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:

Single-cell multi-omics sequencing of human early embryos

A Publisher Correction to this article was published on 18 July 2018

Abstract

DNA methylation, chromatin states and their interrelationships represent critical epigenetic information, but these are largely unknown in human early embryos. Here, we apply single-cell chromatin overall omic-scale landscape sequencing (scCOOL-seq) to generate a genome-wide map of DNA methylation and chromatin accessibility at single-cell resolution during human preimplantation development. Unlike in mice, the chromatin of the paternal genome is already more open than that of the maternal genome at the mid-zygote stage in humans, and this state is maintained until the 4-cell stage. After fertilization, genes with high variations in DNA methylation, and those with high variations in chromatin accessibility, tend to be two different sets. Furthermore, 1,797 out of 5,155 (35%) widely open chromatin regions in promoters closed when transcription activity was inhibited, indicating a feedback mechanism between transcription and open chromatin maintenance. Our work paves the way for dissecting the complex, yet highly coordinated, epigenetic reprogramming during human preimplantation development.

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: Chromatin accessibility of human early embryos.
Fig. 2: Features of open chromatin and nucleosomes in mammalian development.
Fig. 3: Heterogeneity of TSS NDRs among individual cells in hESCs and from oocyte to the 4-cell stage.
Fig. 4: Differential DNA methylation and chromatin accessibility between parental genomes within each individual blastomere during human preimplantation development.
Fig. 5: Distinct features of chromatin accessibility between human and mouse embryos.
Fig. 6: Variance of DNA methylation and chromatin accessibility during human preimplantation development.
Fig. 7: Chromatin accessibility of repeat elements and the regulation network in human early embryos.
Fig. 8: Continual transcription is crucial for human preimplantation chromatin remodelling.

Similar content being viewed by others

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

    Article  CAS  PubMed  Google Scholar 

  3. Rossant, J. & Tam, P. P. New insights into early human development: lessons for stem cell derivation and differentiation. Cell Stem Cell 20, 18–28 (2017).

    Article  CAS  PubMed  Google Scholar 

  4. Rugg-Gunn, P. J., Cox, B. J., Ralston, A. & Rossant, J. Distinct histone modifications in stem cell lines and tissue lineages from the early mouse embryo. Proc. Natl Acad. Sci. USA 107, 10783–10790 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Saitou, M., Kagiwada, S. & Kurimoto, K. Epigenetic reprogramming in mouse pre-implantation development and primordial germ cells. Development 139, 15–31 (2012).

    Article  CAS  PubMed  Google Scholar 

  6. Guo, F. et al. Single-cell multi-omics sequencing of mouse early embryos and embryonic stem cells. Cell Res. 27, 967–988 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Lu, F. et al. Establishing chromatin regulatory landscape during mouse preimplantation development. Cell 165, 1375–1388 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  9. 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 

  10. Dahl, J. & Jung, I. Broad histone H3K4me3 domains in mouse oocytes modulate maternal-to-zygotic transition. Nature 537, 548–552 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Zhang, B., Zheng, H., Huang, B. & Li, W. Allelic reprogramming of the histone modification H3K4me3 in early mammalian development. Nature 537, 553–557 (2016).

    Article  CAS  PubMed  Google Scholar 

  12. Guo, H. et al. The DNA methylation landscape of human early embryos. Nature 511, 606–610 (2014).

    Article  CAS  PubMed  Google Scholar 

  13. Smith, Z. D. et al. DNA methylation dynamics of the human preimplantation embryo. Nature 511, 611–615 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Okae, H. et al. Genome-wide analysis of DNA methylation dynamics during early human development. PLoS Genet. 10, e1004868 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Fulka, H., Mrazek, M., Tepla, O. & Fulka, J. Jr. DNA methylation pattern in human zygotes and developing embryos. Reproduction 128, 703–708 (2004).

    Article  CAS  PubMed  Google Scholar 

  16. Molaro, A. et al. Sperm methylation profiles reveal features of epigenetic inheritance and evolution in primates. Cell 146, 1029–1041 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Hatada, I. et al. Genome-wide profiling of promoter methylation in human. Oncogene 25, 3059–3064 (2006).

    Article  CAS  PubMed  Google Scholar 

  18. Fang, F., Hodges, E., Molaro, A. & Dean, M. Genomic landscape of human allele-specific DNA methylation. Proc. Natl Acad. Sci. USA 109, 7332–7337 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Zhu, P. et al. Single-cell DNA methylome sequencing of human preimplantation embryos. Nat. Genet. 50, 12–19 (2018).

    Article  CAS  PubMed  Google Scholar 

  20. Ambartsumyan, G. & Clark, A. T. Aneuploidy and early human embryo development. Hum. Mol. Genet. 17, R10–R15 (2008).

    Article  CAS  PubMed  Google Scholar 

  21. Vanneste, E. et al. Chromosome instability is common in human cleavage-stage embryos. Nat. Med. 15, 577–583 (2009).

    Article  CAS  PubMed  Google Scholar 

  22. Bolton, H. et al. Mouse model of chromosome mosaicism reveals lineage-specific depletion of aneuploid cells and normal developmental potential. Nat. Commun. 7, 11165 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Clark, S. J. et al. scNMT-seq enables joint profiling of chromatin accessibility DNA methylation and transcription in single cells. Nat. Commun. 9, 781 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Pott, S. Simultaneous measurement of chromatin accessibility, DNA methylation, and nucleosome phasing in single cells. eLife 6, e23203 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Taberlay, P. C. et al. Polycomb-repressed genes have permissive enhancers that initiate reprogramming. Cell 147, 1283–1294 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Kelly, T. K. et al. Genome-wide mapping of nucleosome positioning and DNA methylation within individual DNA molecules. Genome Res. 22, 2497–2506 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Nabilsi, N. H. et al. Multiplex mapping of chromatin accessibility and DNA methylation within targeted single molecules identifies epigenetic heterogeneity in neural stem cells and glioblastoma. Genome Res. 24, 329–339 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Taberlay, P. C., Statham, A. L., Kelly, T. K., Clark, S. J. & Jones, P. A. Reconfiguration of nucleosome-depleted regions at distal regulatory elements accompanies DNA methylation of enhancers and insulators in cancer. Genome Res. 24, 1421–1432 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Lay, F. D. et al. The role of DNA methylation in directing the functional organization of the cancer epigenome. Genome Res. 25, 467–477 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Miura, F., Enomoto, Y., Dairiki, R. & Ito, T. Amplification-free whole-genome bisulfite sequencing by post-bisulfite adaptor tagging. Nucleic Acids Res. 40, e136 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Smallwood, S. A. et al. Single-cell genome-wide bisulfite sequencing for assessing epigenetic heterogeneity. Nat. Methods 11, 817–820 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Guo, H. et al. DNA methylation and chromatin accessibility profiling of mouse and human fetal germ cells. Cell Res. 27, 165–183 (2017).

    Article  CAS  PubMed  Google Scholar 

  33. Tolstorukov, M. Y., Volfovsky, N., Stephens, R. M. & Park, P. J. Impact of chromatin structure on sequence variability in the human genome. Nat. Struct. Mol. Biol. 18, 510–515 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Fincher, J. A., Tyson, G. S. & Dennis, J. H. DNA-encoded chromatin structural intron boundary signals identify conserved genes with common function. Int. J. Genomics 2015, 167578 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Schwartz, S., Meshorer, E. & Ast, G. Chromatin organization marks exon–intron structure. Nat. Struct. Mol. Biol. 16, 990–995 (2009).

    Article  CAS  PubMed  Google Scholar 

  36. ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74 (2012).

    Article  CAS  Google Scholar 

  37. Yan, L. et al. Single-cell RNA-seq profiling of human preimplantation embryos and embryonic stem cells. Nat. Struct. Mol. Biol. 20, 1131–1139 (2013).

    Article  CAS  PubMed  Google Scholar 

  38. Okamoto, I. et al. Eutherian mammals use diverse strategies to initiate X-chromosome inactivation during development. Nature 472, 370–374 (2011).

    Article  CAS  PubMed  Google Scholar 

  39. Petropoulos, S. et al. Single-cell RNA-seq reveals lineage and X chromosome dynamics in human preimplantation embryos. Cell 165, 1012–1026 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Shlyueva, D., Stampfel, G. & Stark, A. Transcriptional enhancers: from properties to genome-wide predictions. Nat. Rev. Genet. 15, 272–286 (2014).

    Article  CAS  PubMed  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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Tsankov, A. M. et al. Transcription factor binding dynamics during human ES cell differentiation. Nature 518, 344–349 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Stadler, M. B. et al. DNA-binding factors shape the mouse methylome at distal regulatory regions. Nature 480, 490–495 (2011).

    Article  CAS  PubMed  Google Scholar 

  44. Gao, L. et al. Chromatin accessibility landscape in human early embryos and its association with evolution. Cell 173, 248–259 (2018).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  46. Williams, B. R. et al. Aneuploidy affects proliferation and spontaneous immortalization in mammalian cells. Nat. Genet. 322, 703–709 (2008).

    CAS  Google Scholar 

  47. Rivera, C. M. & Ren, B. Mapping human epigenomes. Cell 155, 39–55 (2013).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  49. Mulqueen, R. M. et al. Highly scalable generation of DNA methylation profiles in single cells. Nat. Biotechnol. 36, 428–431 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Olova, N. et al. Comparison of whole-genome bisulfite sequencing library preparation strategies identifies sources of biases affecting DNA methylation data. Genome Biol. 19, 33 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Li, R., Qiao, J., Wang, L., Zhen, X. & Lu, Y. Serum progesterone concentration on day of HCG administration and IVF outcome. Reprod. Biomed. Online 16, 627–631 (2008).

    Article  CAS  PubMed  Google Scholar 

  52. Niakan, K. K., Han, J., Pedersen, R. A., Simon, C. & Pera, R. A. Human pre-implantation embryo development. Development 139, 829–841 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Krueger, F. & Andrews, S. R. Bismark: a flexible aligner and methylation caller for bisulfite-seq applications. Bioinformatics 27, 1571–1572 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Eichten, S. R., Stuart, T., Srivastava, A., Lister, R. & Borevitz, J. O. DNA methylation profiles of diverse Brachypodium distachyon align with underlying genetic diversity. Genome Res. 26, 1520–1531 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. 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 

  57. Ha, G. et al. Integrative analysis of genome-wide loss of heterozygosity and monoallelic expression at nucleotide resolution reveals disrupted pathways in triple-negative breast cancer. Genome Res. 22, 1995–2007 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Statham, A. L., Taberlay, P. C., Kelly, T. K., Jones, P. A. & Clark, S. J. Genome-wide nucleosome occupancy and DNA methylation profiling of four human cell lines. Genom. Data 3, 94–96 (2015).

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Thurman, R. E. et al. The accessible chromatin landscape of the human genome. Nature 489, 75–82 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. 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 

  62. Van der Auwera, G. A. et al. From FastQ data to high confidence variant calls: the Genome Analysis Toolkit best practices pipeline. Curr. Protoc. Bioinformatics 43, 11.10.1–11.10.33 (2013).

    Google Scholar 

  63. Li, H. Toward better understanding of artifacts in variant calling from high-coverage samples. Bioinformatics 30, 2843–2851 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Falcon, S. & Gentleman, R. Using GOstats to test gene lists for GO term association. Bioinformatics 23, 257–258 (2007).

    Article  CAS  PubMed  Google Scholar 

  65. Burger, L., Gaidatzis, D., Schubeler, D. & Stadler, M. B. Identification of active regulatory regions from DNA methylation data. Nucleic Acids Res. 41, e155 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by grants from the Ministry of Science and Technology of China (2017YFA0102702 and 2017YFA0103402) and the National Natural Science Foundation of China (81521002 and 31625018). F.T. and J.Q. were also supported by a grant from the Beijing Municipal Science and Technology Commission (D151100002415000). This work was also supported by the Beijing Advanced Innovation Center for Genomics at Peking University. Some of the bioinformatics analyses were conducted on the Computing Platform at the Center for Life Science.

Author information

Authors and Affiliations

Authors

Contributions

F.T. and J.Q. conceived the project. F.G., Y.G., Y.R., P.Y., L.Y., R.L., Y.L., J.L. and J.G. performed the experiments. L.L. and B.H. conducted the bioinformatics analyses. F.T., L.L. and Y.G. wrote the manuscript with help from all authors.

Corresponding authors

Correspondence to Fuchou Tang or Jie Qiao.

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 CNV and quality control of scCOOL-seq samples.

(a) Summary of CNV of each chromosome in single gametes, blastomeres and hESCs. The number of cells, including euploid and aneuploid cells, in each stage are listed on the left. Twenty-three α-Amanitin treated 8-cell blastomeres are not included. (b) Barplot showing the number of single cells with CNV on each chromosome. (c) Comparison analysis of average chromatin accessibility between euploid and aneuploid cells from zygote to TE. A two-tailed Student’s t-test was used to calculate the significance between two groups based on the normalized GCH methylation, that is, the average GCH methylation of each euploid or aneuploid cell is normalized by the mean value of all euploid cells within that stage. The mean value of all euploid cells in each stage was normalized to 1. P = 0.20. ns, not significant. The sample size in this panel corresponds to the euploid and aneuploid sample sizes provided in Fig. 1a (expect for euploid 8-cell blastomeres, n = 31 cells). The center represents mean, whereas error bars represent 1*SEM. (d) Boxplot showing the mapping rate of scCOOL-seq sample across each stage. (e-f) Boxplots showing the number of WCG (ACG/TCG) (e) and GCH (GCA/GCC/GCT) sites (f) covered in a single-cell sample across each stage at 1x depth. Each box in d-f represents median and 25% and 75% quantiles, while whiskers indicate 1.5 times of IQR. (g-h) Chromatin accessibility and DNA methylation coverage of RefSeq gene promoters (g) and CGIs (h) of scCOOL-seq libraries. Only regions covering 5 GCH or 3 WCG sites were calculated. The last row named ‘Mean’ is the mean coverage of all analyzed single cells. The sample size in d-h corresponds to the euploid sample sizes provided in Fig. 1a (expect for 8-cell, n = 31 cells).

Supplementary Figure 2 Chromatin accessibility around transcription start sites.

(a) Chromatin accessibility 2 kb upstream of the TSS, within the gene body, and 2 kb downstream of the TES of pools of samples across each stage. (b) Chromatin accessibility (upper panel) and DNA methylation level (lower panel) of H3K4me3 marked (in red) and unmarked (in blue) promoters. (c) Chromatin accessibility and DNA methylation levels of promoters with different expression levels. Genes were divided into 4 groups, including highly expressed (RPKM > 10), intermediately expressed (1 < RPKM ≤ 10), and lowly expressed genes (0.1 < RPKM ≤ 1) and genes that were not expressed (RPKM ≤ 0.1). The sample size in b-c corresponds to 3 euploid hESCs. (d) GO analysis of 8,385 genes that turned into an open state after fertilization (Related to Fig. 1d). The sample size in a, d corresponds to the euploid sample sizes provided in Fig. 1a (expect for 8-cell, n = 31 cells).

Supplementary Figure 3 Chromatin accessibility and DNA methylation level around the center of NDRs and NORs.

(a) Diagram of the number of TSS NDRs (NDRs containing TSS), proximal NDRs (NDRs within 2 kb upstream and downstream of the TSS) and distal NDRs (NDRs at least 2 kb away from the TSS) called at each stage from pools of samples. The last row named ‘Total’ is the total number of NDRs called from oocytes to hESCs. Overlapping NDRs longer than 10 bp were elongated and combined as one NDR. (b) Boxplot showing the dynamics of the length of TSS NDR distributions during human preimplantation development. The number of TSS NDR in each stage are shown in Supplementary Fig. 3a. Each box represents median and 25% and 75% quantiles, while whiskers indicate 1.5 times of IQR. (c) Chromatin accessibility and DNA methylation levels around the center of proximal NDRs and distal NDRs. (d) Chromatin accessibility and DNA methylation levels around the center of NORs. The sample size in a-d corresponds to the euploid sample sizes provided in Fig. 1a (expect for 8-cell, n = 31 cells).

Supplementary Figure 4 Heterogeneity of TSS NDRs among individual blastomeres from the 8-cell to blastocyst stage.

(a) RNA expression levels of 3 classes of genes during human preimplantation development. A two-tailed Student’s t-test was used to calculate the significance between comparisons. ns, not significant. The numbers of gene promoters in 3 classes defined in each stage were shown on the left of heat map in Fig. 3e and Supplementary Fig. 4c. (b) Coefficient of variation (CV) of expression levels of homogeneously open, divergent and homogeneously closed genes among individual cells during human preimplantation development. A two-tailed Student’s t-test was used to calculate the significance between comparisons. ns, not significant. The numbers of gene promoters in 3 classes defined in each stage were shown on the left of heat map in Fig. 3e and Supplementary Fig. 4c. Each box in a-b represents median and 25% and 75% quantiles, while whiskers indicate 1.5 times of IQR. (c) Heat map showing chromatin accessibility, DNA methylation levels and enriched GO terms of homogeneously open, divergent and homogeneously closed genes from the 8-cell to blastocyst stage. “n” on the left represents the number of promoters in each class. The sample size in a-c corresponds to the euploid sample sizes provided in Fig. 1a (expect for 8-cell, n = 31 cells).

Supplementary Figure 5 Differential DNA methylation and chromatin accessibility between parental genomes within each aneuploid blastomere during human preimplantation development.

(a) The global pattern of differential DNA methylation levels (left panel) and chromatin accessibility (right panel) between parental genomes within individual aneuploid blastomeres. The green bar represents the difference between euploid oocytes and sperm. (b) The difference in DNA methylation levels and chromatin accessibility between parental genomes within individual aneuploid blastomeres in different genomic regions. The green bar represents the difference between euploid oocytes and sperm. The sample size in a-b corresponds to the aneuploid sample sizes provided in Fig. 1a.

Supplementary Figure 6 Chromatin accessibility evolutionary comparison and the linkage between DNA methylation variance and DNA methylation level of gene promoter regions during human preimplantation development.

(a) Chromatin accessibility evolutionary comparison between human and mouse preimplantation embryos was performed using t-SNE (t-Distributed Stochastic Neighbor Embedding) analysis based on 15,057 human-mouse homologous proximal NDRs longer than 100 bp (NDRs within 2 kb upstream and downstream of the TSS) called from oocytes to ESCs. (b) Density plot of the relationship between the DNA methylation variance of the promoter regions (1 kb upstream and 0.5 kb downstream of the TSS) and the DNA methylation level of the corresponding regions among individual cells in each stage. The sample size in a-b corresponds to the euploid sample sizes provided in Fig. 1a (expect for 8-cell, n = 31 cells).

Supplementary Figure 7 The linkage between DNA methylation reprogramming and chromatin remodeling of gene promoter regions during human preimplantation development.

(a) Density plot of the relationship between the DNA methylation variance of the promoter regions (1 kb upstream and 0.5 kb downstream of the TSS) and the chromatin accessibility of the corresponding regions (200 bp upstream and 100 bp downstream of the TSS) among individual cells in each stage. (b) Density plot of the relationship between the chromatin accessibility variance of the promoter regions and the DNA methylation level of the corresponding regions among individual cells in each stage. The sample size in a-b corresponds to the euploid sample sizes provided in Fig. 1a (expect for 8-cell, n = 31 cells).

Supplementary Figure 8 Chromatin accessibility of repeat elements, subfamilies and around putative TF binding sites.

(a) The chromatin accessibility (left panel) and DNA methylation level (right panel) of SVAs. (b) The chromatin accessibility (left panel) and DNA methylation level (right panel) of the subfamilies of LINEs. L1, red line; L2, blue line. (c) The chromatin accessibility (left panel) and DNA methylation level (right panel) of the subfamilies of SINEs. Alu, red line; MIR, blue line. In a-c, the center represents mean, whereas error bars represent 1*SEM. (d) Boxplot showing the chromatin accessibility of repeat elements during human preimplantation development. Each box represents median and 25% and 75% quantiles, while whiskers indicate 1.5 times of IQR. (e) Heat map showing the openness of SOX2 and NANOG putative binding sites identified in hESCs during human preimplantation development (see the legend of Fig. 7d for details.). The sample size in a-e corresponds to the euploid sample sizes provided in Fig. 1a (expect for 8-cell, n = 31 cells). (f) Venn diagram shows the overlap between human-mouse homologous de-novo predicted enhancers based on open chromatin and low-methylated regions (LMRs). De-novo predicted enhancers in human preimplantation development and hESCs are listed in the Supplementary Table 4.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, L., Guo, F., Gao, Y. et al. Single-cell multi-omics sequencing of human early embryos. Nat Cell Biol 20, 847–858 (2018). https://doi.org/10.1038/s41556-018-0123-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-018-0123-2

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