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 spermatogenesis reveals a DNA demethylation event associated with male meiotic recombination

Abstract

Human spermatogenesis is a highly ordered process; however, the roles of DNA methylation and chromatin accessibility in this process remain largely unknown. Here by simultaneously investigating the chromatin accessibility, DNA methylome and transcriptome landscapes using the modified single-cell chromatin overall omic-scale landscape sequencing approach, we revealed that the transcriptional changes throughout human spermatogenesis were correlated with chromatin accessibility changes. In particular, we identified a set of transcription factors and cis elements with potential functions. A round of DNA demethylation was uncovered upon meiosis initiation in human spermatogenesis, which was associated with male meiotic recombination and conserved between human and mouse. Aberrant DNA hypermethylation could be detected in leptotene spermatocytes of certain nonobstructive azoospermia patients. Functionally, the intervention of DNA demethylation affected male meiotic recombination and fertility. Our work provides multi-omics landscapes of human spermatogenesis at single-cell resolution and offers insights into the association between DNA demethylation and male meiotic recombination.

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 chromatin accessibility, DNA methylation and transcriptome of human testicular germ cells.
Fig. 2: Defining epigenetic regulatory networks underlying human spermatogenesis.
Fig. 3: Identification of potential regulators for cell-fate transition during human spermatogenesis.
Fig. 4: A round of DNA demethylation occurs upon human male meiosis initiation.
Fig. 5: DNA demethylation upon human male meiosis initiation is associated with meiotic recombination.
Fig. 6: deMRs exhibit enrichment of human PRDM9 motif and higher recombination rate.
Fig. 7: Aberrant DNA methylation status in nonobstructive azoospermia patients.
Fig. 8: The intervention of DNA demethylation affects male meiotic recombination and fertility.

Similar content being viewed by others

Data availability

All raw sequence data reported in this study have been deposited in the Genome Sequence Archive of the National Genomics Data Center, China National Center for Bioinformation/Beijing Institute of Genomics, Chinese Academy of Sciences. The accession numbers are listed as below: the single-cell multi-omics sequencing raw data are under HRA000148 (https://ngdc.cncb.ac.cn/gsa-human/browse/HRA000148), the scRNA-seq raw data of testicular cells from NOA3-NOA8 patients are under HRA004917 (https://ngdc.cncb.ac.cn/gsa-human/browse/HRA004917), the HEK293T cell line sequencing raw data are under HRA004922 (https://ngdc.cncb.ac.cn/gsa-human/browse/HRA004922) and the Uhrf1-cKO and control mice sequencing raw data are under CRA011561 (https://ngdc.cncb.ac.cn/gsa/browse/CRA011561). All processed data reported in this study have been deposited in the Gene Expression Omnibus (https://www.ncbi.nlm.nih.gov/geo) under accession number GSE235324.

Human reference genome hg38, mouse reference genome mm10 and genomic-region annotations were obtained from UCSC genome browser (https://genome.ucsc.edu/). The union of H3K27ac and H3K4me1 ChIP-seq peaks from GSE143117 was performed to identify human enhancers96. The datasets of previous publication are publicly available under accession numbers: scRNA-seq data of testicular cells from human normal samples (GSE106487 and GSE109037)8,10 and NOA9 patient (GSE157421)64; scCOOL-seq data (GSE100272)22; DNA methylation and chromatin accessibility data of hFGCs (GSE79552)28 and scRNA-seq data of hFGCs (GSE86146)27; DNA methylation data of spermatogenic cells from spermatogenesis-synchronized mice (GSE132446)17; mouse DSB hotspots (SPO11 hotspots, GSE84689; DMC1 hotspots, GSE35498)52,53; mouse PRDM9 binding sites (GSE61613)54; human meiotic DSB hotspots (GSE59836)45. Human recombination hotspots are from ref. 46,47. Source data are provided with this paper.

Code availability

The code used in this study is deposited at: https://github.com/YangXinyan/HumanSperm_MultiOmics (https://doi.org/10.5281/zenodo.8214258).

References

  1. Handel, M. A. & Schimenti, J. C. Genetics of mammalian meiosis: regulation, dynamics and impact on fertility. Nat. Rev. Genet. 11, 124–136 (2010).

    CAS  PubMed  Google Scholar 

  2. Godmann, M., Lambrot, R. & Kimmins, S. The dynamic epigenetic program in male germ cells: Its role in spermatogenesis, testis cancer, and its response to the environment. Microsc. Res. Tech. 72, 603–619 (2009).

    CAS  PubMed  Google Scholar 

  3. Jan, S. Z. et al. Molecular control of rodent spermatogenesis. Biochim. Biophys. Acta 1822, 1838–1850 (2012).

    CAS  PubMed  Google Scholar 

  4. Baudat, F., Imai, Y. & de Massy, B. Meiotic recombination in mammals: localization and regulation. Nat. Rev. Genet. 14, 794–806 (2013).

    CAS  PubMed  Google Scholar 

  5. Baudat, F. et al. PRDM9 is a major determinant of meiotic recombination hotspots in humans and mice. Science 327, 836–840 (2010).

    CAS  PubMed  Google Scholar 

  6. Parvanov, E. D., Petkov, P. M. & Paigen, K. Prdm9 controls activation of mammalian recombination hotspots. Science 327, 835 (2010).

    CAS  PubMed  Google Scholar 

  7. Diagouraga, B. et al. PRDM9 methyltransferase activity is essential for meiotic DNA double-strand break formation at its binding sites. Mol. Cell 69, 853–865.e6 (2018).

    CAS  PubMed  Google Scholar 

  8. Hermann, B. P. et al. The mammalian spermatogenesis single-cell transcriptome, from spermatogonial stem cells to spermatids. Cell Rep. 25, 1650–1667.e8 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Sohni, A. et al. The neonatal and adult human testis defined at the single-cell level. Cell Rep. 26, 1501–1517.e4 (2019).

    PubMed  PubMed Central  Google Scholar 

  10. Wang, M. et al. Single-cell RNA sequencing analysis reveals sequential cell fate transition during human spermatogenesis. Cell Stem Cell 23, 599–614.e4 (2018).

    CAS  PubMed  Google Scholar 

  11. Dai, H. Q. et al. TET-mediated DNA demethylation controls gastrulation by regulating Lefty-Nodal signalling. Nature 538, 528–532 (2016).

    PubMed  Google Scholar 

  12. Markenscoff-Papadimitriou, E. et al. A chromatin accessibility atlas of the developing human telencephalon. Cell 182, 754–769.e8 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Izzo, F. et al. DNA methylation disruption reshapes the hematopoietic differentiation landscape. Nat. Genet. 52, 378–387 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  15. Maezawa, S., Yukawa, M., Alavattam, K. G., Barski, A. & Namekawa, S. H. Dynamic reorganization of open chromatin underlies diverse transcriptomes during spermatogenesis. Nucleic Acids Res. 46, 593–608 (2018).

    CAS  PubMed  Google Scholar 

  16. Gaysinskaya, V. et al. Transient reduction of DNA methylation at the onset of meiosis in male mice. Epigenetics Chromatin 11, 15 (2018).

    PubMed  PubMed Central  Google Scholar 

  17. Chen, Y. et al. Refined spatial temporal epigenomic profiling reveals intrinsic connection between PRDM9-mediated H3K4me3 and the fate of double-stranded breaks. Cell Res. 30, 256–268 (2020).

  18. Guo, J. et al. Chromatin and single-cell RNA-seq profiling reveal dynamic signaling and metabolic transitions during human spermatogonial stem cell development. Cell Stem Cell 21, 533–546.e6 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Guo, J. et al. The adult human testis transcriptional cell atlas. Cell Res. 28, 1141–1157 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  21. 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).

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    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).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Li, L. et al. Single-cell RNA-seq analysis maps development of human germline cells and gonadal niche interactions. Cell Stem Cell 20, 858–873.e4 (2017).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  29. Ren, B. & Yue, F. Transcriptional enhancers: bridging the genome and phenome. Cold Spring Harb. Symp. Quant. Biol. 80, 17–26 (2015).

    PubMed  Google Scholar 

  30. Tan, K. et al. Transcriptome profiling reveals signaling conditions dictating human spermatogonia fate in vitro. Proc. Natl Acad. Sci. USA 117, 17832–17841 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Tassinari, V. et al. Fgf9 inhibition of meiotic differentiation in spermatogonia is mediated by Erk-dependent activation of Nodal-Smad2/3 signaling and is antagonized by Kit Ligand. Cell Death Dis. 6, e1688 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Matson, C. K. et al. The mammalian doublesex homolog DMRT1 is a transcriptional gatekeeper that controls the mitosis versus meiosis decision in male germ cells. Dev. Cell 19, 612–624 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Zhang, T., Murphy, M. W., Gearhart, M. D., Bardwell, V. J. & Zarkower, D. The mammalian Doublesex homolog DMRT6 coordinates the transition between mitotic and meiotic developmental programs during spermatogenesis. Development 141, 3662–3671 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Kistler, W. S. et al. RFX2 is a major transcriptional regulator of spermiogenesis. PLoS Genet. 11, e1005368 (2015).

    PubMed  PubMed Central  Google Scholar 

  35. Hai, T. & Curran, T. Cross-family dimerization of transcription factors Fos/Jun and ATF/CREB alters DNA binding specificity. Proc. Natl Acad. Sci. USA 88, 3720–3724 (1991).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. De Cesare, D. & Sassone-Corsi, P. Transcriptional regulation by cyclic AMP-responsive factors. Prog. Nucleic Acid Res. Mol. Biol. 64, 343–369 (2000).

    PubMed  Google Scholar 

  37. Smith, Z. D. & Meissner, A. DNA methylation: roles in mammalian development. Nat. Rev. Genet. 14, 204–220 (2013).

    CAS  PubMed  Google Scholar 

  38. Kagiwada, S., Kurimoto, K., Hirota, T., Yamaji, M. & Saitou, M. Replication-coupled passive DNA demethylation for the erasure of genome imprints in mice. EMBO J. 32, 340–353 (2013).

    CAS  PubMed  Google Scholar 

  39. Sharif, J. et al. The SRA protein Np95 mediates epigenetic inheritance by recruiting Dnmt1 to methylated DNA. Nature 450, 908–912 (2007).

    CAS  PubMed  Google Scholar 

  40. Bostick, M. et al. UHRF1 plays a role in maintaining DNA methylation in mammalian cells. Science 317, 1760–1764 (2007).

    CAS  PubMed  Google Scholar 

  41. Zhang, J. et al. S phase-dependent interaction with DNMT1 dictates the role of UHRF1 but not UHRF2 in DNA methylation maintenance. Cell Res. 21, 1723–1739 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Li, T. et al. Structural and mechanistic insights into UHRF1-mediated DNMT1 activation in the maintenance DNA methylation. Nucleic Acids Res. 46, 3218–3231 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Dong, J. et al. UHRF1 suppresses retrotransposons and cooperates with PRMT5 and PIWI proteins in male germ cells. Nat. Commun. 10, 4705 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Pan, H. et al. UHRF1-repressed 5’-hydroxymethylcytosine is essential for the male meiotic prophase I. Cell Death Dis. 11, 142 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Pratto, F. et al. DNA recombination. Recombination initiation maps of individual human genomes. Science 346, 1256442 (2014).

    PubMed  PubMed Central  Google Scholar 

  46. The 1000 Genomes Project Consortium A map of human genome variation from population-scale sequencing. Nature 467, 1061–1073 (2010).

    PubMed Central  Google Scholar 

  47. Halldorsson, B. V. et al. Characterizing mutagenic effects of recombination through a sequence-level genetic map. Science 363, eaau1043 (2019).

    CAS  PubMed  Google Scholar 

  48. Myers, S., Freeman, C., Auton, A., Donnelly, P. & McVean, G. A common sequence motif associated with recombination hot spots and genome instability in humans. Nat. Genet. 40, 1124–1129 (2008).

    CAS  PubMed  Google Scholar 

  49. Arnheim, N., Calabrese, P. & Tiemann-Boege, I. Mammalian meiotic recombination hot spots. Annu. Rev. Genet 41, 369–399 (2007).

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

    Google Scholar 

  51. Dreau, A., Venu, V., Avdievich, E., Gaspar, L. & Jones, F. C. Genome-wide recombination map construction from single individuals using linked-read sequencing. Nat. Commun. 10, 4309 (2019).

    PubMed  PubMed Central  Google Scholar 

  52. Brick, K., Smagulova, F., Khil, P., Camerini-Otero, R. D. & Petukhova, G. V. Genetic recombination is directed away from functional genomic elements in mice. Nature 485, 642–645 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Lange, J. et al. The landscape of mouse meiotic double-strand break formation, processing, and repair. Cell 167, 695–708.e6 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Walker, M. et al. Affinity-seq detects genome-wide PRDM9 binding sites and reveals the impact of prior chromatin modifications on mammalian recombination hotspot usage. Epigenetics Chromatin 8, 31 (2015).

    PubMed  PubMed Central  Google Scholar 

  55. Jiao, Y. et al. A TOP6BL mutation abolishes meiotic DNA double-strand break formation and causes human infertility. Sci. Bull. 65, 2120–2129 (2020).

    CAS  Google Scholar 

  56. Yuan, Y. et al. Generation of fertile offspring from Kitw/Kitwv mice through differentiation of gene corrected nuclear transfer embryonic stem cells. Cell Res. 25, 851–863 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Crespo, M. et al. Multi-omic analysis of gametogenesis reveals a novel signature at the promoters and distal enhancers of active genes. Nucleic Acids Res. 48, 4115–4138 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  59. Melamed-Bessudo, C. & Levy, A. A. Deficiency in DNA methylation increases meiotic crossover rates in euchromatic but not in heterochromatic regions in Arabidopsis. Proc. Natl Acad. Sci. USA 109, E981–E988 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Zamudio, N. et al. DNA methylation restrains transposons from adopting a chromatin signature permissive for meiotic recombination. Genes Dev. 29, 1256–1270 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Gonzalo, S. et al. DNA methyltransferases control telomere length and telomere recombination in mammalian cells. Nat. Cell Biol. 8, 416–424 (2006).

    CAS  PubMed  Google Scholar 

  62. Liu, Y., Sarkar, A., Kheradpour, P., Ernst, J. & Kellis, M. Evidence of reduced recombination rate in human regulatory domains. Genome Biol. 18, 193 (2017).

    PubMed  PubMed Central  Google Scholar 

  63. Altemose, N. et al. A map of human PRDM9 binding provides evidence for novel behaviors of PRDM9 and other zinc-finger proteins in meiosis. eLife 6, e28383 (2017).

    PubMed  PubMed Central  Google Scholar 

  64. Wang, M. et al. Deciphering the autophagy regulatory network via single-cell transcriptome analysis reveals a requirement for autophagy homeostasis in spermatogenesis. Theranostics 11, 5010–5027 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Sadate-Ngatchou, P. I., Payne, C. J., Dearth, A. T. & Braun, R. E. Cre recombinase activity specific to postnatal, premeiotic male germ cells in transgenic mice. Genesis 46, 738–742 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Zhao, J. et al. Cell-fate transition and determination analysis of mouse male germ cells throughout development. Nat. Commun. 12, 6839 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  70. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

  72. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902.e21 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Cao, J. et al. The single-cell transcriptional landscape of mammalian organogenesis. Nature 566, 496–502 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. 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).

    CAS  PubMed  Google Scholar 

  75. McLean, C. Y. et al. GREAT improves functional interpretation of cis-regulatory regions. Nat. Biotechnol. 28, 495–501 (2010).

  76. Liu, Y., Siegmund, K. D., Laird, P. W. & Berman, B. P. Bis-SNP: combined DNA methylation and SNP calling for Bisulfite-seq data. Genome Biol. 13, R61 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Danecek, P. et al. The variant call format and VCFtools. Bioinformatics 27, 2156–2158 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Bansal, V. & Bafna, V. HapCUT: an efficient and accurate algorithm for the haplotype assembly problem. Bioinformatics 24, i153–i159 (2008).

    PubMed  Google Scholar 

  79. Van der Auwera, G. A. et al. From FastQ data to high confidence variant calls: the Genome Analysis Toolkit best practices pipeline. Curr. Protoc. Bioinforma. 43, 11.10.11–11.10.33 (2013).

    Google Scholar 

  80. Gaysinskaya, V., Soh, I. Y., van der Heijden, G. W. & Bortvin, A. Optimized flow cytometry isolation of murine spermatocytes. Cytom. A 85, 556–565 (2014).

    Google Scholar 

  81. Lima, A. C. et al. A standardized approach for multispecies purification of mammalian male germ cells by mechanical tissue dissociation and flow cytometry. J. Vis. Exp. https://doi.org/10.3791/55913 (2017).

  82. Chen, Y. et al. Single-cell RNA-seq uncovers dynamic processes and critical regulators in mouse spermatogenesis. Cell Res. 28, 879–896 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Hogarth, C. A. et al. Turning a spermatogenic wave into a tsunami: synchronizing murine spermatogenesis using WIN 18,446. Biol Reprod. 88, 40 (2013).

  84. Bilichak, A. & Kovalchuk, I. The combined bisulfite restriction analysis (COBRA) assay for the analysis of locus-specific changes in methylation patterns. Methods Mol Biol. 1456, 63–71 (2017).

  85. Yao, X. et al. Homology-mediated end joining-based targeted integration using CRISPR/Cas9. Cell Res. 27, 801–814 (2017).

  86. Ran, F. A. et al. Genome engineering using the CRISPR-Cas9 system. Nat. Protoc. 8, 2281–2308 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Wang, X. et al. The chromatin accessibility landscape reveals distinct transcriptional regulation in the induction of human primordial germ cell-like cells from pluripotent stem cells. Stem Cell Rep. 16, 1245–1261 (2021).

    CAS  Google Scholar 

  89. Bolger, A. M., Lohse, M. & Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Ramirez, F. et al. deepTools2: a next generation web server for deep-sequencing data analysis. Nucleic Acids Res. 44, W160–W165 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Acquaviva, L. et al. Ensuring meiotic DNA break formation in the mouse pseudoautosomal region. Nature 582, 426–431 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Li, M. et al. The histone modification reader ZCWPW1 is required for meiosis prophase I in male but not in female mice. Sci. Adv. 5, eaax1101 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Wang, Y. et al. Single-cell multiomics sequencing reveals the functional regulatory landscape of early embryos. Nat. Commun. 12, 1247 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Yan, R. et al. Decoding dynamic epigenetic landscapes in human oocytes using single-cell multi-omics sequencing. Cell Stem Cell 28, 1641–1656 (2021).

  96. Consortium, E. P. An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74 (2012).

    Google Scholar 

Download references

Acknowledgements

We thank M. Tong from the Shanghai Institute of Biochemistry and Cell Biology for providing the Prdm9–/– (Prdm9tm1Ymat) mice, Q. Shi from the University of Science and Technology of China for kindly providing the Top6bl–/– mice and S. Yuan’s laboratory at Huazhong University of Science and Technology for providing the Uhrf1flox/flox mice. We thank F. Tang from Peking University and S. Rao from Nanfang Hospital for helpful discussion and suggestions regarding the paper. This work was supported by grants from the National Key R&D Programme of China (2022YFA0806303 and 2022YFC2702603 to X.-Y.Z., 2021YFA1102700 to L.L., 2020YFA0113300 and 2019YFA0801802 to M.W., and 2019YFE0109500 to G.A.), the National Natural Science Foundation of China (U22A20278 and 82071711 to X.-Y.Z., 31970814 to S.G., 31970787 and 32170869 to G.C., 32070833 and 82101745 to L.L., and 32170866 to M.W.), the Natural Science Funds for Distinguished Young Scholar of Guangdong Province (2022B1515020110 to L.L), the Natural Science Foundation of Shenzhen (JCYJ20210324120212033 to G.C.) and the Key-Area Research and Development Programme of Guangdong Province Modernization of Chinese medicine in Lingnan (2020B1111100011 to L.L.).

Author information

Authors and Affiliations

Authors

Contributions

X.-Y.Z., S.G. and G.C. conceived and supervised the project. X.-Y.Z., G.C. and Y.H. designed the experiments. Y.H., S.G., G.A., M.C., J.L., X.Z., Z.Y., C.W., C.Z., K.S., S.R., and X.X. carried out the experiments. J.Z., X.F., Y.L., X.H., W.W., M.W. and Y.Z. helped with sample collection. With the help of A.P.H., X.B. and K.M., L.L., X.Y. and X.S. performed bioinformatics analyses. X.Z., S.G., G.C., Y.H., L.L., X.Y. and M.C. wrote the paper with help from all authors.

Corresponding authors

Correspondence to Gang Chang, Shuai Gao or Xiao-Yang Zhao.

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 Cell identity assignment and quality control of single-cell multi-omics sequencing datasets.

a, UMAP plots showing transcriptome data of adult human testicular cells in this study (1,097 cells) integrated with the cells (2,854 cells) from Wang et al.10. Cells are coloured by the origin (donors, left) or cluster (right). b, The expression patterns of representative marker genes on the UMAP plots of this study. A gradient of grey to purple indicates gene expression level from low to high. SPG, spermatogonia; SPC, spermatocytes. c, Box plot showing the number of genes detected in transcriptome data in each individual cell of Wang et al.10 and this study. The cell number at each stage of this study is indicated above each box. d,e, Box plots showing the number of WCG sites (d) and GCH sites (e) covered in a single cell at the indicated stage at 1× depth. f, Box plot showing the mapping rate of DNA data (chromatin accessibility and DNA methylation) at each stage. In c-f, each box represents the median and the 25% and 75% quartiles, and the whiskers indicate 1.5 times the interquartile range. All the data of this study represented in this figure are from four biologically independent samples (integration of all samples).

Source data

Extended Data Fig. 2 The relationship between DNA methylation, chromatin accessibility and RNA expression of the corresponding genes during cell-fate transition in human spermatogenesis.

a, Volcano plots showing the relationship between RNA expression and chromatin accessibility of proximal NDRs as cell fate transitioning during human spermatogenesis. The x axis represents the differential RNA expression (log(fold change)) between Undiff.SPGs and Diff.ing SPG (left), Diff.ing SPG and preL-2 (middle), S1 and S2 (right), respectively. Positive values indicate higher expression in the latter stage. The y axis represents the significance of differential RNA expression (two-sided Wilcoxon rank sum test with Bonferroni correction). Number of DEGs in these three groups are shown at the top of each panel. Red dots represent genes whose RNA expression are significantly higher and the proximal NDRs are significantly more open in the latter stage (two-sided multiple t test with Benjamini-Hochberg adjustment for chromatin accessibility). Blue dots are vice versa. b, Violin plots showing the distribution of gene expression, chromatin accessibility and DNA methylation level of correlated proximal NDRs at the indicated stages. Genomic track is shown on top, and the location of correlated proximal NDRs is labelled with red box. Each box within violin plots represents the median and the 25% and 75% quartiles, and the whiskers indicate 1.5 times the interquartile range. Each dot corresponds to a single cell. c, Genome browser views of chromatin accessibility at the ID4 locus in spermatogonia. Heatmap showing RNA expression level of ID4 at each stage (left). Coloured bars indicate methylation level at GCH sites calculated based on single-cell multi-omics data. Red boxes under each track indicate the location of NDRs. The shadow region represents the specific proximal NDR and its mean level of chromatin accessibility at the indicated stage is shown as heatmap on the right of IGV. d, Heatmap showing the K-means clustering (expected clusters = 3) of DNA methylation level (mean WCG methylation) at the promoter regions (1 kb upstream and 0.5 kb downstream of the TSS). All the data represented in this figure are from four biologically independent samples (integration of all samples).

Source data

Extended Data Fig. 3 The epigenetic modification differences between hFGCs and adult spermatogenic cells.

a, Heatmaps showing DNA methylation, chromatin accessibility of promoters and corresponding relative RNA expression level of representative pluripotency genes throughout human male germline development. White indicates missing values (undetected). b, Genome browser views showing DNA methylation levels of representative pluripotency genes loci in hFGCs and spermatogonia. Coloured bars indicate the DNA methylation level of WCG sites calculated based on the NOMe-seq data from ref. 28 (hFGCs) and single-cell multi-omics data in this study (adult testicular cells). Colour bars above the horizontal line indicate the DNA methylation level (higher than 0.1) at individual WCG site, and the black bars indicate the horizontal line denote WCG site with DNA methylation level lower than 0.1, to discriminate from undetected WCG sites. The shadow region represents the promoter of each gene and its mean DNA methylation level at the indicated stage is shown as heatmap on the right of IGV. c, Heatmaps showing DNA methylation and chromatin accessibility of promoters as well as corresponding relative RNA expression of representative meiosis-specific genes throughout human male germline development. d, Genome browser views of DNA methylation and chromatin accessibility at the representative meiosis-specific gene (SPO11) locus in hFGCs and spermatogenic cells. The shadow region represents the promoter of SPO11 and the mean levels of DNA methylation and chromatin accessibility at the indicated stage are shown as heatmaps on the right of IGV, respectively. The DNA methylation and chromatin accessibility data of hFGCs are from ref. 28 and scRNA-seq data of hFGCs are from ref. 27. All the data of this study represented in this figure are from four biologically independent samples (integration of all samples).

Source data

Extended Data Fig. 4 The potential distal cis-regulatory elements and transcription factors involved in the human spermatogenesis.

a, Heatmap showing the pairwise correlations of cells from P to S1 based on the transcriptome data. Unsupervised hierarchical clustering indicates that SPC7 are more similar to S1. b, Chromatin accessibility of the distal cis-regulatory elements of SPC7 and spermatids (S1-S3). The number of cis-regulatory element at the indicated stages is shown on the left of the heatmap. Representative GO terms (GREAT analysis) and genes corresponding to the distal cis-regulatory elements of each cluster are shown on the right of the heatmap. GO terms are coloured by membership of sub-group (early round spermatids: SPC7 and S1; late round spermatids: S2 and S3). c, Immunofluorescence of KLF6 co-staining with germ cells marker DDX4 in adult human testicular paraffin sections from normal sample. White triangles indicate SPG and yellow arrows indicate Sertoli cells. Scale bar, 10 μm. d,e, The relative expression of KLF6 (d) or representative hSSC marker genes (e) after transfection with the negative control (NC) siRNA or KLF6 siRNA (1/2) into the sorted human undifferentiated spermatogonia. Data are represented as mean with data points (n = 3 biologically independent replicates obtained from two healthy donors with normal fertility). f, Immunofluorescence of KLF4 co-staining with spermatid marker PNA in adult human testicular paraffin sections from normal samples. White arrows indicate S1, yellow arrows indicate S2 and white triangles indicate elongating spermatids. Scale bar, 20 μm. g, Transcription factor motif enrichment at all cis-regulatory elements. The size of circle represents P value (-log10(P value)) of the motif-enrichment and the colour represents the RNA expression level. Only motifs with P ≤ 1×10-10 and TPM ≥ 10 in at least one stage are shown. Significance is calculated with one-sided binomial test using HOMER. Cis-regulatory elements are combined for motif enrichment by sub-groups, including Undiff.SPG (Undiff.SPG-1/2), Diff.ing SPG, early primary spermatocytes (Early SPC; preL-Z), late primary spermatocytes (Late SPC; preP-D), early round spermatids (Early SPD; SPC7-S1) and late round spermatids (Late SPC; S2-S3). Data in a,b and g are from four biologically independent samples (integration of all samples).

Source data

Extended Data Fig. 5 DNA methylation changes in human spermatogenesis and the expression patterns of DNA demethylation related genes.

a, Quantification of the relative fluorescence intensity of 5mC to Hoechst at the indicated stages from normal samples (Donor 1: 28, 36, 32 cells; Donor 2: 29, 38, 39 cells; Donor 3: 25, 38, 35 cells; Donor 4: 24, 36, 32 cells at Diff.ing SPG, preL/L and P stage). Data are represented as mean with data points. b, PCA analysis of stages from Diff.ing SPG to L1/2 based on tiles’ DNA methylation level. c, Box plots showing the expression dynamics of DNA methylation-related genes during human spermatogenesis. d, Negative controls (no primary antibody) of immunofluorescence in adult human testicular paraffin sections from normal sample. Scale bar, 10 μm. e, Immunofluorescence of 5hmC and DDX4 in adult human testicular paraffin sections from normal sample. Scale bar, 10 μm. f, Boxplots showing the expression levels of G1/S phase specific genes at the indicated stages. Each box in c and f represents the median and the 25% and 75% quartiles, and the whiskers indicate 1.5 times the interquartile range. Data in b,c and f are from four biologically independent samples (integration of all samples).

Source data

Extended Data Fig. 6 Cell type annotation and characterization in NOA patients.

a, Immunofluorescence of SYCP3 co-staining with γH2AX and PNA in adult human testicular paraffin sections from normal sample (top), NOA1 (middle) and NOA2 patients (bottom). Scale bar, 10 μm. b,c, UMAP plots showing the transcriptome data of NOA1 (176 cells, b) and NOA2 (130 cells, c) in this study integrated with the data from Wang et al. (2,854 cells from nine biologically independent samples; integration of all samples)10. Cells are coloured by sample origins (left) or clusters (right). d,e, UMAP plots showing the cell types detected in NOA1 (d) and NOA2 (e) patients. The number of each cell type is indicated in the parenthesis. f,g, UMAP plots showing the expression patterns of representative marker genes of NOA1 (f) and NOA2 (g) patients. A gradient of grey to purple indicates expression level from low to high.

Extended Data Fig. 7 Abnormal DNA methylation patterns in the testicular germ cells of NOA patients.

a, Dimensionality reduction of the DNA methylation on 500-bp non-overlapping tiles by PCA analysis. Cells are coloured by stage. b, Bar plot showing the number of DEGs with or without differentially methylated promoters at the indicated stages of NOA1 or NOA2 compared with normal samples. c, Enrichment analysis (-log10(P value), one-sided hypergeometric test) for the hypo-DMRs at Undiff.SPG-1 (left), L3 (middle) and preP (right) stages of NOA1/NOA2 patient compared with the normal control. d, Relative expression levels of SVA, ERV1 and ERVK at the indicated stages of NOA patients and normal samples (1,097, 176, 130 cells in normal, NOA1, NOA2 samples). e, Global pattern of DNA methylation level in NOA patients and normal samples at the indicated stages before (left) and after (right) down-sampling (1,097, 176, 130 cells in normal, NOA1, NOA2 samples). In b-e, Undiff.SPG-out1/2 of NOA1 patient is compared with the Undiff.SPG-1 of normal samples. Normal samples in this figure are the integration of four biologically independent samples.

Source data

Extended Data Fig. 8 The DNA demethylation at the onset of meiosis is conserved between human and mouse.

a, Immunofluorescence performed in testicular sections from PND 15 mice. White triangles indicate epL (STRA8 dispersed SYCP3), yellow arrows indicate mpL (STRA8+ dispersed SYCP3) and white arrows indicate P (threads SYCP3). Scale bar, 10 μm. b, Quantification of the relative fluorescence density of 5mC to Hoechst at the indicated stages from PND 15 mice (174, 162, 159 cells from left to right). c, Quantification of fluorescence density of UHRF1 at the indicated stages from PND 15 mice (120, 185, 140 cells from left to right). Statistics of b and c are performed in three independent mice and data are represented as mean ± SD; unpaired two-sided Student’s t test. d,e, ECDF plots showing the differential DNA methylation level of mouse hotspots (d) or PRDM9 binding sites (e) or random regions between the stage when demethylation occurs and the previous stage in mice (mpL versus type B of spermatogonia (BS)). Random regions are randomly selected genomic regions with equal GC content, number of CpG sites and sequence length distribution as that of hotspots (d) and PRDM9 binding sites (e). DMC1 hotspots: 18,291 regions; SPO11 hotspots: 9,714 regions; PRDM9 binding sites: 32,406 regions. f,g, DNA methylation dynamics of known crossover hotspots during mouse (f) and human (g) spermatogenesis. The bottom three hotspots (A1, A2, A4) are previously identified by pedigree analysis and others are identified by sperm typing53. * indicates the locus selected for bisulfite genomic PCR-based Sanger sequencing assay in this study. In d-f, the DNA methylation dataset of spermatogenic cells from spermatogenesis-synchronized mice is from ref. 17 (n = 2 independent replicates). h, COBRA analysis of the representative crossover hotspots in the sorted cell populations of juvenile, adult and spermatogenesis-synchronized mice. P/D, pachytene/diplotene spermatocytes; BS, type B of spermatogonia; mP, middle pachytene spermatocytes; U, uncut; C, cut.

Source data

Extended Data Fig. 9 The binding sites of PRDM9 in HEK293T cells with DNA methylation inhibitor treatment.

a, Bisulfite genomic PCR-based Sanger sequencing showing the DNA methylation patterns from the representative crossover hotspots in the sorted cell populations of juvenile, adult and spermatogenesis-synchronized mice. Top genome browser view showing the DNA methylation level of each locus in the indicated cell populations from spermatogenesis-synchronized mice17. b, Whole-genome bisulfite sequencing showing the DNA methylation level of HEK293T cells treated with dimethylsulfoxide (DMSO, control) or Decitabine for 60 h. Data are represented as mean ± SD; unpaired two-sided Student’s t test. c, Venn diagram showing the overlap of the hypo-DMRs in Decitabine-treated HKE293T cells (Deci hypo-DMRs) with human recombination hotspots. hypo Hotspots represent the human recombination hotspots overlapped with Deci hypo-DMRs; Hotspot_DMRs represent the Deci hypo-DMRs overlapped with human recombination hotspots. d, Genome browser views of the hPRDM9 signal level and DNA methylation level around human recombination hotspots in Decitabine-treated HEK293T cells and the control group. The mean DNA methylation levels of crossover regions (shadow regions) are shown as heatmaps on the right. Data in b-d are from three biologically independent replicates in each group; integration of all samples.

Source data

Extended Data Fig. 10 Male meiotic recombination and fertility are affected by the intervention of DNA methylation.

a, Immunofluorescence of RAD51 co-staining with SYCP3 for surface-spread spermatocytes from Uhrf1 cKO and control mice. Scale bar, 10 μm. b, The foci number of RAD51 per spermatocyte from Uhrf1 cKO and control mice (30, 43, 49, 68, 45, 53 cells from left to right). c, Schematic illustration of the donor constructs for knocking the CAG-the indicated genes’ coding sequence (CDS)-P2A-Tdtomato cassette into the Rosa26 locus. d, The relative expression of Uhrf1 or Dnmt1 in the control, Uhrf1-TG and UD-TG EGFP-labelled mSSCs. e, Representative cell morphology of the control, Uhrf1-TG and UD-TG EGFP-mSSCs. Scale bar, 100 μm. f, Growth curves of the control, Uhrf1-TG and UD-TG EGFP-mSSCs. In d and f, data are represented as mean ± SD (n = 3 independent mice per group). g, Immunofluorescence of mouse spermatogonial markers PLZF, OCT4 and ID4 in the control, Uhrf1-TG and UD-TG EGFP-mSSCs, respectively. Scale bar, 20 μm. h, Immunofluorescence of RAD51 co-staining with SYCP3 and GFP for surface-spread spermatocytes from recipient testes transplanted with the control, Uhrf1-TG and UD-TG EGFP-mSSCs. Scale bar, 5 μm. i, The foci number of RAD51 per spermatocyte from recipient testes transplanted with the control, Uhrf1-TG and UD-TG EGFP-mSSCs (54, 46, 60, 70, 44, 45 cells from left to right). j, Immunofluorescence of SYCP3 co-staining with the spermatid marker PNA on the testicular sections from Uhrf1 cKO and control mice at PND 30. Scale bar, 50 μm. k, Flow cytometry analysis of DNA content revealing the ratio of haploid spermatid (1 C) in the testes of Uhrf1 cKO and control mice at PND 30. Statistics of b (n = 3) and i (n = 5) are performed in at least three independent mice per group and data are represented as mean ± SD; unpaired two-sided Student’s t test.

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–6.

Reporting Summary

Supplementary Tables

Supplementary Tables 1–7.

Supplementary Data 1

Source data for Supplementary Figs. 1–3.

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 Fig. 8

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. 4

Statistical source data.

Source Data Extended Data Fig. 5

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. 8

Unprocessed gels.

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

Huang, Y., Li, L., An, G. 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). https://doi.org/10.1038/s41556-023-01232-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-023-01232-7

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