support from the animal core facility, the sequencing core facility and biocomputing facility at Tsinghua University. This work was funded by the Beijing Municipal Science & Technology Commission (Z181100001318006 to W.X.), National Natural Science Foundation of China (31422031 and 31725018 to W.X.), Beijing Advanced Innovation Center for Structural Biology (100300001 to W.X.), the National Basic Research Program of China (2015CB856201 to W.X.) and the THU-PKU Center for Life Sciences (W.X.). W.X. is a recipient of an HHMI International Research Scholar.

Author information

Authors and Affiliations

Authors

Contributions

Y.X., Y.Z. and W.X. conceived and designed the project. Y.X. dissected early lineages and performed STAR ChIP–seq and RNA-seq library construction. Y.Z. and Y.X. analyzed NGS data. Y.Z. performed STEM-seq experiments. Q.X. collected early embryos, performed in vitro transcription of sgRNAs and electroporation experiments. C.Z. designed and established the Kmt2b zygotic knockout system. B.L. conducted miniATAC-seq. B.Z. conducted STAR ChIP–seq. K.Z. performed sisHi-C library construction of Kmt2b−/− embryos and Z.D. generated sisHi-C libraries of EpiLC. S.G. prepared EpiLC supervised by S.K. Y.W., X.W. and L. Li helped with the preparation of the manuscript. L. Liu performed the genotyping of Kmt2b KO mice. Y.L. and Q.W. performed NGS sequencing. W.X. supervised the project and related experiments. Y.Z, Y.X. and W.X. prepared the manuscript and figures with help from all the authors.

Corresponding author

Correspondence to Wei Xie.

Ethics declarations

Competing interests

The authors declare no competing interests.

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 Global view of histone modifications and chromatin accessibility in mouse early lineages.

a, Snapshots showing two replicates of H3K4me3, H3K27ac, H3K27me3, and ATAC-seq in E6.5Epi, E6.5VE, Ect, PS, Mes, and End at E7.5. mESC data from ENCODE47 are also shown for comparison. The genome browser view scales were adjusted based on the global data range. b, The Pearson correlation between two replicates for H3K4me3, H3K27me3, H3K27ac, and ATAC-seq in post-implantation lineages. The correlation between our dataset and a published dataset73 for H3K27me3 in E6.5 epiblast is also shown.

Extended Data Fig. 2 Global view of histone modifications, chromatin accessibility, and dynamics of putative enhancers during mouse early lineage specification.

a, Hierarchical clustering of two replicates for histone modifications and ATAC-seq data at promoters among post-implantation tissues. b, The heatmap showing the Spearman correlation between gene expression and enrichment of histone modifications and open chromatin at promoters in the post-implantation embryos. c, Heatmaps showing gene expression and related histone modifications/open chromatin enrichment at promoters for lineage-specific genes. d, Heatmaps showing tissue-specific enhancers from mesoderm to heart (left), and endoderm to liver (right). GO terms enriched for each tissue-specific enhancers are also listed. Somatic enhancers were obtained from a previous study47. e, Boxplots showing the dynamics of DNA methylation for tissue-specific enhancers34,47 during development. The median of each dataset is shown by the center line. The bottom, top edges and whiskers represent the twenty-fifth and seventy-fifth percentiles, and 1.5 times the interquartile range (IQR), respectively.

Extended Data Fig. 3 Bivalency establishment in early embryo.

a, Snapshots showing H3K4me3 and H3K27me3 enrichment at developmental genes Hoxd cluster and Gata2 in various cell types (n=2). Gene expression is also shown (log2 transformed FPKM). mESC and somatic tissue data from ENCODE47 are shown. Published data in 8-cell embryos and ICM28,29 are also shown for comparison. The genome browser view scales were adjusted based on the global data range. b, Average plots showing H3K4me3 and H3K27me3 enrichment at the Hox genes, bivalent genes inactive at all stages examined (8-cell, ICM, E5.5Epi, E6.5Epi, E6.5VE, and mESC), and housekeeping genes. The H3K4me3 enrichment is normalized against H3K4me3 signals at housekeeping (HK) gene promoters for each lineage. Arrows indicate E6.5 Epi. c, Average plots showing H3K4me3 and H3K27me3 enrichment at inactive bivalent genes (inactive at E6.5Epi, Ect, PS, Mes, and End), and housekeeping genes. The H3K4me3 enrichment is normalized against H3K4me3 signals at housekeeping (HK) gene promoters for each lineage. Arrows indicate E6.5 Epi.

Extended Data Fig. 4 Bivalency states in E6.5Epi and E6.5VE.

a, The UCSC genome browser views showing the enrichment of H3K4me3 (n=2), H3K27me3 (n=2) and H3 (n=1) at developmental genes Hoxa cluster, Pax6, as well as a housekeeping gene Rpn1 in mouse E6.5Epi and E6.5VE. b, Average plots showing H3K4me3 (n=2), H3K27me3 (n=2) and H3 (n=1) enrichment at the Hox gene cluster, inactive bivalent genes (inactive in E6.5Epi and E6.5VE), and housekeeping genes in E6.5Epi and E6.5VE. c, The scatter plot showing the enrichment of H3K4me3 (ULI-NChIP–seq), either done in this study (left) or in a previous study50 (right), and H3K27me3 (STAR ChIP–seq) of all bivalent genes (n=3,992) for E6.5 epiblast. The average H3K4me3 enrichment of housekeeping gene (HK.ave) is shown for each tissue. The number of super bivalent genes (top right) and Pearson correlation of H3K4me3 and H3K27me3 for each tissue (bottom right) are shown. d, The Venn diagram shows the overlap of super bivalent genes identified by ULI-NChIP-seq (Uli) and STAR ChIP-seq in E6.5 epiblast. The P-values was calculated by Fisher’s test.

Extended Data Fig. 5 Super bivalency marks primed pluripotent state in early lineages and somatic tissues.

a, Snapshots comparing the enrichment of H3K4me3, H3K27me3, and H3K27ac in E6.5 epiblast and somatic tissues from ENCODE47 at developmental genes Neurod1, Pcp4l1, and Foxa1. The heatmaps showing related gene expression levels. The genome browser view scales were adjusted based on the global data range. b, Left, the boxplot showing the expression levels of super bivalent genes and housekeeping genes in E6.5Epi34 and cortex47. The median of each dataset is shown by the center line. The bottom, top edges and whiskers represent the twenty-fifth and seventy-fifth percentiles, and 1.5 times the interquartile range (IQR), respectively. Two-sided P-values calculated by t-test are also shown. Super bivalent genes are identified in E6.5Epi (inactive). C-active, E6.5Epi super bivalent genes that become active in cortex (FPKM > 5); C-inactive, E6.5Epi super bivalent genes that remain inactive in cortex (FPKM < 2). Right, average plots showing the enrichment of H3K4me3 around the active and inactive super bivalent genes and housekeeping genes in E6.5Epi and cortex. c, A similar analysis as b in E6.5Epi and heart. H-active, E6.5Epi super bivalent genes that become active in heart (FPKM > 5); H-inactive, E6.5Epi super bivalent genes that remain inactive in heart (FPKM < 2).

Extended Data Fig. 6 Loss of super bivalent H3K4me3 is associated with aberrant developmental gene activation.

a, Gene expression of Kmt2b in pre- and post-implantation embryos is shown as bar graphs using previously published datasets28,34. b, Left, a schematic showing Kmt2b knockout strategy using Cas9/CRISPR as previously described61. Inner and outer primers for genotyping are shown. Right, the genotyping results of identified wild-type and Kmt2b-/- embryos using extra-embryonic tissues at E6.5. One representative image from three independent experiments is shown. Uncropped gel is also shown in Source data. M, DNA ladder; +/+, wild-type; +/-, heterozygote; -/-, homozygote. c, Average plots showing H3K4me3 enrichment at super bivalent/non-super bivalent/housekeeping genes (defined in E6.5 epiblast) for wild-type (n=2) and Kmt2b-/- E6.5Epi (n=3) and E8.5 head (n=2). The H3K4me3 enrichment is normalized against H3K4me3 signals at housekeeping (HK) gene promoters for each lineage. d, The morphology of wild-type and Kmt2b KO embryos from E6.5 to E9.5. Three independent experiments were performed. e, Hierarchical clustering based on gene expression in wild-type and Kmt2b KO embryos from E6.5 to E9.5. Arrow indicates E9.5 Kmt2b-/- embryo.

Source data

Extended Data Fig. 7 WT vs. Tet1/2 DKO embryos in E6.5 epiblasts.

a, Bar chart showing the expression levels of Kmt2b in wild-type (n=3) and Tet1/2 DKO (n=2) E6.5 epiblasts. The error bar represents the S.D with the barplot showing the the mean value. b, The morphology of wild type and Tet1/2 DKO embryos at E8.75. Two independent experiments were performed. c, Hierarchical clustering based on gene expression in wild-type and Tet1/2 DKO embryos from E6.5 to E8.5.

Extended Data Fig. 8 Spatial interactions of super bivalent genes in E6.5 epiblast.

a, The snapshots showing the ‘Virtual 4C’ (converted from Hi-C datasets) among bivalent genes in 8-cell, ICM, E6.5Epi, E6.5VE, mESC, and fibroblast. The enrichment of H3K4me3 and H3K27me3 in E6.5Epi (n=2) and E6.5VE (n=2) are shown below. Magnified views of interactions between bivalent genes are also shown. b, Heatmaps showing the inter-chromosomal interactions between chromosomes 2 and 11. Boxes show the zoomed-in views of interactions between Dlx, Hoxd cluster (chr2) and Hoxb clusters (chr11). A UCSC genome browser of H3K27me3 enrichment in wild-type E6.5Epi is shown on the top to indicate the positions of Polycomb targets.

Extended Data Fig. 9 Super bivalent genes show strong spatial clustering.

a, A Venn diagram shows the overlap between ELRI genes10 (n=108), top 100 Polycomb nucleation genes55, and super bivalent genes identified in E6.5Epi (see Methods). The P-values showing the overlap among pairwise comparison were calculated by Fisher test. b, Boxplots showing the normalized interaction frequencies among different gene groups in 8-cell, ICM, E6.5Epi, E6.5VE, mESC, EpiSC, EpiLC and fibroblast for ELRI bivalent genes(top) and non-ELRI bivalent genes (bottom). Super, super bivalent genes; Non-super, non-super bivalent genes; HK, housekeeping genes; Inactive, non-bivalent inactive genes (Methods). The median of each dataset is shown by the center line. The bottom, top edges and whiskers represent the twenty-fifth and seventy-fifth percentiles, and 1.5 times the interquartile range (IQR), respectively. Two-sided P-values calculated by t-test are also shown.

Extended Data Fig. 10 Strong spatial clustering of developmental genes in E6.5 epiblast.

a, Boxplots showing the normalized interaction frequencies among genes in each gene group in wild-type (n = 5) and Kmt2b-/- (n=3) E6.5 Epi. P-values calculated by two-sided t-test are also shown. b, Left, a schematic diagram shows the relative spatial position of a selected gene defined by its interactions with developmental genes divided by its interactions with housekeeping genes. Right, boxplots showing the log ratios of such bivalent/housekeeping gene interactions for different gene groups (inactive, active developmental genes, housekeeping genes) in E6.5 epiblast.

Supplementary information

Reporting Summary

Supplementary Tables

Supplementary Tables 1–4

Source data

Source Data Extended Data Fig. 6

Original uncropped gel of Extended Data Figure 6b. Boxes indicate the cropped regions

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xiang, Y., Zhang, Y., Xu, Q. et al. Epigenomic analysis of gastrulation identifies a unique chromatin state for primed pluripotency. Nat Genet 52, 95–105 (2020). https://doi.org/10.1038/s41588-019-0545-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-019-0545-1

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