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:

Polymerase δ replicates both strands after homologous recombination–dependent fork restart

Abstract

To maintain genetic stability, DNA must be replicated only once per cell cycle, and replication must be completed even when individual replication forks are inactivated. Because fork inactivation is common, passive convergence of an adjacent fork is insufficient to rescue all inactive forks. Thus, eukaryotic cells have evolved homologous recombination–dependent mechanisms to restart persistent inactive forks. Completing DNA synthesis via homologous recombination–restarted replication (HoRReR) ensures cell survival, but at a cost. One such cost is increased mutagenesis because HoRReR is more error prone than canonical replication. This increased error rate implies the HoRReR mechanism is distinct from that of a canonical fork. Here we demonstrate, in Schizosaccharomyces pombe, that a DNA sequence duplicated by HoRReR during S phase is replicated semiconservatively, but both the leading and lagging strands are synthesized by DNA polymerase δ.

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

Figure 1: Polδ, but not Polɛ, participates in bulk DNA synthesis after HR restart.
Figure 2: Polδ usage is relatively uniform, and Polα usage decreases.
Figure 3: HR-restarted replication is semiconservative.
Figure 4: Instability resulting from HR-restarted replication is not intrinsic to leading-strand synthesis by Polδ.
Figure 5: Polδ usage correlates with fragile sites.

Similar content being viewed by others

References

  1. Lambert, S. & Carr, A.M. Replication stress and genome rearrangements: lessons from yeast models. Curr. Opin. Genet. Dev. 23, 132–139 (2013).

    CAS  PubMed  Google Scholar 

  2. Liu, P., Carvalho, C.M., Hastings, P.J. & Lupski, J.R. Mechanisms for recurrent and complex human genomic rearrangements. Curr. Opin. Genet. Dev. 22, 211–220 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Sun, Z. et al. Replicative mechanisms of CNV formation preferentially occur as intrachromosomal events: evidence from Potocki-Lupski duplication syndrome. Hum. Mol. Genet. 22, 749–756 (2013).

    CAS  PubMed  Google Scholar 

  4. Carvalho, C.M. et al. Replicative mechanisms for CNV formation are error prone. Nat. Genet. 45, 1319–1326 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Bignell, G.R. et al. Architectures of somatic genomic rearrangement in human cancer amplicons at sequence-level resolution. Genome Res. 17, 1296–1303 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Greenman, C. et al. Patterns of somatic mutation in human cancer genomes. Nature 446, 153–158 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Barlow, J.H. et al. Identification of early replicating fragile sites that contribute to genome instability. Cell 152, 620–632 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Lambert, S. & Carr, A.M. Impediments to replication fork movement: stabilisation, reactivation and genome instability. Chromosoma 122, 33–45 (2013).

    CAS  PubMed  Google Scholar 

  9. Errico, A. & Costanzo, V. Mechanisms of replication fork protection: a safeguard for genome stability. Crit. Rev. Biochem. Mol. Biol. 47, 222–235 (2012).

    CAS  PubMed  Google Scholar 

  10. Lopes, M. et al. The DNA replication checkpoint response stabilizes stalled replication forks. Nature 412, 557–561 (2001).

    CAS  PubMed  Google Scholar 

  11. Higgs, M.R. et al. BOD1L is required to suppress deleterious resection of stressed replication forks. Mol. Cell 59, 462–477 (2015).

    CAS  PubMed  Google Scholar 

  12. 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 

  13. Lambert, S., Watson, A., Sheedy, D.M., Martin, B. & Carr, A.M. Gross chromosomal rearrangements and elevated recombination at an inducible site-specific replication fork barrier. Cell 121, 689–702 (2005).

    CAS  PubMed  Google Scholar 

  14. Anand, R.P., Lovett, S.T. & Haber, J.E. Break-induced DNA replication. Cold Spring Harb. Perspect. Biol. 5, a010397 (2013).

    PubMed  PubMed Central  Google Scholar 

  15. Jain, S. et al. A recombination execution checkpoint regulates the choice of homologous recombination pathway during DNA double-strand break repair. Genes Dev. 23, 291–303 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Malkova, A. & Ira, G. Break-induced replication: functions and molecular mechanism. Curr. Opin. Genet. Dev. 23, 271–279 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Lambert, S. et al. Homologous recombination restarts blocked replication forks at the expense of genome rearrangements by template exchange. Mol. Cell 39, 346–359 (2010).

    CAS  PubMed  Google Scholar 

  18. Mohebi, S., Mizuno, K., Watson, A., Carr, A.M. & Murray, J.M. Checkpoints are blind to replication restart and recombination intermediates that result in gross chromosomal rearrangements. Nat. Commun. 6, 6357 (2015).

    CAS  PubMed  Google Scholar 

  19. Nguyen, M.O., Jalan, M., Morrow, C.A., Osman, F. & Whitby, M.C. Recombination occurs within minutes of replication blockage by RTS1 producing restarted forks that are prone to collapse. eLife 4, e04539 (2015).

    PubMed  PubMed Central  Google Scholar 

  20. Mizuno, K., Lambert, S., Baldacci, G., Murray, J.M. & Carr, A.M. Nearby inverted repeats fuse to generate acentric and dicentric palindromic chromosomes by a replication template exchange mechanism. Genes Dev. 23, 2876–2886 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Smith, C.E., Llorente, B. & Symington, L.S. Template switching during break-induced replication. Nature 447, 102–105 (2007).

    CAS  PubMed  Google Scholar 

  22. Deem, A. et al. Break-induced replication is highly inaccurate. PLoS Biol. 9, e1000594 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Costantino, L. et al. Break-induced replication repair of damaged forks induces genomic duplications in human cells. Science 343, 88–91 (2014).

    CAS  PubMed  Google Scholar 

  24. Iraqui, I. et al. Recovery of arrested replication forks by homologous recombination is error-prone. PLoS Genet. 8, e1002976 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Mizuno, K., Miyabe, I., Schalbetter, S.A., Carr, A.M. & Murray, J.M. Recombination-restarted replication makes inverted chromosome fusions at inverted repeats. Nature 493, 246–249 (2013).

    CAS  PubMed  Google Scholar 

  26. Eydmann, T. et al. Rtf1-mediated eukaryotic site-specific replication termination. Genetics 180, 27–39 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Calzada, A., Hodgson, B., Kanemaki, M., Bueno, A. & Labib, K. Molecular anatomy and regulation of a stable replisome at a paused eukaryotic DNA replication fork. Genes Dev. 19, 1905–1919 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Miyabe, I., Kunkel, T.A. & Carr, A.M. The major roles of DNA polymerases epsilon and delta at the eukaryotic replication fork are evolutionarily conserved. PLoS Genet. 7, e1002407 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Daigaku, Y. et al. A global profile of replicative polymerase usage. Nat. Struct. Mol. Biol. 22, 192–198 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Kai, M. & Wang, T.S. Checkpoint activation regulates mutagenic translesion synthesis. Genes Dev. 17, 64–76 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Donnianni, R.A. & Symington, L.S. Break-induced replication occurs by conservative DNA synthesis. Proc. Natl. Acad. Sci. USA 110, 13475–13480 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Saini, N. et al. Migrating bubble during break-induced replication drives conservative DNA synthesis. Nature 502, 389–392 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Wilson, M.A. et al. Pif1 helicase and Polδ promote recombination-coupled DNA synthesis via bubble migration. Nature 502, 393–396 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Meselson, M. & Stahl, F.W. The replication of DNA in Escherichia coli. Proc. Natl. Acad. Sci. USA 44, 671–682 (1958).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Kesti, T., Flick, K., Keranen, S., Syvaoja, J.E. & Wittenberg, C. DNA polymerase epsilon catalytic domains are dispensable for DNA replication, DNA repair, and cell viability. Mol. Cell 3, 679–685 (1999).

    CAS  PubMed  Google Scholar 

  36. Waga, S. & Stillman, B. Anatomy of a DNA replication fork revealed by reconstitution of SV40 DNA replication in vitro. Nature 369, 207–212 (1994).

    CAS  PubMed  Google Scholar 

  37. Handa, T., Kanke, M., Takahashi, T.S., Nakagawa, T. & Masukata, H. DNA polymerization-independent functions of DNA polymerase epsilon in assembly and progression of the replisome in fission yeast. Mol. Biol. Cell 23, 3240–3253 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Schlacher, K., Wu, H. & Jasin, M. A distinct replication fork protection pathway connects Fanconi anemia tumor suppressors to RAD51-BRCA1/2. Cancer Cell 22, 106–116 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Petermann, E., Orta, M.L., Issaeva, N., Schultz, N. & Helleday, T. Hydroxyurea-stalled replication forks become progressively inactivated and require two different RAD51-mediated pathways for restart and repair. Mol. Cell 37, 492–502 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Carr, A.M. & Lambert, S. Replication stress-induced genome instability: the dark side of replication maintenance by homologous recombination. J. Mol. Biol. 425, 4733–4744 (2013).

    CAS  PubMed  Google Scholar 

  41. Kunkel, T.A. & Burgers, P.M. Dividing the workload at a eukaryotic replication fork. Trends Cell Biol. 18, 521–527 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Georgescu, R.E. et al. Mechanism of asymmetric polymerase assembly at the eukaryotic replication fork. Nat. Struct. Mol. Biol. 21, 664–670 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Reijns, M.A. et al. Lagging-strand replication shapes the mutational landscape of the genome. Nature 518, 502–506 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Clausen, A.R. et al. Tracking replication enzymology in vivo by genome-wide mapping of ribonucleotide incorporation. Nat. Struct. Mol. Biol. 22, 185–191 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Lujan, S.A. et al. Heterogeneous polymerase fidelity and mismatch repair bias genome variation and composition. Genome Res. 24, 1751–1764 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Bartkova, J. et al. DNA damage response as a candidate anti-cancer barrier in early human tumorigenesis. Nature 434, 864–870 (2005).

    CAS  PubMed  Google Scholar 

  47. Gorgoulis, V.G. et al. Activation of the DNA damage checkpoint and genomic instability in human precancerous lesions. Nature 434, 907–913 (2005).

    CAS  PubMed  Google Scholar 

  48. Bartkova, J. et al. Oncogene-induced senescence is part of the tumorigenesis barrier imposed by DNA damage checkpoints. Nature 444, 633–637 (2006).

    CAS  PubMed  Google Scholar 

  49. Bester, A.C. et al. Nucleotide deficiency promotes genomic instability in early stages of cancer development. Cell 145, 435–446 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Dominguez-Sola, D. et al. Non-transcriptional control of DNA replication by c-Myc. Nature 448, 445–451 (2007).

    CAS  PubMed  Google Scholar 

  51. Jones, R.M. et al. Increased replication initiation and conflicts with transcription underlie Cyclin E-induced replication stress. Oncogene 32, 3744–3753 (2013).

    CAS  PubMed  Google Scholar 

  52. Di Micco, R. et al. Oncogene-induced senescence is a DNA damage response triggered by DNA hyper-replication. Nature 444, 638–642 (2006).

    CAS  PubMed  Google Scholar 

  53. Neelsen, K.J., Zanini, I.M., Herrador, R. & Lopes, M. Oncogenes induce genotoxic stress by mitotic processing of unusual replication intermediates. J. Cell Biol. 200, 699–708 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Moreno, S., Klar, A. & Nurse, P. Molecular genetic analysis of fission yeast Schizosaccharomyces pombe. Methods Enzymol. 194, 795–823 (1991).

    CAS  PubMed  Google Scholar 

  55. Watson, A.T., Garcia, V., Bone, N., Carr, A.M. & Armstrong, J. Gene tagging and gene replacement using recombinase-mediated cassette exchange in Schizosaccharomyces pombe. Gene 407, 63–74 (2008).

    CAS  PubMed  Google Scholar 

  56. Lea, D.E. & Coulson, C.A. The distribution of the numbers of mutants in bacterial populations. J. Genet. 49, 264–285 (1949).

    CAS  PubMed  Google Scholar 

  57. Watson, A.T. et al. Optimisation of the Schizosaccharomyces pombe urg1 expression system. PLoS ONE 8, e83800 (2013).

    PubMed  PubMed Central  Google Scholar 

  58. Arcangioli, B. Fate of mat1 DNA strands during mating-type switching in fission yeast. EMBO Rep. 1, 145–150 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by Project Z01 ES065070 (T.A.K.) from the Division of Intramural Research of the US National Institutes of Health, by Medical Research Council (UK) grants G0801078 and G1100074 (A.M.C.) and by European Research Council grant 268788-SMI-DDR (A.M.C.). We thank H. Masukata (Osaka University) for the cdc20::hphMX6-Pnmt1-cdc20CTD strain.

Author information

Authors and Affiliations

Authors

Contributions

A.M.C. conceived the study. A.M.C., T.A.K., I.M. and J.M.M. designed the experimental approach. I.M., K'I.M., S.M., Y.D., A.K. and M.S. performed experiments and interpreted data. I.M. and A.M.C. wrote the manuscript. T.A.K. and J.M.M. edited the manuscript.

Corresponding author

Correspondence to Antony M Carr.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Data Set 1

Full autoradiographs of all blots used in the figures (PDF 13572 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Miyabe, I., Mizuno, K., Keszthelyi, A. et al. Polymerase δ replicates both strands after homologous recombination–dependent fork restart. Nat Struct Mol Biol 22, 932–938 (2015). https://doi.org/10.1038/nsmb.3100

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nsmb.3100

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