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.

  • Letter
  • Published:

Distinct features of H3K4me3 and H3K27me3 chromatin domains in pre-implantation embryos

Abstract

Histone modifications have critical roles in regulating the expression of developmental genes during embryo development in mammals1,2. However, genome-wide analyses of histone modifications in pre-implantation embryos have been impeded by the scarcity of the required materials. Here, by using a small-scale chromatin immunoprecipitation followed by sequencing (ChIP–seq) method3, we map the genome-wide profiles of histone H3 lysine 4 trimethylation (H3K4me3) and histone H3 lysine 27 trimethylation (H3K27me3), which are associated with gene activation and repression4,5, respectively, in mouse pre-implantation embryos. We find that the re-establishment of H3K4me3, especially on promoter regions, occurs much more rapidly than that of H3K27me3 following fertilization, which is consistent with the major wave of zygotic genome activation at the two-cell stage. Furthermore, H3K4me3 and H3K27me3 possess distinct features of sequence preference and dynamics in pre-implantation embryos. Although H3K4me3 modifications occur consistently at transcription start sites, the breadth of the H3K4me3 domain is a highly dynamic feature. Notably, the broad H3K4me3 domain (wider than 5 kb) is associated with higher transcription activity and cell identity not only in pre-implantation development but also in the process of deriving embryonic stem cells from the inner cell mass and trophoblast stem cells from the trophectoderm. Compared to embryonic stem cells, we found that the bivalency (that is, co-occurrence of H3K4me3 and H3K27me3) in early embryos is relatively infrequent and unstable. Taken together, our results provide a genome-wide map of H3K4me3 and H3K27me3 modifications in pre-implantation embryos, facilitating further exploration of the mechanism for epigenetic regulation 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

Figure 1: Dynamics of the histone modification landscape in early mouse embryos.
Figure 2: Establishment of H3K4me3 and H3K27me3 in early mouse embryos.
Figure 3: Broad H3K4me3 is associated with transcription regulation and cell identity.
Figure 4: Bivalent chromatin domains in the early mouse embryos.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Data deposits

The ULI-NChIP–seq dataset has been deposited in the Gene Expression Omnibus (GEO) under accession number GSE73952. The gene expression and DNA methylation data are deposited under accession number GSE70608.

References

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

    Article  CAS  PubMed  Google Scholar 

  2. Bogliotti, Y. S. & Ross, P. J. Mechanisms of histone H3 lysine 27 trimethylation remodeling during early mammalian development. Epigenetics 7, 976–981 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. 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  ADS  PubMed  CAS  Google Scholar 

  4. Schuettengruber, B., Chourrout, D., Vervoort, M., Leblanc, B. & Cavalli, G. Genome regulation by polycomb and trithorax proteins. Cell 128, 735–745 (2007)

    Article  CAS  PubMed  Google Scholar 

  5. Ringrose, L. & Paro, R. Epigenetic regulation of cellular memory by the Polycomb and Trithorax group proteins. Annu. Rev. Genet. 38, 413–443 (2004)

    Article  CAS  PubMed  Google Scholar 

  6. Erhardt, S. et al. Consequences of the depletion of zygotic and embryonic enhancer of zeste 2 during preimplantation mouse development. Development 130, 4235–4248 (2003)

    Article  CAS  PubMed  Google Scholar 

  7. 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 

  8. Zhang, M., Wang, F., Kou, Z., Zhang, Y. & Gao, S. Defective chromatin structure in somatic cell cloned mouse embryos. J. Biol. Chem. 284, 24981–24987 (2009)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Wang, H. & Dey, S. K. Roadmap to embryo implantation: clues from mouse models. Nat. Rev. Genet. 7, 185–199 (2006)

    Article  PubMed  CAS  Google Scholar 

  10. Chang, G. et al. High-throughput sequencing reveals the disruption of methylation of imprinted gene in induced pluripotent stem cells. Cell Res. 24, 293–306 (2014)

    Article  CAS  PubMed  Google Scholar 

  11. Soshnikova, N. & Duboule, D. Epigenetic temporal control of mouse Hox genes in vivo. Science 324, 1320–1323 (2009)

    Article  ADS  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Plath, K. et al. Role of histone H3 lysine 27 methylation in X inactivation. Science 300, 131–135 (2003)

    Article  ADS  CAS  PubMed  Google Scholar 

  14. Mak, W. et al. Mitotically stable association of polycomb group proteins Eed and Enx1 with the inactive X chromosome in trophoblast stem cells. Curr. Biol. 12, 1016–1020 (2002)

    Article  CAS  PubMed  Google Scholar 

  15. Thomson, J. P. et al. CpG islands influence chromatin structure via the CpG-binding protein Cfp1. Nature 464, 1082–1086 (2010)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  17. Brandeis, M. et al. Sp1 elements protect a CpG island from de novo methylation. Nature 371, 435–438 (1994)

    Article  ADS  CAS  PubMed  Google Scholar 

  18. Hashimshony, T., Zhang, J., Keshet, I., Bustin, M. & Cedar, H. The role of DNA methylation in setting up chromatin structure during development. Nat. Genet. 34, 187–192 (2003)

    Article  CAS  PubMed  Google Scholar 

  19. Lande-Diner, L. et al. Role of DNA methylation in stable gene repression. J. Biol. Chem. 282, 12194–12200 (2007)

    Article  CAS  PubMed  Google Scholar 

  20. Wang, L. et al. Programming and inheritance of parental DNA methylomes in mammals. Cell 157, 1735–1735 (2014)

    Article  CAS  Google Scholar 

  21. Benayoun, B. A. et al. H3K4me3 breadth is linked to cell identity and transcriptional consistency. Cell 158, 673–688 (2014)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Greer, E. L. & Shi, Y. Histone methylation: a dynamic mark in health, disease and inheritance. Nat. Rev. Genet. 13, 343–357 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Huang, J. et al. Impairment of preimplantation porcine embryo development by histone demethylase KDM5B knockdown through disturbance of bivalent H3K4me3-H3K27me3 modifications. Biol. Reprod. 92, 72 (2015)

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  27. Vastenhouw, N. L. et al. Chromatin signature of embryonic pluripotency is established during genome activation. Nature 464, 922–926 (2010)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  28. Sachs, M. et al. Bivalent chromatin marks developmental regulatory genes in the mouse embryonic germline in vivo. Cell Reports 3, 1777–1784 (2013)

    Article  CAS  PubMed  Google Scholar 

  29. Paranjpe, S. S. & Veenstra, G. J. Establishing pluripotency in early development. Biochim. Biophys. Acta 1849, 626–636 (2015)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Pérez-Lluch, S. et al. Absence of canonical marks of active chromatin in developmentally regulated genes. Nat. Genet. 47, 1158–1167 (2015)

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  32. 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 

  33. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows–Wheeler transform. Bioinformatics 25, 1754–1760 (2009)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. 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 

  36. 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 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  39. Weber, M. et al. Distribution, silencing potential and evolutionary impact of promoter DNA methylation in the human genome. Nat. Genet. 39, 457–466 (2007)

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  42. Cabili, M. N. et al. Integrative annotation of human large intergenic noncoding RNAs reveals global properties and specific subclasses. Genes Dev. 25, 1915–1927 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Barrett, T. et al. NCBI GEO: archive for functional genomics data sets–update. Nucleic Acids Res. 41, D991–D995 (2013)

    Article  CAS  PubMed  Google Scholar 

  44. Liu, W. et al. Identification of key factors conquering developmental arrest of somatic cell cloned embryos by combining embryo biopsy and single-cell sequencing. Cell Discovery 2, 16010 (2016)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors would like to thank S. Liu from the University of British Columbia for advice with the ULI-NChIP–seq experiments. This work was supported by the National Natural Science Foundation of China (31325019, 31430056, 91319306, 31401266, 31501196, 31401247, 31501197, 31322031, 31371288 and 31571365), the Ministry of Science and Technology of China (2015CB964503, 2016YFA0100400 and 2015CB964800), the Science and Technology Commission of Shanghai Municipality (14CG16 and 15XD1503500), and the Major Program of Development Fund for Zhangjiang National Innovation Demonstration Zone (ZJ2014-ZD-002).

Author information

Authors and Affiliations

Authors

Contributions

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

Corresponding authors

Correspondence to Yong Zhang, Yawei Gao or Shaorong Gao.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

Reviewer Information

Nature thanks R. Schultz and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Extended data figures and tables

Extended Data Figure 1 Ultra-low-input ChIP–seq in ES cells and pre-implantation embryos.

a, Sample correlation for the ChIP–seq experiments using 200, 1,000 and bulk mES cells. Each point represents the normalized reads count of a RefSeq promoter for corresponding samples. Pearson’s correlation coefficients of the two experiments were calculated using all the RefSeq promoters. b, Genome browser view of the H3K4me3 and H3K27me3 RPM (reads per million reads) at the Fgf4 and Fbln2 gene regions for the mES cells in duplicate. The ChIP–seq RPM (reads per million mapped reads) was smoothened by the mean of five pixels using the UCSC genome browser. c, Correlation of the H3K4me3 samples for embryo developmental stages and cell lines. Each point represents the normalized reads count of a RefSeq promoter for the replicates. Pearson’s correlation coefficients of the two replicates were calculated using all the RefSeq promoters. d, Correlation of the H3K27me3 samples for embryo developmental stages and cell lines. Each point represents the normalized reads count of a RefSeq promoter for the replicates. Pearson’s correlation coefficients of the two replicates were calculated using all the RefSeq promoters. e, f, Genome browser view of the H3K4me3 RPM at the Fgf4 (e) and the H3K27me3 RPM at the Fbln2 and Gata6 (f) gene regions for embryos and cell lines. The ChIP–seq RPM was smoothened by the mean of five pixels using the UCSC genome browser.

Extended Data Figure 2 Features of the chromatin states in pre-implantation embryos.

a. Number of high-enrichment and low-enrichment H3K4me3-only regions during embryogenesis. The high-enrichment H3K4me3-only domains were defined as segments retained using a stringent ChromHMM threshold (fourfold over input file) and the low-enrichment H3K4me3-only domains were the additional segments when the stringent threshold was not used. b, Emission parameters of the ChromHMM segmentations. Colour indicates the enrichment of chromatin modifications in each specific segmentation. The whole genome was classified into: 1, non-marked regions; 2, H3K4me3-only regions; 3, H3K27me3-only regions; and 4, bivalent regions. c, Genome distribution of the different classes of the ChromHMM segmentations across the mouse developmental stages and cell lines. The analysed regions are labelled to the left of the heat map, and segmentations are labelled at the top of the heat map. Colour represents relative percentage scaled by row. d, RNA expression level and relative protein level of the H3K4me3 and H3K27me3 writers. RNA-seq data and mass spectrometric data on early mouse embryos (unpublished) were used in this analysis. The expression level was evaluated using a log-scaled FPKM, and the protein level was evaluated using the relative ratio over all the stages. In this figure, H3K4me3 writers included Rbbp5, Dpy30, Ash2l, Wdr5 and Setd1a. H3K27me3 writers included Jarid2, Ezh2, Eed, Suz12 and Rnf2.

Extended Data Figure 3 Dynamics of the histone-modification landscape in embryogenesis and during differentiation.

a, b, Left panel, alluvial plots showing the global dynamics of the bivalent (a) and non-marked (b) regions during early embryo development. Each line represents a 200-bp bin defined based on the ChromHMM categories. For each panel, the total regions were those classified as this state in at least one analysed stage. Right panel, the percentage of the analysed states (that is, bivalent or non-marked) in each stage compared to the total regions. The global dynamics (solid line) and chromosome X dynamics (dashed line) are plotted separately. c, Averaged distance to the nearest TSS of the stable and dynamic H3K4me3-only marked regions. The stable H3K4me3-only regions were defined as those regions that retained the H3K4me3 mark in the next stage, while the dynamic H3K4me3-only regions were defined as those regions that lost the modification in the next stage. d, Averaged domain length of the stable and dynamic H3K4me3-only marked regions. e, Averaged signal density of the stable and dynamic H3K4me3-only marked regions. The signal density was calculated as H3K4me3 ChIP–seq RPKM (reads per million reads per kb). fi, Left panel, alluvial plots showing the global dynamics of H3K4me3-only (f), bivalent (g), H3K27me3-only (h) and non-marked (i) regions during the first cell lineage differentiation and cell line derivation. Each line represents a 200-bp bin defined according to the ChromHMM categories. For each state, the total regions were those classified as this state in at least one analysed stage. The morula stage was plotted in the middle, and the two differentiated lineages were plotted on either side. Right panel, the percentage of the analysed states in each stage compared to the total regions. The global dynamics (solid line) and chromosome X dynamics (dashed line) are plotted separately.

Extended Data Figure 4 Establishment and dynamics of H3K4me3 and H3K27me3 on promoters.

a, b, Heat map of the H3K4me3-marked (a) and H3K27me3-marked (b) transcript dynamic during early mouse embryo development and differentiation. The H3K4me3-marked transcripts were defined with an overlap of at least 200 bp with the ChromHMM H3K4me3 segmentations, including the H3K4me3-only and bivalent segmentations. H3K27me3-marked transcripts were defined as having an overlap of at least 200 bp with the ChromHMM H3K27me3 segmentations, including the H3K27me3-only and bivalent segmentations. The transcript clustering was based on the binary score; see the Methods section for the detailed clustering method. c, d, Correlation between the H3K4me3 (c) or H3K27me3 (d) signal intensity and local CpG ratio in all pre-implantation developmental stages and cell lines. ChIP–seq signal intensity was evaluated using a weighted ChIP–seq signal of ±2 kb around the TSS. Pearson correlation coefficients were calculated and are shown in the top right of each panel. e, f, Averaged H3K4me3 (e) and H3K27me3 (f) signal intensity on the HCPs, ICPs and LCPs throughout all pre-implantation developmental stages and cell lines. The definition of the three class promoters is described in the Methods.

Extended Data Figure 5 Distinct features of the establishment of H3K4me3 and H3K27me3 in embryogenesis and differentiation.

a, Profiles of the H3K27me3 signal density on the HCPs, ICPs and LCPs from MII to morula stage. The H3K27me3 signal density was calculated as H3K27me3 RPM with 50-bp resolution. b, Averaged local max CpG ratio of the new H3K27me3-marked transcripts. The newly established H3K27me3-marked transcripts were defined as transcripts belongs to non-marked state in the previous stage and marked by H3K27me3 in the present stage. c, Averaged H3K27me3 signals of the HCPs, ICPs and LCPs in the two-cell stage, classified by the methylation level. The published whole-genome bisulphite sequencing data was used in this analysis20. The methylated genes were defined as genes with an averaged promoter methylation level ≥0.6, and the unmethylated genes were defined as genes with an averaged promoter methylation level ≤0.25. d, e, Profiles of the H3K4me3 (d) and H3K27me3 (e) signal density on the HCPs, ICPs and LCPs in ICM, TE, ES cells and TSCs. The H3K4me3 and H3K27me3 signal density was calculated as H3K27me3 RPM with 50-bp resolution. f, g, Scatter plot of the signal intensities of H3K4me3 (f) and H3K27me3 (g) with the promoter regions and DNA methylation levels. Our reduced representation bisulphite sequencing (RRBS) data published in another project44 was used in this analysis. The red and blue fitting curves represent the ChIP–seq intensity and methylation level in the corresponding regions, respectively. The horizontal axis, from left to right, represents the transcripts ranked from high to low according to their promoter H3K4me3 or H3K27me3 intensities, and the y axis indicates the averaged methylation level of corresponding promoter Pearson correlations of the H3K4me3 intensities and DNA methylation levels were calculated and included in the panel f and Pearson correlations of the H3K27me3 intensities and DNA methylation levels in panel g. h, Correlation of H3K4me3 and H3K27me3 signal intensity throughout early embryo developmental stages and cell lines.

Extended Data Figure 6 Identification and regulation of H3K4me3 broad domains on promoters.

a, Bar plot showing the distribution of the H3K4me3 domain length at the promoter regions during mouse development. The two dashed lines represent the cut-offs between the plotted narrow, medium and broad domains. The percentages of transcripts classified into these three groups and control are listed in the top right of the panel. The representative genes Ndrg2, Ctcf and Ldb1, whose promoters were covered by narrow, medium and broad domains, respectively, are displayed. b, Distribution of the H3K4me3 domain length at the promoter regions in all pre-implantation developmental stages and cell lines. The dashed line represents the cut-off between the medium and broad domains. c, Heat map showing the H3K4me3 RPM on all the RefSeq transcripts of the four states of H3K4me3 domains for the morula, ICM and ES cell samples. Each row represents a promoter region with ±10 kb around a TSS. d, Genome browser view of H3K4me3 broad domains establishment at Nanog and Pou5f1 genes from the two-cell stage to the ICM stage. ChIP–seq RPM was smoothed by the mean of 5 pixels using the UCSC genome browser and visualized. e, Depletion of H3K4me3 demethylase inhibited the formation of blastocysts. The siRNA mixture of each predicated H3K4me3 demethylase was injected into MII oocytes separately or mixed together. The injected oocytes were then performed ICSI. The data are represented as the mean ± s.d. (n = 3). f, Knockdown of Kdm5b (si-Kdm5b) inhibited the formation of expanding and hatching of blastocysts. Injection of Kdm5b mRNA (oe-Kdm5b) as well as control RNA was also performed for comparison. The data are represented as the mean ± s.d. (n = 3). g, Heat map showing the breadth of H3K4me3 domains on promoters in control, oe-Kdm5b and si-Kdm5b samples. The H3K4me3 domains were classified into four groups: broad domains (dark red), medium domains (red), narrow domains (pink) and control domains (grey). The dashed lines represent the classification based on control samples. h, Box plot showing the averaged H3K4me3 domain breadth in different samples on promoter covered by broad (left), medium (middle) and control (right) H3K4me3 domains in control samples. i, Genome browser view of control, oe-Kdm5b and si-Kdm5b samples at Zfp85 gene locus. ChIP–seq RPM was smoothed by the mean of 5 pixels using the UCSC genome browser and visualized.

Extended Data Figure 7 H3K4me3 breadth is highly associated with transcription activity and cell identity.

a, Box plot showing the averaged expression level of the broad, medium, narrow and control H3K4me3 domains in all pre-implantation developmental stages and cell lines. The number of the transcript is listed with the group name. b, Heat map showing the gain and loss of the broad domains during the transition from the morula stage to the ICM and TE. The transcripts were clustered using a k-means function into seven groups. c, Averaged expression level of the transcript in the morula, ICM and TE samples for the clusters in b. The expression level was evaluated using the averaged log2 FPKM. d, Tendencies of the four types of H3K4me3 domains in the next stage during stable cell line derivation. Each panel represents a specific type of H3K4me3 domain. Each bar represents the types and percentages of the transcripts in the next stage. e, Broad domains inherited from the TE to TSCs. A gene ontology analysis was performed on the overlapped genes from the TE to TSCs, and representative genes were labelled. The P value was calculated using multiple Student’s t-tests. f, g, Bar plot of the H3K4me3 domain length at the Id2 (f) and Elf5 (g) promoter regions (left panel) and the corresponding expression level (right panel) in embryo and cell lines. The dashed line represents the cut-off for broad domains.

Extended Data Figure 8 Dynamics of the bivalent promoters in embryogenesis and differentiation.

a, Heat map showing the dynamics of the bivalent genes during early mouse embryo development. Each row represents one RefSeq transcript. Red indicates a transcript that is only marked by H3K4me3, blue represents a transcript that is marked by H3K27me3, and dark green indicates a transcript in a bivalent state. b, Number of bivalent transcripts in MII oocytes, pre-implantation embryos and cell lines. c, Tendencies of the H3K4me3-only promoters, H3K27me3-only promoters, bivalent promoters and non-marked promoters in the next stage during early mouse embryo development. Each bar represents the type and percentage of the transcript in the next stage. d, Bar plot showing the possibility of the H3K27me3-marked condition for transcripts with a broad, medium or narrow H3K4me3 domains. Red represents transcripts without the H3K27me3 mark, and dark green represents transcripts with the H3K27me3 mark, indicating a bivalent state.

Extended Data Figure 9 Inheritance of the ICM-bivalent promoters in ES cell derivation.

a, Tendencies of the H3K4me3-only promoters, H3K27me3-only promoters, bivalent promoters and non-marked promoters in the next stage during cell line derivation. Each bar represents the type and percentage of the transcript in the next stage. The ICM-bivalent genes, which are covered only by H3K4me3 signals, or only by H3K27me3 signals or maintain bivalent states in the ES cell samples, were defined as ES-cell-activated, ES-cell-repressed and ES-cell-maintained, respectively. b, c, Genome browser view of the H3K4me3 and H3K27me3 density at activated bivalent gene Jade1 (b) and repressed bivalent gene Cxcl10 (c) in ES cell and ICM samples. The ChIP–seq RPM was smoothed by the mean of 5 pixels using the UCSC genome browser. d, H3K4me3 and H3K27me3 signal intensities of the three types of ICM-bivalent transcripts, ES-cell-activated, ES-cell-repressed and ES-cell-maintained, defined in a. e, Expression level during early mouse embryo development of the three types of ICM bivalent transcripts, ES-cell-activated, ES-cell-repressed and ES-cell-maintained, defined in a.

Related audio

Supplementary information

Supplementary Data

This file contains Supplementary Table 1, a summary of chromatin state of TSS in MII oocytes, pre-implantation embryos, ESCs and TSCs. (XLSX 1751 kb)

Supplementary Data

This file contains Supplementary Table 2, functional annotation of transcripts that maintain H3K27me3 from MII Oocyte to embryo development. (XLSX 17 kb)

Supplementary Data

This file contains Supplementary Table 3, a summary of genes covered by broad H3K4me3 in MII oocytes, pre-implantation embryos, ESCs and TSCs. (XLSX 1834 kb)

Supplementary Data

This file contains Supplementary Table 4, a list of siRNA used in the experiment. (PDF 101 kb)

Supplementary Data

This file contains Supplementary Table 5, a summary of quality information of ChIP data and correlation of replicates in each stage. (XLSX 23 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, X., Wang, C., Liu, W. et al. Distinct features of H3K4me3 and H3K27me3 chromatin domains in pre-implantation embryos. Nature 537, 558–562 (2016). https://doi.org/10.1038/nature19362

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature19362

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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