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 profiling of human preimplantation embryos identifies cytoskeletal defects during embryonic arrest

Abstract

Human in vitro fertilized embryos exhibit low developmental capabilities, and the mechanisms that underlie embryonic arrest remain unclear. Here using a single-cell multi-omics sequencing approach, we simultaneously analysed alterations in the transcriptome, chromatin accessibility and the DNA methylome in human embryonic arrest due to unexplained reasons. Arrested embryos displayed transcriptome disorders, including a distorted microtubule cytoskeleton, increased genomic instability and impaired glycolysis, which were coordinated with multiple epigenetic reprogramming defects. We identified Aurora A kinase (AURKA) repression as a cause of embryonic arrest. Mechanistically, arrested embryos induced through AURKA inhibition resembled the reprogramming abnormalities of natural embryonic arrest in terms of the transcriptome, the DNA methylome, chromatin accessibility and H3K4me3 modifications. Mitosis-independent sequential activation of the zygotic genome in arrested embryos showed that YY1 contributed to human major zygotic genome activation. Collectively, our study decodes the reprogramming abnormalities and mechanisms of human embryonic arrest and the key regulators of zygotic genome activation.

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: Overview of multi-omics reprogramming defects in human arrested embryos.
Fig. 2: ZGA is linked to epigenetic reprogramming defects in human arrested embryos.
Fig. 3: YY1 is crucial for human ZGA.
Fig. 4: AURKA repression results in human and mouse embryonic arrest through the induction of microtubule cytoskeletal abnormalities.
Fig. 5: AURKA triggers genomic instability in human and mouse arrested embryos.
Fig. 6: AURKA impedes glycolysis in human arrested embryos.
Fig. 7: Human arrested embryos from the AURKA-inhibited group resemble the reprogramming defects of natural embryonic arrest as revealed by single-cell multi-omics data.

Similar content being viewed by others

Data availability

All raw sequencing data of human embryos reported in this study have been deposited into the Genome Sequence Archive (GSA) for human database with the accession number HRA003366. All raw sequencing data of mouse embryos reported in this study have been deposited into the GSA database with the accession number CRA012505. All processed data of human embryos generated in this study have been deposited into the Gene Expression Omnibus (GEO; https://www.ncbi.nlm.nih.gov/geo/) under accession number GSE247678. All processed data of mouse embryos generated in this study have been deposited in the OMIX database (China National Center for Bioinformation/Beijing Institute of Genomics, Chinese Academy of Sciences) with accession number OMIX004867. Previously published data that were re-analysed in this study are available under the following accession codes from the GEO database: GSE36552 and GSE133856 (scRNA-seq datasets of human early embryos); GSE100272 (scCOOL-seq datasets of human early embryos); GSE124718 (H3K4me3 ChIP–seq datasets of human 4-cell embryos and 8-cell embryos, then converted to hg38 reference using UCSC liftOver tool)92; GSM803513 (ChIP–seq datasets of YY1 in H1 hESCs); GSM3738360 (ChIP–seq datasets of KLF4 in naive hESCs); GSM2026849 (ChIP–seq datasets of ZFP42 in HEK 293T cells); GSE76495 (RNA-seq data of YY1 in HEK 293T cells); and GSE214608 (RNA-seq data of wild type HEK293 cells). For Extended Data Fig. 3b and Supplementary Fig. 4b, raw data were downloaded from the GEO (identifier PRJNA603589 (single-embryo RNA-seq datasets of TUBB8 mutant human arrested zygote)) and were processed according to the methods of the original article4, and the European Bioinformatics Institute (identifier E-MTAB-3929 (scRNA-seq datasets of day 5 blastocyst))105. Further information of resources is available from the lead contact Lin Li on reasonable request. Source data are provided with this paper.

References

  1. Keiding, N. et al. The use of time to pregnancy for estimating and monitoring human fecundity from demographic and health surveys. Epidemiology 32, 27 (2021).

    Article  PubMed  Google Scholar 

  2. Liu, L., Cai, J., Chen, P., Sha, A. & Ren, J. Clinical outcome of IVF/ICSI cycles with an arrested embryo on day 3. Int. J. Clin. Exp. Med. 9, 16414–16424 (2016).

    Google Scholar 

  3. Vastenhouw, N. L., Cao, W. X. & Lipshitz, H. D. The maternal-to-zygotic transition revisited. Development 146, dev161471 (2019).

    Article  CAS  PubMed  Google Scholar 

  4. Sha, Q.-Q. et al. Dynamics and clinical relevance of maternal mRNA clearance during the oocyte-to-embryo transition in humans. Nat. Commun. 11, 4917 (2020).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  5. Hernandez Mora, J. R. et al. Single-cell multi-omic analysis profiles defective genome activation and epigenetic reprogramming associated with human pre-implantation embryo arrest. Cell Rep. 42, 112100 (2023).

    Article  CAS  PubMed  Google Scholar 

  6. Favetta, L. A., St John, E. J., King, W. A. & Betts, D. H. High levels of p66shc and intracellular ROS in permanently arrested early embryos. Free Radic. Biol. Med. 42, 1201–1210 (2007).

    Article  CAS  PubMed  Google Scholar 

  7. Yang, Y. et al. Metabolic and epigenetic dysfunctions underlie the arrest of in vitro fertilized human embryos in a senescent-like state. PLoS Biol. 20, e3001682 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Chen, B. et al. Novel mutations and structural deletions in TUBB8: expanding mutational and phenotypic spectrum of patients with arrest in oocyte maturation, fertilization or early embryonic development. Hum. Reprod. 32, 457–464 (2017).

    Article  CAS  PubMed  Google Scholar 

  9. Zhang, H. et al. Dominant mutations in CHK1 cause pronuclear fusion failure and zygote arrest that can be rescued by CHK1 inhibitor. Cell Res. 31, 814–817 (2021).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  10. Wang, W. et al. FBXO43 variants in patients with female infertility characterized by early embryonic arrest. Hum. Reprod. 36, 2392–2402 (2021).

    Article  CAS  PubMed  Google Scholar 

  11. Gao, L. et al. Chromatin accessibility landscape in human early embryos and its association with evolution. Cell 173, 248–259.e15 (2018).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  ADS  Google Scholar 

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

    Article  CAS  PubMed  ADS  Google Scholar 

  14. Li, L. et al. Single-cell multi-omics sequencing of human early embryos. Nat. Cell Biol. 20, 847–858 (2018).

    Article  CAS  PubMed  Google Scholar 

  15. Snider, A. C. et al. The chromatin remodeling factor Chd1l is required in the preimplantation embryo. Biol. Open 2, 121–131 (2013).

    Article  CAS  PubMed  Google Scholar 

  16. Arand, J., Reijo Pera, R. A. & Wossidlo, M. Reprogramming of DNA methylation is linked to successful human preimplantation development. Histochem. Cell Biol. 156, 197–207 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Wang, J. et al. Single-cell multiomics sequencing reveals the reprogramming defects in embryos generated by round spermatid injection. Sci. Adv. 8, eabm3976 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Huang, Y. et al. Single-cell multi-omics sequencing of human spermatogenesis reveals a DNA demethylation event associated with male meiotic recombination. Nat. Cell Biol. 25, 1520–1534 (2023).

    Article  CAS  PubMed  Google Scholar 

  19. 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 

  20. Leng, L. et al. Single-cell transcriptome analysis of uniparental embryos reveals parent-of-origin effects on human preimplantation development. Cell Stem Cell 25, 697–712.e6 (2019).

    Article  CAS  PubMed  Google Scholar 

  21. Djekidel, M. N. et al. Reprogramming of chromatin accessibility in somatic cell nuclear transfer is DNA replication independent. Cell Rep. 23, 1939–1947 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Chen, X. et al. Key role for CTCF in establishing chromatin structure in human embryos. Nature 576, 306–310 (2019).

    Article  CAS  PubMed  Google Scholar 

  23. Liu, L. et al. An integrated chromatin accessibility and transcriptome landscape of human pre-implantation embryos. Nat. Commun. 10, 364 (2019).

    Article  PubMed  PubMed Central  ADS  Google Scholar 

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

    Article  CAS  PubMed  ADS  Google Scholar 

  25. Bu, G. et al. Coordination of zygotic genome activation entry and exit by H3K4me3 and H3K27me3 in porcine early embryos. Genome Res. 32, 1487–1501 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  27. Klemm, S. L., Shipony, Z. & Greenleaf, W. J. Chromatin accessibility and the regulatory epigenome. Nat. Rev. Genet. 20, 207–220 (2019).

    Article  CAS  PubMed  Google Scholar 

  28. Bevilacqua, A., Fiorenza, M. T. & Mangia, F. A developmentally regulated GAGA box-binding factor and Sp1 are required for transcription of the hsp70.1 gene at the onset of mouse zygotic genome activation. Development 127, 1541–1551 (2000).

    Article  CAS  PubMed  Google Scholar 

  29. Gassler, J. et al. Zygotic genome activation by the totipotency pioneer factor Nr5a2. Science 378, 1305–1315 (2022).

    Article  CAS  PubMed  ADS  Google Scholar 

  30. Zou, Z. et al. Translatome and transcriptome co-profiling reveals a role of TPRXs in human zygotic genome activation. Science 378, abo7923 (2022).

    Article  PubMed  Google Scholar 

  31. Yuan, S. et al. Human zygotic genome activation is initiated from paternal genome. Cell Discov. 9, 13 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Gertz, J. et al. Distinct properties of cell-type-specific and shared transcription factor binding sites. Mol. Cell 52, 25–36 (2013).

    Article  CAS  PubMed  Google Scholar 

  33. Pontis, J. et al. Hominoid-specific transposable elements and KZFPs facilitate human embryonic genome activation and control transcription in naive human ESCs. Cell Stem Cell 24, 724–735.e5 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Schmitges, F. W. et al. Multiparameter functional diversity of human C2H2 zinc finger proteins. Genome Res. 26, 1742–1752 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Zhang, P., Frederick, M. I. & Heinemann, I. U. Terminal uridylyltransferases TUT4/7 regulate microRNA and mRNA homeostasis. Cells 11, 3742 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Nikonova, A. S., Astsaturov, I., Serebriiskii, I. G., Dunbrack, R. L. Jr & Golemis, E. A. Aurora A kinase (AURKA) in normal and pathological cell division. Cell. Mol. Life Sci. 70, 661–687 (2013).

    Article  CAS  PubMed  Google Scholar 

  37. Ding, J., Swain, J. E. & Smith, G. D. Aurora kinase‐A regulates microtubule organizing center (MTOC) localization, chromosome dynamics, and histone‐H3 phosphorylation in mouse oocytes. Mol. Reprod. Dev. 78, 80–90 (2011).

    Article  CAS  PubMed  Google Scholar 

  38. Wang, X., Baumann, C., De La Fuente, R. & Viveiros, M. M. CEP215 and AURKA regulate spindle pole focusing and aMTOC organization in mouse oocytes. Reproduction 159, 261–274 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Kovarikova, V. et al. Aurora kinase A is essential for correct chromosome segregation in mouse zygote. Zygote 24, 326–337 (2016).

    Article  CAS  PubMed  Google Scholar 

  40. Guo, M. et al. Precise temporal regulation of Dux is important for embryo development. Cell Res. 29, 956–959 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Nguyen, T. T. T. et al. PLEKHG3 enhances polarized cell migration by activating actin filaments at the cell front. Proc. Natl Acad. Sci. USA 113, 10091–10096 (2016).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  42. Kosla, J., Dvorak, M. & Cermak, V. Molecular analysis of the TGF-β controlled gene expression program in chicken embryo dermal myofibroblasts. Gene 513, 90–100 (2013).

    Article  CAS  PubMed  Google Scholar 

  43. Wang, B. et al. Acyl‐CoA thioesterase 9 promotes tumour growth and metastasis through reprogramming of fatty acid metabolism in hepatocellular carcinoma. Liver Int. 42, 2548–2561 (2022).

    Article  CAS  PubMed  Google Scholar 

  44. Toyo-oka, K. et al. Loss of the Max-interacting protein Mnt in mice results in decreased viability, defective embryonic growth and craniofacial defects: relevance to Miller–Dieker syndrome. Hum. Mol. Genet. 13, 1057–1067 (2004).

    Article  CAS  PubMed  Google Scholar 

  45. Yanagisawa, H., Schluterman, M. K. & Brekken, R. A. Fibulin-5, an integrin-binding matricellular protein: its function in development and disease. J. Cell Commun. Signal. 3, 337–347 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Kuht, H. J. et al. SLC38A8 mutations result in arrested retinal development with loss of cone photoreceptor specialization. Hum. Mol. Genet. 29, 2989–3002 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Tong, H. et al. Transcriptomic analysis of gene expression profiles of stomach carcinoma reveal abnormal expression of mitotic components. Life Sci. 170, 41–49 (2017).

    Article  CAS  PubMed  Google Scholar 

  48. Wang, Y. et al. iTALK: an R package to characterize and illustrate intercellular communication. Preprint at bioRxiv https://doi.org/10.1101/507871 (2019).

  49. Jalalirad, M. et al. Aurora-A kinase oncogenic signaling mediates TGF-β-induced triple-negative breast cancer plasticity and chemoresistance. Oncogene 40, 2509–2523 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Connell, M. et al. HMMR acts in the PLK1-dependent spindle positioning pathway and supports neural development. eLife 6, e28672 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Pujana, M. A. et al. Network modeling links breast cancer susceptibility and centrosome dysfunction. Nat. Genet. 39, 1338–1349 (2007).

    Article  CAS  PubMed  Google Scholar 

  52. Younger, S. T. & Rinn, J. L. p53 regulates enhancer accessibility and activity in response to DNA damage. Nucleic Acids Res. 45, 9889–9900 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Toiber, D. et al. SIRT6 recruits SNF2H to DNA break sites, preventing genomic instability through chromatin remodeling. Mol. Cell 51, 454–468 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Mantikou, E., Wong, K. M., Repping, S. & Mastenbroek, S. Molecular origin of mitotic aneuploidies in preimplantation embryos. Biochim. Biophys. Acta 1822, 1921–1930 (2012).

    Article  CAS  PubMed  Google Scholar 

  55. Vera-Rodriguez, M., Chavez, S. L., Rubio, C., Pera, R. A. R. & Simon, C. Prediction model for aneuploidy in early human embryo development revealed by single-cell analysis. Nat. Commun. 6, 7601 (2015).

    Article  PubMed  ADS  Google Scholar 

  56. Janssen, A., van der Burg, M., Szuhai, K., Kops, G. J. P. L. & Medema, R. H. Chromosome segregation errors as a cause of DNA damage and structural chromosome aberrations. Science 333, 1895–1898 (2011).

    Article  CAS  PubMed  ADS  Google Scholar 

  57. Sheltzer, J. M. et al. Aneuploidy drives genomic instability in yeast. Science 333, 1026–1030 (2011).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  58. Qi, S.-T., Liang, L.-F., Xian, Y.-X., Liu, J.-Q. & Wang, W. Arrested human embryos are more likely to have abnormal chromosomes than developing embryos from women of advanced maternal age. J. Ovarian Res. 7, 65 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Maurer, M. et al. Chromosomal aneuploidies and early embryonic developmental arrest. Int. J. Fertil. Steril. 9, 346–353 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Birsoy, K. et al. An essential role of the mitochondrial electron transport chain in cell proliferation is to enable aspartate synthesis. Cell 162, 540–551 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Nguyen, T. T. T. et al. Aurora kinase A inhibition reverses the Warburg effect and elicits unique metabolic vulnerabilities in glioblastoma. Nat. Commun. 12, 5203 (2021).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  62. Milazzotto, M. P., Noonan, M. J. & Ferraz, M. D. A. M. M. Mining RNAseq data reveals dynamic metaboloepigenetic profiles in human, mouse and bovine pre-implantation embryos. iScience 25, 103904 (2022).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  63. Zhao, J. et al. Metabolic remodelling during early mouse embryo development. Nat. Metab. 3, 1372–1384 (2021).

    Article  CAS  PubMed  Google Scholar 

  64. Donohoe, M. E. et al. Targeted disruption of mouse Yin Yang 1 transcription factor results in peri-implantation lethality. Mol. Cell. Biol. 19, 7237–7244 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Ding, Y.-H. et al. Alisertib, an Aurora kinase A inhibitor, induces apoptosis and autophagy but inhibits epithelial to mesenchymal transition in human epithelial ovarian cancer cells. Drug. Des. Devel. Ther. 9, 425–464 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Dong, J. et al. Single-cell RNA-seq analysis unveils a prevalent epithelial/mesenchymal hybrid state during mouse organogenesis. Genome Biol. 19, 31 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  67. Behringer, R., Gertsenstein, M., Nagy, K. V. & Nagy, A. Manipulating the Mouse Embryo: A Laboratory Manual (Cold Spring Harbor Lab. Press, 2014).

  68. Navarro Gonzalez, J. et al. The UCSC genome browser database: 2021 update. Nucleic Acids Res. 49, D1046–D1057 (2021).

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  70. Anders, S., Pyl, P. T. & Huber, W. HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).

    Article  CAS  PubMed  Google Scholar 

  71. Krueger, F. Trim Galore: a wrapper tool around Cutadapt and FastQC to consistently apply quality and adapter trimming to FastQ files, with some extra functionality for MspI-digested RRBS-type (Reduced Representation Bisufite-Seq) libraries. http://www.bioinformatics.babraham.ac.uk/projects/trim_galore/ (2012).

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Li, H. et al. The sequence alignment/map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  74. Ranjan, B. et al. DUBStepR is a scalable correlation-based feature selection method for accurately clustering single-cell data. Nat. Commun. 12, 5849 (2021).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  75. Angerer, P. et al. destiny: diffusion maps for large-scale single-cell data in R. Bioinformatics 32, 1241–1243 (2016).

    Article  CAS  PubMed  Google Scholar 

  76. De Vries, A., Ripley, B. D. & de Vries, M. A. ggdendro: Create dendrograms and tree diagrams using ggplot2[J]. R package version 0.1-22 (2020).

  77. Leek, J. T., Johnson, W. E., Parker, H. S., Jaffe, A. E. & Storey, J. D. The sva package for removing batch effects and other unwanted variation in high-throughput experiments. Bioinformatics 28, 882–883 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Park, S.-J., Shirahige, K., Ohsugi, M. & Nakai, K. DBTMEE: a database of transcriptome in mouse early embryos. Nucleic Acids Res. 43, D771–D776 (2015).

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  80. Zhou, Y. et al. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat. Commun. 10, 1523 (2019).

    Article  PubMed  PubMed Central  ADS  Google Scholar 

  81. Wickham, H., Chang, W. & Wickham, M. H. ggplot2: create elegant data visualisations using the grammar of graphics. R package version 3.3.5,1-189 (2016).

  82. Korotkevich, G. et al. Fast gene set enrichment analysis. Preprint at bioRxiv https://doi.org/10.1101/060012 (2021).

  83. Yu, G., Wang, L.-G., Han, Y. & He, Q.-Y. clusterProfiler: an R package for comparing biological themes among gene clusters. Omics 16, 284–287 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Liberzon, A. et al. Molecular signatures database (MSigDB) 3.0. Bioinformatics 27, 1739–1740 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Jin, Y., Tam, O. H., Paniagua, E. & Hammell, M. TEtranscripts: a package for including transposable elements in differential expression analysis of RNA-seq datasets. Bioinformatics 31, 3593–3599 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Kumar, N., Mishra, B., Athar, M. & Mukhtar, S. Inference of gene regulatory network from single-cell transcriptomic data using pySCENIC. Methods Mol. Biol. 2328, 171–182 (2021).

    Article  CAS  PubMed  Google Scholar 

  87. Moerman, T. et al. GRNBoost2 and Arboreto: efficient and scalable inference of gene regulatory networks. Bioinformatics 35, 2159–2161 (2019).

    Article  CAS  PubMed  Google Scholar 

  88. Shannon, P. et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 13, 2498–2504 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Gu, Z., Gu, L., Eils, R., Schlesner, M. & Brors, B. circlize implements and enhances circular visualization in R. Bioinformatics 30, 2811–2812 (2014).

  90. 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  ADS  Google Scholar 

  91. Lai, D. & Ha, G. HMMcopy: a package for bias-free copy number estimation and robust CNA detection in tumour samples from WGS HTS data. R package version 1.30.0 (2013).

  92. Kuhn, R. M., Haussler, D. & Kent, W. J. The UCSC genome browser and associated tools. Brief. Bioinform. 14, 144–161 (2013).

    Article  CAS  PubMed  Google Scholar 

  93. 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 

  94. Ramírez, F., Dündar, F., Diehl, S., Grüning, B. A. & Manke, T. deepTools: a flexible platform for exploring deep-sequencing data. Nucleic Acids Res. 42, W187–W191 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  95. Stacklies, W., Redestig, H., Scholz, M., Walther, D. & Selbig, J. pcaMethods—a bioconductor package providing PCA methods for incomplete data. Bioinformatics 23, 1164–1167 (2007).

    Article  CAS  PubMed  Google Scholar 

  96. Street, K. et al. Slingshot: cell lineage and pseudotime inference for single-cell transcriptomics. BMC Genomics 19, 477 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  97. Danese, A. et al. EpiScanpy: integrated single-cell epigenomic analysis. Nat. Commun. 12, 5228 (2021).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  98. McLeay, R. C. & Bailey, T. L. Motif enrichment analysis: a unified framework and an evaluation on ChIP data. BMC Bioinformatics 11, 165 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  99. Madsen, J. G. S. et al. Integrated analysis of motif activity and gene expression changes of transcription factors. Genome Res. 28, 243–255 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  ADS  Google Scholar 

  101. Gu, Z., Eils, R. & Schlesner, M. Complex heatmaps reveal patterns and correlations in multidimensional genomic data. Bioinformatics 32, 2847–2849 (2016).

    Article  CAS  PubMed  Google Scholar 

  102. Wybrow, M., Rodgers, P. & Dib, F. K. Euler diagrams drawn with ellipses area-proportionally (Edeap). BMC Bioinformatics 22, 214 (2021).

  103. Gao, Y. et al. 5-Formylcytosine landscapes of human preimplantation embryos at single-cell resolution. PLoS Biol. 18, e3000799 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  105. 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 

Download references

Acknowledgements

We thank X. Zhao from Southern Medical University and G. Chang from Shenzhen University Medical School for suggestions and discussion. This study was supported by grants from the National Key R&D Program of China (2021YFA1102700 to Lin Li), the National Natural Science Foundation of China (82101745 and 32070833 to Lin Li, 81871211 to Lei Li, 81901566 to R.H. and 81971452 to J.L), the Natural Science Funds for Distinguished Young Scholar of Guangdong province (2022B1515020110 to Lin Li), and the Key-Area Research and Development Program of Guangdong Province, Modernization of Chinese medicine in Lingnan (2020B1111100011 to Lin Li).

Author information

Authors and Affiliations

Authors

Contributions

Lin Li conceived and supervised the project. T.W., J.F. and J.P. facilitated its designs. N.T., X.Z., Y.B., X.K., J.L., L.Z. and Lei Li collected the human arrested and 3PN embryos. J.P. performed library constructions of the single-cell/embryo multi-omics sequencing data. X.Z. performed the siRNA microinjection experiments in human early embryos. J.P., R.H., Zhihao Wang, L.W., Y.B., X.Q., Zimeng Wang, C.L. and W.Z. performed the other experiments. T.W. and J.F. conducted the bioinformatics analyses. Lin Li, T.W., J.F. and J.P. interpreted the data. Lin Li, T.W., J.F. and J.P. wrote the paper with assistance from all the authors.

Corresponding authors

Correspondence to Lei Li or Lin Li.

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.

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 The developmental characteristics and cell morphologies of human IVF embryos.

a. Bar graph showing developmental outcomes of 610 human 2PN embryos underwent IVF procedure. Human arrested embryos are those without further development for another 2 days at the zygote and cleavage stages. Embryos that were developmentally delayed, fragmented or dead were excluded. b. Bar graph showing the number of human embryos arrested at different developmental stages from a. c. Representative bright-field images of human normal and arrested embryos at different developmental stages. Scale bars, 20 μm. d. Chromatin accessibility around the promoter regions (TSS ± 2 kb) in human arrested embryos at each stage. Lines are colored according to developmental stage. TSS, transcription start site. e. The average DNA methylation level along the intragenic regions and their flanking regions in human arrested embryos at each stage. Lines are colored according to the developmental stage. TES, transcription end site. f, g. PCA projections of human normal and arrested embryos at each stage with RNA-part data from single-cell multi-omics sequencing, integrated with the cells from Yan et al.19 and Leng et al.20 In f, Single cells are colored according to data source. In g, single cells are colored according to stage. h. Diffusion map constructing the putative trajectory of human normal and arrested embryos at each stage with RNA-part data from single-cell multi-omics sequencing, integrated with the cells from Yan et al.19 and Leng et al.20. Single cells were colored according to data source.

Extended Data Fig. 2 The reprogramming defects in human arrested embryos.

a. The transcriptional activity (mean expression levels of all genes in each single cell) in human normal and arrested embryos. The number of biologically independent samples is 10, 10, 11, 10, 9 for human normal zygote to morula; 18, 13, 8, 7 for human arrested zygote to 8-cell embryos. b. The number of DEGs between human arrested and normal embryos. c. Hierarchical clustering of human normal and arrested embryos using RNA-part data from single-cell multi-omics sequencing. d. The number of arrested-specific hyper-DMRs (red) and arrested-specific hypo-DMRs (blue) located within promoter regions between human arrested and normal embryos. DMRs, differentially methylated regions. e. Lollipop graphs showing the proportion of arrested-specific hypo-DMRs in each stage of human arrested embryos overlapped with hypo-DMRs between consecutive stages in human normal embryos. f. The number of normal-specific NDRs (red) and arrested-specific NDRs (purple) between human arrested and normal embryos. NDRs, nucleosome-depleted regions. g. The expression levels of CTCF in human normal and arrested embryos. The number of biologically independent samples is 9, 10, 10, 11, 10, 9 for human normal MII to morula; 18, 13, 8, 7 for human arrested zygote to 8-cell embryos. h. RT-qPCR results showing the relative expression levels of CTCF in human normal (3PN) and arrested 2-cell or 8-cell embryos (2PN). Data are represented as mean ± SEM (3, 5, 4, 3 biologically independent samples); two-sided Mann-Whitney U test. In g, h, error bars represent mean ± SEM. i. The average expression levels of major ZGA genes in human normal 8-cell embryos (left) and human arrested zygotes to 8-cell embryos (right) that located within or outside topologically associating domains (TADs) identified from Hi-C data in human early embryos from Chen et al.22. In a, i, each box represents median, 25% and 75% quartiles; whiskers indicate 1.5 times the interquartile range; unpaired two-sided Student’s t-test without adjustment. j. GO terms of up-regulated (left) and down-regulated genes (right) in human arrested embryos compared to human normal embryos with similar morphologies. Hypergeometric test without adjustment.

Source data

Extended Data Fig. 3 The maternal-to-zygote transition is disorganized in human arrested embryos.

a. Venn plots showing the overlap between major ZGA genes and up-regulated genes in human arrested zygotes to 4-cell embryos. b. Radar chart showing the average expression levels of representative major ZGA genes in human TUBB8mutant arrested zygotes from Sha et al.4 and human normal zygotes from Yan et al.19 from 2, 3 biologically independent samples; integration of all samples. c. Expression patterns and GO terms of each cluster of up-regulated major ZGA genes in human arrested embryos. Left, heatmap showing k-means clustering (n = 4) of major ZGA genes that are commonly up-regulated in human arrested zygotes to 4-cell embryos. The color key from blue to red indicates the expression level from low to high. Middle, line plots showing the average expression patterns of genes in each cluster. Right, GO terms of genes in each cluster. In a, c, hypergeometric test without adjustment. d-f. Line plots showing the expression levels of d. SVA, e. representative TEs and f. M-decay genes in human normal and arrested embryos at each stage. TEs, transposable elements. CPM, count-per-million. The number of biologically independent samples is 9, 10, 10, 11, 10, 9 for human normal MII to morula; 18, 13, 8, 7 for human arrested zygote to 8-cell embryos. g, h. g. RT-qPCR results showing the relative expression levels of M-decay gene EIF3K in human normal (3PN) and arrested zygotes or 2-cell embryos (2PN). Each dot represents an embryo. Data are from 4, 4, 6, 3, 5 biologically independent samples. h. RT-qPCR results showing the relative expression levels of M-decay genes PABPC1L, TLE6, ZP2 in human normal (3PN) and arrested zygotes (2PN). Data are from 4, 4, 6 biologically independent samples. In c-h, error bars represent the mean ± SEM. In g, h, two-sided Mann-Whitney U test.

Source data

Extended Data Fig. 4 DNA hyper-methylation (intragenic regions) and excessive open chromatins link with up-regulated major ZGA genes in human arrested embryos.

a, f. Relative enrichment analysis of a. DMRs, f. differential NDRs between human normal and arrested embryos in different genomic regions. b. The expression levels of genes associated with arrested-specific hypo-DMRs and hyper-DMRs (promoter) in human arrested embryos. c. Heatmap showing the expression levels of DEGs in human arrested embryos compared with human normal embryos (left). Pie charts showing the proportion of DEGs associated with DMRs (promoter or intragenic, right). d, e. The relationship between gene expression and DNA methylation level of DMRs (intragenic) at d. zygote and e. 2-cell stages of major ZGA genes. Up-regulated major ZGA genes with significant changes in DNA methylation levels are labeled in red. g. The spearman correlation coefficients between the normalized chromatin accessibility (core promoter regions) and gene expression levels. The number of biologically independent samples is 20, 20, 22, 22 for human normal zygote to 8-cell embryos; 15, 13, 7, 6 biologically independent samples for human arrested ones. Error bars represent mean ± SEM. h. The average expression levels of genes associated with arrested-specific proximal-merged NDRs (mNDRs) and that associated with normal-specific proximal mNDRs in human arrested embryos. In b, h, data are from 10, 10, 11, 12 biologically independent samples for human normal zygote to 8-cell embryos; 15, 13, 7, 6 biologically independent samples for human arrested ones; each box represents the median, 25% and 75% quartiles; whiskers indicate 1.5 times the interquartile range; unpaired two-sided Student’s t-tests without adjustment. i. Heatmap showing the proximal NDRs that were specifically open in human normal or arrested embryos (left). Pie charts showing the proportion of DEGs associated with arrested or normal-specific mNDRs (proximal, right). j, k. The association between major ZGA genes and differential NDRs (proximal) in j. human arrested zygotes and k. 2-cell embryos. Red dots represent up-regulated major ZGA genes in human arrested embryos occupied with arrested-specific NDRs (proximal). In d, e, j, k, representative major ZGA genes are labeled in purple.

Source data

Extended Data Fig. 5 KDM5 or H3K4me3 erasure regulates human zygotic genome activation.

a. Line plot showing the average expression levels of KDM5B in human normal MII to morula and arrested embryos. The number of biologically independent samples is 9, 10, 10, 11, 10, 9 for human normal MII to morula; 18, 13, 8, 7 for human arrested zygote to 8-cell embryos. Error bars represent the mean ± SEM. b. RT-qPCR results showing the relative expression levels of KDM5B in human normal (3PN) and arrested zygotes (2PN). Data are represented as mean ± SEM from 4, 4, 6 biologically independent samples; two-sided Mann-Whitney U test. c. Flowchart of the experimental scheme for KDM5 inhibitor treatment of human zygote (3PN). Human 8-cell embryos (3PN) were collected on day 3 post in vitro fertilization (IVF). d. Immunofluorescence of H3K4me3 in human 8-cell embryos (developed from 3PN zygotes) from control and KDM5 inhibited group. Data are from 7, 10 biologically independent samples. Scale bar, 20 μm. Data are represented as mean ± SEM; unpaired two-sided Student’s t-test. e. Boxplots showing the expression levels of TEs in human control and KDM5 inhibited 8-cell embryos (developed from 3PN zygote). Each dot represents an embryo. Data are from 4, 5 biologically independent samples. Each box represents the median, 25% and 75% quartiles; whiskers indicate 1.5 times the interquartile range. Unpaired two-sided Student’s t-test. TEs, transposable elements. CPM, count-per million. f-h. Genome browser views showing the normalized chromatin accessibility of major ZGA genes accompanied with H3K4me3 decreases from human normal 4-cell (broad peak) to normal 8-cell (narrow peak) embryos in human normal and arrested f. zygotes, g. 2-cell embryos and h. 4-cell embryos. The expression levels of major ZGA genes are shown at right. Black bars represent GCH sites with normalized chromatin accessibility less than 1/10 of the normalized data range (but are detected). Bar plot under ChIP-seq signals represents peaks of H3K4me3 from Xia et al.13.

Source data

Extended Data Fig. 6 YY1 promotes human zygotic genome activation.

a. Regulatory network of key transcription factors (TFs) that are shared or unique among human arrested zygotes to 4-cell embryos predicted by SCENIC analysis. b. Bar graph showing the number of target genes that are regulated by TFs in human arrested zygotes to 4-cell embryos predicted by SCENIC analysis. Bars are colored according to developmental stage; integration of all samples. c. Line plots showing the average expression levels of KLF4 and ZFP42 in human normal MII to morula and arrested embryos at each stage. The number of biologically independent samples is 9, 10, 10, 11, 10, 9 for human normal MII to morula; 18, 13, 8, 7 for human arrested zygote to 8-cell embryos. d. Venn plot showing the overlap between major ZGA genes and the target genes of KLF4 or ZFP42 identified from public ChIP-seq data in human embryonic stem cells (hESCs)33,34. Hypergeometric test without adjustment. e. Venn plot showing the overlap among target genes that are regulated by YY1, KLF4 or ZFP42 identified from public ChIP-seq datasets in hESCs or HEK 293T cells32,33,34. f. Boxplots showing the average expression levels of major ZGA genes in human 2PN (from Yan et al.19, n = 16) and 3PN 8-cell embryos (n = 16). Data are from 3, 4 biologically independent samples. Each box represents the median, 25% and 75% quartiles; whiskers indicate 1.5 times the interquartile range. Unpaired two-sided Student’s t-test. g. Bar plot showing the expression levels of YY1 among YY1 KD and control human 8-cell embryos (developed from human 3PN zygotes). Each point represents an embryo. Data are from 3, 3, 2 biologically independent samples. In c, g, error bars represent mean ± SEM. h. Genome browser views showing YY1 signals from public ChIP-seq data in hESCs32.

Source data

Extended Data Fig. 7 Human and mouse arrested embryos display abnormal microtubule cytoskeleton and mitosis defects.

a. GSEA analysis showed the enriched terms that were down-regulated in human arrested embryos. Circle size: normalized enrichment score (NES); color: FDR. b, c. The average expression levels of b. representative cytoskeletal or mitotic genes, and c. ORC5. Data in b, c are from 9, 10, 10, 11, 10, 9 biologically independent samples for human normal MII to morula; 18, 13, 8, 7 for human arrested zygote to 8-cell embryos. d. RT-qPCR results showing the relative expression levels of ORC5 in human normal (3PN) and arrested zygotes (2PN) from 5, 11 biologically independent samples. e. Immunofluorescence of γ-tubulin (green) co-stained with β-tubulin (red) in human arrested zygotes (2PN, with magnified images showing nucleus). Scale bar, 20 μm. f. GO terms of down-regulated genes in mouse arrested zygotes from MLN8237 treated group. MLN8237, AURKA inhibitor. g. Immunofluorescence and h. statistical analysis of γ-tubulin signal intensity in mouse control (KSOM) and arrested zygotes (7 biologically independent samples). Scale bar, 20 μm. In b-d, h, error bars represent mean ± SEM. i. The average expression levels of representative cytoskeletal and mitotic genes in mouse arrested zygotes and control zygotes. j. Hierarchical clustering of mouse arrested zygotes and control zygotes, early two-cell (E2C) as well as late two-cell (L2C) embryos. k. The average expression levels of major ZGA genes in mouse arrested zygotes and mouse control embryos. Data are from 17, 10, 30, 17 biologically independent samples. l. The overlap between up-regulated genes in mouse arrested zygotes and mouse major ZGA genes. In f, l, hypergeometric test without adjustment. m. The expression levels of representative major ZGA genes in mouse arrested zygotes and control embryos. Data are from 17, 10, 30, 17 biologically independent samples. In k, m, each box represents the median, 25% and 75% quartiles; whiskers indicate 1.5 times the interquartile range. n. Scatter plots of TEs expression in mouse arrested zygotes compared with control zygotes. Blue dots represent differentially expressed TEs with representative ZGA TEs shown in red. In d, h, k, m, n, unpaired two-sided Student’s t-test without adjustment.

Source data

Extended Data Fig. 8 Aberrant reprogramming of DNA methylation and chromatin accessibility link with cytoskeleton defects in human arrested embryos.

a. Venn plot showing the overlap of genes associated with arrested-specific hypo-DMRs (promoter) among human arrested zygotes to 8-cell embryos. b. Differences in the DNA methylation level of autophagy and RNA splicing genes (promoter) between human arrested and normal embryos at each stage. Data are from biologically independent samples as indicated in Fig. 1f; integration of all samples. c. Genome browser views showing the DNA methylation levels of cytoskeletal and mitotic related genes in human normal and arrested embryos at each stage. The expression levels of genes are shown (right). The WCG sites detected with DNA methylation lower than 1/10 of the data range (but are detected) are shown as black bars. d. GO terms of genes associated with arrested-specific NDRs (see details in the Method section) and normal-specific NDRs (promoter) in human arrested embryos at each stage. Bars are colored according to developmental stage. Genes in GO terms that are consistent with gene expression levels are colored in red. Hypergeometric test without adjustment.

Source data

Extended Data Fig. 9 Human arrested 2-cell embryos present increased intra-embryonic heterogeneity.

a. Heatmap showing the variance of gene expression level in each blastomeres within an embryo in human normal and arrested 2-cell to 8-cell embryos (no more than one blastomere failed quality control within an embryo are allowed for 4-cell and 8-cell embryos). The color key from blue to red indicates the variance from low to high. The number of biologically independent samples is 6, 5, 2 for human normal 2-cell to 8-cell embryos; 4, 7, 3 for human arrested 2-cell to 8-cell embryos. b. GO terms of up-regulated genes between two blastomeres of human arrested 2-cell embryos. Hypergeometric test without adjustment. c. The expression levels of up-regulated genes between two blastomeres of human normal and arrested 2-cell embryos. Blastomeres are colored by transcription activity (blastomeres 1: higher transcription activity; blastomeres 2: lower transcription activity). The dispersion degree along the y-axis can be visualized in each embryo and represents the overall heterogeneity of gene expression levels between two blastomeres within each embryo. Error bars represent mean ± SEM. In a, c, unpaired two-sided Student’s t-test is used for statistical analysis. d, e. iTALK analysis of cell-cell communication between two blastomeres in d. human normal (left) or e. human arrested (right) 2-cell embryos. Data in c-e are from biologically independent samples as indicated in a.

Source data

Extended Data Fig. 10 Human arrested embryos display increased genomic instability.

a. Boxplots showing the gene expression levels (left), the DNA methylation level of WCG sites within DMRs (intragenic, middle), and chromatin accessibility of arrested-specific NDRs (promoter, right) in individual cell of human normal and arrested 4-cell embryos. Data are from biologically independent samples as indicated in Fig. 1f (RNA-part data) and Fig. 3b (DNA-part data). Each box represents median, 25% and 75% quartiles; whiskers indicate 1.5 times the interquartile range; one side Wilcoxon test. b. Bar graphs showing the number of CNVs on 22 autosomes in human arrested embryos at each stage. Data are from 10, 10, 11, 12 biologically independent samples for human normal zygote to 8-cell embryos; 15, 13, 7, 6 biologically independent samples for human arrested zygote to 8-cell embryos; integration of all samples. c. Representative examples of euploid and aneuploid cells in intact human arrested embryos. CNVs that are complementary in blastomeres within a complete human embryo are highlighted by a black dotted frame. The CNV patterns were profiled using DNA-part data at 1 M resolution. Windows with normalized count data ≥ 3 or ≤ 1 are marked in green.

Source data

Supplementary information

Supplementary Figures

Supplementary Figs. 1–7.

Reporting Summary

Supplementary Tables

Supplementary Tables 1–10. Supplementary Table 1: Quality assessment of the single-cell multi-omics data in human arrested embryos. Supplementary Table 2: DEGs in human arrested embryos compared with normal ones (two-sided t-test with Benjamin and Hochberg adjustment). Supplementary Table 3: NDR- and DMR-associated DEGs in human arrested embryos. Supplementary Table 4: Quality assessment of the single-embryo RNA-seq data in human 3PN 8-cell embryos from the control group and the KDOAM25-treated group. Supplementary Table 5: The overlap between upregulated major ZGA genes in human arrested embryos and targeted genes of YY1, KLF4 and ZFP42 with publicly available ChIP–seq data from hESCs or HEK 293T cells. Supplementary Table 6: Quality assessment of the single-embryo multi-omics data in human 3PN 8-cell embryos from the negative control group and the YY1 knockdown group. Supplementary Table 7: Quality assessment of the single cell RNA-seq data in mouse control embryos and arrested zygotes from the AURKA-inhibited group. Supplementary Table 8: DEGs between mouse arrested zygotes from the AURKA-inhibited group and the control group. Supplementary Table 9: Quality assessment of the single-cell multi-omics data in human arrested zygotes from the AURKA-inhibited group and control zygotes. Supplementary Table 10: RT–qPCR primer sequences.

Supplementary Data 1

Source data for Supplementary Figs. 1–7.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

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 Fig. 7

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. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, T., Peng, J., Fan, J. et al. Single-cell multi-omics profiling of human preimplantation embryos identifies cytoskeletal defects during embryonic arrest. Nat Cell Biol 26, 263–277 (2024). https://doi.org/10.1038/s41556-023-01328-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-023-01328-0

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