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.

  • Article
  • Published:

Genome-wide stability of the DNA replication program in single mammalian cells

Abstract

Here, we report a single-cell DNA replication sequencing method, scRepli-seq, a genome-wide methodology that measures copy number differences between replicated and unreplicated DNA. Using scRepli-seq, we demonstrate that replication-domain organization is conserved among individual mouse embryonic stem cells (mESCs). Differentiated mESCs exhibited distinct profiles, which were also conserved among cells. Haplotype-resolved scRepli-seq revealed similar replication profiles of homologous autosomes, while the inactive X chromosome was clearly replicated later than its active counterpart. However, a small degree of cell-to-cell replication-timing heterogeneity was present, which was smallest at the beginning and the end of S phase. In addition, developmentally regulated domains were found to deviate from others and showed a higher degree of heterogeneity, thus suggesting a link to developmental plasticity. Moreover, allelic expression imbalance was found to strongly associate with replication-timing asynchrony. Our results form a foundation for single-cell-level understanding of DNA replication regulation and provide insights into three-dimensional genome organization.

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: Establishment of a single-cell RT profiling method, scRepli-seq.
Fig. 2: Single-cell replication profiles of mESCs before and after 7-d differentiation.
Fig. 3: Stability and heterogeneity of RT regulation in single cells.
Fig. 4: Developmentally regulated sequences exhibit RT heterogeneity in mESCs.
Fig. 5: Haplotype-resolved scRepli-seq analysis of X chromosomes during mESC differentiation.
Fig. 6: Haplotype-resolved scRepli-seq analysis of autosomes.
Fig. 7: Single-cell RT data correlate well with Hi-C A/B compartments.

Similar content being viewed by others

Data availability

All RT profiles (BrdU-IP, 100 cells and single-cell Repli-seq) and RNA-seq data generated in this study have been deposited in the NCBI Gene Expression Omnibus (GEO) database under accession code GSE108556.

References

  1. Berezney, R., Dubey, D. D. & Huberman, J. A. Heterogeneity of eukaryotic replicons, replicon clusters, and replication foci. Chromosoma. 108, 471–484 (2000).

    Article  CAS  Google Scholar 

  2. Rhind, N. & Gilbert, D. M. DNA Replication timing. Cold Spring Harb. Perspect. Biol. 5, 1–26 (2013).

    Article  Google Scholar 

  3. Prioleau, M. N. & MacAlpine, D. M. DNA replication origins: where do we begin? Gene. Dev. 30, 1683–1697 (2016).

    Article  CAS  Google Scholar 

  4. Rivera-Mulia, J. C. & Gilbert, D. M. Replication timing and transcriptional control: beyond cause and effect: part III. Curr. Opin. Cell Biol. 40, 168–178 (2016).

    Article  CAS  Google Scholar 

  5. Hiratani, I. et al. Genome-wide dynamics of replication timing revealed by in vitro models of mouse embryogenesis. Genome Res. 20, 155–169 (2010).

    Article  CAS  Google Scholar 

  6. Lieberman-Aiden, E. et al. Comprehensive mapping of long-range interactions reveals folding principles of the human genome. Science 326, 289–293 (2009).

    Article  CAS  Google Scholar 

  7. Ryba, T. et al. Evolutionarily conserved replication timing profiles predict long-range chromatin interactions and distinguish closely related cell types. Genome Res. 20, 761–770 (2010).

    Article  CAS  Google Scholar 

  8. Ryba, T., Battaglia, D., Pope, B. D., Hiratani, I. & Gilbert, D. M. Genome-scale analysis of replication timing: from bench to bioinformatics. Nat. Protoc. 6, 870–895 (2011).

    Article  CAS  Google Scholar 

  9. Hansen, R. S. et al. Sequencing newly replicated DNA reveals widespread plasticity in human replication timing. Proc. Natl Acad. Sci. USA 107, 139–144 (2010).

    Article  CAS  Google Scholar 

  10. Takebayashi, S.-I., Manders, E. M. M., Kimura, H., Taguchi, H. & Okumura, K. Mapping sites where replication initiates in mammalian cells using dna fibers. Exp. Cell Res. 271, 263–268 (2001).

    Article  CAS  Google Scholar 

  11. Norio, P. et al. Progressive activation of DNA replication initiation in large domains of the immunoglobulin heavy chain locus during B cell development. Mol. Cell 20, 575–587 (2005).

    Article  CAS  Google Scholar 

  12. Lebofsky, R., Heilig, R., Sonnleitner, M., Weissenbach, J. & Bensimon, A. DNA replication origin interference increases the spacing between initiation events in human cells. Mol. Biol. Cell. 17, 5337–5345 (2006).

    Article  CAS  Google Scholar 

  13. Dileep, V. & Gilbert, D. M. Single-cell replication profiling to measure stochastic variation in mammalian replication timing. Nat. Commun. 9, 427 (2018).

    Article  Google Scholar 

  14. Hiratani, I. et al. Global reorganization of replication domains during embryonic stem cell differentiation. PLoS Biol. 6, 2220–2236 (2008).

    Article  CAS  Google Scholar 

  15. Woodfine, K. et al. Replication timing of the human genome. Hum. Mol. Genet. 13, 191–202 (2004).

    Article  CAS  Google Scholar 

  16. Koren, A. et al. Genetic variation in human DNA replication timing. Cell 159, 1015–1026 (2014).

    Article  CAS  Google Scholar 

  17. Mukhopadhyay, R. et al. Allele-specific genome-wide profiling in human primary erythroblasts reveal replication program organization. PLoS Genet. 10, e1004319 (2014).

    Article  Google Scholar 

  18. Jiang, X. R. et al. Telomerase expression in human somatic cells does not induce changes associated with a transformed phenotype. Nat. Genet. 21, 111–114 (1999).

    Article  CAS  Google Scholar 

  19. Baslan, T. et al. Optimizing sparse sequencing of single cells for highly multiplex copy number profiling. Genome Res. 125, 714–724 (2015).

    Article  Google Scholar 

  20. Morgani, S., Nichols, J. & Hadjantonakis, A.-K. The many faces of pluripotency: in vitro adaptations of a continuum of in vivo states. BMC Dev. Biol. 17, 7 (2017).

    Article  Google Scholar 

  21. Takahashi, S., Kobayashi, S. & Hiratani, I. Epigenetic differences between naïve and primed pluripotent stem cells. Cell. Mol. Life Sci. 75, 1191–1203 (2018).

    Article  CAS  Google Scholar 

  22. Rivera-Mulia, J. C. et al. Allele-specific control of replication timing and genome organization during development. Genome Res. 28, 800–811 (2018).

    Article  CAS  Google Scholar 

  23. Dileep, V. et al. Topologically associating domains and their long-range contacts are established during early G1 coincident with the establishment of the replication timing program. Genome Res. 25, 1104–1113 (2015).

    Article  CAS  Google Scholar 

  24. Besnard, E. et al. Unraveling cell type-specific and reprogrammable human replication origin signatures associated with G-quadruplex consensus motifs. Nat. Struct. Mol. Biol. 19, 837–844 (2012).

    Article  CAS  Google Scholar 

  25. Murakami, K., Araki, K., Ohtsuka, S., Wakayama, T. & Niwa, H. Choice of random rather than imprinted X inactivation in female embryonic stem cell-derived extra-embryonic cells. Development 138, 197–202 (2011).

    Article  CAS  Google Scholar 

  26. Takada, T. et al. The ancestor of extant Japanese fancy mice contributed to the mosaic genomes of classical inbred strains. Genome Res. 23, 1329–1338 (2013).

    Article  CAS  Google Scholar 

  27. Taylor, J. H. Asynchronous duplication of chromosomes in cultured cells of chinese hamster. J. Cell. Biol. 7, 455–463 (1960).

    Article  CAS  Google Scholar 

  28. Morishima, A., Grumbach, M. M. & Taylor, J. H. Asynchronous duplication of human chromosomes and the origin of sex chromatin. Proc. Natl Acad. Sci. USA 48, 756–763 (1962).

    Article  CAS  Google Scholar 

  29. Hiratani, I. & Gilbert, D. M. Autosomal lyonization of replication domains during early mammalian development. Adv. Exp. Med. Biol. 695, 41–58 (2010).

    Article  CAS  Google Scholar 

  30. Hiratani, I., Takebayashi, S., Lu, J. & Gilbert, D. M. Replication timing and transcriptional control: beyond cause and effect-part II. Curr. Opin. Genet. Dev. 19, 142–149 (2009).

    Article  CAS  Google Scholar 

  31. Gimelbrant, A. A. & Chess, A. An epigenetic state associated with areas of gene duplication. Genome Res. 16, 723–729 (2006).

    Article  CAS  Google Scholar 

  32. Nagano, T. et al. Cell-cycle dynamics of chromosomal organization at single-cell resolution. Nature 547, 61–67 (2017).

    Article  CAS  Google Scholar 

  33. Flyamer, I. M. et al. Single-cell Hi-C reveals unique chromatin reorganization at oocyte-tozygote transition. Nature 544, 110–114 (2017).

    Article  CAS  Google Scholar 

  34. Stevens, T. J. et al. 3D structures of individual mammalian genomes studied by single-cell Hi-C. Nature 544, 59–64 (2017).

    Article  CAS  Google Scholar 

  35. Takebayashi, S. I., Ogata, M. & Okumura, K. Anatomy of mammalian replication domains. Genes 8, 110 (2017).

    Article  Google Scholar 

  36. Yamazaki, S., Hayano, M. & Masai, H. Replication timing regulation of eukaryotic replicons: Rif1 as a global regulator of replication timing. Trends Genet. 29, 449–460 (2013).

    Article  CAS  Google Scholar 

  37. Niwa, H. How is pluripotency determined and maintained? Development 134, 635–646 (2007).

    Article  CAS  Google Scholar 

  38. Meshorer, E. et al. Hyperdynamic plasticity of chromatin proteins in pluripotent embryonic stem cells. Dev. Cell. 10, 105–116 (2006).

    Article  CAS  Google Scholar 

  39. Fussner, E. et al. Constitutive heterochromatin reorganization during somatic cell reprogramming. EMBO J. 30, 1778–1789 (2011).

    Article  CAS  Google Scholar 

  40. van Steensel, B. & Belmont, A. S. Lamina-associated domains: links with chromosome architecture, heterochromatin, and gene repression. Cell 169, 780–791 (2017).

    Article  Google Scholar 

  41. Kind, J. et al. Genome-wide maps of nuclear lamina interactions in single human cells. Cell 163, 134–147 (2015).

    Article  CAS  Google Scholar 

  42. Das, S. P. et al. Replication timing is regulated by the number of MCMs loaded at origins. Genome Res. 25, 1886–1892 (2015).

    Article  CAS  Google Scholar 

  43. Dixon, J. R. et al. Topological domains in mammalian genomes identified by analysis of chromatin interactions. Nature 485, 376–380 (2012).

    Article  CAS  Google Scholar 

  44. Dimitrova, D. S. & Gilbert, D. M. The spatial position and replication timing of chromosomal domains are both established in early G1 phase. Mol. Cell 4, 983–993 (1999).

    Article  CAS  Google Scholar 

  45. Rao, S. S. P. et al. Cohesin loss eliminates all loop domains. Cell 171, 305–320 (2017).

    Article  CAS  Google Scholar 

  46. Nora, E. P. et al. Targeted degradation of ctcf decouples local insulation of chromosome domains from genomic compartmentalization. Cell 169, 930–944 (2017).

    Article  CAS  Google Scholar 

  47. Bolzer, A. et al. Three-dimensional maps of all chromosomes in human male fibroblast nuclei and prometaphase rosettes. PLoS Biol. 3, e157 (2005).

    Article  Google Scholar 

  48. Pope, B. D. et al. Replication-timing boundaries facilitate cell-type and species-specific regulation of a rearranged human chromosome in mouse. Hum. Mol. Genet. 21, 4162–4170 (2012).

    Article  CAS  Google Scholar 

  49. Macaulay, I. C., Ponting, C. P. & Voet, T. Single-cell multiomics: multiple measurements from single cells. Trends Genet. 33, 155–168 (2017).

    Article  CAS  Google Scholar 

  50. Calaway, J. D. et al. Genetic architecture of skewed X inactivation in the laboratory mouse. PLoS Genet. 9, e1003853 (2013).

    Article  Google Scholar 

  51. Hayashi, K. & Saitou, M. Generation of eggs from mouse embryonic stem cells and induced pluripotent stem cells. Nat. Protoc. 8, 1513–1524 (2013).

    Article  CAS  Google Scholar 

  52. Eiraku, M. & Sasai, Y. Mouse embryonic stem cell culture for generation of three-dimensional retinal and cortical tissues. Nat. Protoc. 7, 69–79 (2012).

    Article  CAS  Google Scholar 

  53. Rathjen, J. & Rathjen, P. D. Lineage specific differentiation of mouse ES cells: formation and differentiation of early primitive ectoderm-like (EPL) cells. Methods Enzymol. 365, 3–25 (2003).

    PubMed  Google Scholar 

  54. Kadota, M. et al. CTCF binding landscape in jawless fish with reference to Hox cluster evolution. Sci. Rep. 7, 4957 (2017).

    Article  Google Scholar 

  55. Martin, M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet J 17, 10–12 (2011).

    Article  Google Scholar 

  56. Sakata, Y. et al. Defects in dosage compensation impact global gene regulation in the mouse trophoblast. Development 144, 2784–2797 (2017).

    Article  CAS  Google Scholar 

  57. Keane, T. M. et al. Mouse genomic variation and its effect on phenotypes and gene regulation. Nature 477, 289–294 (2011).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  Google Scholar 

  60. Bakker, B. et al. Single-cell sequencing reveals karyotype heterogeneity in murine and human malignancies. Genome. Biol. 17, 1–15 (2016).

    Article  Google Scholar 

  61. Cayrou, C. et al. The chromatin environment shapes DNA replication origin organization and defines origin classes. Genome Res. 25, 1873–1885 (2015).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank S. Kuraku and members of his laboratory for assistance with NGS, F. Matsuzaki for the use of FACS and A. Tanigawa and Y. Kondo for technical assistance. We also thank D. M. Gilbert for exchanging unpublished observations; H. Niwa and K. Araki for CBMS1 mESCs; and B. D. Pope, M. Sugimoto and H. Masai for helpful discussions. This work was supported by a RIKEN CDB/BDR intramural grant to I.H., the Special Postdoctoral Researcher (SPDR) Program of RIKEN to S.T. and a Grant-in-Aid for Scientific Research on Innovative Areas (16H01405) to S-i.T., 18H05530 to I.H., 18K14681 to S.T., 15K06942, 15H01462 and 17H06426 to K.N., from the Ministry of Education, Culture, Sports, Science, and Technology (MEXT).

Author information

Authors and Affiliations

Authors

Contributions

S.T., H.M., T.S., S-i.T. and I.H. conceived the project. S.T., T.S. and S-i.T. developed and conducted scRepli-seq and BrdU-IP experiments. S.T. and I.H. performed mESC culture, differentiation and sample collection. T.S. and S-i.T. performed hTERT-RPE1 cell culture and sample collection. K.N. and C.O. constructed diploid reference genome and helped with the haplotype-resolved analysis pipeline setup. H.M. and S-i.T. performed bioinformatics analyses. K.O. and M.O. supported for the design and execution of the project. S.T., H.M., S-i.T. and I.H. wrote the manuscript.

Corresponding authors

Correspondence to Shin-ichiro Takebayashi or Ichiro Hiratani.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Takahashi, S., Miura, H., Shibata, T. et al. Genome-wide stability of the DNA replication program in single mammalian cells. Nat Genet 51, 529–540 (2019). https://doi.org/10.1038/s41588-019-0347-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-019-0347-5

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