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.

Allele-specific H3K9me3 and DNA methylation co-marked CpG-rich regions serve as potential imprinting control regions in pre-implantation embryo

Abstract

Parental DNA methylation and histone modifications undergo distinct global reprogramming in mammalian pre-implantation embryos, but the landscape of epigenetic crosstalk and its effects on embryogenesis are largely unknown. Here we comprehensively analyse the association between DNA methylation and H3K9me3 reprogramming in mouse pre-implantation embryos and reveal that CpG-rich genomic loci with high H3K9me3 signal and DNA methylation level (CHM) are hotspots of DNA methylation maintenance during pre-implantation embryogenesis. We further profile the allele-specific epigenetic map with unprecedented resolution in gynogenetic and androgenetic embryos, respectively, and identify 1,279 allele-specific CHMs, including 19 known imprinting control regions (ICRs). Our study suggests that 22 ICR-like regions (ICRLRs) may regulate allele-specific transcription similarly to known ICRs, and five of them are confirmed to be important for mouse embryo development. Taken together, our study reveals the widespread existence of allele-specific CHMs and largely extends the scope of allele-specific regulation in mammalian pre-implantation embryos.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: CHMs are hotspots of DNA methylation maintenance in pre-implantation embryos.
Fig. 2: Computational framework of PCAR.
Fig. 3: Widespread allele-specific CHMs in pre-implantation embryos.
Fig. 4: DNA methylation asymmetry at allele-specific CHMs are partially determined by H3K9me3.
Fig. 5: H3K9me3 is involved in the repressive functions of ICRLRs.
Fig. 6: ICRLRs are critical for early embryo development.

Data availability

All the ChIP-seq, WGBS and RNA-seq datasets generated in this study are summarized in Supplementary Table 2 and have been deposited in the Genome Sequence Archive (https://bigd.big.ac.cn/gsa/) under accession no. CRA004123. All public ChIP-seq, DNase-seq, ATAC-seq, WGBS and RNA-seq datasets used in this study are summarized in Supplementary Table 1. 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 computational pipeline for CHM calling and scoring for allele-specific regulatory potentials (PCAR) is available at https://github.com/TongjiZhanglab/PCAR.

References

  1. 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 

  2. 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 

  3. Wu, X. & Zhang, Y. TET-mediated active DNA demethylation: mechanism, function and beyond. Nat. Rev. Genet. 18, 517–534 (2017).

    Article  CAS  PubMed  Google Scholar 

  4. Zeng, Y. & Chen, T. DNA methylation reprogramming during mammalian development. Genes (Basel) 10, 257 (2019).

    Article  CAS  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  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. Rose, N. R. & Klose, R. J. Understanding the relationship between DNA methylation and histone lysine methylation. Biochim. Biophys. Acta 1839, 1362–1372 (2014).

    Article  CAS  PubMed  Google Scholar 

  8. Lehnertz, B. et al. Suv39h-mediated histone H3 lysine 9 methylation directs DNA methylation to major satellite repeats at pericentric heterochromatin. Curr. Biol. 13, 1192–1200 (2003).

    Article  CAS  PubMed  Google Scholar 

  9. Matsui, T. et al. Proviral silencing in embryonic stem cells requires the histone methyltransferase ESET. Nature 464, 927–931 (2010).

    Article  CAS  PubMed  Google Scholar 

  10. Dong, K. B. et al. DNA methylation in ES cells requires the lysine methyltransferase G9a but not its catalytic activity. EMBO J. 27, 2691–2701 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Leung, D. et al. Regulation of DNA methylation turnover at LTR retrotransposons and imprinted loci by the histone methyltransferase Setdb1. Proc. Natl Acad. Sci. USA 111, 6690–6695 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Li, Y. & Sasaki, H. Genomic imprinting in mammals: its life cycle, molecular mechanisms and reprogramming. Cell Res. 21, 466–473 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Morgan, H. D., Santos, F., Green, K., Dean, W. & Reik, W. Epigenetic reprogramming in mammals. Hum. Mol. Genet. 14 Spec No 1, R47–R58 (2005).

    Article  PubMed  CAS  Google Scholar 

  14. Howell, C. Y. et al. Genomic imprinting disrupted by a maternal effect mutation in the Dnmt1 gene. Cell 104, 829–838 (2001).

    Article  CAS  PubMed  Google Scholar 

  15. Duffie, R. et al. The Gpr1/Zdbf2 locus provides new paradigms for transient and dynamic genomic imprinting in mammals. Genes Dev. 28, 463–478 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Wijchers, P. J. et al. Characterization and dynamics of pericentromere-associated domains in mice. Genome Res. 25, 958–969 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Proudhon, C. et al. Protection against de novo methylation is instrumental in maintaining parent-of-origin methylation inherited from the gametes. Mol. Cell 47, 909–920 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Strogantsev, R. et al. Allele-specific binding of ZFP57 in the epigenetic regulation of imprinted and non-imprinted monoallelic expression. Genome Biol. 16, 112 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Li, Y. et al. Precise allele-specific genome editing by spatiotemporal control of CRISPR-Cas9 via pronuclear transplantation. Nat. Commun. 11, 4593 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Liu, X. et al. UHRF1 targets DNMT1 for DNA methylation through cooperative binding of hemi-methylated DNA and methylated H3K9. Nat. Commun. 4, 1563 (2013).

    Article  PubMed  CAS  Google Scholar 

  21. Rothbart, S. B. et al. Association of UHRF1 with methylated H3K9 directs the maintenance of DNA methylation. Nat. Struct. Mol. Biol. 19, 1155–1160 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. 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 

  23. Quenneville, S. et al. In embryonic stem cells, ZFP57/KAP1 recognize a methylated hexanucleotide to affect chromatin and DNA methylation of imprinting control regions. Mol. Cell 44, 361–372 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Hirasawa, R. et al. Maternal and zygotic Dnmt1 are necessary and sufficient for the maintenance of DNA methylation imprints during preimplantation development. Genes Dev. 22, 1607–1616 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Ren, W. et al. Direct readout of heterochromatic H3K9me3 regulates DNMT1-mediated maintenance DNA methylation. Proc. Natl Acad. Sci. USA 117, 18439–18447 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Zuo, X. et al. Zinc finger protein ZFP57 requires its co-factor to recruit DNA methyltransferases and maintains DNA methylation imprint in embryonic stem cells via its transcriptional repression domain. J. Biol. Chem. 287, 2107–2118 (2012).

    Article  CAS  PubMed  Google Scholar 

  27. 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 

  28. Duffie, R. & Bourc’his, D. Parental epigenetic asymmetry in mammals. Curr. Top. Dev. Biol. 104, 293–328 (2013).

    Article  CAS  PubMed  Google Scholar 

  29. Picelli, S. et al. Smart-seq2 for sensitive full-length transcriptome profiling in single cells. Nat. Methods 10, 1096–1098 (2013).

    Article  CAS  PubMed  Google Scholar 

  30. Picelli, S. Full-length single-cell RNA sequencing with Smart-seq2. Methods Mol. Biol. 1979, 25–44 (2019).

    Article  CAS  PubMed  Google Scholar 

  31. Tang, F. et al. mRNA-seq whole-transcriptome analysis of a single cell. Nat. Methods 6, 377–382 (2009).

    Article  CAS  PubMed  Google Scholar 

  32. Tang, F. et al. RNA-seq analysis to capture the transcriptome landscape of a single cell. Nat. Protoc. 5, 516–535 (2010).

    Article  CAS  PubMed  Google Scholar 

  33. 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  CAS  Google Scholar 

  34. Guo, H. et al. Single-cell methylome landscapes of mouse embryonic stem cells and early embryos analyzed using reduced representation bisulfite sequencing. Genome Res. 23, 2126–2135 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Langdon, W. B. Performance of genetic programming optimised Bowtie2 on genome comparison and analytic testing (GCAT) benchmarks. BioData Min. 8, 1 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Kim, D., Paggi, J. M., Park, C., Bennett, C. & Salzberg, S. L. Graph-based genome alignment and genotyping with HISAT2 and HISAT-genotype. Nat. Biotechnol. 37, 907–915 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  40. Sing, T., Sander, O., Beerenwinkel, N. & Lengauer, T. ROCR: visualizing classifier performance in R. Bioinformatics 21, 3940–3941 (2005).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Xie, W. et al. Base-resolution analyses of sequence and parent-of-origin dependent DNA methylation in the mouse genome. Cell 148, 816–831 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Babak, T. et al. Genetic conflict reflected in tissue-specific maps of genomic imprinting in human and mouse. Nat. Genet. 47, 544–549 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Schulz, R. et al. WAMIDEX: a web atlas of murine genomic imprinting and differential expression. Epigenetics 3, 89–96 (2008).

    Article  PubMed  Google Scholar 

  45. Morison, I. M., Ramsay, J. P. & Spencer, H. G. A census of mammalian imprinting. Trends Genet. 21, 457–465 (2005).

    Article  CAS  PubMed  Google Scholar 

  46. Wei, Y. et al. MetaImprint: an information repository of mammalian imprinted genes. Development 141, 2516–2523 (2014).

    Article  CAS  PubMed  Google Scholar 

  47. Harrow, J. et al. GENCODE: the reference human genome annotation for The ENCODE Project. Genome Res. 22, 1760–1774 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Navarro Gonzalez, J. et al. The UCSC Genome Browser database: 2021 update. Nucleic Acids Res. 49, D1046–D1057 (2021).

    Article  PubMed  CAS  Google Scholar 

  49. Zheng, R. et al. Cistrome Data Browser: expanded datasets and new tools for gene regulatory analysis. Nucleic Acids Res. 47, D729–D735 (2019).

    Article  CAS  PubMed  Google Scholar 

  50. 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 

  51. Shao, W. & Wang, T. Transcript assembly improves expression quantification of transposable elements in single-cell RNA-seq data. Genome Res. 31, 88–100 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  53. Niknafs, Y. S., Pandian, B., Iyer, H. K., Chinnaiyan, A. M. & Iyer, M. K. TACO produces robust multisample transcriptome assemblies from RNA-seq. Nat. Methods 14, 68–70 (2017).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank C. Zhao and W. Wang for assistance with data analysis. This work was supported by the National Key Research and Development Program of China (2017YFA0102600 (Y.Z.) and 2016YFA0100400 (S.G.)), the National Natural Science Foundation of China (32030022 (Y.Z.), 31970642 (Y.Z.), 31721003 (S.G.), 31820103009 (S.G.), 31871448 (W.L.) and 31801059 (C.W.)) and the Peak Disciplines (Type IV) of Institutions of Higher Learning in Shanghai (Y.Z. and S.G.).

Author information

Authors and Affiliations

Authors

Contributions

S.G. and Y.Z. conceived and designed the research. H.Y., Z.Y., C.W. and Y.Z. designed and performed computational analysis. W.L., D.B., Y.L. and Y.S. performed experiments. H.Y., W.L., Z.Y. and Y.Z. wrote the manuscript.

Corresponding authors

Correspondence to Wenqiang Liu, Shaorong Gao or Yong Zhang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Cell Biology thanks the anonymous reviewers for their contribution to the peer review of this work. Peer reviewer reports are available.

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 Features of stable CHMs in mouse pre-implantation embryos.

a, Line chart of the percentage of 1-kb bins escaping DNA demethylation at each given stage (that is, DNA methylation level ≥ 0.5 at the given stage and all previous stages). b, c, Boxplots comparing DNA methylation level (b) and H3K9me3 signals (c) at stable CHMs (n = 6,655), stage-specific CHMs (n = 16,461) and highly methylated non-CHM CpG islands (CGIs) (n = 1,632). Significance between stable CHMs and highly methylated non-CHM CGIs was evaluated by two-sided Wilcoxon rank sum test, *** indicates a p-value < 0.001 (p-value for DNA methylation level at 2-cell: 2.3×10−243; p-value for DNA methylation level at other stages and for H3K9me3 signals at all stages: < 2.2×10−308). In the boxplots, the centre lines mark the median, the box limits indicate the 25th and 75th percentiles, and the whiskers extend to 1.5 × the interquartile range from the 25th and 75th percentiles. d, Pie plot showing status of stable CHMs in gametes. e, f, Scatter plots showing DNA methylation level (e) and H3K9me3 signals (f) of stable CHMs in gametes. g, Boxplots comparing the expression levels between potential target genes of CHMs (n = 1,129) and other genes (n = 34,538). FPKM, fragments per kilobase of exon per million mapped fragments. Potential target genes were defined as the nearest genes within 300 kb of CHMs located in promoter and potential enhancers. CHMs located in PADs were removed. Significance between the groups was evaluated by two-sided Wilcoxon rank sum test, *** indicates a p-value < 0.001 and n.s. indicates not significant (p-value at 2-cell, 8-cell, Morula, ICM: 0.33, 4.9×10−19, 2.6×10−25 and 2.8×10−12). The meaning of boxplots is identical to that in b, c. h, Gene Ontology analysis of potential target genes of CHMs. The p-values were calculated based on a one-sided Fisher’s exact test.

Source data

Extended Data Fig. 2 CHMs are hotspots of DNA methylation maintenance in GG and AG pre-implantation embryos.

a, Line charts showing DNA methylation level from SNP-trackable data (C57BL: maternal, DBA: paternal) around hypermethylated CpG-rich regions in GG and AG embryos. b, Line charts showing H3K9me3 signals from SNP-trackable data around H3K9me3-marked CpG-rich regions in GG and AG embryos. c, d, Scatter plots showing the Pearson’s correlation of allelic DNA methylation level (c) and H3K9me3 signals (d) between GG / AG embryos and strain-hybrid embryos in SNP-trackable CpG-rich 1 kb bins, covering ≥ 100 SNP-trackable reads of corresponding WGBS data. e, f, i, j, Bar plot showing the percentage of highly methylated CpGs (DNA methylation level ≥ 0.5) at 2-cell stage maintaining high methylation status during GG (e) and AG (f) pre-implantation embryogenesis and in maternal (C57BL) (i) and paternal (DBA) (j) allele. g, h, k, l, Boxplots comparing DNA methylation level (left) and H3K9me3 signals (right) at stable CHMs (n = 8,384 / 6,433), stage-specific CHMs (n = 15,171 / 19,082) and highly methylated non-CHM CGIs (n = 731 / 703) in GG (g) / AG (h) embryos, or at SNP-trackable (that is, covering ≥ 20 SNP-trackable reads of corresponding WGBS data) stable CHMs (n = 2,072 / 1,499), stage-specific CHMs (n = 3,474 / 4,300) and highly methylated non-CHM CGIs (n = 94 / 96) in maternal (C57BL) (k) and paternal (DBA) (l) allele. Significance between stable CHMs and highly methylated non-CHM CGIs was evaluated by two-sided Wilcoxon rank sum test, ***: p-value < 0.001, **: p-value < 0.01, *: p-value < 0.05. p-values in each panel (from left to right): 4.5×10−44, 4.8×10−5, 3.3×10−69, < 2.2×10−308, < 2.2×10−308, < 2.2×10−308 (g); 4.3×10−126, 3.5×10−239, < 2.2×10−308, < 2.2×10−308, < 2.2×10−308, 1.0 × 10−255 (h); 0.012, 7.2×10−19, 5.6×10−12, 2.8×10−35 (k); 2.3×10−14, 6.8×10−22, 4.5×10−3, 1.4×10−9 (l). The meaning of boxplots is identical to that in Extended Data Fig. 1.

Source data

Extended Data Fig. 3 Status of allele-specific CHMs in gametes and E6.5 embryos.

a, Pie plots showing status of allele-specific CHMs in public strain-hybrid 2-cell (upper) and ICM (bottom) embryos. SNP-trackable allele-specific CHMs are required to cover ≥ 100 SNP-trackable reads of corresponding WGBS data. b, The UCSC Genome Browser view showing DNA methylation level and H3K9me3 signals in pre-implantation GG and AG embryos surrounding known gamete ICR Zrsr1/Commd (highlighted by blue), which is not identified as an allele-specific CHM. CHMs in GG and AG embryos are indicated by orange and green rectangles respectively. c, The UCSC Genome Browser view showing status of allele-specific CHMs showed consistent parent-of-origin DNA methylation and H3K9me3 asymmetry in gametes. The genomic location of the mCHM and pCHM are indicated by orange and green rectangle respectively. d, Scatter plots with distribution information showing DNA methylation level and H3K9me3 signals of mCHMs (upper) and pCHMs (bottom) in gametes according to their presence and absence in corresponding gamete. ICRLRs selected for validation were labelled and highlighted as orange and green dots respectively. e, The UCSC Genome Browser view showing status of allele-specific CHMs not identified as CHMs in corresponding gametes. The genomic location of mCHM and pCHM are indicated by orange and green rectangle respectively. f, The UCSC Genome Browser view showing status of allele-specific CHMs in E6.5 embryos. The genomic location of mCHM and pCHM are indicated by orange and green rectangle respectively.

Source data

Extended Data Fig. 4 DNA methylation asymmetry at allele-specific CHMs influenced by H3K9me3 depletion.

a, Representative immunofluorescence staining for H3K9me3 (green), DAPI (grey) and merged images in control and H3K9me3-depleted embryos at the zygote stage. Data is reproducible in three independent experiments. Scale bar, 20 μm. b, The UCSC Genome Browser view of a representative mCHM showing the decreased DNA methylation level in H3K9me3-depleted embryos at the morula stage. mCHM is indicated with orange rectangles. DNA methylation change after H3K9me3-depleted represents smoothed DNA methylation level change (H3K9me3-depleted - WT) in 200-bp windows.

Extended Data Fig. 5 Allele-specific expressed genes and transposable elements surrounding ICRs and ICRLRs.

a, b, Heatmap showing expression pattern of allele-specific expressed gene (a) and transposable element (b) surrounding ICRs and ICRLRs within 300 kb in pre-implantation embryos. Maternal-specific expressed genes (a) and transposable element (b) are indicated with orange and paternal-specific expressed genes (a) and transposable element (b) are indicated with green. ICRs are highlighted in red and transient gDMRs reported are highlighted in blue. Known imprinted genes are marked with asterisk. c, d, Pie plots showing allele-specific status of monoallelic expressed genes and transposable elements nearby ICRs (c) and ICRLRs (d) in hybrid pre-implantation embryos. SNP-trackable monoallelic expressed genes and transposable elements are required to cover ≥ 100 SNP-trackable reads of corresponding RNA-seq data. ‘Consistent’ indicates SNP-trackable monoallelic expressed genes and transposable elements showing similar allele preference in hybrid embryos. ‘Inconsistent’ indicates SNP-trackable monoallelic expressed genes and transposable elements showing no or contrast allele preference in hybrid embryos.

Source data

Extended Data Fig. 6 Functions of 5 identified ICRLRs in the development of early embryos.

a, UCSC Genome Browser view of ICRLRs selected for validation. The maternal and paternal ICRLRs are indicated by orange and green rectangles respectively. b, Box plots showing the expression levels of genes (n = 6) harbouring validated maternal ICRLRs in pre-implantation GG/AG embryos. The meaning of boxplots is identical to that in Extended Data Fig. 1. c, Representative images of blastocyst-stage embryos produced from control, mCHM_245-KO, mCHM_177-KO, mCHM_328-KO, mCHM_4-KO and pCHM_824-KO embryos. Data is reproducible in two independent experiments. Scale bar, 150 μm. d, Schematic diagram of generating ICRLR-KO GG and AG embryos. e, DNA sequencing showing effective paternal-specific (left) and maternal-specific (right) mCHM_177-KO. f, Bar plot showing the ratios of decidua at the E9.5 stage in the control, maternal-specific and paternal-specific mCHM_177-KO groups. Control embryos (two biologically independent experiments, sample size: 48, 46), maternal-specific (three biologically independent experiments, sample size: 56, 36, 45) and paternal-specific (three biologically independent experiments, sample size: 68, 40, 31) mCHM_177-KO are indicated in grey, orange and green respectively. Significance between groups was evaluated by two-sided Welch’s t-test, n.s. indicates not significant (p-value between maternal-specific and paternal-specific mCHM_177-KO: 0.36). g, DNA sequencing showing effective maternal-specific (upper) and paternal-specific (bottom) Brd2-KO. h, Bar plot for the ratios of blastocyst of control and BC051142-KO embryos. Control (two biologically independent experiments, sample size: 36, 30) and BC051142-KO embryos (two biologically independent experiments, sample size: 33, 36) are indicated in grep and black. Sample size refers to the number of 2-cell stage embryos. i, Genomic schematic diagram showing expression details of genes expressed in at least 1 stage in GG and AG pre-implantation embryos within 300 kb of mCHM_177. The gene Brd2 was indicated as green. Allele-specific score was calculated by PCAR.

Source data

Supplementary information

Reporting Summary

Peer Review File

Supplementary Tables

Supplementary Table 1 Summary of the public ChIP-seq, DNase-seq, ATAC-seq, WGBS and RNA-seq datasets used in this study. Supplementary Table 2 Summary of the ChIP-seq, WGBS and RNA-seq datasets generated in this study. Supplementary Table 3 Summary of the allele-specific CHMs identified in pre-implantation embryos. The scores were calculated based on features of known ICRs. Allele-specific CHMs with equal or higher scores than any known ICR were regarded as ICRLRs. ICRLRs with functional validation in this study were marked with green. Supplementary Table 4 Sequences for siRNAs, PCR primers and sgRNAs; mutants information

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yang, H., Bai, D., Li, Y. et al. Allele-specific H3K9me3 and DNA methylation co-marked CpG-rich regions serve as potential imprinting control regions in pre-implantation embryo. Nat Cell Biol 24, 783–792 (2022). https://doi.org/10.1038/s41556-022-00900-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-022-00900-4

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