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.

  • Review Article
  • Published:

Control of DNA replication timing in the 3D genome

Abstract

The 3D organization of mammalian chromatin was described more than 30 years ago by visualizing sites of DNA synthesis at different times during the S phase of the cell cycle. These early cytogenetic studies revealed structurally stable chromosome domains organized into subnuclear compartments. Active-gene-rich domains in the nuclear interior replicate early, whereas more condensed chromatin domains that are largely at the nuclear and nucleolar periphery replicate later. During the past decade, this spatiotemporal DNA replication programme has been mapped along the genome and found to correlate with epigenetic marks, transcriptional activity and features of 3D genome architecture such as chromosome compartments and topologically associated domains. But the causal relationship between these features and DNA replication timing and the regulatory mechanisms involved have remained an enigma. The recent identification of cis-acting elements regulating the replication time and 3D architecture of individual replication domains and of long non-coding RNAs that coordinate whole chromosome replication provide insights into such mechanisms.

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: Replication timing relationship to 3D chromatin structure.
Fig. 2: 3D chromatin structure and replication timing are dynamic during cell differentiation and during the cell cycle.
Fig. 3: Epigenetic versus sequence-specific regulation of replication timing.
Fig. 4: Cis- and trans-acting elements regulating replication timing.
Fig. 5: Proposed model of organization within the nucleus.

Similar content being viewed by others

References

  1. Taylor, J. H. Asynchronous duplication of chromosomes in cultured cells of Chinese hamster. J. Biophys. Biochem. Cytol. 7, 455–464 (1960).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Desprat, R. et al. Predictable dynamic program of timing of DNA replication in human cells. Genome Res. 19, 2288–2299 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Farkash-Amar, S. et al. Global organization of replication time zones of the mouse genome. Genome Res. 18, 1562–1570 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Hiratani, I. et al. Global reorganization of replication domains during embryonic stem cell differentiation. PLOS Biol. 6, e245 (2008).

    PubMed  PubMed Central  Google Scholar 

  5. MacAlpine, D. M., Rodríguez, H. K. & Bell, S. P. Coordination of replication and transcription along a Drosophila chromosome. Genes Dev. 18, 3094–3105 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Schübeler, D. et al. Genome-wide DNA replication profile for Drosophila melanogaster: a link between transcription and replication timing. Nat. Genet. 32, 438–442 (2002).

    PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  8. White, E. J. et al. DNA replication-timing analysis of human chromosome 22 at high resolution and different developmental states. Proc. Natl Acad. Sci. USA 101, 17771–17776 (2004).

    CAS  PubMed  Google Scholar 

  9. Rhind, N. & Gilbert, D. M. DNA replication timing. Cold Spring Harb. Perspect. Biol. 5, a010132 (2013).

    PubMed  PubMed Central  Google Scholar 

  10. Müller, C. A. & Nieduszynski, C. A. Conservation of replication timing reveals global and local regulation of replication origin activity. Genome Res. 22, 1953–1962 (2012).

    PubMed  PubMed Central  Google Scholar 

  11. Yang, Y. et al. Continuous-trait probabilistic model for comparing multi-species functional genomic data. Cell Syst. 7, 208–218 (2018).

    PubMed  PubMed Central  Google Scholar 

  12. Ryba, T. et al. Replication timing: a fingerprint for cell identity and pluripotency. PLOS Comput. Biol. 7, e1002225 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Farkash-Amar, S. & Simon, I. Genome-wide analysis of the replication program in mammals. Chromosome Res. 18, 115–125 (2010).

    CAS  PubMed  Google Scholar 

  15. Xu, J. et al. Genome-wide identification and characterization of replication origins by deep sequencing. Genome Biol. 13, R27 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Agier, N. et al. The evolution of the temporal program of genome replication. Nat. Commun. 9, 2199 (2018).

    PubMed  PubMed Central  Google Scholar 

  17. Mantiero, D., Mackenzie, A., Donaldson, A. & Zegerman, P. Limiting replication initiation factors execute the temporal programme of origin firing in budding yeast. EMBO J. 30, 4805–4814 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Rivera-Mulia, J. C. & Gilbert, D. M. Replicating large genomes: divide and conquer. Mol. Cell 62, 756–765 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Müller, C. A. & Nieduszynski, C. A. DNA replication timing influences gene expression level. J. Cell Biol. 216, 1907–1914 (2017).

    PubMed  PubMed Central  Google Scholar 

  20. Voichek, Y., Bar-Ziv, R. & Barkai, N. Expression homeostasis during DNA replication. Science 351, 1087–1090 (2016).

    CAS  PubMed  Google Scholar 

  21. Padovan-Merhar, O. et al. Single mammalian cells compensate for differences in cellular volume and DNA copy number through independent global transcriptional mechanisms. Mol. Cell 58, 339–352 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Sima, J. & Gilbert, D. M. Complex correlations: replication timing and mutational landscapes during cancer and genome evolution. Curr. Opin. Genet. Dev. 25, 93–100 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Supek, F. & Lehner, B. Differential DNA mismatch repair underlies mutation rate variation across the human genome. Nature 521, 81–84 (2015).

    CAS  Google Scholar 

  24. Kazanov, M. D. et al. APOBEC-induced cancer mutations are uniquely enriched in early-replicating, gene-dense, and active chromatin regions. Cell Rep. 13, 1103–1109 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Blumenfeld, B., Ben-Zimra, M. & Simon, I. Perturbations in the replication program contribute to genomic instability in cancer. Int. J. Mol. Sci. 18, E1138 (2017).

  26. Lu, J. et al. The distribution of genomic variations in human iPSCs is related to replication-timing reorganization during reprogramming. Cell Rep. 7, 70–78 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Lu, J. et al. Influence of ATM-mediated DNA damage response on genomic variation in human induced pluripotent stem cells. Stem Cells Dev. 25, 740–747 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Dileep, V. & Gilbert, D. M. Single-cell replication profiling to measure stochastic variation in mammalian replication timing. Nat. Commun. 9, 427 (2018). This article develops methods to quantify the degree of cell-to-cell and homologue-to-homologue variation in the replication timing programme in single-haploid and inter-species F1 hybrid mouse cells, demonstrating high conservation of replication timing and chromatin compartments, conclusions that were later confirmed in Takahashi et al. (2019).

    PubMed  PubMed Central  Google Scholar 

  29. Takahashi, S. et al. Genome-wide stability of the DNA replication program in single mammalian cells. Nat. Genet. 51, 529–540 (2019).

    CAS  PubMed  Google Scholar 

  30. Ozgyin, L., Horvath, A., Hevessy, Z. & Balint, B. L. Extensive epigenetic and transcriptomic variability between genetically identical human B-lymphoblastoid cells with implications in pharmacogenomics research. Sci. Rep. 9, 4889 (2019).

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Rivera-Mulia, J. C. et al. Dynamic changes in replication timing and gene expression during lineage specification of human pluripotent stem cells. Genome Res. 25, 1091–1103 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Wilson, K. A., Elefanty, A. G., Stanley, E. G. & Gilbert, D. M. Spatio-temporal re-organization of replication foci accompanies replication domain consolidation during human pluripotent stem cell lineage specification. Cell Cycle 15, 2464–2475 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  35. Xiang, W. et al. Correlative live and super-resolution imaging reveals the dynamic structure of replication domains. J. Cell Biol. 217, 1973–1984 (2018). This article labels replication foci and tracks their dynamic movements through the cell cycle at super-resolution in living cells, demonstrating that replication foci, consisting of several simultaneously labelled replication forks clustered in space, have properties resembling the behaviour of self-associating chromatin units or TADs, consistent with the genomic studies in Pope et al. (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Ryba, T. et al. Abnormal developmental control of replication-timing domains in pediatric acute lymphoblastic leukemia. Genome Res. 22, 1833–1844 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Rivera-Mulia, J. C. et al. DNA replication timing alterations identify common markers between distinct progeroid diseases. Proc. Natl Acad. Sci. USA 114, E10972–E10980 (2017).

    CAS  PubMed  Google Scholar 

  38. Sasaki, T. et al. Stability of patient-specific features of altered DNA replication timing in xenografts of primary human acute lymphoblastic leukemia. Exp. Hematol. 51, 71–82 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. 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).

    CAS  PubMed  Google Scholar 

  40. Stoffregen, E. P., Donley, N., Stauffer, D., Smith, L. & Thayer, M. J. An autosomal locus that controls chromosome-wide replication timing and mono-allelic expression. Hum. Mol. Genet. 20, 2366–2378 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Donley, N., Stoffregen, E. P., Smith, L., Montagna, C. & Thayer, M. J. Asynchronous replication, mono-allelic expression, and long range cis-effects of ASAR6. PLOS Genet. 9, e1003423 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Donley, N., Smith, L. & Thayer, M. J. ASAR15, A cis-acting locus that controls chromosome-wide replication timing and stability of human chromosome 15. PLOS Genet. 11, e1004923 (2015).

    PubMed  PubMed Central  Google Scholar 

  43. Platt, E. J., Smith, L. & Thayer, M. J. L1 retrotransposon antisense RNA within ASAR lncRNAs controls chromosome-wide replication timing. J. Cell Biol. 217, 541–553 (2018). This article demonstrates that the activity of ASARs, mono-allelically expressed lncRNAs, coating the chromosome from which they are expressed and required for coordination of replication timing between chromosome homologues, can be traced to much smaller L1 retrotransposons expressed in the antisense direction within the ASAR lncRNAs.

    CAS  PubMed  PubMed Central  Google Scholar 

  44. zur Hausen, H. Chromosomal changes of similar nature in seven established cell lines derived from the peripheral blood of patients with leukemia. J. Natl Cancer Inst. 38, 683–696 (1967).

    CAS  PubMed  Google Scholar 

  45. Smith, L., Plug, A. & Thayer, M. Delayed replication timing leads to delayed mitotic chromosome condensation and chromosomal instability of chromosome translocations. Proc. Natl Acad. Sci. USA 98, 13300–13305 (2001).

    CAS  PubMed  Google Scholar 

  46. Sparvoli, E., Levi, M. & Rossi, E. Replicon clusters may form structurally stable complexes of chromatin and chromosomes. J. Cell Sci. 107, 3097–3103 (1994).

    CAS  PubMed  Google Scholar 

  47. Ferreira, J., Paolella, G., Ramos, C. & Lamond, A. I. Spatial organization of large-scale chromatin domains in the nucleus: a magnified view of single chromosome territories. J. Cell Biol. 139, 1597–1610 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Jackson, D. A. & Pombo, A. Replicon clusters are stable units of chromosome structure: evidence that nuclear organization contributes to the efficient activation and propagation of S phase in human cells. J. Cell Biol. 140, 1285–1295 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. 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).

    CAS  PubMed  Google Scholar 

  50. Cremer, T. & Cremer, C. Chromosome territories, nuclear architecture and gene regulation in mammalian cells. Nat. Rev. Genet. 2, 292–301 (2001).

    CAS  PubMed  Google Scholar 

  51. Nakamura, H., Morita, T. & Sato, C. Structural organizations of replicon domains during DNA synthetic phase in the mammalian nucleus. Exp. Cell Res. 165, 291–297 (1986).

    CAS  PubMed  Google Scholar 

  52. O’Keefe, R. T., Henderson, S. C. & Spector, D. L. Dynamic organization of DNA replication in mammalian cell nuclei: spatially and temporally defined replication of chromosome-specific α-satellite DNA sequences. J. Cell Biol. 116, 1095–1110 (1992).

    PubMed  Google Scholar 

  53. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Solovei, I. et al. Nuclear architecture of rod photoreceptor cells adapts to vision in mammalian evolution. Cell 137, 356–368 (2009).

    CAS  PubMed  Google Scholar 

  55. Abney, J. R., Cutler, B., Fillbach, M. L., Axelrod, D. & Scalettar, B. A. Chromatin dynamics in interphase nuclei and its implications for nuclear structure. J. Cell Biol. 137, 1459–1468 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Yaffe, E. et al. Comparative analysis of DNA replication timing reveals conserved large-scale chromosomal architecture. PLOS Genet. 6, e1001011 (2010).

    PubMed  PubMed Central  Google Scholar 

  58. Rao, S. S. P. et al. A 3D map of the human genome at kilobase resolution reveals principles of chromatin looping. Cell 159, 1665–1680 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Rowley, M. J. et al. Evolutionarily conserved principles predict 3D chromatin organization. Mol. Cell 67, 837–852 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Cremer, T. et al. Role of chromosome territories in the functional compartmentalization of the cell nucleus. Cold Spring Harb. Symp. Quant. Biol. 58, 777–792 (1993).

    CAS  PubMed  Google Scholar 

  61. Diaz-Perez, S. V. et al. A deletion at the mouse Xist gene exposes trans-effects that alter the heterochromatin of the inactive X chromosome and the replication time and DNA stability of both X chromosomes. Genetics 174, 1115–1133 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Diaz-Perez, S. et al. The element(s) at the nontranscribed Xist locus of the active X chromosome controls chromosomal replication timing in the mouse. Genetics 171, 663–672 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Vogel, M. J., Peric-Hupkes, D. & van Steensel, B. Detection of in vivo protein–DNA interactions using DamID in mammalian cells. Nat. Protoc. 2, 1467–1478 (2007).

    CAS  PubMed  Google Scholar 

  64. Farkash-Amar, S. et al. Systematic determination of replication activity type highlights interconnections between replication, chromatin structure and nuclear localization. PLOS ONE 7, e48986 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Ragoczy, T., Telling, A., Scalzo, D., Kooperberg, C. & Groudine, M. Functional redundancy in the nuclear compartmentalization of the late-replicating genome. Nucleus 5, 626–635 (2014).

    PubMed  PubMed Central  Google Scholar 

  66. Pope, B. D. et al. Topologically associating domains are stable units of replication-timing regulation. Nature 515, 402–405 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Peric-Hupkes, D. et al. Molecular maps of the reorganization of genome–nuclear lamina interactions during differentiation. Mol. Cell 38, 603–613 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Kind, J. et al. Single-cell dynamics of genome–nuclear lamina interactions. Cell 153, 178–192 (2013).

    CAS  PubMed  Google Scholar 

  69. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Nora, E. P. et al. Spatial partitioning of the regulatory landscape of the X-inactivation centre. Nature 485, 381–385 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Moindrot, B. et al. 3D chromatin conformation correlates with replication timing and is conserved in resting cells. Nucleic Acids Res. 40, 9470–9481 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Rao, S. S. P. et al. Cohesin loss eliminates all loop domains. Cell 171, 305–320.e24 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Rodríguez-Carballo, E. et al. The HoxD cluster is a dynamic and resilient TAD boundary controlling the segregation of antagonistic regulatory landscapes. Genes Dev. 31, 2264–2281 (2017).

    PubMed  PubMed Central  Google Scholar 

  75. Sima, J. et al. Identifying cis elements for spatiotemporal control of mammalian DNA replication. Cell 176, 816–830 (2019). This article describes the discovery and characterization of cis-acting elements controlling early replication, chromatin compartment, TAD architecture and transcription of entire replication domains in mouse ESCs.

    CAS  PubMed  Google Scholar 

  76. Ma, J. & Duan, Z. Replication timing becomes intertwined with 3D genome organization. Cell 176, 681–684 (2019).

    CAS  PubMed  Google Scholar 

  77. Oldach, P. & Nieduszynski, C. A. Cohesin-mediated genome architecture does not define DNA replication timing domains. Genes 10, 196 (2019).

  78. Bintu, B. et al. Super-resolution chromatin tracing reveals domains and cooperative interactions in single cells. Science 362, eaau1783 (2018). This article demonstrates that TAD boundaries are variable from cell to cell, and that when cohesin is depleted, which has no effect on replication timing, TAD boundaries are still detectable in single cells but their positions are too stochastic to be detectable in ensemble/population genomics studies.

    Google Scholar 

  79. Kimura, A. & Horikoshi, M. Partition of distinct chromosomal regions: negotiable border and fixed border. Genes Cells 9, 499–508 (2004).

    CAS  PubMed  Google Scholar 

  80. Schwarzer, W. et al. Two independent modes of chromatin organization revealed by cohesin removal. Nature 551, 51–56 (2017).

    PubMed  PubMed Central  Google Scholar 

  81. Bonev, B. et al. Multiscale 3D genome rewiring during mouse neural development. Cell 171, 557–572 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Stadhouders, R. et al. Transcription factors orchestrate dynamic interplay between genome topology and gene regulation during cell reprogramming. Nat. Genet. 50, 238–249 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Dixon, J. R. et al. Chromatin architecture reorganization during stem cell differentiation. Nature 518, 331–336 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Pope, B. D. & Gilbert, D. M. The replication domain model: regulating replicon firing in the context of large-scale chromosome architecture. J. Mol. Biol. 425, 4690–4695 (2013).

    CAS  PubMed  Google Scholar 

  85. Takebayashi, S., Dileep, V., Ryba, T., Dennis, J. H. & Gilbert, D. M. Chromatin–interaction compartment switch at developmentally regulated chromosomal domains reveals an unusual principle of chromatin folding. Proc. Natl Acad. Sci. USA 109, 12574–12579 (2012).

    CAS  PubMed  Google Scholar 

  86. Fisher, C. L. & Fisher, A. G. Chromatin states in pluripotent, differentiated, and reprogrammed cells. Curr. Opin. Genet. Dev. 21, 140–146 (2011).

    CAS  PubMed  Google Scholar 

  87. Dileep, V. et al. Rapid irreversible transcriptional reprogramming in human stem cells accompanied by discordance between replication timing and chromatin compartment. Stem Cell Rep. 13, 193–206 (2019).

    CAS  Google Scholar 

  88. Ke, Y. et al. 3D Chromatin structures of mature gametes and structural reprogramming during mammalian embryogenesis. Cell 170, 367–381.e20 (2017).

    CAS  PubMed  Google Scholar 

  89. Ferreira, J. & Carmo-Fonseca, M. Genome replication in early mouse embryos follows a defined temporal and spatial order. J. Cell Sci. 110, 889–897 (1997).

    CAS  PubMed  Google Scholar 

  90. Siefert, J. C., Georgescu, C., Wren, J. D., Koren, A. & Sansam, C. L. DNA replication timing during development anticipates transcriptional programs and parallels enhancer activation. Genome Res. 27, 1406–1416 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Kaaij, L. J. T., van der Weide, R. H., Ketting, R. F. & de Wit, E. Systemic loss and gain of chromatin architecture throughout zebrafish development. Cell Rep. 24, 1–10 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Hug, C. B., Grimaldi, A. G., Kruse, K. & Vaquerizas, J. M. Chromatin architecture emerges during zygotic genome activation independent of transcription. Cell 169, 216–228 (2017).

    CAS  PubMed  Google Scholar 

  93. Sasaki, T., Sawado, T., Yamaguchi, M. & Shinomiya, T. Specification of regions of DNA replication initiation during embryogenesis in the 65-kilobase DNApolα-dE2F locus of Drosophila melanogaster. Mol. Cell. Biol. 19, 547–555 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Hyrien, O., Maric, C. & Méchali, M. Transition in specification of embryonic metazoan DNA replication origins. Science 270, 994–997 (1995).

    CAS  PubMed  Google Scholar 

  95. Seller, C. A., Cho, C.-Y. & O’Farrell, P. H. Rapid embryonic cell cycles defer the establishment of heterochromatin by Eggless/SetDB1 in Drosophila. Genes Dev. 33, 403–417 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Hiratani, I. & Takahashi, S. DNA replication timing enters the single-cell era. Genes 10, 221 (2019).

  97. Nagano, T. et al. Single-cell Hi-C reveals cell-to-cell variability in chromosome structure. Nature 502, 59–64 (2013).

    CAS  PubMed  Google Scholar 

  98. Raghuraman, M. K., Brewer, B. J. & Fangman, W. L. Cell cycle-dependent establishment of a late replication program. Science 276, 806–809 (1997).

    CAS  PubMed  Google Scholar 

  99. Lu, J., Li, F., Murphy, C. S., Davidson, M. W. & Gilbert, D. M. G2 phase chromatin lacks determinants of replication timing. J. Cell Biol. 189, 967–980 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Wu, J. R. & Gilbert, D. M. Lovastatin arrests CHO cells between the origin decision point and the restriction point. FEBS Lett. 484, 108–112 (2000).

    CAS  PubMed  Google Scholar 

  103. Li, F., Chen, J., Solessio, E. & Gilbert, D. M. Spatial distribution and specification of mammalian replication origins during G1 phase. J. Cell Biol. 161, 257–266 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Demczuk, A. et al. Regulation of DNA replication within the immunoglobulin heavy-chain locus during B cell commitment. PLOS Biol. 10, e1001360 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Kitsberg, D. et al. Allele-specific replication timing of imprinted gene regions. Nature 364, 459–463 (1993).

    CAS  PubMed  Google Scholar 

  106. Kawame, H., Gartler, S. M. & Hansen, R. S. Allele-specific replication timing in imprinted domains: absence of asynchrony at several loci. Hum. Mol. Genet. 4, 2287–2293 (1995).

    CAS  PubMed  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Masika, H. et al. Programming asynchronous replication in stem cells. Nat. Struct. Mol. Biol. 24, 1132–1138 (2017).

    CAS  PubMed  Google Scholar 

  110. Li, J., Santoro, R., Koberna, K. & Grummt, I. The chromatin remodeling complex NoRC controls replication timing of rRNA genes. EMBO J. 24, 120–127 (2005).

    PubMed  Google Scholar 

  111. Schlesinger, S., Selig, S., Bergman, Y. & Cedar, H. Allelic inactivation of rDNA loci. Genes Dev. 23, 2437–2447 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  112. Yokochi, T. et al. G9a selectively represses a class of late-replicating genes at the nuclear periphery. Proc. Natl Acad. Sci. USA 106, 19363–19368 (2009).

    CAS  PubMed  Google Scholar 

  113. Yue, F. et al. A comparative encyclopedia of DNA elements in the mouse genome. Nature 515, 355–364 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Chen, C. C. L. et al. H3S10ph broadly marks early-replicating domains in interphase ESCs and shows reciprocal antagonism with H3K9me2. Genome Res. 28, 37–51 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  115. Brustel, J. et al. Histone H4K20 tri-methylation at late-firing origins ensures timely heterochromatin replication. EMBO J. 36, 2726–2741 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Alabert, C. et al. Two distinct modes for propagation of histone PTMs across the cell cycle. Genes Dev. 29, 585–590 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Schübeler, D. et al. Nuclear localization and histone acetylation: a pathway for chromatin opening and transcriptional activation of the human β-globin locus. Genes Dev. 14, 940–950 (2000).

    PubMed  PubMed Central  Google Scholar 

  118. Epner, E., Forrester, W. C. & Groudine, M. Asynchronous DNA replication within the human beta-globin gene locus. Proc. Natl Acad. Sci. USA 85, 8081–8085 (1988).

    CAS  PubMed  Google Scholar 

  119. Kemp, M. G., Ghosh, M., Liu, G. & Leffak, M. The histone deacetylase inhibitor trichostatin A alters the pattern of DNA replication origin activity in human cells. Nucleic Acids Res. 33, 325–336 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Goren, A., Tabib, A., Hecht, M. & Cedar, H. DNA replication timing of the human β-globin domain is controlled by histone modification at the origin. Genes Dev. 22, 1319–1324 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Casas-Delucchi, C. S. et al. Histone hypoacetylation is required to maintain late replication timing of constitutive heterochromatin. Nucleic Acids Res. 40, 159–169 (2012).

    CAS  PubMed  Google Scholar 

  122. Yoshida, K. et al. The histone deacetylases sir2 and rpd3 act on ribosomal DNA to control the replication program in budding yeast. Mol. Cell 54, 691–697 (2014).

    CAS  PubMed  Google Scholar 

  123. Lande-Diner, L., Zhang, J. & Cedar, H. Shifts in replication timing actively affect histone acetylation during nucleosome reassembly. Mol. Cell 34, 767–774 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Zhang, J., Xu, F., Hashimshony, T., Keshet, I. & Cedar, H. Establishment of transcriptional competence in early and late S phase. Nature 420, 198–202 (2002).

    CAS  PubMed  Google Scholar 

  125. Gindin, Y., Valenzuela, M. S., Aladjem, M. I., Meltzer, P. S. & Bilke, S. A chromatin structure-based model accurately predicts DNA replication timing in human cells. Mol. Syst. Biol. 10, 722 (2014).

    PubMed  PubMed Central  Google Scholar 

  126. Takebayashi, S., Ryba, T. & Gilbert, D. M. Developmental control of replication timing defines a new breed of chromosomal domains with a novel mechanism of chromatin unfolding. Nucleus 3, 500–507 (2012).

    PubMed  PubMed Central  Google Scholar 

  127. 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).

    CAS  PubMed  Google Scholar 

  128. Mesner, L. D. et al. Bubble-seq analysis of the human genome reveals distinct chromatin-mediated mechanisms for regulating early- and late-firing origins. Genome Res. 23, 1774–1788 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  129. Petryk, N. et al. Replication landscape of the human genome. Nat. Commun. 7, 10208 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  130. Langley, A. R., Gräf, S., Smith, J. C. & Krude, T. Genome-wide identification and characterisation of human DNA replication origins by initiation site sequencing (ini-seq). Nucleic Acids Res. 44, 10230–10247 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Dellino, G. I. et al. Genome-wide mapping of human DNA-replication origins: levels of transcription at ORC1 sites regulate origin selection and replication timing. Genome Res. 23, 1–11 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  132. Miotto, B., Ji, Z. & Struhl, K. Selectivity of ORC binding sites and the relation to replication timing, fragile sites, and deletions in cancers. Proc. Natl Acad. Sci. USA 113, E4810–E4819 (2016).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  134. Cadoret, J.-C. et al. Genome-wide studies highlight indirect links between human replication origins and gene regulation. Proc. Natl Acad. Sci. USA 105, 15837–15842 (2008).

    CAS  PubMed  Google Scholar 

  135. Picard, F. et al. The spatiotemporal program of DNA replication is associated with specific combinations of chromatin marks in human cells. PLOS Genet. 10, e1004282 (2014).

    PubMed  PubMed Central  Google Scholar 

  136. Bartholdy, B., Mukhopadhyay, R., Lajugie, J., Aladjem, M. I. & Bouhassira, E. E. Allele-specific analysis of DNA replication origins in mammalian cells. Nat. Commun. 6, 7051 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  137. Stinchcomb, D. T., Struhl, K. & Davis, R. W. Isolation and characterisation of a yeast chromosomal replicator. Nature 282, 39–43 (1979).

    CAS  PubMed  Google Scholar 

  138. Gilbert, D. M. Making sense of eukaryotic DNA replication origins. Science 294, 96–100 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  139. Fang, D. et al. Dbf4 recruitment by forkhead transcription factors defines an upstream rate-limiting step in determining origin firing timing. Genes Dev. 31, 2405–2415 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  140. Ostrow, A. Z. et al. Conserved forkhead dimerization motif controls DNA replication timing and spatial organization of chromosomes in S. cerevisiae. Proc. Natl Acad. Sci. USA 114, E2411–E2419 (2017). This article demonstrates the separation of transcription and replication timing functions of forkhead proteins; mutations that disrupt the dimerization of forkhead proteins disrupt the 3D clustering of early replication origins and their early replication but do not affect transcriptional control by these proteins.

    CAS  PubMed  Google Scholar 

  141. Simon, I. et al. Developmental regulation of DNA replication timing at the human beta globin locus. EMBO J. 20, 6150–6157 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  142. Hassan-Zadeh, V. et al. USF binding sequences from the HS4 insulator element impose early replication timing on a vertebrate replicator. PLOS Biol. 10, e1001277 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  143. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  145. Wutz, A. & Jaenisch, R. A shift from reversible to irreversible X inactivation is triggered during ES cell differentiation. Mol. Cell 5, 695–705 (2000).

    CAS  PubMed  Google Scholar 

  146. Friedman, K. L. et al. Multiple determinants controlling activation of yeast replication origins late in S phase. Genes Dev. 10, 1595–1607 (1996).

    CAS  PubMed  Google Scholar 

  147. Yompakdee, C. & Huberman, J. A. Enforcement of late replication origin firing by clusters of short G-rich DNA sequences. J. Biol. Chem. 279, 42337–42344 (2004).

    CAS  PubMed  Google Scholar 

  148. Forrester, W. C. et al. A deletion of the human β-globin locus activation region causes a major alteration in chromatin structure and replication across the entire β-globin locus. Genes Dev. 4, 1637–1649 (1990).

    CAS  PubMed  Google Scholar 

  149. Aladjem, M. I. et al. Participation of the human β-globin locus control region in initiation of DNA replication. Science 270, 815–819 (1995).

    CAS  PubMed  Google Scholar 

  150. Cimbora, D. M. et al. Long-distance control of origin choice and replication timing in the human β-globin locus are independent of the locus control region. Mol. Cell. Biol. 20, 5581–5591 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  152. Tanaka, S. et al. CDK-dependent phosphorylation of Sld2 and Sld3 initiates DNA replication in budding yeast. Nature 445, 328–332 (2007).

    CAS  PubMed  Google Scholar 

  153. Mailand, N. & Diffley, J. F. X. CDKs promote DNA replication origin licensing in human cells by protecting Cdc6 from APC/C-dependent proteolysis. Cell 122, 915–926 (2005).

    CAS  PubMed  Google Scholar 

  154. Owens, J. C., Detweiler, C. S. & Li, J. J. CDC45 is required in conjunction with CDC7/DBF4 to trigger the initiation of DNA replication. Proc. Natl Acad. Sci. USA 94, 12521–12526 (1997).

    CAS  PubMed  Google Scholar 

  155. Jiang, W., McDonald, D., Hope, T. J. & Hunter, T. Mammalian Cdc7–Dbf4 protein kinase complex is essential for initiation of DNA replication. EMBO J. 18, 5703–5713 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  156. Moyer, S. E., Lewis, P. W. & Botchan, M. R. Isolation of the Cdc45/Mcm2-7/GINS (CMG) complex, a candidate for the eukaryotic DNA replication fork helicase. Proc. Natl Acad. Sci. USA 103, 10236–10241 (2006).

    CAS  PubMed  Google Scholar 

  157. Kumagai, A., Shevchenko, A., Shevchenko, A. & Dunphy, W. G. Treslin collaborates with TopBP1 in triggering the initiation of DNA replication. Cell 140, 349–359 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  158. Guo, C. et al. Interaction of Chk1 with Treslin negatively regulates the initiation of chromosomal DNA replication. Mol. Cell 57, 492–505 (2015).

    CAS  PubMed  Google Scholar 

  159. Tazumi, A. et al. Telomere-binding protein Taz1 controls global replication timing through its localization near late replication origins in fission yeast. Genes Dev. 26, 2050–2062 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  160. Hayashi, M. T., Takahashi, T. S., Nakagawa, T., Nakayama, J. & Masukata, H. The heterochromatin protein Swi6/HP1 activates replication origins at the pericentromeric region and silent mating-type locus. Nat. Cell Biol. 11, 357–362 (2009).

    CAS  PubMed  Google Scholar 

  161. Natsume, T. et al. Kinetochores coordinate pericentromeric cohesion and early DNA replication by Cdc7–Dbf4 kinase recruitment. Mol. Cell 50, 661–674 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  162. Utani, K. et al. Phosphorylated SIRT1 associates with replication origins to prevent excess replication initiation and preserve genomic stability. Nucleic Acids Res. 45, 7807–7824 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  163. Fatoba, S. T. et al. Human SIRT1 regulates DNA binding and stability of the Mcm10 DNA replication factor via deacetylation. Nucleic Acids Res. 41, 4065–4079 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  164. Cornacchia, D. et al. Mouse Rif1 is a key regulator of the replication-timing programme in mammalian cells. EMBO J. 31, 3678–3690 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  165. Yamazaki, S. et al. Rif1 regulates the replication timing domains on the human genome. EMBO J. 31, 3667–3677 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  166. Foti, R. et al. Nuclear architecture organized by Rif1 underpins the replication-timing program. Mol. Cell 61, 260–273 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  167. Hiraga, S.-I. et al. Human RIF1 and protein phosphatase 1 stimulate DNA replication origin licensing but suppress origin activation. EMBO Rep. 18, 403–419 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  168. Hiraga, S.-I. et al. Rif1 controls DNA replication by directing protein phosphatase 1 to reverse Cdc7-mediated phosphorylation of the MCM complex. Genes Dev. 28, 372–383 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  169. Sreesankar, E., Bharathi, V., Mishra, R. K. & Mishra, K. Drosophila Rif1 is an essential gene and controls late developmental events by direct interaction with PP1-87B. Sci. Rep. 5, 10679 (2015).

    PubMed  PubMed Central  Google Scholar 

  170. Sukackaite, R. et al. Mouse Rif1 is a regulatory subunit of protein phosphatase 1 (PP1). Sci. Rep. 7, 2119 (2017).

    PubMed  PubMed Central  Google Scholar 

  171. Hiraga, S.-I. et al. Budding yeast Rif1 binds to replication origins and protects DNA at blocked replication forks. EMBO Rep. 19, e46222 (2018).

  172. Ogawa, S. et al. Shelterin promotes tethering of late replication origins to telomeres for replication-timing control. EMBO J. 37, e98997 (2018).

  173. Knott, S. R. V. et al. Forkhead transcription factors establish origin timing and long-range clustering in S. cerevisiae. Cell 148, 99–111 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  174. Sansam, C. G. et al. A mechanism for epigenetic control of DNA replication. Genes Dev. 32, 224–229 (2018). This article shows that human Treslin directly interacts with the histone acetylation readers BRD2/BRD4 and this interaction is necessary for the normal spatiotemporal distribution of replication foci, providing a clear link between histone acetylation, pre-RC activation and replication timing.

    CAS  PubMed  PubMed Central  Google Scholar 

  175. Goldman, M. A., Holmquist, G. P., Gray, M. C., Caston, L. A. & Nag, A. Replication timing of genes and middle repetitive sequences. Science 224, 686–692 (1984).

    CAS  PubMed  Google Scholar 

  176. Hansen, R. S., Canfield, T. K., Fjeld, A. D. & Gartler, S. M. Role of late replication timing in the silencing of X-linked genes. Hum. Mol. Genet. 5, 1345–1353 (1996).

    CAS  PubMed  Google Scholar 

  177. Woodfine, K. et al. Replication timing of human chromosome 6. Cell Cycle 4, 172–176 (2005).

    CAS  PubMed  Google Scholar 

  178. Therizols, P. et al. Chromatin decondensation is sufficient to alter nuclear organization in embryonic stem cells. Science 346, 1238–1242 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  179. Hatton, K. S. et al. Replication program of active and inactive multigene families in mammalian cells. Mol. Cell. Biol. 8, 2149–2158 (1988).

    CAS  PubMed  PubMed Central  Google Scholar 

  180. Raghuraman, M. K. et al. Replication dynamics of the yeast genome. Science 294, 115–121 (2001).

    CAS  PubMed  Google Scholar 

  181. Reik, A. et al. The locus control region is necessary for gene expression in the human β-globin locus but not the maintenance of an open chromatin structure in erythroid cells. Mol. Cell. Biol. 18, 5992–6000 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  182. Blin, M. et al. Transcription-dependent regulation of replication dynamics modulates genome stability. Nat. Struct. Mol. Biol. 26, 58–66 (2019).

    CAS  PubMed  Google Scholar 

  183. Pourkarimi, E., Bellush, J. M. & Whitehouse, I. Spatiotemporal coupling and decoupling of gene transcription with DNA replication origins during embryogenesis in C. elegans. eLife 5, e21728 (2016).

    PubMed  PubMed Central  Google Scholar 

  184. Seller, C. A. & O’Farrell, P. H. Rif1 prolongs the embryonic S phase at the Drosophila mid-blastula transition. PLOS Biol. 16, e2005687 (2018). This article presents an elegant study of how the onset of a replication timing programme during Drosophila mid-blastula transition is regulated through the interplay of Rif1 and S-phase kinases.

    PubMed  PubMed Central  Google Scholar 

  185. Rivera-Mulia, J. C., Kim, S., Gabr, H., Kahveci, T. & Gilbert, D. M. Replication timing networks: a novel class of gene regulatory networks. Genome Res. in the press (2019).

    Article  Google Scholar 

  186. Sabari, B. R. et al. Coactivator condensation at super-enhancers links phase separation and gene control. Science 361, eaar3958 (2018).

    Google Scholar 

  187. Denker, A. & de Laat, W. The second decade of 3C technologies: detailed insights into nuclear organization. Genes Dev. 30, 1357–1382 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  189. Phillips-Cremins, J. E. et al. Architectural protein subclasses shape 3D organization of genomes during lineage commitment. Cell 153, 1281–1295 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  190. Battulin, N. et al. Comparison of the three-dimensional organization of sperm and fibroblast genomes using the Hi-C approach. Genome Biol. 16, 77 (2015).

    PubMed  PubMed Central  Google Scholar 

  191. Berlivet, S. et al. Clustering of tissue-specific sub-TADs accompanies the regulation of HoxA genes in developing limbs. PLOS Genet. 9, e1004018 (2013).

    PubMed  PubMed Central  Google Scholar 

  192. Méndez, J. & Stillman, B. Chromatin association of human origin recognition complex, cdc6, and minichromosome maintenance proteins during the cell cycle: assembly of prereplication complexes in late mitosis. Mol. Cell. Biol. 20, 8602–8612 (2000).

    PubMed  PubMed Central  Google Scholar 

  193. Okuno, Y., McNairn, A. J., den Elzen, N., Pines, J. & Gilbert, D. M. Stability, chromatin association and functional activity of mammalian pre-replication complex proteins during the cell cycle. EMBO J. 20, 4263–4277 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  194. Dimitrova, D. S., Prokhorova, T. A., Blow, J. J., Todorov, I. T. & Gilbert, D. M. Mammalian nuclei become licensed for DNA replication during late telophase. J. Cell Sci. 115, 51–59 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  195. Im, J.-S. et al. Assembly of the Cdc45–Mcm2-7–GINS complex in human cells requires the Ctf4/And-1, RecQL4, and Mcm10 proteins. Proc. Natl Acad. Sci. USA 106, 15628–15632 (2009).

    CAS  PubMed  Google Scholar 

  196. Douglas, M. E., Ali, F. A., Costa, A. & Diffley, J. F. X. The mechanism of eukaryotic CMG helicase activation. Nature 555, 265–268 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  197. Diffley, J. F. Once and only once upon a time: specifying and regulating origins of DNA replication in eukaryotic cells. Genes Dev. 10, 2819–2830 (1996).

    CAS  PubMed  Google Scholar 

  198. Anglana, M., Apiou, F., Bensimon, A. & Debatisse, M. Dynamics of DNA replication in mammalian somatic cells: nucleotide pool modulates origin choice and interorigin spacing. Cell 114, 385–394 (2003).

    CAS  PubMed  Google Scholar 

  199. Sasaki, T. et al. The Chinese hamster dihydrofolate reductase replication origin decision point follows activation of transcription and suppresses initiation of replication within transcription units. Mol. Cell. Biol. 26, 1051–1062 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  200. Woodward, A. M. et al. Excess Mcm2-7 license dormant origins of replication that can be used under conditions of replicative stress. J. Cell Biol. 173, 673–683 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  201. Ge, X. Q., Jackson, D. A. & Blow, J. J. Dormant origins licensed by excess Mcm2-7 are required for human cells to survive replicative stress. Genes Dev. 21, 3331–3341 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  202. Maya-Mendoza, A. & Jackson, D. A. Labeling DNA. replication foci to visualize chromosome territories in vivo. Curr. Protoc. Cell Biol. 75, 22.21.1–22.21.16 (2017).

    Google Scholar 

  203. Maya-Mendoza, A., Olivares-Chauvet, P., Shaw, A. & Jackson, D. A. S phase progression in human cells is dictated by the genetic continuity of DNA foci. PLOS Genet. 6, e1000900 (2010).

    PubMed  PubMed Central  Google Scholar 

  204. Marchal, C. et al. Genome-wide analysis of replication timing by next-generation sequencing with E/L Repli-seq. Nat. Protoc. 13, 819–839 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  205. Zuin, J. et al. Cohesin and CTCF differentially affect chromatin architecture and gene expression in human cells. Proc. Natl Acad. Sci. USA 111, 996–1001 (2014).

    CAS  PubMed  Google Scholar 

  206. Geeven, G. et al. Local compartment changes and regulatory landscape alterations in histone H1-depleted cells. Genome Biol. 16, 289 (2015).

    PubMed  PubMed Central  Google Scholar 

  207. Takebayashi, S.-I. et al. Murine esBAF chromatin remodeling complex subunits BAF250a and Brg1 are necessary to maintain and reprogram pluripotency-specific replication timing of select replication domains. Epigenetics Chromatin 6, 42 (2013).

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by National Institutes of Health (NIH) grants GM083337 and DK107965 to D.M.G. The authors thank M. Thayer for helpful discussions and critique of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

C.M. and D.M.G. have contributed equally to all stages of writing and revision of the article and display items; J.S. contributed to the initial draft of the main text.

Corresponding author

Correspondence to David M. Gilbert.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information

Nature Reviews Molecular Cell Biology thanks Sara Buonomo, Carl Schildkraut and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Glossary

Constant timing regions

(CTRs). Regions of the chromatin containing one or more adjacent replication domains that are replicating at nearly the same time.

Replicons

Units of replication comprising a replication origin and the DNA being replicated.

Timing transition regions

(TTRs). Regions of the chromatin between two replications domains replicating at different times.

Replication forks

Complexes comprising template DNA, oligonucleotide primers and proteins necessary for DNA replication. Two ‘sister’ replication forks are assembled at each replication origin and may be associated in 3D space.

Replication stress

Stalling of replication forks, which can be caused by chemical interference or radiation, or can be the result of a lack of nucleotides or proteins necessary for DNA replication.

Mismatch repair

(MMR). DNA repair mechanism occurring at the replication fork involved in the repair of base mismatch and small insertions/deletions occurring during DNA replication or recombination.

Hi-C

A genome-wide chromatin conformation capture technology that uses chromatin digestion, re-ligation of sequences in close proximity and sequencing to identify chromatin pairwise 3D chromatin interactions in the nucleus.

Principal component analysis

(PCA). Mathematical transformation applied on complex (with multiple variables) datasets, to extract values describing the data called PC1, PC2 and so forth, with PC1 the value best describing the data.

Xist

Long non-coding RNA that is expressed in female cells from only one X chromosome. Xist inactivates the X chromosome from which it is expressed, enabling dosage compensation (similar X-linked gene expression levels in male and female cells).

Asynchronous replication and autosomal RNAs

(ASARs). Long non-coding RNAs that are essential for the timely replication and condensation of the entire chromosome from which they are expressed.

Lamina-associated domains

(LADs). Domains of chromatin that come in close proximity to the nuclear lamina.

Inter-LADs

Domains between two adjacent lamina-associated domains.

Topologically associated domains

(TADs). Domains of chromatin enriched for 3D interactions within the domain as compared with between domains.

Correlative live and super-resolution microscopy

Combines the temporal resolution of time-lapse fluorescence microscopy with the spatial resolution of super-resolution microscopy.

CCCTC-binding factor

(CTCF). Protein highly conserved in eukaryotes that associates with chromatin to mediate transcription and chromatin insulation.

Cohesin

Protein complex composed of SMC1 and SMC3 that forms a large ring structure that accommodates two strands of chromatin. Cohesin has a key architectural role, forming chromatin loops and maintaining sister chromatids tied together after DNA replication.

Early replication control elements

(ERCEs). DNA sequences necessary for the early replication of entire replication domains.

Super-enhancers

Chromatin region rich in enhancers, with a high level of transcription associated factors and acetylated histones.

Chromatin conformation capture

Technologies that assess 3D interactions within chromatin. These technologies are based on the cutting and re-ligation of chromatin immobilized in intact nuclei and identification of pairwise interactions by quantitative PCR or sequencing.

Zygotic genome activation

(ZGA). Stage of embryonic development at which the transcription of the zygotic genome becomes activated.

Imprinted genes

Genes that are expressed only from one chromosome homologue, the choice of which is dependent on its parental origin. The mechanism behind genomic imprinting relies on DNA methylation.

β-Globin domain

A megabase-sized locus containing the β-globin gene, which is expressed only in erythroid cells. DNA replication properties of this domain have been extensively studied as it replicates early in erythroid cells and late in other cell types.

Ribosomal DNA

(rDNA). Part of the genome coding for ribosomal RNA. rDNA is constituted of several copies of the same genes, the expression of each varying depending on cell type.

Indels

Insertions or deletions in the genome of a cell or an individual.

Long interspersed nuclear element 1

(LINE1). Class of transposable elements estimated to be present at around 500,000 copies in the human and mouse genomes.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Marchal, C., Sima, J. & Gilbert, D.M. Control of DNA replication timing in the 3D genome. Nat Rev Mol Cell Biol 20, 721–737 (2019). https://doi.org/10.1038/s41580-019-0162-y

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41580-019-0162-y

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