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.

  • Resource
  • Published:

Reprogramming of H3K9me3-dependent heterochromatin during mammalian embryo development

Abstract

H3K9me3-dependent heterochromatin is a major barrier of cell fate changes that must be reprogrammed after fertilization. However, the molecular details of these events are lacking in early embryos. Here, we map the genome-wide distribution of H3K9me3 modifications in mouse early embryos. We find that H3K9me3 exhibits distinct dynamic features in promoters and long terminal repeats (LTRs). Both parental genomes undergo large-scale H3K9me3 reestablishment after fertilization, and the imbalance in parental H3K9me3 signals lasts until blastocyst. The rebuilding of H3K9me3 on LTRs is involved in silencing their active transcription triggered by DNA demethylation. We identify that Chaf1a is essential for the establishment of H3K9me3 on LTRs and subsequent transcriptional repression. Finally, we find that lineage-specific H3K9me3 is established in post-implantation embryos. In summary, our data demonstrate that H3K9me3-dependent heterochromatin undergoes dramatic reprogramming during early embryonic development and provide valuable resources for further exploration of the epigenetic mechanism in early embryos.

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: Genome-wide profiling of H3K9me3 in mouse gametes and early embryos.
Fig. 2: Dynamics of H3K9me3-dependent heterochromatin in mouse early embryos.
Fig. 3: Reprogramming of maternal and paternal H3K9me3 during fertilization.
Fig. 4: Landscape of allelic-specific H3K9me3 during early embryonic development.
Fig. 5: Epigenetic switching of DNA methylation with H3K9me3 on LTR regions.
Fig. 6: Chaf1a is involved in LTR silencing by H3K9me3 and is crucial for normal embryonic development.
Fig. 7: Lineage-specific H3K9me3 is established at promoter regions after implantation.

Similar content being viewed by others

References

  1. Hackett, J. A. & Surani, M. A. Beyond DNA: programming and inheritance of parental methylomes. Cell 153, 737–739 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Li, E. Chromatin modification and epigenetic reprogramming in mammalian development. Nat. Rev. Genet. 3, 662–673 (2002).

    Article  CAS  PubMed  Google Scholar 

  3. Burton, A. & Torres-Padilla, M. E. Epigenetic reprogramming and development: a unique heterochromatin organization in the preimplantation mouse embryo. Brief. Funct. Genomics 9, 444–454 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Okamoto, I., Otte, A. P., Allis, C. D., Reinberg, D. & Heard, E. Epigenetic dynamics of imprinted X inactivation during early mouse development. Science 303, 644–649 (2004).

    Article  CAS  PubMed  Google Scholar 

  5. Feldman, N. et al. G9a-mediated irreversible epigenetic inactivation of Oct-3/4 during early embryogenesis. Nat. Cell Biol. 8, 188–194 (2006).

    Article  CAS  PubMed  Google Scholar 

  6. Fukuda, A. et al. The role of maternal-specific H3K9me3 modification in establishing imprinted X-chromosome inactivation and embryogenesis in mice. Nat. Commun. 5, 5464 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Tachibana, M. et al. G9a histone methyltransferase plays a dominant role in euchromatic histone H3 lysine 9 methylation and is essential for early embryogenesis. Genes Dev. 16, 1779–1791 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Messerschmidt, D. M. et al. Trim28 is required for epigenetic stability during mouse oocyte to embryo transition. Science 335, 1499–1502 (2012).

    Article  CAS  PubMed  Google Scholar 

  9. Liu, H., Kim, J. M. & Aoki, F. Regulation of histone H3 lysine 9 methylation in oocytes and early pre-implantation embryos. Development 131, 2269–2280 (2004).

    Article  CAS  PubMed  Google Scholar 

  10. Santos, F., Peters, A. H., Otte, A. P., Reik, W. & Dean, W. Dynamic chromatin modifications characterise the first cell cycle in mouse embryos. Dev. Biol. 280, 225–236 (2005).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Wang, L. et al. Programming and inheritance of parental DNA methylomes in mammals. Cell 157, 979–991 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  15. Kigami, D., Minami, N., Takayama, H. & Imai, H. MuERV-L is one of the earliest transcribed genes in mouse one-cell embryos. Biol. Reprod. 68, 651–654 (2003).

    Article  CAS  PubMed  Google Scholar 

  16. Leung, D. C. & Lorincz, M. C. Silencing of endogenous retroviruses: when and why do histone marks predominate? Trends Biochem. Sci. 37, 127–133 (2012).

    Article  CAS  PubMed  Google Scholar 

  17. Bulut-Karslioglu, A. et al. Suv39h-dependent H3K9me3 marks intact retrotransposons and silences LINE elements in mouse embryonic stem cells. Mol. Cell 55, 277–290 (2014).

    Article  CAS  PubMed  Google Scholar 

  18. Yang, B. X. et al. Systematic identification of factors for provirus silencing in embryonic stem cells. Cell 163, 230–245 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. 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).

    Article  PubMed  Google Scholar 

  20. 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 

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

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Becker, J. S., Nicetto, D. & Zaret, K. S. H3K9me3-dependent heterochromatin: barrier to cell fate changes. Trends Genet. 32, 29–41 (2016).

    Article  CAS  PubMed  Google Scholar 

  25. Surface, L. E., Thornton, S. R. & Boyer, L. A. Polycomb group proteins set the stage for early lineage commitment. Cell Stem Cell 7, 288–298 (2010).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  27. 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 

  28. Reik, W. & Walter, J. Genomic imprinting: parental influence on the genome. Nat. Rev. Genet. 2, 21–32 (2001).

    Article  CAS  PubMed  Google Scholar 

  29. 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 

  30. Lewis, A. et al. Imprinting on distal chromosome 7 in the placenta involves repressive histone methylation independent of DNA methylation. Nat. Genet. 36, 1291–1295 (2004).

    Article  CAS  PubMed  Google Scholar 

  31. Umlauf, D. et al. Imprinting along the Kcnq1 domain on mouse chromosome 7 involves repressive histone methylation and recruitment of Polycomb group complexes. Nat. Genet. 36, 1296–1300 (2004).

    Article  CAS  PubMed  Google Scholar 

  32. Choi, Y. J. et al. Deficiency of microRNA miR-34a expands cell fate potential in pluripotent stem cells. Science 355, eaag1927 (2017).

    Article  PubMed  Google Scholar 

  33. Fort, A. et al. Deep transcriptome profiling of mammalian stem cells supports a regulatory role for retrotransposons in pluripotency maintenance. Nat. Genet. 46, 558–566 (2014).

    Article  CAS  PubMed  Google Scholar 

  34. Nakamura, T. et al. PGC7 binds histone H3K9me2 to protect against conversion of 5mC to 5hmC in early embryos. Nature 486, 415–419 (2012).

    Article  CAS  PubMed  Google Scholar 

  35. Zhou, L. Q. & Dean, J. Reprogramming the genome to totipotency in mouse embryos. Trends Cell Biol. 25, 82–91 (2015).

    Article  CAS  PubMed  Google Scholar 

  36. Macfarlan, T. S. et al. Embryonic stem cell potency fluctuates with endogenous retrovirus activity. Nature 487, 57–63 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Hatanaka, Y. et al. Histone chaperone CAF-1 mediates repressive histone modifications to protect preimplantation mouse embryos from endogenous retrotransposons. Proc. Natl Acad. Sci. USA 112, 14641–14646 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Ishiuchi, T. et al. Early embryonic-like cells are induced by downregulating replication-dependent chromatin assembly. Nat. Struct. Mol. Biol. 22, 662–671 (2015).

    Article  CAS  PubMed  Google Scholar 

  39. Burton, A. & Torres-Padilla, M. E. Chromatin dynamics in the regulation of cell fate allocation during early embryogenesis. Nat. Rev. Mol. Cell Biol. 15, 723–734 (2014).

    Article  CAS  PubMed  Google Scholar 

  40. Albert, M. & Peters, A. H. Genetic and epigenetic control of early mouse development. Curr. Opin. Genet. Dev. 19, 113–121 (2009).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. 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 

  43. Adenot, P. G., Mercier, Y., Renard, J. P. & Thompson, E. M. Differential H4 acetylation of paternal and maternal chromatin precedes DNA replication and differential transcriptional activity in pronuclei of 1-cell mouse embryos. Development 124, 4615–4625 (1997).

    CAS  PubMed  Google Scholar 

  44. Kimura, Y. & Yanagimachi, R. Intracytoplasmic sperm injection in the mouse. Biol. Reprod. 52, 709–720 (1995).

    Article  CAS  PubMed  Google Scholar 

  45. Chatot, C. L., Ziomek, C. A., Bavister, B. D., Lewis, J. L. & Torres, I. An improved culture medium supports development of random-bred 1-cell mouse embryos in vitro. J. Reprod. Fertil. 86, 679–688 (1989).

    Article  CAS  PubMed  Google Scholar 

  46. Li, H. & Durbin, R. Fast and accurate long-read alignment with Burrows–Wheeler transform. Bioinformatics 26, 589–595 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Trapnell, C., Pachter, L. & Salzberg, S. L. TopHat: discovering splice junctions with RNA-seq. Bioinformatics 25, 1105–1111 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Trapnell, C. et al. Transcript assembly and quantification by RNA-seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat. Biotechnol. 28, 511–515 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Xi, Y. & Li, W. BSMAP: whole genome bisulfite sequence MAPping program. BMC Bioinformatics 10, 232 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Sun, D. et al. MOABS: model based analysis of bisulfite sequencing data. Genome Biol. 15, R38 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Huang, D. W., Sherman, B. T. & Lempicki, R. A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 4, 44–57 (2009).

    Article  CAS  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  53. 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 

  54. Angermueller, C. et al. Parallel single-cell sequencing links transcriptional and epigenetic heterogeneity. Nat. Methods 13, 229–232 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. 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 

  56. Johnson, W. E., Li, C. & Rabinovic, A. Adjusting batch effects in microarray expression data using empirical Bayes methods. Biostatistics 8, 118–127 (2007).

    Article  PubMed  Google Scholar 

  57. Mei, S. et al. Cistrome Data Browser: a data portal for ChIP-seq and chromatin accessibility data in human and mouse. Nucleic Acids Res. 45, 658–662 (2017).

    Article  Google Scholar 

Download references

Acknowledgements

This work was primarily supported by the National Key R&D Program of China (2016YFA0100400) and the National Natural Science Foundation of China (31721003). This work was also supported by the Ministry of Science and Technology of China (2017YFA0102602 and 2015CB964800), the National Natural Science Foundation of China (31430056, 81630035, 31401266, 31771646, 31701262, 31401247, 31501196, 31501183, 31571365 and 31501197), the Shanghai Subject Chief Scientist Program (15XD1503500 and 17XD1403600), the Shanghai Rising-Star Program (17QA1402700 and 17QA1404200), the Shanghai Chenguang Program (16CG17, 16CG19 and 15CG19), the Shanghai municipal medical and health discipline construction projects (2017ZZ02015) and the National Postdoctoral Program for Innovative Talents (BX20170174).

Author information

Authors and Affiliations

Authors

Contributions

Y.G. and S.G. conceived and designed the experiments. C.W. performed computational analysis. X.L. and L.Y. performed the ChIP experiments. C.W., X.L., Y.Z. and Y.G. designed and performed the data analysis. W.L., C.C., X.K., J.C., Y.H.Z., Y.W., R.L., H.W. and T.D. assisted with the sample preparation. C.W., X.L., Y.G., Y.Z. and S.G. wrote the manuscript.

Corresponding authors

Correspondence to Yawei Gao, Yong Zhang or Shaorong Gao.

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.

Integrated supplementary information

Supplementary Figure 1 Distinct establishment patterns of H3K9me3 on genes and LTRs in mouse early embryos.

a, Scatter plot shows the enrichment of H3K9me3 in union peaks for mESC samples of this study and ENCODE data. Pearson's correlation coefficient is also shown. b, Barplot shows the Pearson's correlation coefficients between the replicates of embryo H3K9me3 samples in union peaks. c, PCA analysis of H3K9me3, H3K4me3 and H3K27me3 signals in union peaks (n = 45 biologically independent samples). d, Barplots show the Pearson's correlation coefficients between H3K9me3 and other epigenetic modifications in promoter regions. e, The UCSC genome browser view of H3K9me3 and H3K27me3 signal around Hoxa Clusters of mouse gametes and early embryos. Signals represent log2-transformed ChIP / input ratio. f, Graphs show the averaged expression level of H3K9me3 writers, including Setdb1, Suv39h1, Suv39h2 and Urf1. g, Graphs show the averaged expression level of H3K9me3 erasers, including Kdm4a, Kdm4b, Kdm4c and Kdm4d. In f and g, RNA-seq has been performed 2 times for MII oocyte, morula, E5.5 Epi, E6.5 Epi and E6.5 Exe, 3 times for 8-cell and 4 times for 2-cell, 4-cell, ICM and TE. Results for each experiment are plotted as individual points with the average value indicated by the black line.

Supplementary Figure 2 Dynamics of H3K9me3 during mouse early embryo development.

a, Boxplots show the H3K9me3, H3K27me3 and DNA methylation level of Oocyte specific, Cleavage specific and Blastocyst specific H3K9me3 domains. Statistical significance between stages was evaluated based on one-sided Wilcoxon test, * represent p-value < 0.05, *** represent p-value < 0.001. Oocyte specific (n = 38996 H3K9me3 domains), Cleavage specific (n = 114033 H3K9me3 domains), Blastocyst specific (n = 119837 H3K9me3 domains). The center represents the median value and the upper/lower line represent 5% and 95% quantile for the boxplots. b, Heatmaps show the dynamic of H3K9me3, H3K27me3 and DNA methylation level in blastocysts and post-implanted embryos. The heatmaps were generated using the same order of pre-implantation H3K9me3 heatmap. Colors represent log2-transformed ChIP / input ratio scaled by row, or absolute DNA methylation level. ChIP-seq for H3K9me3 has been performed twice for each sample and data shown represent averages for these independent experiments. H3K27me3 ChIP seq data for zygote was performed three times and data for pre-implantation embryos were from our previous publication (GSE73952). WGBS has been performed once for each sample.

Supplementary Figure 3 Landscape of allelic specific H3K9me3 during early embryo development.

a, Heatmap shows the Pearson's correlation coefficients of BDF1 H3K9me3 samples and C57BL/DBA H3K9me3 samples. b, The UCSC genome browser view of allelic H3K9me3, H3K4me3, H3K27me3 and DNA methylation level near gene Etv6. Signals represent ChIP-seq RPM for histone modifications and absolute level for DNA methylation. c, Boxplots show the allelic H3K9me3 signal of different clusters in Fig. 4c. n = number of H3K9me3 domains between different groups. The center represents the median value and the upper/lower line represent 5% and 95% quantile for the boxplots. d, Bar plot show the fraction of maternally-expressed (left) and paternally-expressed (right) imprinted genes that regulated by allelic specific H3K9me3, H3K27me3 and DNA methylation. ChIP-seq for H3K9me3 has been performed 2 time for each stage except late 2-cell (3 times) and data shown represent average values. WGBS for each sample was performed once. e, Pie chart show the overlap of maternal specific H3K9me3, H3K27me3 and DNA methylation regulated imprinted genes at ICM stage. H3K27me3 ChIP-seq data for MII oocyte, sperm, late 2cell and E3.5 ICM were from public data (GSE76687).

Supplementary Figure 4 Epigenetic switch of DNA methylation with H3K9me3 on LTR regions.

a, Scatterplots show the association tests between log2-transformed H3K27me3 / input ratio and DNA methylation level during pre-implantation embryos on specific LTR families. Different colors represent four major family of the LTRs, including ERVK, ERV1, MaLR and ERVL (two-sided association test, n = 7 biologically independent samples. The x-axis represents the Pearson's correlation coefficients and the y-axis represents P-value of the tests. The blue horizontal line in each plot correspond p-value of 0.05 significance threshold. The total number of significant positive (+) and negative (−) correlations (P-value < 0.05) for each annotation is shown at the top of each plot. b, Boxplots show the log2-transformed H3K9me3 / input ratio, log2-transformed H3K27me3 / input ratio, DNA methylation, ATAC signal and expression level of the 28 H3K27me3 increased LTR during pre-implantation development. Statistical significance between stages was evaluated based on one-sided Wilcoxon test, * represent p-value < 0.05, *** represent p-value < 0.001. n = 28 different LTRs. The center represents the median value and the upper/lower line represent 5% and 95% quantile for the boxplots. c, Scatterplots show the association tests between log2-transformed H3K27me3 / input ratio and DNA methylation level during pre-implantation embryos on specific LTR families (two-sided association test, n = 7 biologically independent samples). Different colors represent four major family of the LTRs, including ERVK, ERV1, MaLR and ERVL. The total number of significant positive (+) and negative (−) correlations (P-value < 0.05) for each family is shown at the top of each plot. d, Graphs show the averaged expression level of Chaf1a, Sumo2, Trim28 and Zfp809. RNA-seq has been performed 2 times for MII oocyte, morula, 3 times for 8-cell and 4 times for 2-cell, 4-cellICM and TE. Results for each experiment are plotted as individual points with the average value indicated by the black line.

Supplementary Figure 5 Chaf1a is involved in LTR silencing by H3K9me3 and is crucial for normal embryo development.

a, Gene expression levels were reduced by corresponding siRNA injection into embryos. RT-qPCR analysis was performed 48 h after injection. The relative expression levels of corresponding genes relative to Gapdh were compared with control embryos. Data shown represent the mean (indicated by the line in the figure) of 2–4 independent experiments: in the order of control siRNA and specific siRNA: Chaf1a (n = 2, 2), Sumo2 (n = 2, 2), Setdb1 (n = 4, 4), Trim28 (n = 2, 4), Ube2i (n = 4, 4) Zfp809 (n = 2, 2). “n” represents number of samples repeats in RT-qPCR. b, Heatmap shows the Pearson's correlation coefficients of replicates for control and siRNA knockdown H3K9me3 samples. c, Scatterplots show the comparison of control and siRNA knockdown H3K9me3 samples. The x-axis stands for log2-transformed H3K9me3 / input ratio on LTRs for control samples (log2), and the y-axis stands for log2-transformed H3K9me3 / input ratio on LTRs for siRNA knockdown experiments. The diagonal dashed line means LTRs with significant H3K9me3 change (log2-transformed absolute fold change > 0.25). d, Scatterplots show the comparison of control and siRNA knockdown expression samples. The x-axis stands for normalized RNA-seq reads count on LTRs for control samples (log2), and the y-axis stands for normalized RNA-seq reads count on LTRs for siRNA knockdown experiments. The diagonal dashed line means LTRs with significant expression level change (log2-transformed absolute fold change > 1). e, Boxplots show the H3K9me3 and expression level of all LTRs (left two panel), 50 H3K9me3 increased LTRs (middle two panel) and 8 H3K9me3 decreased LTRs (right two panel). The center represents the median value and the upper/lower line represent 5% and 95% quantile for the boxplots. All LTRs (n = 471 LTRs), Increased LTRs (n = 50 LTRs), Decreased LTRs (n = 8 LTRs).

Supplementary Figure 6 Lineage specific H3K9me3 is established at promoter regions after implantation.

a, Boxplots show the expression level of E6.5 Epi specific H3K9me3 marked genes (left panel) and E6.5 Exe specific H3K9me3 marked genes (right panel). Statistical significance between groups was evaluated based on one-sided Wilcoxon test, * represent p-value < 0.05,*** represent p-value < 0.001, n = number of H3K9me3 domains between groups. The center represents the median value and the upper/lower line represent 5% and 95% quantile for the boxplots. b, Scatterplots showing the preference of epigenetic factor binding in Epi/Exe-specific H3K9me3 domains based on mESC ChIP-seq peaks. The x-axis represents the P-value of Fisher's exact tests, and the y-axis represents the fold-change of binding sites in Epi/Exe-specific H3K9me3 peaks. Factors with significant preferences are labeled on the graph. c, Boxplots show the H3K9me3 and H3K27me3 signal of E7.5 newly established Epi/Exe specific H3K9me3 marked genes. Statistical significance between groups was evaluated based on one-sided Wilcoxon test, * represent p-value < 0.05,*** represent p-value < 0.001, n = number of H3K9me3 domains between groups. The center represents the median value and the upper/lower line represent 5% and 95% quantile for the boxplots. d, Graphs show the expression level of identified lineage specific TFs in E6.5 Epi and Exe. Color represent FPKM.

Supplementary information

Supplementary Information

Supplementary Figures 1–6 and legends for Supplementary Tables 1–3

Reporting Summary

Supplementary Table 1

Supplementary Table 2

Supplementary Table 3

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, C., Liu, X., Gao, Y. et al. Reprogramming of H3K9me3-dependent heterochromatin during mammalian embryo development. Nat Cell Biol 20, 620–631 (2018). https://doi.org/10.1038/s41556-018-0093-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-018-0093-4

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